15.12.2012 Views

2D PAGE: Sample Preparation and Fractionation - Mycobacteriology ...

2D PAGE: Sample Preparation and Fractionation - Mycobacteriology ...

2D PAGE: Sample Preparation and Fractionation - Mycobacteriology ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>


METHODS IN MOLECULAR BIOLOGY TM<br />

John M. Walker, SERIES EDITOR<br />

460. Essential Concepts in Toxicogenomics, edited by<br />

Donna L. Mendrick <strong>and</strong> William B. Mattes, 2008<br />

459. Prion Protein Protocols, edited by Andrew F. Hill,<br />

2008<br />

458. Artificial Neural Networks: Methods <strong>and</strong><br />

Applications, edited by David S. Livingstone, 2008<br />

457. Membrane Trafficking, edited by Ales Vancura,<br />

2008<br />

456. Adipose Tissue Protocols, Second Edition, edited by<br />

Kaiping Yang, 2008<br />

455. Osteoporosis, edited by Jennifer J. Westendorf, 2008<br />

454. SARS- <strong>and</strong> Other Coronaviruses: Laboratory<br />

Protocols, edited by Dave Cavanagh, 2008<br />

453. Bioinformatics, Volume 2: Structure, Function, <strong>and</strong><br />

Applications, edited by Jonathan M. Keith, 2008<br />

452. Bioinformatics, Volume 1: Data, Sequence Analysis,<br />

<strong>and</strong> Evolution, edited by Jonathan M. Keith, 2008<br />

451. Plant Virology Protocols: From Viral Sequence to<br />

Protein Function, edited by Gary Foster, Elisabeth<br />

Johansen, Yiguo Hong, <strong>and</strong> Peter Nagy, 2008<br />

450. Germline Stem Cells, edited by Steven X. Hou <strong>and</strong><br />

Shree Ram Singh, 2008<br />

449. Mesenchymal Stem Cells: Methods <strong>and</strong> Protocols,<br />

edited by Darwin J. Prockop, Douglas G. Phinney,<br />

<strong>and</strong> Bruce A. Brunnell, 2008<br />

448. Pharmacogenomics in Drug Discovery <strong>and</strong><br />

Development, edited by Qing Yan, 2008<br />

447. Alcohol: Methods <strong>and</strong> Protocols, edited by<br />

Laura E. Nagy, 2008<br />

446. Post-translational Modification of Proteins: Tools<br />

for Functional Proteomics, Second Edition, edited by<br />

Christoph Kannicht, 2008<br />

445. Autophagosome <strong>and</strong> Phagosome, edited by<br />

Vojo Deretic, 2008<br />

444. Prenatal Diagnosis, edited by Sinhue Hahn <strong>and</strong><br />

Laird G. Jackson, 2008<br />

443. Molecular Modeling of Proteins, edited by<br />

Andreas Kukol, 2008<br />

442. RNAi: Design <strong>and</strong> Application, edited by Sailen<br />

Barik, 2008<br />

441. Tissue Proteomics: Pathways, Biomarkers, <strong>and</strong> Drug<br />

Discovery, edited by Brian Liu, 2008<br />

440. Exocytosis <strong>and</strong> Endocytosis, edited by<br />

Andrei I. Ivanov, 2008<br />

439. Genomics Protocols, Second Edition, edited by<br />

Mike Starkey <strong>and</strong> Ramnanth Elaswarapu, 2008<br />

438. Neural Stem Cells: Methods <strong>and</strong> Protocols, Second<br />

Edition, edited by Leslie P. Weiner, 2008<br />

437. Drug Delivery Systems, edited by Kewal K. Jain,<br />

2008<br />

436. Avian Influenza Virus, edited by Erica Spackman,<br />

2008<br />

435. Chromosomal Mutagenesis, edited by Greg Davis<br />

<strong>and</strong> Kevin J. Kayser, 2008<br />

434. Gene Therapy Protocols: Volume 2, Design <strong>and</strong><br />

Characterization of Gene Transfer Vectors, edited by<br />

Joseph M. LeDoux, 2008<br />

433. Gene Therapy Protocols: Volume 1, Production <strong>and</strong><br />

In Vivo Applications of Gene Transfer Vectors, edited<br />

by Joseph M. LeDoux, 2008<br />

432. Organelle Proteomics, edited by Delphine Pflieger<br />

<strong>and</strong> Jean Rossier, 2008<br />

431. Bacterial Pathogenesis: Methods <strong>and</strong> Protocols,<br />

edited by Frank DeLeo <strong>and</strong> Michael Otto, 2008<br />

430. Hematopoietic Stem Cell Protocols, edited by Kevin<br />

D. Bunting, 2008<br />

429. Molecular Beacons: Signalling Nucleic Acid Probes,<br />

Methods <strong>and</strong> Protocols, edited by Andreas Marx <strong>and</strong><br />

Oliver Seitz, 2008<br />

428. Clinical Proteomics: Methods <strong>and</strong> Protocols, edited<br />

by Antonio Vlahou, 2008<br />

427. Plant Embryogenesis, edited by Maria Fern<strong>and</strong>a<br />

Suarez <strong>and</strong> Peter Bozhkov, 2008<br />

426. Structural Proteomics: High-Throughput Methods,<br />

edited by Bostjan Kobe, Mitchell Guss, <strong>and</strong> Huber<br />

Thomas, 2008<br />

425. <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>,<br />

Volume 2, edited by Anton Posch, 2008<br />

424. <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>,<br />

Volume 1, edited by Anton Posch, 2008<br />

423. Electroporation Protocols, edited by Shulin Li, 2008<br />

422. Phylogenomics, edited by William J. Murphy, 2008<br />

421. Affinity Chromatography, Methods <strong>and</strong> Protocols,<br />

Second Edition, edited by Michael Zachariou, 2007<br />

420. Drosophila, Methods <strong>and</strong> Protocols, edited by<br />

Christian Dahmann, 2008<br />

419. Post-Transcriptional Gene Regulation, edited by<br />

Jeffrey Wilusz, 2008<br />

418. Avidin-Biotin Interactions, Methods <strong>and</strong><br />

Applications, edited by Robert J. McMahon, 2008<br />

417. Tissue Engineering, Second Edition, edited by<br />

Hannsjörg Hauser <strong>and</strong> Martin Fussenegger, 2007<br />

416. Gene Essentiality: Protocols <strong>and</strong> Bioinformatics,<br />

edited by Andrei L. Osterman, 2008<br />

415. Innate Immunity, edited by Jonathan Ewbank <strong>and</strong><br />

Eric Vivier, 2007<br />

414. Apoptosis <strong>and</strong> Cancer: Methods <strong>and</strong> Protocols,<br />

edited by Gil Mor <strong>and</strong> Ayesha B. Alvero, 2008<br />

413. Protein Structure Prediction, Second Edition,<br />

edited by Mohammed Zaki <strong>and</strong> Chris Bystroff, 2008<br />

412. Neutrophil Methods <strong>and</strong> Protocols, edited by Mark<br />

T. Quinn, Frank R. DeLeo, <strong>and</strong> Gary M. Bokoch,<br />

2007<br />

411. Reporter Genes for Mammalian Systems, edited by<br />

Don Anson, 2007<br />

410. Environmental Genomics, edited by Cristofre C.<br />

Martin, 2007<br />

409. Immunoinformatics: Predicting Immunogenicity In<br />

Silico, edited by Darren R. Flower, 2007


METHODS IN MOLECULAR BIOLOGY TM<br />

<strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong><br />

<strong>Preparation</strong><br />

<strong>and</strong> <strong>Fractionation</strong><br />

Volume 2<br />

Edited by<br />

Anton Posch<br />

Bio-Rad Laboratories GmbH, Munich, Germany


Editor<br />

Anton Posch<br />

Bio-Rad Laboratories GmbH<br />

Munich, Germany<br />

Series Editor<br />

John M. Walker<br />

School of Life Sciences<br />

University of Hertfordshire<br />

Hatfield, Herts., UK<br />

ISBN: 978-1-60327-209-4 e-ISBN: 978-1-60327-210-0<br />

ISSN: 1064-3745<br />

Library of Congress Control Number: 2007943017<br />

©2008 Humana Press, a part of Springer Science+Business Media, LLC<br />

All rights reserved. This work may not be translated or copied in whole or in part without the written<br />

permission of the publisher (Humana Press, 999 Riverview Drive, Suite 208, Totowa, NJ 07512 USA),<br />

except for brief excerpts in connection with reviews or scholarly analysis. Use in connection with any form<br />

of information storage <strong>and</strong> retrieval, electronic adaptation, computer software, or by similar or dissimilar<br />

methodology now known or hereafter developed is forbidden.<br />

The use in this publication of trade names, trademarks, service marks, <strong>and</strong> similar terms, even if they are<br />

not identified as such, is not to be taken as an expression of opinion as to whether or not they are subject to<br />

proprietary rights.<br />

While the advice <strong>and</strong> information in this book are believed to be true <strong>and</strong> accurate at the date of going to<br />

press, neither the authors nor the editors nor the publisher can accept any legal responsibility for any errors<br />

or omissions that may be made. The publisher makes no warranty, express or implied, with respect to the<br />

material contained herein.<br />

Cover illustration: Figure 3, Chapter 14, “The Terminator: A Device for High Throughput Extraction of<br />

Plant Material,” by B. M. van den Berg.<br />

Printed on acid-free paper<br />

987654321<br />

springer.com


Preface<br />

This book, split into two volumes, presents a broad coverage of the principles<br />

<strong>and</strong> recent developments of sample preparation <strong>and</strong> fractionation tools in<br />

Expression Proteomics in general <strong>and</strong> for two-dimensional electrophoresis<br />

(2-DE) in particular. 2-DE, with its unique capacity to resolve thous<strong>and</strong>s of<br />

proteins in a single run, is still a fundamental research tool for nearly all<br />

protein-related scientific projects. The methods described here in detail are not<br />

limited to 2-DE <strong>and</strong> can also be applied to other protein separation techniques.<br />

Because each biological sample is unique, a suited sample preparation<br />

strategy has to consider the type of sample as well as the type of biological<br />

question being addressed. The complex nature of proteins often requires a<br />

multitude of sample preparation options. In addition, sample preparation is<br />

not only a prerequisite for a successful <strong>and</strong> reproducible Proteomics experiment,<br />

but also the key factor to meaningful data evaluation. Interestingly, not<br />

much attention was paid to this area during Proteomics methodology development<br />

<strong>and</strong> therefore this book is intended to explain in depth how proteins<br />

from various sources can be properly isolated <strong>and</strong> prepared for reproducible<br />

Proteome analysis.<br />

The application of fractionation <strong>and</strong> enrichment strategies has become a<br />

major part of sample preparation. The number of possible different proteins in<br />

a cell or tissue sample is believed to be in the several hundreds of thous<strong>and</strong>s,<br />

spanning concentration ranges from the level of a single molecule to micromolar<br />

amounts, <strong>and</strong> no single analytical method developed today is capable of<br />

resolving <strong>and</strong> detecting such a diverse sample. <strong>Sample</strong> fractionation reduces the<br />

overall complexity of the sample, <strong>and</strong> enriches low abundance proteins relative<br />

to the original sample. Proteins that may originally have been undetectable are<br />

thus rendered amenable to analysis by 2-DE <strong>and</strong> a broad variety of gel-free<br />

mass spectrometry-based technologies.<br />

This book is for students of Biochemistry, Biomedicine, Biology, <strong>and</strong><br />

Genomics <strong>and</strong> will be an invaluable source for the experienced, practicing<br />

scientist, too.<br />

Anton Posch<br />

v


Contents<br />

Preface ................................................................ v<br />

Contributors ........................................................... xi<br />

1. Application of Fluorescence Dye Saturation Labeling<br />

for Differential Proteome Analysis of 1,000 Microdissected<br />

Cells from Pancreatic Ductal Adenocarcinoma Precursor<br />

Lesions .........................................................<br />

Barbara Sitek, Bence Sipos, Günter Klöppel, Wolff Schmiegel,<br />

Stephan A. Hahn, Helmut E. Meyer, <strong>and</strong> Kai Stühler<br />

1<br />

2. Albumin <strong>and</strong> Immunoglobulin Depletion of Human Plasma ........<br />

Rosalind E. Jenkins, Neil R. Kitteringham,<br />

Carrie Greenough, <strong>and</strong> B. Kevin Park<br />

15<br />

3. Multi-Component Immunoaffinity Subtraction<br />

<strong>and</strong> Reversed-Phase Chromatography of Human Serum ..........<br />

James Martosella <strong>and</strong> Nina Zolotarjova<br />

27<br />

4. Immunoaffinity <strong>Fractionation</strong> of Plasma Proteins by Chicken IgY<br />

Antibodies......................................................<br />

Lei Huang <strong>and</strong> Xiangming Fang<br />

41<br />

5. Proteomics of Cerebrospinal Fluid:<br />

Methods for <strong>Sample</strong> Processing .................................<br />

John E. Hale, Valentina Gelfanova, Jin-Sam You,<br />

Michael D. Knierman, <strong>and</strong> Robert A. Dean<br />

53<br />

6. <strong>Sample</strong> <strong>Preparation</strong> of Bronchoalveolar Lavage Fluid ...............<br />

Baptiste Leroy, Paul Falmagne, <strong>and</strong> Ruddy Wattiez<br />

67<br />

7. <strong>Preparation</strong> of Nasal Secretions for Proteome Analysis ..............<br />

Begona Casado, Paolo Iadarola, <strong>and</strong> Lewis K. Pannell<br />

77<br />

8. <strong>Preparation</strong> of Urine <strong>Sample</strong>s for Proteomic Analysis ...............<br />

Rembert Pieper<br />

89<br />

9. Isolation of Cytoplasmatic Proteins from Cultured Cells for<br />

Two-Dimensional Gel Electrophoresis ........................... 101<br />

Ying Wang, Jen-Fu Chiu, <strong>and</strong> Qing-Yu He<br />

vii


viii Contents<br />

10. <strong>Sample</strong> <strong>Preparation</strong> of Culture Medium from Madin-Darby<br />

Canine Kidney Cells ............................................ 113<br />

Daniel Ambort, Daniel Lottaz, <strong>and</strong> Erwin Sterchi<br />

11. <strong>Sample</strong> <strong>Preparation</strong> for Mass Spectrometry Analysis<br />

of Formalin-Fixed Paraffin-Embedded Tissue: Proteomic<br />

Analysis of Formalin-Fixed Tissue ............................... 131<br />

Nicolas A. Stewart <strong>and</strong> Timothy D. Veenstra<br />

12. Metalloproteomics in the Molecular Study of Cell Physiology<br />

<strong>and</strong> Disease .................................................... 139<br />

Hermann-Josef Thierse, Stefanie Helm,<br />

<strong>and</strong> Patrick Pankert<br />

13. Protein Extraction from Green Plant Tissue ......................... 149<br />

Ragnar Flengsrud<br />

14. The Terminator: A Device for High-Throughput Extraction of<br />

Plant Material .................................................. 153<br />

B. M. van den Berg<br />

15. Isolation of Mitochondria from Plant Cell Culture...................163<br />

Etienne H. Meyer <strong>and</strong> A. Harvey Millar<br />

16. Isolation <strong>and</strong> <strong>Preparation</strong> of Chloroplasts from Arabidopsis<br />

thaliana Plants .................................................. 171<br />

Sybille E. Kubis, Kathryn S. Lilley, <strong>and</strong> Paul Jarvis<br />

17. Isolation of Plant Cell Wall Proteins ................................187<br />

Elisabeth Jamet, Georges Boudart, Gisèle Borderies,<br />

Stephane Charmont, Claude Lafitte, Michel Rossignol,<br />

Herve Canut, <strong>and</strong> Rafael Pont-Lezica<br />

18. Isolation <strong>and</strong> <strong>Fractionation</strong> of the Endoplasmic Reticulum from<br />

Castor Bean (Ricinus communis) Endosperm for Proteomic<br />

Analyses........................................................203<br />

William J. Simon, Daniel J. Maltman<br />

<strong>and</strong> Antoni R. Slabas<br />

19. Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics ........... 217<br />

Aida Pitarch, César Nombela, <strong>and</strong> Concha Gil<br />

20. Collection of Proteins Secreted from Yeast Protoplasts in Active<br />

Cell Wall Regeneration ......................................... 241<br />

Aida Pitarch, César Nombela, <strong>and</strong> Concha Gil<br />

21. <strong>Sample</strong> <strong>Preparation</strong> Procedure for Cellular Fungi ................... 265<br />

Alois Harder


Contents ix<br />

22. Isolation <strong>and</strong> Enrichment of Secreted Proteins from Filamentous<br />

Fungi ...........................................................275<br />

Martha L. Medina <strong>and</strong> Wilson A. Francisco<br />

23. Isolation <strong>and</strong> Solubilization of Cellular Membrane Proteins from<br />

Bacteria ........................................................ 287<br />

Kheir Zuobi-Hasona <strong>and</strong> L. Jeannine Brady<br />

24. Isolation <strong>and</strong> Solubilization of Gram-Positive Bacterial Cell<br />

Wall-Associated Proteins........................................295<br />

Jason N. Cole, Steven P. Djordjevic, <strong>and</strong> Mark J. Walker<br />

25. Cell <strong>Fractionation</strong> of Parasitic Protozoa ............................ 313<br />

W<strong>and</strong>erley de Souza, José Andrés Morgado-Diaz,<br />

<strong>and</strong> Narcisa L. Cunha-e-Silva<br />

Index..................................................................333


Contributors<br />

Daniel Ambort • University of Berne, Berne, Switzerl<strong>and</strong><br />

Leroy Baptiste • University of Mons-Hainaut, Mons, Belgium<br />

Gisèle Borderies • UMR 5546 CNRS-Université Paul Sabatier-Toulouse III,<br />

Castanet-Tolosan, France<br />

Georges Boudart • UMR 5546 CNRS-Université Paul Sabatier-Toulouse<br />

III, Castanet-Tolosan, France<br />

L. Jeannine Brady • University of Florida, Gainesville, Florida<br />

Herve Canut • UMR 5546 CNRS-Université Paul Sabatier-Toulouse III,<br />

Castanet-Tolosan, France<br />

Begona Casado • Swiss Federal Institute of Technology, Zurich, Switzerl<strong>and</strong><br />

Stephane Charmont • Novartis Pharma AG, Basel, Switzerl<strong>and</strong><br />

Jen-Fu Chiu • The University of Hong Kong, Hong Kong, China<br />

Jason N. Cole • University of Wollongong, Wollongong, Australia<br />

Narcisa L. Cunha-e-Silva • Universidade Federal do Rio de Janeiro,<br />

Rio de Janeiro, Brazil<br />

W<strong>and</strong>erley de Souza • Universidade Federal do Rio de Janeiro, Rio de<br />

Janeiro, Brazil<br />

Robert A. Dean • Lilly Research Laboratories, Indianapolis, Indiana<br />

Steven P. Djordjevic • Elizabeth Macarthur Agricultural Institute,<br />

Menangle, Australia<br />

Paul Falmagne • University of Mons-Hainaut, Mons, Belgium<br />

Xiangming Fang • GenWay Biotech, Inc., San Diego, California<br />

Ragnar Flengsrud • Norwegian University of Life Sciences, Ås, Norway<br />

Wilson A. Francisco • Arizona State University, Tempe, Arizona<br />

Valentina Gelfanova • Lilly Research Laboratories, Greenfield, Indiana<br />

Concha Gil • Complutense University of Madrid, Madrid, Spain<br />

Carrie Greenough • University of Liverpool, Liverpool, Great Britain<br />

Stephan A. Hahn • Ruhr-University Bochum, Bochum, Germany<br />

John E. Hale • Lilly Research Laboratories, Greenfield, Indiana<br />

Alois Harder • Toplab GmbH, Martinsried, Germany<br />

Qing-Yu He • The University of Hong Kong, Hong Kong, China<br />

Stefanie Helm • University of Heidelberg, Mannheim, Germany<br />

Lei Huang • GenWay Biotech Inc., San Diego, California<br />

xi


xii Contributors<br />

Paolo Iadarola • University of Pavia, Pavia, Italy<br />

Elisabeth Jamet • UMR 5546 CNRS-Université Paul Sabatier-Toulouse III,<br />

Castanet-Tolosan, France<br />

Paul Jarvis • University of Leicester, Leicester, Great Britain<br />

Rosalind E. Jenkins • University of Liverpool, Liverpool, Great Britain<br />

Neil R. Kitteringham • University of Liverpool, Liverpool, Great Britain<br />

Günter Klöppel • Christian Albrechts University, Kiel, Germany<br />

Michael D. Knierman • Lilly Research Laboratories, Greenfield, Indiana<br />

Sybille E. Kubis • University of Leicester, Leicester, Great Britain<br />

Claude Lafitte • UMR 5546 CNRS-Université Paul Sabatier-Toulouse III,<br />

Castanet-Tolosan, France<br />

Baptiste Leroy • University of Mons-Hainaut, Mons, Belgium<br />

Kathryn S. Lilley • University of Cambridge, Cambridge, Great Britain<br />

Daniel Lottaz • University of Berne, Berne, Switzerl<strong>and</strong><br />

Daniel J. Maltman • University of Durham, Durham, Great Britain<br />

James Martosella • Agilent Technologies Inc., Wilmington, Delaware<br />

Martha L. Medina • Arizona State University, Tempe, Arizona<br />

Helmut E. Meyer • Ruhr-University Bochum, Bochum, Germany<br />

Etienne H. Meyer • The University of Western Australia, Perth, Australia<br />

A. Harvey Millar • The University of Western Australia, Perth, Australia<br />

José Andrés Morgado-Diaz • Instituto Nacional de Câncer, Rio de<br />

Janeiro, Brazil<br />

César Nombela • Complutense University of Madrid, Madrid, Spain<br />

Patrick Pankert • University of Heidelberg, Mannheim, Germany<br />

Lewis K. Pannell • University of South Alabama, Mobile, Alabama<br />

B. Kevin Park • University of Liverpool, Liverpool, Great Britain<br />

Falmagne Paul • University of Mons-Hainaut, Mons, Belgium<br />

Rembert Pieper • The Institute for Genomic Research, Rockville, Maryl<strong>and</strong><br />

Aida Pitarch • Complutense University of Madrid, Madrid, Spain<br />

Rafael Pont-Lezica • UMR 5546 CNRS-Universitè Paul Sabatier-Toulouse<br />

III, Castanet-Tolosan, France<br />

Michel Rossignol • UMR 5546 CNRS-Universitè Paul Sabatier-Toulouse<br />

III, Castanet-Tolosan, France<br />

Wolff Schmiegel • Ruhr-University Bochum, Bochum, Germany<br />

William J. Simon • University of Durham, Durham, Great Britain<br />

Bence Sipos • Christian Albrechts University, Kiel, Germany<br />

Barbara Sitek • Ruhr-University Bochum, Bochum, Germany<br />

Antoni R. Slabas • University of Durham, Durham, Great Britain<br />

Erwin Sterchi • University of Berne, Berne, Switzerl<strong>and</strong><br />

Nicolas A. Stewart • National Cancer Institute at Frederick, Frederick,<br />

Maryl<strong>and</strong>


Contributors xiii<br />

Kai Stühler • Ruhr-University Bochum, Bochum, Germany<br />

Hermann-Josef Thierse • University of Heidelberg, Mannheim, Germany<br />

B. M. van den Berg • Elexa, Enkhuizen, The Netherl<strong>and</strong>s<br />

Timothy D. Veenstra • National Cancer Institute at Frederick, Frederick,<br />

Maryl<strong>and</strong><br />

Mark J. Walker • University of Wollongong, Wollongong, Australia<br />

Ying Wang • The University of Hong Kong, Hong Kong, China<br />

Ruddy Wattiez • University of Mons-Hainaut, Mons, Belgium<br />

Jin-Sam You • Indiana Centers for Applied Protein Sciences, Indianapolis,<br />

Indiana<br />

Nina Zolotarjova • Agilent Technologies Inc., Wilmington, Delaware<br />

Kheir Zuobi-Hasona • University of Florida, Gainesville, Florida


1<br />

Application of Fluorescence Dye Saturation Labeling<br />

for Differential Proteome Analysis of 1,000<br />

Microdissected Cells from Pancreatic Ductal<br />

Adenocarcinoma Precursor Lesions<br />

Barbara Sitek, Bence Sipos, Günter Klöppel, Wolff Schmiegel,<br />

Stephan A. Hahn, Helmut E. Meyer, <strong>and</strong> Kai Stühler<br />

Summary<br />

The identification of molecular changes underlying clinical pathogenic processes is<br />

often hampered by significant cellular diversity of the tissue. Pathogenic aberrant cells<br />

are surrounded by cells originating e.g., from stroma, the vascular system or other neighbouring<br />

cell types, which lead to under-representation of interesting cells when analysing<br />

whole tissue specimen. Therefore, selection of relevant cell types for detailed analysis<br />

is an absolute prerequisite for in depth elucidation of underlying biological processes.<br />

Microdissection offers the advantage to select for a biologically relevant cell type which<br />

is often in low abundance. Here, we present a proteomics approach allowing us to analyse<br />

1,000 microdissected cells stemming from pancreatic carcinoma precursor lesions applying<br />

fluorescence dye saturation labeling.<br />

Key Words: Difference gel electrophoresis; DIGE; fluorescence dye saturation<br />

labeling; pancreatic ductal adenocarcinoma; panin, tumor marker; two-dimensional gel<br />

electrophoresis.<br />

1. Introduction<br />

To identify new c<strong>and</strong>idate molecular markers for pancreatic ductal adenocarcinoma<br />

we established a proteomics approach analysing microdissected<br />

cells from precursor lesions, the so called pancreatic intraepithelial neoplasia<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

1


2 Sitek et al.<br />

(PanIN) (1). Due to a limited amount of proteins available from microdissection<br />

we developed a procedure which included fluorescence dye saturation labeling<br />

in combination with high resolution two-dimensional gel electrophoresis<br />

(2-DE) (2). With this procedure we were able to analyse proteins extracted<br />

from 1,000 microdissected cells with a high resolution of up to 2,500 protein<br />

spots. For differential proteome analysis we analysed microdissected cells<br />

from 9 patients. We compared the protein expression of the different PanIN<br />

grades (PanIN 1A/B, PanIN 2, PanIN 3) <strong>and</strong> carcinoma cells related to normal<br />

epithelial cells <strong>and</strong> found 86 significantly regulated spots (p < 0.05, ratio >1.6).<br />

Using protein lysates from pancreatic carcinoma tissue as a reference proteome<br />

we were able to successfully identify the proteins after tryptic in-gel digestion.<br />

2. Materials<br />

2.1. Microdissection <strong>and</strong> <strong>Sample</strong> <strong>Preparation</strong><br />

1. Microscope: BH2 (Olympus, Wetzla, Germany).<br />

2. Cryostat: Cryotom SME (Sh<strong>and</strong>on).<br />

3. Ultrasonic bath (VWR).<br />

4. H<strong>and</strong> homogenisator.<br />

5. Injection needle: 0.65 mm × 25 mm (Braun, Melsungen, Germany).<br />

6. Lysis buffer: 2 M thiourea, 7 M urea, 4% 3-[(3-cholamidopropyl)<br />

dimethylammino]-1-propane sulfonate (CHAPS), 30 mM Tris-HCl, pH 8.0.<br />

7. Hematoxylin stain: 25% (v/v) hematoxylin according to Mayer (Merck).<br />

8. Eosin stain: 1.7% (w/v) eosin (Merck), diluted in 96% ethanol.<br />

2.2. Cysteine-Specific Protein Labeling Using CyDye DIGE<br />

Fluor Saturation Dyes<br />

1. Dye stock solution: CyDye DIGE Fluor saturation dyes solid compounds are<br />

reconstituted in dimethylformamide (DMF) giving a concentration of 2 mM (50<br />

μL DMF to 100 nM of dye) for analytical gels <strong>and</strong> 20 mM (20 μL DMF to 400<br />

nM of dye) for preparative gels. It is stable at –20°C for 3 mo.<br />

2. 50 mM NaOH solution (for pH adjustment).<br />

3. 2 mM TCEP (triscarboxethylphosphine) solution for analytical gels <strong>and</strong> 20 mM<br />

TCEP solution for preparative gels.<br />

4. Image software: ImageQuantTM (GE Healthcare Bioscience).<br />

5. Differential image analysis software: Decyder v5.02 (GE Healthcare Bioscience).<br />

6. Fluorescence Scanner: Typhoon 9400 (GE Healthcare Bioscience).<br />

2.3. Isoelectric Focusing<br />

1. Seperation gel buffer: 3.5% (w/v) acrylamide, 0.3% (w/v) piperazindiacrylamide,<br />

4% (v/v) carrier ampholines mixture (pH 2–11), 9.0 M urea, 5% glycerol, 0.06%<br />

(v/v) TEMED.


Application of Fluorescence Dye Saturation Labeling 3<br />

2. Cap gel buffer: 12.3% (w/v) acrylamide, 0.13% (w/v) piperazindiacrylamide, 4%<br />

(v/v) carrier ampholyte mixture, 9.0 M urea, 5% glycerol, 0.06% (v/v) TEMED<br />

3. 1.2% (w/v) APS (Ammoniumpersulfat).<br />

4. Anodic buffer: 3 M urea, 7.3% (w/v) phosphoric acid.<br />

5. Cathodic buffer: 9 M urea, 5% (w/v) glycerol, 0.75 M ethylendiamine.<br />

6. Sephadex solution: 270 mg Sephadex suspension (20 g Sephadex was swollen in<br />

500 mL water, resuspended into 1Lof25%glycerol solution <strong>and</strong> filtered), plus<br />

233 mg urea, plus 98 mg thiourea, <strong>and</strong> 25 μL ampholine mixture, pH 2–11 <strong>and</strong><br />

25 μL DTT (1.08 g/5 mL).<br />

7. Protection solution: 30% urea (w/v), 5% glycerol (w/v), 2% carrier ampholytes,<br />

pH 2–4.<br />

8. Equilibration solution: 125 mM Tris-base, 40% glycerol, 65 mM DTT, 3% SDS.<br />

2.4. Two-dimensional Polyacrylamid Gel Electrophoresis (<strong>2D</strong>-<strong>PAGE</strong>)<br />

1. Glass plates compatible with fluorescence imaging (25 × 30 × 0.4 cm).<br />

2. Plastic spacer (30 ×1×0.15 cm).<br />

3. Apparatus for vertical SDS-<strong>PAGE</strong>. (System VA Sarstedt, Nümbrecht, Germany)<br />

4. Gel carrier grooves (self made).<br />

5. Gel solution: 570 mM Tris-base <strong>and</strong> 180 mM Tris-HCL, 0.06% TEMED, 0.2%<br />

SDS, 15% acrylamide, 0.2% bisacrylamide.<br />

6. Running buffer: 0.2 M Tris, 1.92 mM glycine, 0.1% (w/v) SDS.<br />

7. 40% (w/w) APS (Ammoniumpersulfat).<br />

8. Protection solution: 285 mM Tris-base <strong>and</strong> 90 mM Tris-HCL, 0.1% SDS.<br />

9. Agarose solution: 1% (w/w) agarose (dissolved in running buffer), 0.001% (w/w)<br />

bromphenol blue.<br />

3. Methods<br />

The number of cells available by manual microdissection is rather<br />

limited. When analysing precursor lesions of the pancreatic adenocarcinoma<br />

only 1,000–5,000 cells can be provided in a reasonable time window (3–4 h).<br />

Due to the scarce protein amount (1,000 cells are equivalent to approx 2 μg<br />

protein) applying classical 2-DE techniques in combination with silver staining<br />

(loading amount 100 μg) or difference gel electrophoresis (DIGE) minimal<br />

labeling (50 μg) is not feasible. Therefore, DIGE saturation labeling which<br />

has a 50-fold higher sensitivity for protein detection must be applied when<br />

analysing such scarce sample amount cells (3).<br />

DIGE saturation labeling is based on covalent attachment of all protein<br />

cysteine residues prior to 2-DE (4). In contrast to DIGE minimal labeling<br />

where only one lysine residue of approx 3% of a protein species is labeled by<br />

a fluorescence dye (Cy2, Cy3 or Cy5) (5), DIGE saturation dyes leads to a<br />

complete labeling of all proteins (3).


4 Sitek et al.<br />

Moreover, in contrast to DIGE minimal labeling multiplexing within one gel<br />

for direct differential analysis is not feasible when using saturation labeling.<br />

Effects of fluorescence resonance energy transfer (FRET), by which the fluorescence<br />

emission of a CyDye can be enhanced or quenched, respectively, have to<br />

be considered (6). Therefore, only two saturation dyes are available, whereof<br />

one CyDye (mostly Cy5) is taken for differential analysis whereas the other<br />

CyDye (mostly Cy3) is taken as an internal st<strong>and</strong>ard for controlling system<br />

variation that ultimately provides a more accurate quantification of relative<br />

protein abundance.<br />

Furthermore, preparative protein amounts (300–500 μg) for protein identification<br />

can not be provided by microdissection. Therefore, a reference proteome<br />

stemming from a comparable source (e.g., whole tissue or cell culture lysate)<br />

must be determined. This reference proteome should match the proteome of<br />

the microdissected samples to a high degree (>90%) <strong>and</strong> thus facilitates protein<br />

identification in subsequent in-gel digestion using mass spectrometry (1).<br />

However, before commencing DIGE analysis one must optimally determine<br />

the labeling conditions i.e., the proportion of fluorescent dye to microdissected<br />

cells. It has been shown (own observations) that due to inherent differences<br />

in the cysteine content of a given proteome, coupled with variable sample<br />

conditions, one has to independently develop an optimisation procedure for each<br />

sample. This allows a high performance <strong>and</strong> high quality proteomic analysis.<br />

3.1. Optimised Manual Microdissection for the Proteome Analysis<br />

of Pancreatic Adenocarcinoma Cells<br />

1. Froze tissue resections containing pancreatic carcinoma on liquid nitrogen <strong>and</strong><br />

store at –80°C (see Note 1). For histological classification of normal ducts <strong>and</strong><br />

different PanIN stages prepare 5 μm frozen sections of peritumoral pancreatic<br />

parenchyma using a cryostat <strong>and</strong> stain with hematoxylin <strong>and</strong> eosin.<br />

2. For microdissection in subsequent tissue slides prepare 10-μm frozen section from<br />

tissue blocks containing the required grades <strong>and</strong> only stain with haematoxylin<br />

(Fig. 1) It has been shown that eosin interferes with 2-DE leading to reduced<br />

protein recovery (Fig. 2).<br />

3. Under microscopic observation harvest the required number of cells using a sterile<br />

injection needle.<br />

4. Lyse the microdissected cells in 100 μL lysis buffer (4°C), sonicate (6 × 10 sec<br />

pulses on ice) after each collection step <strong>and</strong> finally centrifuge (12,000g for 5 min)<br />

the lysate <strong>and</strong> store the supernatant at –80°C.<br />

3.2. Determination of the Minimal Number of Microdissected Cells<br />

1. Because microdissection is very time consuming <strong>and</strong> the number of available<br />

sample material is limited it is necessary to find the minimal number of cells


Application of Fluorescence Dye Saturation Labeling 5<br />

Fig. 1. Manual microdissection. Subsequent histological classification the<br />

hematoxylin stained (10 μm serial sections) PanIN lesions (A) were microdissected<br />

under a microscope using a sterile injection needle (B).<br />

Fig. 2. Compatibility of H&E staining with 2-DE. Influence of H&E staining<br />

was investigated applying 7000 microdissected cells, each stained with H&E (A) or<br />

hematoxylin only (B). After microdissection <strong>and</strong> labeling with Cy3 the samples were<br />

separated by 2-DE <strong>and</strong> scanned using fluorescence scanner. Due to the weak protein<br />

recovery shown in (A) it is obvious that eosin interferes with proteome analysis <strong>and</strong><br />

that hematoxylin staining is compatible with 2-DE analysis.


6 Sitek et al.<br />

Fig. 3. Determination of the minimal number of microdissected cells required for<br />

<strong>2D</strong>E. Aliquots with 1000 <strong>and</strong> 7000 cells were labeled with Cy3 <strong>and</strong> analysed using<br />

2-DE under the same conditions. The spots in the gels were detected using DIA mode<br />

of DeCyder software. In the gel with 1000 cells 2500 spots (A) <strong>and</strong> in the gel with<br />

7000 cells 2600 spots (B) could be detected.<br />

sufficient for a differential proteome analysis. Therefore, a pool of approx 30,000<br />

microdissected cells is required allowing to determine the optimal number of<br />

spots by a dilution experiment.<br />

2. Prepare four aliquots containing 1,000, 2,500, 5,000, <strong>and</strong> 7,000 cells in the volume<br />

of 100 μL lysis buffer, respectively (see Note 2).<br />

3. For protein labeling use the st<strong>and</strong>ard protocol according to user manual. Briefly,<br />

add 4 nM TCEP to each sample <strong>and</strong> incubate for 1hat37°C. Label the samples<br />

with 8 nM saturation dyes for 30 min at 37°C. For stopping the labeling reaction<br />

add 10 μL DTT <strong>and</strong> 10 μL Ampholytes, pH 2–4, before 2-DE.<br />

4. Subsequent to 2-DE scan the gels as described elsewhere (see Note 3) <strong>and</strong> detect<br />

the number of spots using DIA module of DeCyder software (see Note 4). The<br />

optimal number of cells considered for subsequent analyses results from highest<br />

number of protein spots per analysed number of cells (Fig. 3).<br />

3.3. Optimisation of Fluorescence Dye Labeling<br />

1. Having established the optimal number of cells for comprehensive proteome<br />

coverage, one must subsequently empirically optimise the labeling conditions.<br />

This avoids any effects of over- <strong>and</strong> under-labeling. Therefore, a so called


Application of Fluorescence Dye Saturation Labeling 7<br />

Fig. 4. Effects of different DIGE saturation labeling conditions detected by<br />

same/same experiment. For the optimal application of DIGE saturation labeling the<br />

labeling conditions have to be optimized. (A) If too much dye amount has been applied<br />

over-labeling leads to a horizontal shift detected in CyDye-dependent manner (arrows).<br />

(B) Under-labeling of proteins with CyDye results in vertical streaking. (C) The optimal<br />

label condition is reached if an exact overlay of both fluorescence images can be<br />

detected.<br />

“same/same” experiment titrating different dye amounts is performed. Thus,<br />

prepare six aliquots containing the optimal number of cells (see Chapter 3.2) from<br />

the pool of 30,000 cells.<br />

2. Label three of them with 2, 4, <strong>and</strong> 8 nM of Cy3 <strong>and</strong> another three with 2, 4, <strong>and</strong><br />

8nM of Cy5.<br />

3. Mix samples with equal dye amount <strong>and</strong> perform 2-DE for same/same experiment.<br />

4. After gel scanning analyse the overlay images in order to find the optimal ratio<br />

of protein <strong>and</strong> dye showing an accurate overlay of Cy3 <strong>and</strong> Cy5 images using<br />

ImageQuant TM . As shown in Fig. 4, applying too much dye results in horizontal<br />

shifts could occur due to additional labeling (e.g., -amino group of lysines),<br />

whereas an insufficient amount of dye causes vertical streaks due to inadequate<br />

protein labeling.<br />

3.4. Reference Proteome for Internal St<strong>and</strong>ardization<br />

<strong>and</strong> Protein Identification<br />

1. 2-DE analysis of 1,000 microdissected cells (∼2 μg) does not deliver sufficient<br />

sample material for protein identification using mass spectrometry. Therefore,<br />

a reference proteome from a closely related source has to be defined allowing<br />

protein identification by protein spot assignment. Additionally, this reference<br />

proteome may be considered as an internal st<strong>and</strong>ard (labeled with Cy3) which<br />

reduces consumption of microdissected samples.<br />

2. It is therefore imperative that a suitable reference proteome is identified i.e.<br />

one which not only demonstrates a high correlation to microdissected cells, but<br />

also provides an adequate protein concentration for subsequent identification.


8 Sitek et al.<br />

Therefore freeze several tissues containing pancreatic carcinoma on liquid<br />

nitrogen directly after resection <strong>and</strong> store at –80°C.<br />

3. For homogenization use 100 mg of the tissue on liquid nitrogen in 148 μL lysis<br />

buffer using a h<strong>and</strong> homogeniser <strong>and</strong> sonicate the sample 6 times for 10 sec<br />

on ice.<br />

4. To remove insoluble debris centrifuge (12,000g for 5 min) the homogenisate.<br />

5. Label 3 μg of each lysate with 2 nM Cy3 according to the st<strong>and</strong>ard labeling<br />

protocol according to user manual.<br />

6. Label aliquots of 1,000 microdissected cells with 4 nM Cy5.<br />

7. Pair-wise mix the labeled tissue lysates with labeled cells <strong>and</strong> process by 2-DE.<br />

8. After scanning, analyze the images using DIA module of DeCyder (Fig. 5) <strong>and</strong><br />

calculate a matching rate between microdissected cells <strong>and</strong> carcinoma tissue (see<br />

Note 5).<br />

9. Optimize the labeling conditions for the tissue lysate showing the highest matching<br />

rate with the microdissected cells also by same/same experiment (see Chapter 3.3).<br />

Fig. 5. Protein spot pattern of carcinoma tissue (reference proteome) <strong>and</strong> microdissected<br />

cells after 2-DE. For protein identification <strong>and</strong> internal st<strong>and</strong>ardization a<br />

reference proteome with high degree in protein spot matching is necessary. For the<br />

proteome of a pancreatic tissue lysate a high matching rate (>90%) has been determined.<br />

In section A all detected protein spots of carcinoma tissue <strong>and</strong> microdissected<br />

cells, respectively, can be matched (letters are shown for better assignment). Whereas<br />

in section B beside a number of assignable protein spots (numbers) a protein spot<br />

unique for microdissected cells (arrows) was revealed.


Application of Fluorescence Dye Saturation Labeling 9<br />

3.5. Two-Dimensional Gel Electrophoresis<br />

1. These instructions are modified from the 2-DE technique as described by Klose<br />

<strong>and</strong> Kobalz (2). This technique is based on isoelectric focusing employing carrier<br />

ampholyte tube gels. It can be easily adapted to the DIGE technique <strong>and</strong> other<br />

formats, including analytical as well as preparative gels. In spite of the fact<br />

that most of the instruments were constructed in-house, equivalent equipment<br />

is commercially available. For the second dimension the Desaga VA300 gel<br />

system is applied.<br />

2. Two days before running isoelectric focusing (IEF) tube gels (Ø 1.5 mm, 20 cm)<br />

are prepared. Add 45 μL of APS solution to 2 mL of separations gel solution<br />

<strong>and</strong> fill tube to first mark (20 cm) using a syringe. Now, add 14 μL APS solution<br />

to 0.7 mL cap gel solution <strong>and</strong> cast cap gel to second mark (20.5 cm) behind<br />

the separation gel. To prevent urea crystallisation place an air cushion under the<br />

cap gel to third mark (21 cm).<br />

3. Before starting IEF apply 2 mm sephadex solution to prevent protein precipitation<br />

onto the separation gel. Then load the sample (dye-labeled) <strong>and</strong> overlay<br />

the sample with protection solution (approx 5 mm) to prevent direct contact of<br />

the acidic cathodic buffer (see Note 6).<br />

4. Fill anodic (bottom) <strong>and</strong> cathodic buffer (top) into the IEF chambers. Ensure<br />

that no air bubbles hamper IEF (see Note 6). Start isoelectric focusing applying<br />

a step-wise voltage program (100V for 1 h, 200 V/1 h, 400 V/17.5 h, 650 V/1<br />

h, 1,000 V/30 min, 1,500 V/10 min, 2,000 V/5 min).<br />

5. While the 21.5 h IEF is running, gels for the second dimension should be<br />

prepared. Clean the glass plates thrice (gel side) using a lint-free paper towel—<br />

for the first wash use doubled distilled water followed by 100% ethanol <strong>and</strong><br />

finally 70% ethanol. To ensure correct gel dimension two 1.5 mm plastic spacers<br />

are placed between two plates sealed by silicon.<br />

6. Add 288 μL APS solution into 144 mL gel buffer, cast the gel <strong>and</strong> overlay with<br />

water-saturated isobutanol.<br />

7. After polymerization (45 min) remove isobutanol <strong>and</strong> wash the surface with a<br />

protection solution. To protect gel drying place 2-DE protection solution onto<br />

the gel <strong>and</strong> store the gels at 4°C.<br />

8. After IEF extrude the gel by means of inserting a nylon fiber (see Note 7) into<br />

the gel groove of the IEF gel carrier <strong>and</strong> incubate with equilibration solution for<br />

15 min to load proteins with SDS.<br />

9. Wash the gel three times with running buffer before applying to the second<br />

dimension.<br />

10. For the transfer of the IEF gel into SDS-<strong>PAGE</strong> gel hold the groove with gel<br />

in contact with the edge of the glass plate <strong>and</strong> slide the gel between the glass<br />

plates using a wire suitably formed.<br />

11. Overlay the IEF gels with agarose solution, add the running buffer to the upper<br />

<strong>and</strong> lower (15°C) chambers <strong>and</strong> start the electrophoresis. For the entrance of the<br />

proteins into the SDS-<strong>PAGE</strong> gel apply low current (75 mA) for 15min. When the<br />

proteins have entered increase the current to 200 mA for approximately 5–7 h.


10 Sitek et al.<br />

3.6. Differential Proteome Analysis of 1000 Microdissected Cells<br />

from Different PanIN Stages<br />

1. This instruction comprised all steps obtained during optimisation procedure as<br />

described above.<br />

2. Harvest 1000 cells by manual microdissection <strong>and</strong> incubate the cells in 100 μL<br />

lysis buffer.<br />

3. For protein labeling according to the optimised protocol reduce the proteins with<br />

2 nM TCEP <strong>and</strong> label the proteins of the microdissected cells with 4 nM Cy5.<br />

4. For the preparation of the internal st<strong>and</strong>ard label 3 μg protein from a PDAC<br />

tissue sample lysate with 2 nM Cy3.<br />

5. Add 10 μL DTT to stop labeling reaction <strong>and</strong> add 10 μL Ampholytes, pH 2–4.<br />

Fig. 6. Representative images for each investigated progression step of PDAC. The<br />

lysates from 1000 microdissected cells were labeled with Cy5 mixed with internal<br />

st<strong>and</strong>ard <strong>and</strong> processed by 2-DE. Subsequent image acquisition the spot patterns were<br />

analyzed using DeCyder software <strong>and</strong> 2,000–2,500 protein spots have been detected<br />

for the different PanIN grades. Protein spot 2574 has been detected as differentially<br />

expressed in PanIN 2, PanIN 3 <strong>and</strong> Carcinoma (see Fig.7)


Application of Fluorescence Dye Saturation Labeling 11<br />

6. Mix the sample <strong>and</strong> analyse the mixture using 2-DE.<br />

7. After 2-DE, leave the gels between the glass plates <strong>and</strong> acquire the images<br />

using the Typhoon 9400 scanner (see Note 3). Therefore, choose excitation<br />

wavelengths <strong>and</strong> emission filters specific for each of the CyDyes according to<br />

the Typhoon user guide.<br />

8. Before image analysis with DeCyder software crop the images with<br />

ImageQuant TM software (Fig. 6).<br />

9. For intra-gel spot detection <strong>and</strong> quantification use the Differential In-gel Analysis<br />

(DIA) mode of the DeCyder software. Set the estimated number of spots to<br />

3000. Apply an exclusion filter to remove spots with a slope greater than 1.6<br />

(see Note 4).<br />

10. After spot matching between the different gels using the Biological Variation<br />

Analysis mode (BVA) consider only protein spots with an expression changes<br />

of factor > 1.6 <strong>and</strong> p-value (Student’s t-test) < 0.05 as significantly regulated<br />

(Fig. 7).<br />

Fig. 7. Stage-dependent regulation of protein spot 2574. For each patient <strong>and</strong> PanIN<br />

stage the protein spot intensity is shown. The depicted protein spot 2574 (see Fig. 6)<br />

shows a significant up-regulation (p < 0.05) in the carcinoma stage by a factor of 3.2.<br />

In the PanIN stages 2 <strong>and</strong> 3 a significant down-regulation by a factor of -2.7 <strong>and</strong> -1.8<br />

has been determined.


12 Sitek et al.<br />

3.7. Micropreparation of Significantly Regulated Protein Spots<br />

1. After differential analysis protein spots are identified using mass spectrometry<br />

(MALDI-MS, ESI-MS) subsequent tryptic in-gel digestion. Therefore, 100-fold<br />

protein amount of the internal st<strong>and</strong>ard (reference proteome) must be labeled <strong>and</strong><br />

applied to 2-DE. A preparative label kit with 400 nM Cy3 is available.<br />

2. For preparative gels label 400 μg of tissue lysate with 260 nM of Cy3 (130 nM<br />

TCEP) (see Note 8).<br />

3. Directly after gel scanning isolate the protein spots of interest manually (see<br />

Note 9). Assign the positions of the spots using a printout of the gel placed<br />

underneath the glass plate.<br />

4. Put the isolated protein spots into sample cups (glass) <strong>and</strong> store them at –80°C.<br />

5. For protein identification different mass spectrometric (MS) techniques can<br />

be applied subsequent to enzymatic in-gel digestion. Matrix assisted laser<br />

desorption/ionization MS (MALDI-MS) allows a fast <strong>and</strong> sensitive MS analysis<br />

performing peptide mass fingerprinting (PMF) (7). In cases where more<br />

sequence information is necessary (e.g. posttranslational modification) liquid<br />

chromatography-coupled electrospray ionisation MS (LC-ESI-MS) with its high<br />

performance for peptide fragment mass fingerprinting (PFF) is preferable (8).<br />

4. Notes<br />

1. To protect samples against tissue disruption (freezer burn) after resection the<br />

tissue is placed in tin foil at first <strong>and</strong> then stored in a cryotube.<br />

2. Usually, depending of tube’s diameter not more than 50 μL can be applied.<br />

Therefore, we increased the volume by blowing up the glass tube so that a sample<br />

volume of 100–200 μL can be applied.<br />

3. Before scanning the glass plates have to be cleaned thoroughly. First of all use<br />

ethanol for removing of acrylamide or silicone <strong>and</strong> then clean the glass plates<br />

with water.<br />

4. To avoid the detection of dust particles <strong>and</strong> artifacts as protein spots in the gel,<br />

an exclusion filter concerning slope, area, peak height, or volume can be applied.<br />

For one or more of these characteristics a value that distinguishes dust particles<br />

<strong>and</strong> spots has to be found.<br />

5. For the determination of the matching rate between two images which derived<br />

from the same gel analyze the gel using DIA mode of DeCyder software. After a<br />

spot detection an average ratio of 2.0 could be set in order to find spots occurring<br />

in both images (Average ratio < 2.0).<br />

6. Air bubbles should be avoided by applying the solution slowly under the surface<br />

(1 mm) of the solution which has been applied before. For Sephadex application<br />

the small volume of separation gel buffer generated during polymerisation is<br />

sufficient.<br />

7. To prevent destruction of the IEF gel by the nylon fiber (i) the thermoplastic nylon<br />

fiber should be fitted to the tube inner diameter by melting one end into the tube<br />

<strong>and</strong> (ii) the gel should be extruded using the cap gel (acrylamide concentration


Application of Fluorescence Dye Saturation Labeling 13<br />

12.3%) as a cushion or (iii) another possibility is to polymerise a high concentrated<br />

acrylamide solution (15%) above the extruding side <strong>and</strong> use this gel piece as a<br />

cushion.<br />

8. To avoid a high sample volume for the preparative labeling TCEP <strong>and</strong> Cy3 should<br />

have a concentration of 20 mM respectively instead of 2 mM (see Chapter 2.2).<br />

The amount of TCEP <strong>and</strong> Cy3 for labeling of preparative protein lysate has to be<br />

calculated according to labeling conditions for analytical gels.<br />

9. Before scanning, mark the glass plate from the picking gel with fluorescence<br />

stickers The stickers are necessary for matching between the gel (the proteins are<br />

not visible) <strong>and</strong> the gel image. After scanning print the gel image in the original<br />

size. Put the print under the glass plates, align the position of the stickers on the<br />

glass plate with the image.<br />

Acknowledgments<br />

The authors would like to thank Kathy Pfeiffer, Conny Bieling, Sabine<br />

Burkert <strong>and</strong> Birgit Streletzki for excellent technical assistance <strong>and</strong> Jon Barbour<br />

for critical reading of the manuscript. This work was supported by the grant<br />

from the Deutsche Krebshilfe (B.S., J.L., S.A.H <strong>and</strong> K.S., 70-2988-Schm3),<br />

Bundesministerium für Bildung und Forschung (NGFN, FZ 031U119) <strong>and</strong> the<br />

Nordrhein Westfalen Ministerium für Wissenschaft und Forschung.<br />

References<br />

1. Sitek, B., Lüttges, J., Marcus, K., Klöppel, G., Schmiegel, W., Meyer, H. E., Hahn, S.<br />

A. <strong>and</strong> Stühler, K. (2005) Application of fluorescence difference gel electrophoresis<br />

saturation labeling for the analysis of microdissected precursor lesions of pancreatic<br />

ductal adenocarcinoma. J. Proteomics, 5, 2665–2679.<br />

2. Klose, J. <strong>and</strong> Kobalz, U. (1995) Two-dimensional electrophoresis of proteins: an<br />

updated protocol <strong>and</strong> implications for a functional analysis of the genome. J.<br />

Electrophoresis 6, 1034–1059.<br />

3. Kondo, T., Seike, M., Mori, Y., Fujii,K., Yamada, T., <strong>and</strong> Hirohashi, S. (2003)<br />

Application of sensitive fluorescent dyes in linkage of laser microdissection <strong>and</strong><br />

two-dimensional gel J. Proteomics 3, 1758–1766.<br />

4. Sitek, B., Scheibe, B., Jung, K, Schramm ,A., <strong>and</strong> Stühler, K. (2006) Difference gel<br />

electrophoresis (dige): the next generation of two-dimensional gel electrophoresis<br />

for clinical research, in Proteomics in Drug Research (Hamacher et al., eds), Wiley-<br />

VCH, Weinberg, pp 33–55.<br />

5. Unlü, M., Morgan,M. E., <strong>and</strong> Minden,J. S. (1997) Difference gel electrophoresis:<br />

a single gel method for detecting changes in protein extracts. Electrophoresis 18,<br />

2071–2077.<br />

6. Gruber, H. J., Hahn, C. D., Kada, G., Riener, C. K., Harms, G. S., Ahrer, W.,<br />

Dax, T. G., <strong>and</strong> Knaus, H.-G. (2000) Anomalous fluorescence enhancement of Cy3


14 Sitek et al.<br />

<strong>and</strong> cy3.5 versus anomalous fluorescence loss of Cy5 <strong>and</strong> Cy7 upon covalent linking<br />

to IgG <strong>and</strong> noncovalent binding to avidin. Bioconjug. Chem. 11, 696–704.<br />

7. Stühler, K., <strong>and</strong> Meyer, H. E. (2004) MALDI: more than peptide mass fingerprints.<br />

Curr Opin Mol Ther. 3, 239–248.<br />

8. Marcus, K., Moebius, J., <strong>and</strong> Meyer, H. E. (2003) Differential analysis of phosphorylated<br />

proteins in resting <strong>and</strong> thrombin-stimulated human platelets. Anal Bioanal<br />

Chem. 7, 973–993.


2<br />

Albumin <strong>and</strong> Immunoglobulin Depletion<br />

of Human Plasma<br />

Rosalind E. Jenkins, Neil R. Kitteringham, Carrie Greenough,<br />

<strong>and</strong> B. Kevin Park<br />

Summary<br />

Plasma <strong>and</strong> serum have been the focus of intense study in recent years in the expectation<br />

that they will provide important biomarkers of health <strong>and</strong> disease, without the need for<br />

invasive procedures. This aim has been hindered by the fact that a few highly abundant<br />

proteins dominate the protein profile, masking the lower abundance proteins <strong>and</strong> limiting<br />

our ability to analyse the entire plasma proteome. This chapter details a simple <strong>and</strong><br />

effective method for removal of two of the most dominant proteins in plasma, albumin<br />

<strong>and</strong> immunoglobulin.<br />

Key Words: Affinity chromatography; albumin; depletion; immunoglobulin; plasma<br />

proteome.<br />

1. Introduction<br />

The dynamic range of proteins in human plasma is greater than ten orders<br />

of magnitude, from albumin present at around 30 mg/mL to cytokines such<br />

as interleukin 6 present at basal levels of 1–3 pg/mL (1). This extraordinarily<br />

wide range of protein concentrations within a single compartment makes<br />

detailed analysis of the lower abundance proteins technically dem<strong>and</strong>ing, yet it<br />

is exactly these low level species that are likely to provide insight into disease<br />

states, into the effects of therapeutic intervention, <strong>and</strong> to yield specific <strong>and</strong><br />

sensitive biomarkers. Many methods have been developed to deplete serum <strong>and</strong><br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

15


16 Jenkins et al.<br />

plasma of the high abundance, housekeeping proteins, including alcohol precipitation,<br />

ultracentrifugation, salting in/salting out (2,3), <strong>and</strong> molecular weight<br />

fractionation (4,5). However, removal of proteins by affinity capture is the<br />

most commonly used approach because of the highly selective nature of the<br />

depletion.<br />

Anti-human albumin monoclonal antibody immobilized onto a solid support<br />

such as sepharose forms the basis of many immunoaffinity columns for the<br />

removal of the most abundant protein in human plasma. Clearly the affinity<br />

<strong>and</strong> specificity of the antibody determine the efficiency <strong>and</strong> discrimination of<br />

protein removal. However, the support on which the antibody is immobilized<br />

will also impact on the process, not least in terms of the binding capacity of<br />

the matrix. Affinity matrices based on bacterial protein A, protein G, or protein<br />

L, or molecularly engineered versions of these, are most commonly used to<br />

isolate immunoglobulins (6–11). Sophisticated multicomponent immunoaffinity<br />

matrices that are designed to deplete 10–15 of the most abundant plasma<br />

proteins are available (12) <strong>and</strong> have been used with great success, but they do<br />

have several drawbacks: they are expensive; they have a relatively low binding<br />

capacity so that several aliquots of plasma must be processed individually<br />

<strong>and</strong> then pooled to generate sufficient material for further analysis; <strong>and</strong> the<br />

depletion of the target proteins is frequently far from complete. The method<br />

described in this chapter, although limited to the depletion of only two of the<br />

highest abundance proteins, is simple, rapid, <strong>and</strong> accessible <strong>and</strong> provides a<br />

significant improvement in coverage of the plasma proteome obtainable by <strong>2D</strong><br />

gel electrophoresis.<br />

2. Materials<br />

2.1. Blood Sampling<br />

1. Heparinized tubes.<br />

2. Refrigerated centrifuge (up to 450g).<br />

3. 0.5-mL Eppendorf tubes.<br />

2.2. Albumin Depletion<br />

1. 2-mL cartridge containing POROS® beads coated with monoclonal goat antihuman<br />

serum albumin (HSA) (Applied Biosystems).<br />

2. Loading <strong>and</strong> washing buffer: phosphate buffered saline (PBS): 137 mM NaCl, 1<br />

mM KH2PO4, 5.6 mM Na2HPO4, 2.7 mM KCl, pH 7.4.<br />

3. Elution buffer: 12 mM HCl.<br />

4. HSA column wash buffer: 1 M NaCl.<br />

5. High performance liquid chromatography system capable of flow rates from 0.5–3<br />

mL/min (for automated depletions).


Albumin <strong>and</strong> Immunoglobulin Depletion of Human Plasma 17<br />

6. 2-mL <strong>and</strong> 10-mL plastic syringes (for manual depletions).<br />

7. Blunt ended syringe needle (for manual depletions).<br />

8. 1.5-mL Eppendorf tubes.<br />

9. Spectrophotometer able to read the absorbance at 280 nm (for manual depletions).<br />

2.3. Immunoglobulin Depletion<br />

1. 0.2 mL cartridge containing POROS® beads coated with recombinant protein G<br />

(Applied Biosystems).<br />

2. Loading <strong>and</strong> washing buffer: phosphate buffered saline (PBS).<br />

3. Elution buffer: 12 mM HCl.<br />

4. Protein G column wash buffer: 1 M NaCl 10% acetic acid.<br />

5. High performance liquid chromatography system capable of flow rates from 0.5–3<br />

mL/min (for automated depletions).<br />

6. 2-mL <strong>and</strong> 10-mL plastic syringes (for manual depletions).<br />

7. Blunt ended syringe needle (for manual depletions).<br />

8. 1.5-mL Eppendorf tubes.<br />

9. Spectrophotometer able to read the absorbance at 280 nm (for manual depletions).<br />

2.4. One-Dimensional (1D) gel Electrophoresis<br />

1. St<strong>and</strong>ard 1D gel electrophoresis apparatus.<br />

2. Laemmli sample buffer (1×): 3% SDS, 0.1 M Tris-HCl, 15% glycerol, 0.2%<br />

bromophenol blue, pH 7.6.<br />

2.5. TCA Precipitation of Proteins<br />

1. 20% trichloroacetic acid (TCA) <strong>and</strong> ice-cold acetone.<br />

2. Resuspension buffer: 5% SDS, 1.15% DTT.<br />

3. Refrigerated centrifuge (up to 14,000g).<br />

2.6. <strong>2D</strong> Gel Electrophoresis<br />

1. St<strong>and</strong>ard <strong>2D</strong> gel electrophoresis apparatus.<br />

2. IPG rehydration buffer: 9 M urea, 2% (w/v) CHAPS, bromophenol blue (trace),<br />

2% IPG buffer (Pharmacia), 0.28% DTT<br />

3. Methods<br />

3.1. Collection of <strong>Sample</strong>s<br />

1. Collect the blood into heparinized tubes <strong>and</strong> sediment the red blood cells as soon<br />

after acquisition as possible by centrifugation at 450g for 10 min (see Notes 1,<br />

2, <strong>and</strong> 3).


18 Jenkins et al.<br />

2. Recover the supernatant, divide it into small aliquots <strong>and</strong> store them at –80°C.<br />

Each aliquot should be thawed <strong>and</strong> used only once.<br />

3. Once thawed, centrifuge the aliquot briefly to remove any precipitate that may<br />

have formed at low temperatures <strong>and</strong> that may block the affinity cartridges.<br />

3.2. Albumin Depletion<br />

1. <strong>Sample</strong>s are diluted to 6 mg/mL in PBS <strong>and</strong> 600 μL (equivalent to 3.6 mg protein<br />

or approx 60 μL plasma) are applied to the column (see Notes 4, 5 <strong>and</strong> 6).<br />

2. The POROS column is mounted in a cartridge holder supplied by the manufacturer<br />

with ports for insertion of st<strong>and</strong>ard HPLC fittings, if performing the following<br />

steps robotically, or for a needle port adapter at the top <strong>and</strong> a short piece of PEEK<br />

tubing at the base to help direct the flow-through when performing the steps<br />

manually. The blunt-ended needle attached to a 10-mL syringe is inserted firmly<br />

into the needle port adapter for manual application of the sample <strong>and</strong> buffers<br />

to the column, which should be applied at a flow rate of one drop per second<br />

throughout for manual chromatography. The flow rates for depletions performed<br />

on an HPLC system are given at the appropriate points in the text.<br />

3. The column is equilibrated by applying 10 column volumes (20 mL) of PBS at a<br />

flow rate of 2.4 mL/min.<br />

4. The diluted plasma sample is applied to the column at a flow rate of 1.2 mL/min<br />

<strong>and</strong> the flow-through (containing plasma proteins minus albumin) is collected as<br />

500-μL fractions into 1.5-mL eppendorf tubes.<br />

5. A further 10 column volumes (20 mL) PBS are applied to the column to ensure that<br />

all non-specifically bound proteins are flushed through, with continued collection<br />

of fractions. Collection of a total of ten fractions is generally sufficient to capture<br />

all of the nonbound plasma proteins.<br />

6. Albumin is eluted from the column by the application of 5 column volumes (10<br />

mL) of 12 mM HCl at a flow rate of 2.4 mL/min, with the first five 1-mL fractions<br />

collected into 1.5-mL eppendorf tubes.<br />

7. The column is cleaned by applying 10 column volumes (20 mL) of HSA column<br />

wash buffer (1 M NaCl) at a flow rate of 2.4 mL/min (see Note7).<br />

8. If further samples are to be processed, the column may be equilibrated with PBS<br />

as in point 3. If the column is to be stored for 1–2 d, flushing through with 10<br />

column volumes (20 mL) PBS <strong>and</strong> storage at 4°C will be sufficient. If it is to be<br />

stored for a longer period, the PBS should contain 0.05% sodium azide.<br />

3.3. Assessment of Fractions Containing Albumin-Depleted Plasma<br />

Proteins by 1D Gel Electrophoresis (see Note 8)<br />

1. Aliquots of 10 μL of the flow-through fractions are mixed with 4× Laemmli<br />

sample buffer, boiled <strong>and</strong> loaded onto SDS-<strong>PAGE</strong> (sodium dodecyl sulphate<br />

polyacrylamide gel electrophoresis) minigels.<br />

2. Similarly, 3–6 μL of the eluted HSA fractions, <strong>and</strong> 2 μL of the dilute undepleted<br />

plasma sample, are prepared for gel analysis.


Albumin <strong>and</strong> Immunoglobulin Depletion of Human Plasma 19<br />

Fig. 1. Depletion of albumin from human plasma. Coomassie blue (A) <strong>and</strong> silver (B)<br />

stained 1D gel of undepleted plasma (P), aliquots of the flow-through fractions 1–10<br />

<strong>and</strong> eluted HSA (E1). The undepleted plasma appears to contain a relatively complex<br />

mixture of proteins that is dominated by the albumin b<strong>and</strong> at 66 kDa. The depleted<br />

fractions should contain the same range of proteins but the albumin b<strong>and</strong> should be<br />

completely absent. In contrast, the fraction representing eluted albumin should contain<br />

no other protein b<strong>and</strong>s, even at the level of sensitivity of silver stain. The multiple<br />

b<strong>and</strong>s visible below the major HSA b<strong>and</strong> were determined by mass spectrometry to<br />

be breakdown products, presumably because of the acid conditions under which the<br />

albumin was eluted from the column. (C) UV trace of albumin depletion performed by<br />

HPLC showing that fractions 3–7 contain the protein flow-through from the anti-HSA<br />

column whereas fractions 17 <strong>and</strong> 18 contain the eluted HSA (E1) itself.<br />

3. The samples are loaded onto 10% SDS-<strong>PAGE</strong> minigels <strong>and</strong> the gels stained with<br />

colloidal Coomassie Blue (Fig. 1A) or silver stain (Fig. 1B) after electrophoresis.<br />

3.4. Assessment of Fractions Containing Albumin-Depleted Plasma<br />

Proteins by UV Absorbance<br />

1. Once the depletion method has been optimized, the flow-through fractions may<br />

be assessed by spectrophotometry. If samples have been depleted using an<br />

HPLC system, a real-time UV trace of the chromatographic separation is usually<br />

available. Fig. 1 is an example of such a trace.


20 Jenkins et al.<br />

2. If performing the depletions manually, the fractions may be assessed using an<br />

off-line spectrophotometer. Aliquots of 100 μL of each fraction are placed in<br />

mini-cuvets or wells of a 96-well plate, <strong>and</strong> the absorbance at 280 nm recorded.<br />

3. All flow-through fractions producing an absorbance value are pooled for the next<br />

step in the protocol, the depletion of immunoglobulin (see Note 9).<br />

3.5. Immunoglobulin Depletion<br />

1. The 0.2-mL protein G POROS column is supplied with a smaller cartridge holder<br />

<strong>and</strong> should be mounted as described earlier. The blunt-ended needle attached to a<br />

2-mL syringe is used for manual application of sample <strong>and</strong> buffers to the column.<br />

As for the HSA depletion column, all reagents are applied to the column at a<br />

flow rate of 1 drop per second when the chromatography is performed manually.<br />

Flow rates for depletion using an HPLC system are noted at appropriate points<br />

in the text.<br />

2. The column is equilibrated by applying 10 column volumes (2 mL) of PBS at a<br />

flow rate of 1mL/min.<br />

3. The albumin-depleted plasma sample cannot be applied as a single aliquot but<br />

should be split into two aliquots of 600 μL (see Note 10), each being loaded<br />

at a flow rate of 0.5 mL/min. The flow-through (containing plasma proteins<br />

minus albumin <strong>and</strong> immunoglobulin) is collected as 500-μL fractions into 1.5-mL<br />

eppendorf tubes.<br />

4. A further 10 column volumes (2 mL) PBS are applied to the column at a flow rate<br />

of 1 mL/min to ensure that all nonspecifically bound proteins are flushed through,<br />

with continued collection of fractions. Collection of a total of six fractions is<br />

generally sufficient to capture all the nonbound plasma proteins.<br />

5. Immunoglobulin is eluted from the column by the application of 10 column<br />

volumes (2 mL) of 12 mM HCl at a flow rate of 1 mL/min, with two 1-mL<br />

fractions being collected into 1.5-mL eppendorf tubes.<br />

6. After the first aliquot has been depleted of immunoglobulin, the column is reequilibrated<br />

with PBS as in point 2, <strong>and</strong> the second aliquot processed as in<br />

steps 3–5.<br />

7. After the second aliquot has been processed, 10 column volumes (2 mL) of protein<br />

G column wash buffer (1 M NaCl containing 10% acetic acid) are applied to the<br />

column at a flow rate of 1 mL/min (see Note 11). As for the anti-HSA column,<br />

the protein G column may be stored for 1–2 days with PBS but for longer periods,<br />

the PBS should contain 0.05% sodium azide.<br />

3.6. Assessment of Fractions Containing Albumin<strong>and</strong><br />

Immunoglobulin-Depleted Plasma Proteins by 1D Gel<br />

Electrophoresis (see Note 8)<br />

1. Aliquots of 10 μL of the flow-through fractions are mixed with 4× Laemmli<br />

sample buffer, boiled <strong>and</strong> loaded onto SDS-<strong>PAGE</strong> minigels.


Albumin <strong>and</strong> Immunoglobulin Depletion of Human Plasma 21<br />

Fig. 2. Depletion of immunoglobulin from albumin-depleted human plasma.<br />

Coomassie blue (A) <strong>and</strong> silver (B) stained 1D gel of undepleted plasma (P),<br />

HSA-depleted plasma (H), aliquots of the flow-through fractions 1–6 <strong>and</strong> eluted<br />

immunoglobulin (E2). The depletion of the immunoglobulin b<strong>and</strong>s is more difficult to<br />

discern than the depletion of the very abundant albumin b<strong>and</strong> shown in figure 1, but<br />

the eluted protein should be clearly seen as two b<strong>and</strong>s representing the heavy <strong>and</strong> light<br />

chains of the immunoglobulin (Ig H <strong>and</strong> Ig L). (C) UV trace of immunoglobulin depletion<br />

performed by HPLC showing that fractions 1 <strong>and</strong> 2 contain the protein flow-through<br />

from the protein G column whereas fraction 7 contains the eluted immunoglobulin.<br />

2. Similarly, 3–6 μL of the eluted immunoglobulin fractions, 2 μL of the dilute<br />

undepleted plasma, <strong>and</strong> 10 μL of the albumin-depleted plasma are prepared for<br />

gel analysis.<br />

3. The samples are electrophoresed on 10% SDS-<strong>PAGE</strong> minigels <strong>and</strong> the gels stained<br />

with colloidal Coomassie Blue or silver stain (Fig. 2).<br />

3.7. Assessment of Fractions Containing Albumin- <strong>and</strong><br />

Immunoglobulin-Depleted Plasma Proteins by UV Absorbance<br />

1. Once the depletion method has been optimized, the flow-through fractions may<br />

be assessed by simple spectrophotometry by measuring their absorbance in-line


22 Jenkins et al.<br />

at 214 nm using the UV detector on the HPLC system, or off-line at 280 nm in<br />

a cuvet or microplate format.<br />

2. All flow-through fractions producing an absorbance value are pooled before<br />

processing for <strong>2D</strong> gel electrophoresis or other proteomic analysis (see Note 12).<br />

3.8. TCA Precipitation of Depleted Protein <strong>Sample</strong>s<br />

for <strong>2D</strong> Gel Electrophoresis<br />

1. The depleted samples should be maintained at 4 ° C or on ice throughout this<br />

procedure.<br />

2. A solution of 20% TCA in water is prepared just before use <strong>and</strong> placed on ice to<br />

chill (see Note 13).<br />

3. 2 mL 20% TCA are added to 2 mL of the dilute depleted plasma, <strong>and</strong> the sample<br />

is mixed gently <strong>and</strong> incubated on ice for 30 min.<br />

4. The sample is then divided equally between three 1.5-mL eppendorf tubes <strong>and</strong><br />

centrifuged at 14,000g <strong>and</strong> 4 ° C for 10 min to pellet the proteins.<br />

Fig. 3. <strong>2D</strong> gel images of undepleted (A) <strong>and</strong> depleted (B) plasma. The plasma<br />

proteins were focussed on nonlinear pH 3–10 IPG strips before 2nd dimension<br />

separation on 12% SDS <strong>PAGE</strong> gels. The gels were stained with colloidal Coomassie<br />

blue. Image analysis of the gels shows that the albumin <strong>and</strong> immunoglobulin heavy<br />

chain are 98% <strong>and</strong> 80% depleted, respectively. The resolution of the protein spots is<br />

improved significantly by depletion, particularly for proteins migrating in the top half<br />

of the gel close to the migration position of albumin. Indeed, peptide mass fingerprinting<br />

of the visible features reveals that hemopexin comigrates with albumin, yet it<br />

is undetectable on gels of the undepleted sample. There is an overall increase in the<br />

number of detectable spots of approx 50% when the gels are stained with Coomassie<br />

blue, indicating that the detection of lower abundance proteins has been improved.


Albumin <strong>and</strong> Immunoglobulin Depletion of Human Plasma 23<br />

5. The supernatant is discarded, <strong>and</strong> the pellets washed with 2×1mLofice-cold<br />

acetone to remove all traces of the TCA.<br />

6. The pellets are air-dried briefly (approx 5 min) before resuspension in 12 μL<br />

resuspension buffer (see Note 14).<br />

7. The sample is heated at 95 ° C for 10 min to aid solubilisation followed by the<br />

addition of 350 μL IPG-strip rehydration buffer.<br />

8. The sample is clarified by a high speed centrifugation step before conventional<br />

<strong>2D</strong> electrophoresis (Fig. 3).<br />

4. Notes<br />

1. To make meaningful comparisons between plasma samples, it is vital that the<br />

collection <strong>and</strong> storage is consistent. Plasma is rich in proteases <strong>and</strong> inappropriate<br />

or overlong storage can lead to significant changes in the protein profile.<br />

Whatever procedure is chosen for sample preparation, it must be strictly<br />

adhered to.<br />

2. Serum <strong>and</strong> plasma are almost identical in terms of the protein profile observed<br />

on <strong>2D</strong> gels. However, some of the proteins involved in the clotting process,<br />

such as fibrinogen, are removed when the clotted red blood cells are separated<br />

from the serum by centrifugation. There is also a slightly increased tendency<br />

for lysis of the red blood cells when preparing serum. It is probably easier <strong>and</strong><br />

more consistent to prepare plasma.<br />

3. The anti-HSA column may be used to deplete albumin from the plasma or serum<br />

of various animal species, but lower levels of total protein must be loaded onto<br />

the column. We have depleted albumin from mouse <strong>and</strong> rat serum after loading<br />

1–2 mg total protein. The protein G column works as effectively for animal<br />

immunoglobulins as for human.<br />

4. Most human plasma samples have a protein concentration of 60–70 mg/mL with<br />

approximately half of that comprised of albumin. The binding capacity of a 2<br />

mL anti-HSA POROS cartridge is 1.8–2 mg albumin so the samples will require<br />

a dilution step before loading onto the column.<br />

5. Both the anti-HSA <strong>and</strong> the protein G POROS columns may be used for the<br />

depletion of multiple samples without loss of effectiveness, as long as they are<br />

cleaned <strong>and</strong> stored correctly. The smaller protein G column does have a tendency<br />

to deteriorate before the larger anti-HSA column, but we have successfully<br />

processed at least 50 plasma samples through one pair of columns.<br />

6. Manual depletions are just as effective as those performed on HPLC systems,<br />

but they are rather tedious. A cheap but effective alternative is to use a syringe<br />

pump to apply samples <strong>and</strong> buffers.<br />

7. The anti-HSA column should be cleaned with wash buffer (1 M NaCl) after<br />

every depletion to prevent residual protein build-up that would lead to decreased<br />

efficiency of albumin removal <strong>and</strong> increased pressure during the chromatography<br />

steps.


24 Jenkins et al.<br />

8. Measuring the absorbance at 214/280 nm provides a measure of the total amount<br />

of protein in each fraction, but it does not indicate how much of the target<br />

protein has been removed (efficiency of depletion), nor whether other proteins<br />

have been depleted as well (specificity of depletion). A visual assessment of<br />

these factors can be rapidly achieved by 1D gel electrophoresis. However, once<br />

the protocol has been optimized, it should be necessary to perform this sort of<br />

quality control only if the UV trace looks anomalous, or when a new POROS<br />

cartridge is being employed.<br />

9. At this stage, it is usual to have a total volume of approx 1.5 mL of albumindepleted<br />

plasma containing roughly 900 μg of protein. Because the amount of<br />

protein loaded onto the column was approx 3.6 mg, the reduction in total protein<br />

content is equivalent to 75% suggesting that the depletion of albumin is complete.<br />

10. Immunoglobulin comprises 8–26% of the total protein in plasma <strong>and</strong> is<br />

therefore present at concentrations of 5–18 mg/mL. The sample has already<br />

been diluted at least 1:10 during the albumin depletion, so the concentration is<br />

now approx 0.5–1.8 mg/mL. The binding capacity of the protein G cartridge is<br />

up to 1.8 mg immunoglobulin, so the maximum volume of the pooled fractions<br />

containing HSA-depleted plasma that could be loaded is 1 mL. These fractions<br />

may be stored at 4 ° C for short periods of time before the immunoglobulin<br />

depletion, but should be processed within 8h.<br />

11. The protein G column should be cleaned after 2 immunoglobulin depletions<br />

to keep it functioning at the highest efficiency. After the first depletion, the<br />

column is simply re-equilibrated with PBS as in Section 2.3.5 point 1, but after<br />

the second, it should be exposed to wash buffer (1 M NaCl/10% acetic acid).<br />

12. At this stage, it is usual to have a total volume of approx 2 mL of depleted plasma<br />

containing roughly 500 μg protein. This is equivalent to a further 11% reduction<br />

in protein content of the plasma, which is approximately what would be expected<br />

following removal of the immunoglobulin. However, this is too dilute for most<br />

proteomic analyses so a method to concentrate the proteins must be employed.<br />

13. There are several reagents for precipitating proteins from dilute samples,<br />

including acetone, methanol <strong>and</strong> acetonitrile, but the method that seems to<br />

work most effectively for the samples described here is TCA precipitation. It<br />

is essential that the TCA is prepared immediately before use: even storage for<br />

2–3 h reduces the efficiency of precipitation. The sample may be precipitated<br />

with TCA for longer than 30 min, but there is a risk of protein degradation<br />

or modification on prolonged exposure. Alternative methods for protein<br />

concentration include molecular weight cut-off filters, for which a significant<br />

loss of total protein is a factor, or the use of reversed phase matrices to capture<br />

the proteins, for which the elution buffer must be very carefully selected for its<br />

compatibility with the 1st dimension separation.<br />

14. The volume of resuspension buffer (5% SDS, 1.15% DTT) to be added must<br />

be calculated carefully to ensure that after mixing with IPG-strip rehydration<br />

buffer, the level of SDS is within tolerable levels for the 1st dimension


Albumin <strong>and</strong> Immunoglobulin Depletion of Human Plasma 25<br />

separation, i.e., less than 0.25%. When the sample described here is mixed<br />

with 350-μL IPG-strip rehydration buffer, the final concentration of SDS in the<br />

buffer is 0.17%, well within the tolerance of the 1st dimension separation.<br />

Acknowledgments<br />

This work was supported by the Wellcome Trust. Thanks to Rod Watson of<br />

Applied Biosystems for assistance with the establishment of protocols.<br />

References<br />

1. Anderson, N. L., <strong>and</strong> Anderson, N. G. (2002) The human plasma proteome: history,<br />

character, <strong>and</strong> diagnostic prospects. Mol. Cell Proteomics 1, 845–67.<br />

2. Cohn, E. J. (1941) The properties <strong>and</strong> functions of the plasma proteins with a<br />

consideration of the methods for their separation <strong>and</strong> purification Chem. Rev. 28,<br />

395.<br />

3. Simoni, R. D., Hill, R. L., <strong>and</strong> Vaughan, M. (2002) The beginning of protein<br />

physical chemistry. Determinations of protein molecular weights. The work of<br />

Edwin Joseph Cohn. J. Biol. Chem. 277, 19e.<br />

4. Georgiou, H. M., Rice, G. E., <strong>and</strong> Baker, M. S. (2001) Proteomic analysis of<br />

human plasma: Failure of centrifugal ultrafiltration to remove albumin <strong>and</strong> other<br />

high molecular weight proteins Proteomics 1, 1503–06.<br />

5. Tirumalai, R. S., Chan, K. C., Prieto, D. A., Issaq, H. J., Conrads, T. P., <strong>and</strong><br />

Veenstra, T. D. (2003) Characterization of the low molecular weight human serum<br />

proteome Mol. Cell Proteomics 13, 13.<br />

6. Akerstrom, B., Brodin, T., Reis, K., <strong>and</strong> Bjorck, L. (1985) Protein G: a powerful<br />

tool for binding <strong>and</strong> detection of monoclonal <strong>and</strong> polyclonal antibodies J. Immunol.<br />

135, 2589–92.<br />

7. Forsgren, A., <strong>and</strong> Sjoquist, J. (1966) “Protein A” from S. aureus. I. Pseudo-immune<br />

reaction with human gamma-globulin J. Immunol.97, 822–7.<br />

8. Housden, N. G., Harrison, S., Roberts, S. E., Beckingham, J. A., Graille, M., Stura,<br />

E., <strong>and</strong> Gore, M. G. (2003) Immunoglobulin-binding domains: Protein L from<br />

Peptostreptococcus magnus Biochem. Soc. Trans. 31, 716–8.<br />

9. Kabir, S. (2002) Immunoglobulin purification by affinity chromatography using<br />

protein A mimetic lig<strong>and</strong>s prepared by combinatorial chemical synthesis Immunol.<br />

Invest. 31, 263–78.<br />

10. Svensson, H. G., Hoogenboom, H. R., <strong>and</strong> Sjobring, U. (1998) Protein LA, a novel<br />

hybrid protein with unique single-chain Fv antibody- <strong>and</strong> Fab-binding properties<br />

Eur. J. Biochem. 258, 890–6.<br />

11. Wilchek, M., Miron, T., <strong>and</strong> Kohn, J. (1984) Affinity chromatography Methods<br />

Enzymol. 104, 3–55.<br />

12. Pieper, R., Su, Q., Gatlin, C. L., Huang, S. T., Anderson, N. L., <strong>and</strong><br />

Steiner, S. (2003) Multi-component immunoaffinity subtraction chromatography:<br />

An innovative step towards a comprehensive survey of the human plasma proteome<br />

Proteomics 3, 422–32.


3<br />

Multi-Component Immunoaffinity Subtraction<br />

<strong>and</strong> Reversed-Phase Chromatography of Human Serum<br />

James Martosella <strong>and</strong> Nina Zolotarjova<br />

Summary<br />

Serum analysis represents an extreme challenge because of the dynamic range of<br />

the proteins of interest, <strong>and</strong> the high structural complexity of the constituent proteins.<br />

High-abundant proteins such as albumin, IgG, transferrin, haptoglobin, IgA <strong>and</strong> alpha1anti-trypsin<br />

represent up to 85% of the total protein mass in serum (Fig. 1). These<br />

major protein constituents interfere with identification <strong>and</strong> characterization of important<br />

moderate- <strong>and</strong> low-abundant proteins by limiting the dynamic range of mass spectral<br />

<strong>and</strong> electrophoretic analysis. During protein isolation, separation, <strong>and</strong> analysis, these six<br />

proteins often mask the detection of the more important low-abundant proteins that are<br />

of high interest as biomarkers of disease or drug targets. In one- <strong>and</strong> two-dimensional<br />

gel electrophoresis (1DGE <strong>and</strong> <strong>2D</strong>GE) for example, the spots or b<strong>and</strong>s because of these<br />

six highly abundant proteins, as well as their fragments, often overlap or completely<br />

mask large regions of the gel, making detection of the myriad low-abundant proteins very<br />

difficult, if not impossible. Moreover, proteomic analysis methods commonly include an<br />

electrophoretic or chromatographic separation step which, of course, has a finite mass<br />

loading tolerance. The presence of a large quantity of high-abundant proteins limits the<br />

mass load of targeted proteins that can be initially sampled by these separation methods<br />

<strong>and</strong> thus requires the need for multidimensional separation techniques to reduce sample<br />

complexity.<br />

Herein we describe immunoaffinity depletion combined with reversed-phase separation<br />

modes to reduce the sample complexity of human serum. We selectively immunodepleted<br />

six of the most abundant proteins from human serum, then employed gradient elution<br />

reversed-phase (RP) HPLC to fractionate the remaining serum proteins. The workflow<br />

shown in (Fig. 2) was optimized to process immunodepleted flow-through serum samples<br />

directly to a RP column with minimal sample h<strong>and</strong>ling. The RP operational conditions<br />

permitted robust <strong>and</strong> repeatable separations <strong>and</strong> have been optimized specifically for<br />

immunodepleted serum samples.<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

27


28 Martosella <strong>and</strong> Zolotarjova<br />

Key Words: affinity chromatography; human plasma; human serum; HPLC; immunodepletion;<br />

prefractionation; proteomics; reversed-phase chromatography.<br />

Immunoglobulin G<br />

16.6%<br />

Albumin<br />

54.3%<br />

Alpha-1-antitrypsin 3.8%<br />

Immunoglobulin A 3.4%<br />

Transferrin 3.3%<br />

Haptoglobin 2.9%<br />

Other 15%<br />

Fig. 1. Composition of proteins in human serum. The protein composition is<br />

schematically depicted based on mass abundance in normal human serum. The six<br />

high-abundant proteins removed by the immunoaffinity column comprise approx 85%<br />

of the total protein mass in human serum.<br />

Human Serum or Plasma<br />

Immunodepletion of Six<br />

High-abundant Proteins<br />

High Temp. RP-HPLC<br />

Multiple RP HPLC<br />

Fraction Collection<br />

<strong>2D</strong>-<strong>PAGE</strong><br />

LC/MS/MS<br />

<strong>Sample</strong> Denaturation<br />

3-Step Multi-Segment Gradient<br />

Fig. 2. Multidimensional chromatographic workflow.


Multi-Component Immunoaffinity Subtraction <strong>and</strong> Reversed-Phase 29<br />

1. Introduction<br />

Interest in proteomic analysis of human serum has been greatly elevated during<br />

the past several years as liquid chromatography (LC) <strong>and</strong> mass spectrometry (MS)<br />

methodologies have evolved sufficiently to investigate this challenging sample.<br />

The popularization of multidimensional LC methods, <strong>and</strong> the ever-improving<br />

sensitivity <strong>and</strong> performance of multi-stage MS instruments, combined with highspeed<br />

database searching, is permitting complex protein samples to undergo<br />

analysis by identification of constituent tryptic peptide fragments. Proteomic<br />

analysis of human serum represents an extreme challenge because of the dynamic<br />

rangeoftheproteinsofinterest.Serum(plasma)containsmanyproteins,estimated<br />

to include almost 500,000 molecular species <strong>and</strong> has more than 10 orders of<br />

magnitude concentration range, from mg/mL to pg/mL, <strong>and</strong> possibly less (1).<br />

This wide range of analytical target molecules is currently outside the realm of<br />

the dynamic range of available technologies for proteomic analyses. A means to<br />

address the complexity of these samples is the application of multidimensional<br />

separation techniques (2,3), for example, by multidimensional LC fractionation.<br />

Wedescribeanimmunoaffinity<strong>and</strong>RPLCcolumnapproach,whichreducesthe<br />

complexity of serum or plasma. <strong>Sample</strong>s are first immunodepleted of the six most<br />

abundant proteins using an immunoaffinity LC method. This separation delivers<br />

a flow-through fraction containing low-abundant proteins, whereas the bound<br />

high-abundant proteins are left behind. Second, the immunodepleted samples are<br />

separated under a set of optimized reversed-phase (RP) conditions using a highrecovery<br />

macroporous column material. The conditions <strong>and</strong> protocol have been<br />

designed specifically for immunodepleted human serum or plasma samples. The<br />

combination of sample simplification by immunoaffinity depletion, combined<br />

with robust <strong>and</strong> high recovery RP-HPLC fractionation, yields samples permitting<br />

higher quality protein identifications (4). The approach presented here enables<br />

an exp<strong>and</strong>ed dynamic range for the detection of low-abundant proteins in the<br />

complex proteomic samples <strong>and</strong> thereby assists in the search for novel biomarkers<br />

of disease states <strong>and</strong> intervention strategies.<br />

2. Materials<br />

2.1. Immunoaffinity Depletion of High-Abundant Proteins<br />

2.1.1. Serum Collection<br />

1. Becton Dickinson Vacutainer tubes (VWR International) with SST gel <strong>and</strong> BD<br />

clot activator.<br />

2.1.2. Immunoaffinity Chromatography<br />

1. An 1100 liquid chromatograph (Agilent Technologies, Wilmington, DE.)<br />

consisting of a binary gradient pumping system, a thermostatted autosampler,


30 Martosella <strong>and</strong> Zolotarjova<br />

a variable wavelength absorbance detector, a temperature controlled column<br />

compartment, <strong>and</strong> a thermostatted automated analytical scale fraction collector<br />

(see Note 1).<br />

2. Agilent Chemstation Software version B.01.03.<br />

3. Multiple Affinity Removal System column, 4.6 mm id × 100 mm, Immunodepletion<br />

Buffer A <strong>and</strong> Buffer B (Agilent Technologies, Wilmington, DE).<br />

4. 0.22-μm spin filters.<br />

2.2. Immunodepleted Serum <strong>and</strong> Bound Fraction Processing<br />

1. BCA Assay Kit (Pierce, Rockford, IL).<br />

2. Tris-glycine precast gels (4–20% acrylamide, 10 wells, 1 mm), sample preparation<br />

(loading) <strong>and</strong> running buffers, Mark12 unstained st<strong>and</strong>ards (Invitrogen,<br />

Carlsbad, CA).<br />

3. 4-mL spin concentrators with 5 kDa molecular weight cutoffs (Agilent<br />

Technologies, Wilmington, DE).<br />

4. St<strong>and</strong>ard equipment for two-dimensional electrophoresis (Bio-Rad, Hercules, CA).<br />

5. Rehydration buffer: 8 M urea, 2% CHAPS, 2% ampholytes <strong>and</strong> 20 mM dithiothreitol.<br />

6. 11 cm immobilized pH gradient (IPG), pH 3–10 nonlinear strips (Bio-Rad,<br />

Hercules, CA).<br />

7. 8–16% precast Tris-glycine gels for second dimension separation (Bio-Rad).<br />

8. Gel Code Blue – Coomassie stain (Pierce, Rockford, IL).<br />

2.3. Reversed-phase Separation of Low-abundant Proteins<br />

1. An 1100 liquid chromatograph (Agilent Technologies, Wilmington, DE.)<br />

consisting of a binary gradient pumping system equipped with a 900 μL capillary<br />

injector loop, a thermostated autosampler, a variable wavelength absorbance<br />

detector, a temperature controlled column compartment, <strong>and</strong> a thermostated<br />

automated analytical scale fraction collector (see Note 1).<br />

2. 4.6 mm ID × 50 mm macroporous high recovery reversed-phase C18 column (mRP-<br />

C18)(AgilentTechnologies,Wilmington,DE).Particlecompositionisasilica-based<br />

macroparticulate material with a particle size of 5.0 μm. The column hardware is<br />

made of PEEK composition <strong>and</strong> the frits are 2.0 μm PEEK encapsulated.<br />

3. Methods<br />

3.1. Immunoaffinity Depletion of High-Abundant Proteins<br />

3.1.1. Serum Collection<br />

1. Collect serum samples into a Becton Dickinson Vacutainer tubes (VWR International)<br />

with SST gel <strong>and</strong> BD clot activator. After clot formation, centrifuge<br />

sample at 1000g for 15 min. Remove serum <strong>and</strong> store aliquotes at –80°C. Total<br />

time for serum processing is less than 60 min.


Multi-Component Immunoaffinity Subtraction <strong>and</strong> Reversed-Phase 31<br />

3.1.2. Immunoaffinity Chromatography<br />

The immunoaffinity column depletion technology offers rapid <strong>and</strong> simultaneous<br />

removal of six high-abundant proteins in 28 min per sample (Fig. 3).<br />

The column is based on the rabbit polyclonal antibodies to six major<br />

serum proteins—human serum albumin (HSA), transferrin, alpha1-anti-trypsin,<br />

haptoglobin, immunoglobulin A (IgA), <strong>and</strong> immunoglobulin G (IgG) that were<br />

affinity purified on corresponding protein antigen columns (4). The resulting<br />

affinity-purified antibodies were covalently coupled to porous beads via their<br />

Fc region <strong>and</strong> cross-linked. Spatially controlled cross-linking of the antibodies<br />

resulted in preferential orientation of the antibody binding sites away from the<br />

solid-phase surface, supporting maximum binding capacity of targeted proteins.<br />

Through the series of column loading, washing, collection, <strong>and</strong> reequilibration<br />

steps, multiple serum samples can be processed <strong>and</strong> depleted of<br />

targeted high-abundant proteins. After each pass of serum through the column,<br />

flow-through fractions containing low-abundant proteins can be pooled <strong>and</strong><br />

concentrated for downstream postaffinity processing. The methods outlined<br />

below demonstrate a typical workflow for immunodepleting a given quantity<br />

of human serum samples. Similar procedures have been employed successfully<br />

for cerebrospinal fluid, amniotic fluid <strong>and</strong> for urine analysis.<br />

This methodology is robust, scalable, <strong>and</strong> easily automated for multiple<br />

samples processing, as well as compatible with downstream 1D <strong>and</strong> <strong>2D</strong>-SDS-<br />

<strong>PAGE</strong>, LC, LC/MS, <strong>and</strong>/or enzymatic or chemical fragmentation methods.<br />

1. Purge LC system with Buffer A <strong>and</strong> Buffer B at a flow rate of 1.0 mL/min for<br />

10 min. without column (see Note 2).<br />

Serum<br />

Flow-through<br />

Fraction<br />

Injection Elution<br />

Bound Fraction<br />

Re-equilibration<br />

Fig. 3. Chromatogram for the affinity removal of high-abundant proteins from<br />

human serum. 35 μL of serum was diluted 5× <strong>and</strong> injected on a 4.6 mm id × 100<br />

mm immunoaffinity column (0.50 mL/min) <strong>and</strong> a flow-through peak (3–5.0 min) was<br />

collected for reversed-phase HPLC fractionation. The column was washed with Buffer<br />

A <strong>and</strong> the targeted high-abundant proteins were eluted with Buffer B.


32 Martosella <strong>and</strong> Zolotarjova<br />

Table 1<br />

LC timetable<br />

Time (mins) %B Flow rate Max. pressure<br />

1 0.00 000 0.500 120<br />

2 10.00 000 0.500 120<br />

3 10.01 10000 1.000 120<br />

4 17.00 10000 1.000 120<br />

5 17.01 000 1.000 120<br />

6 28.00 000 1.000 120<br />

2. Run two method blanks by injecting 200 μL of Buffer A without the column<br />

according to the LC timetable (Table 1) (see Note 3). Table 1 here<br />

3. Attach a 4.6 × 100 mm immunoaffinity column <strong>and</strong> equilibrate in Buffer A for 4<br />

min at a flow rate of 1 mL/min.<br />

4. Dilute human serum five times with Buffer A (for example 35 μL of human<br />

serum with 140 μL of Buffer A (see Note 4).<br />

5. Remove sample particulates with a 0.22 μm spin filter <strong>and</strong> centrifuge sample for<br />

1.0 min at 16,000g.<br />

6. Inject 175 μL of the diluted serum at a flow rate of 0.5 mL/min <strong>and</strong> run LC<br />

timetable.<br />

7. Collect the flow-through fraction (appears between 3.0–5.0 min; see Fig. 3 for the<br />

chromatogram) into 1.5-mL microcentrifuge tubes at 4°C. Store collected fraction<br />

at –20°C if not analyzed immediately.<br />

8. Wash column with buffer A (see LC timetable) <strong>and</strong> elute the bound fraction with<br />

Buffer B at a flow rate of 1.0 mL/min for 7.0 min.<br />

9. Regenerate the column by equilibrating it with Buffer A for 11.0 min at a flow<br />

rate of 1.0 mL/min for a total run cycle of 28.0 min. (see Note 5).<br />

3.2. Immunodepleted Serum <strong>and</strong> Bound Fraction Processing<br />

For analyses of immunodepleted serum before RP separation or without<br />

further RP fractionation proceed with the recommendations below. For direct<br />

processing of the flow-through for RP HPLC fractionation of the immunodepleted<br />

serum see Section 3.3.<br />

1. If lyophylization of the immunodepleted serum is desired, buffer exchange to<br />

a volatile buffer (for example, ammonium bicarbonate) because of high salt<br />

concentration in Buffer A.<br />

2. Measure protein concentration in serum, flow-through <strong>and</strong> bound fractions using<br />

BCA protein assay. For 1D-SDS-<strong>PAGE</strong> analysis of the flow-through fraction<br />

(immunodepleted serum) or bound fraction, mix sample aliquots (5–10 μg of<br />

protein) with the equal volume of the loading buffer, boil sample for 3 min. <strong>and</strong>


Multi-Component Immunoaffinity Subtraction <strong>and</strong> Reversed-Phase 33<br />

200.0<br />

116.3<br />

97.4<br />

66.3<br />

55.4<br />

36.5<br />

31.0<br />

21.5<br />

14.4<br />

6.0<br />

3.5<br />

1 2 3 4 5<br />

Fig. 4. 1D SDS gel electrophoresis of human serum protein fractions from an<br />

immunoaffinity column. An equal amount (9 μg) of crude serum (Lane 2), flow-through<br />

(Lane 3) <strong>and</strong> bound fractions (Lane 4) were separated on 4–20% SDS-<strong>PAGE</strong> gel under<br />

nonreducing conditions. Lanes 1 <strong>and</strong> 5 are the molecular weight st<strong>and</strong>ards (Mark12)<br />

from Invitrogen. The proteins were stained with Coomassie Blue dye. Based on the<br />

protein assay of the flow-through fraction, 85% of total protein was removed from the<br />

crude serum.<br />

load on the gel. Fig. 4 shows 1D gel electrophoresis data for crude serum, flowthrough<br />

<strong>and</strong> bound fractions from an immunoaffinity column. Equal amounts<br />

of protein (9 μg) were loaded in each lane. Results show that high-abundant<br />

proteins in serum (Lane 2) are clearly removed <strong>and</strong> are not visible in the flowthrough<br />

fraction (Lane 3). Also, low-abundant proteins that were not visible in<br />

the serum before depletion (Lane 2) became visible in the flow-through fraction<br />

after removal of the high-abundant proteins.<br />

3. For IEF, <strong>2D</strong>-SDS-<strong>PAGE</strong>, <strong>and</strong> MS analysis, it is necessary to buffer<br />

exchange/desalt <strong>and</strong> concentrate fractions to an appropriate buffer. Use 4 mL<br />

spin concentrators with 5 kDa molecular weight cutoffs. Centrifuge samples at<br />

7,500g for 20 min at 4°C. Buffer exchange into 20 mM Tris-HCl, pH 7.4, by<br />

3 rounds of buffer addition, with 20 min centrifugation for each round. Aliquot<br />

the concentrated samples <strong>and</strong> store at –70°C until the analysis. Analyze protein<br />

concentration using a BCA protein assay.<br />

4. Prepare <strong>2D</strong> electrophoresis samples by mixing 250 μg of proteins with 185<br />

μL of rehydration buffer containing 8 M urea, 2% CHAPS, 2% ampholytes,<br />

pH 3–10, <strong>and</strong> 20 mM dithiothreitol. Apply samples on 11-cm immobilized<br />

pH gradient (IPG), pH 3–10 nonlinear strips <strong>and</strong> process them according to


34 Martosella <strong>and</strong> Zolotarjova<br />

the manufacturer’s instructions. Perform the second dimension separation on<br />

8–16% precast Tris-glycine gels. Visualize proteins by Coomassie Blue staining.<br />

Fig. 5 shows the protein pattern of human serum before (Panel A) <strong>and</strong> after<br />

immunodepletion (Panel B). Circles indicate the areas where the targeted highabundant<br />

proteins reside. The depletion of high-abundant proteins unmasks the<br />

low-abundant proteins because of the substantial removal of protein mass from<br />

the sample. More than 85% of total protein was depleted after a single pass<br />

of serum through the immunoaffinity column. This enabled a large increase in<br />

low-abundant protein mass loading onto the gel (up to 10 times). As a result, lowabundant<br />

protein fractions become enriched <strong>and</strong> more easily detectable on the gel,<br />

making the protein spots more amenable to quantitation <strong>and</strong> MS identification.<br />

The immunoaffinity column is highly specific for the removal of highabundant<br />

proteins from serum (5). A small number of nontargeted proteins are<br />

bound to the column. None of the nonspecific proteins are bound quantitatively<br />

to the immunoaffinity column <strong>and</strong> represent only a small percentage of the<br />

total flow-through.<br />

Anti-trypsin IgA<br />

Transferrin<br />

IgG Heavy<br />

Albumin<br />

Chain MW<br />

(kDa)<br />

200<br />

116<br />

97<br />

66<br />

55<br />

Haptoglobin<br />

A, Crude Serum B, Serum after immunodepletion<br />

Ig Light<br />

Chain<br />

37<br />

31<br />

21<br />

14<br />

6<br />

pH 3 –10<br />

MW<br />

(kDa)<br />

200<br />

116<br />

97<br />

66<br />

55<br />

Fig. 5. <strong>2D</strong> gel electrophoresis of human serum before <strong>and</strong> after removal of highabundant<br />

proteins. Panel A. Human serum before depletion. The targeted high abundant<br />

proteins are circled. Panel B. Human serum after depletion of the six targeted high<br />

abundant proteins. The positions of the removed proteins are circled. 250 μg of total<br />

protein loaded on each gel. Molecular weight st<strong>and</strong>ards–Mark12 (Invitrogen). Proteins<br />

were visualized by staining with Coomassie Blue.<br />

37<br />

31<br />

21<br />

14<br />

6


Multi-Component Immunoaffinity Subtraction <strong>and</strong> Reversed-Phase 35<br />

3.3. Reversed-phase Separation of Low-Abundant Proteins<br />

3.3.1. Reversed-Phase Chromatographic Conditions<br />

1. Eluent A: 0.1% TFA in water, Eluent B: 0.08% TFA in acetonitrile.<br />

2. Autosampler <strong>and</strong> fraction collection temperature: 4°C.<br />

3. Column temperature: 80°C. If column oven cannot reach or maintain 80°C, a<br />

lower temperature can be used, however, protein recovery <strong>and</strong> chromatographic<br />

resolution may be increasingly compromised with decreasing temperature.<br />

4. UV absorbance: 280 nm (preferred) or 210 nm.<br />

5. <strong>Sample</strong> flow rate: 0.75 mL/min.<br />

6. Prepare LC gradient elution according to LC Timetable in Table 2.<br />

7. Prepare flow-through sample directly for Reversed-Phase chromatographic<br />

separation (see Note 6). Allow sample to equilibrate at room temperature for at<br />

least 30 min.<br />

8. Inject up to 900 μL of the denatured (6M urea/1.0% acetic acid) flow-through<br />

proteins from immunodepletion. If it is desirable to process the entire flowthrough<br />

sample volume from each immunodepletion run (for example 1.5 mL<br />

of denaturated flow-through) or load multiple flow-throughs, an injector loading<br />

program is needed. This can also be useful for utilizing the columns full loading<br />

capacity when flow-throughs from multiple immunodepletions have been pooled<br />

(see Note 7). Fig. 6 is representative of a RP elution profile of immunodepleted<br />

human serum obtained when using the optimized multi-segmented elution conditions<br />

presented in Table 2. The RP separation can be fractionated <strong>and</strong> the collected<br />

fractions processed for downstream electrophoretic or LC/MS analyses (6).<br />

9. Collect fractions at 1.0 min time intervals from 0–54 min. At the recommended<br />

flow of 0.75 mL/min., the fraction volumes will be 0.75 mL (see Note 8).<br />

10. Dry each fraction in a centrifugal vacuum concentrator. To avoid protein degradation<br />

do not dry overnight. If sample processing is not immediate, store dried<br />

fractions at –80°C.<br />

11. For SDS-<strong>PAGE</strong> analysis of the separation efficiency (an example of the results<br />

produced is shown in Fig. 7), dry fractions in the vacuum concentrator <strong>and</strong><br />

Table 2<br />

LC timetable<br />

Time (mins) %B<br />

1 0.00 3.00<br />

2 6.00 30.0<br />

3 39.0 55.0<br />

4 49.0 100.0<br />

5 53.0 100.0<br />

6 58.00 3.00<br />

7 postrun re-equilibration 68.00 3.00


36 Martosella <strong>and</strong> Zolotarjova<br />

Absorbance @ 280 nm (mAu)<br />

mAU<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

0 5 10 15 20 25<br />

Time (min.)<br />

30 35 40 45<br />

Fig. 6. Representative RP-HPLC elution profile (absorbance at 280 nm) for human<br />

serum depleted of high-abundant proteins. An aqueous TFA <strong>and</strong> ACN (TFA) gradient<br />

was used at 80°C at a flowrate of 0.75 mL/min on a 4.6 mm id × 50mm mRP-C18<br />

column. The sample comprised a total of 270 μg protein in 6 M urea/1% AcOH.<br />

then dissolve each with electrophoresis sample preparation (loading) buffer as<br />

recommended below (see Note 9).<br />

12. For <strong>2D</strong> <strong>PAGE</strong> analysis of the RP fractions, we suggest combining the fractions in<br />

a manner suitable to accommodate specific workflows <strong>and</strong> goals. Fraction pooling<br />

will be required to achieve appropriate protein loads for <strong>2D</strong> <strong>PAGE</strong> analysis. To<br />

process the RP fractions, dry in a vacuum concentrator <strong>and</strong> dilute with the IEF<br />

buffer according to the manufacturers protocol.<br />

13. For consistent <strong>and</strong> repeatable reversed-phase separation consult Note 10.<br />

4. Notes<br />

1. Conventional autosamplers generally do not provide optimum sampling because<br />

of conditions which can lead to extra column b<strong>and</strong> broadening <strong>and</strong> mixing,<br />

thereby reducing resolution.<br />

2. The immunoaffinity column requires a proprietary two buffer system (Buffer<br />

A <strong>and</strong> Buffer B) for operation. The two buffers provide the means to separate<br />

min


Multi-Component Immunoaffinity Subtraction <strong>and</strong> Reversed-Phase 37<br />

Fractions 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26<br />

kDa<br />

200.0<br />

116.3<br />

97.4<br />

66.3<br />

55.4<br />

36.5<br />

31.0<br />

21.5<br />

14.4<br />

Fractions 27 28 29 30 31 32 33 34 35 36<br />

kDa<br />

200.0<br />

116.3<br />

97.4<br />

66.3<br />

55.4<br />

36.5<br />

31.0<br />

21.5<br />

14.4<br />

Fig. 7. SDS-<strong>PAGE</strong> analysis of RP-HPLC fractionated immunodepleted human<br />

serum from an mRP-C18 column (4.6 mm id × 50 mm). A depleted human serum<br />

sample was injected onto the column <strong>and</strong> eluted by a multi-segment gradient. Fifty-four<br />

fractions were collected (29 showing the majority of protein elution) for analysis by<br />

4–20% SDS-<strong>PAGE</strong>.<br />

low-abundant proteins from high-abundant proteins <strong>and</strong> regenerate the column,<br />

all in about 20–30 min per injection. Buffers A <strong>and</strong> B are optimized to minimize<br />

co-adsorption of nontargeted proteins to the column packing, <strong>and</strong> to ensure<br />

reproducibility of column performance <strong>and</strong> long column lifetime. Buffer A is<br />

a salt-containing neutral buffer (pH 7.4) used for loading, washing, <strong>and</strong> reequilibrating<br />

the column. Buffer B is a low pH urea buffer used for eluting<br />

the bound high-abundant proteins from the column. Serum samples are injected<br />

onto the column <strong>and</strong> the high-abundant proteins are simultaneously removed as<br />

low-abundant proteins pass through in the flow-through fraction. After collecting<br />

the low-abundant proteins <strong>and</strong> washing the column, the bound proteins are<br />

eluted with Buffer B <strong>and</strong> the column is re-equilibrated with Buffer A. Do not<br />

expose immunoaffinity column to solvents other than Buffers A <strong>and</strong> B. Organic<br />

solvents (acetonitrile, alcohols, etc.), strong oxidizers, acids, reducing agents,<br />

<strong>and</strong> other protein denaturing agents will cause irreversible column damage.


38 Martosella <strong>and</strong> Zolotarjova<br />

Under optimized operating conditions the column stable <strong>and</strong> robust for at least<br />

200 injections of serum samples.<br />

3. Ensure proper sample loop size in autosampler.<br />

4. Consult column certificate of analysis to verify column capacity. Concentrations<br />

of some high-abundant proteins can vary widely depending on the sample<br />

source. Proteins such as alpha1-antitrypsin, haptoglobin, IgG rise several folds<br />

in response to stress, infection, inflammation, or tissue necrosis <strong>and</strong> are known<br />

as acute phase reactants. Users need to adjust column loading volume accordingly.<br />

It is not recommended to load crude serum onto the column. Addition<br />

of protease inhibitors cocktail (Complete, Roche, IN) in buffer A for sample<br />

dilution helps prevent protein degradation.<br />

5. When not in use, store the column, with the end-caps sealed, in a refrigerator at<br />

2–8°C to minimize losses in column capacity. Do not freeze the column.<br />

6. Flow-through sample preparation: The amount of flow-through sample volume<br />

from a 4.6 mm id × 100 mm immunodepletion column is approx 1.0–1.5 mL.<br />

Immunodepleted serum fractions can either be pooled together or processed<br />

individually. However, by either method, they must first be denatured under<br />

acidic conditions. To denature before RP separation, 480 mg solid urea <strong>and</strong> 13<br />

μL acetic acid (AcOH) are added for every 1.0 mL of flow-through for a final<br />

sample concentration of approx 6 M urea/1.0% AcOH. Calculation is based on<br />

an immunodepletion separation of 35 μL human serum (diluted 5×), in which<br />

BCA protein analysis gave a flow-through protein concentration of 0.38 mg/mL.<br />

It is recommended to measure the actual flow-through protein concentration<br />

for each serum sample lot processed <strong>and</strong> adjust the 6 M urea <strong>and</strong> 1.0% AcOH<br />

concentrations accordingly.<br />

7. For loading sample volumes greater than 900 μL, an isocratic loading method<br />

is required. To load under isocratic conditions set 97% Eluent A (0.1% TFA in<br />

water) <strong>and</strong> Eluent B (0.08% TFA in acetonitrile) at 3.0% for a minimum run time<br />

of 3.0 min (maintain all other chromatographic conditions from Section 3.3).<br />

Perform desired amount of column injections without overloading the column.<br />

The maximum loading capacity for a 4.6 mm id × 50 mm mRP-C18 column<br />

is approx 400 μg of immunodepleted serum. After multiple sample injections<br />

have been loaded, proceed with the elution gradient in Table 2. In Chemstation,<br />

this process can be automated <strong>and</strong> configured under Sequence <strong>and</strong> Sequence<br />

Table.<br />

8. Depending on the user’s capacity for sampling h<strong>and</strong>ling <strong>and</strong>/or processing objectives,<br />

some workflows may require the need to collect more or a lesser amount<br />

of fractions <strong>and</strong> may therefore need to vary the time-based collection. However,<br />

fraction collection greater than 2.0 min time intervals will require collection<br />

tubes larger than 2.0 mL <strong>and</strong> typically require tray or instrument adjustments for<br />

many automated fraction collectors. Fractions collected from 1 to 6 min <strong>and</strong> 37<br />

to 54 min are not shown in Fig. 7. The majority of protein elution as determined<br />

by Commassie blue staining occurs from 7 to 36 min. If a comprehensive MS<br />

analysis is the goal, we recommend fraction collection throughout the entire run.


Multi-Component Immunoaffinity Subtraction <strong>and</strong> Reversed-Phase 39<br />

9. For fractions #1–#12 <strong>and</strong> #37–#54, dissolve each fraction in 30 μL of<br />

electrophoresis sample preparation buffer <strong>and</strong> heat for 3 min at 70°C. If the<br />

entire protein load on the column was 400 μg or less, load the entire 30 μL<br />

of prepared sample directly onto the gel. If protein fractions were pooled from<br />

several chromatographic runs <strong>and</strong> thus exceeded 400 μg of total column load,<br />

remove 15 μL of prepared sample <strong>and</strong> dilute with 15 μL deionized water (30 μL<br />

total) <strong>and</strong> load onto the 1D SDS-<strong>PAGE</strong>.<br />

For fractions #13–#36, dissolve each fraction in 75 μL of sample preparation<br />

buffer <strong>and</strong> heat for 3 min at 70°C. If the entire protein load on the column was<br />

400 μg or less, directly load 15 μL (without water dilution) onto SDS-<strong>PAGE</strong>.<br />

If protein fractions were pooled from several chromatographic runs <strong>and</strong> thus<br />

exceeded 400 μg of total column load, remove 7 μl of prepared sample, dilute<br />

with 7 μL deionized water (14 μL total) <strong>and</strong> load onto the gel.<br />

10. Column runs from the same depleted serum sample, during the same run<br />

progression, should be very repeatable with identical peak shapes <strong>and</strong> intensities.<br />

If variances are occurring the column may need replacing. Column life varies<br />

with use <strong>and</strong> conditions, but should typically last for over 75 injections. It is<br />

recommended to complete each RP separation with a minimum postrun time<br />

of 10.0 min to ensure that the column has re-equilibrated. Periodically perform<br />

blank injections to evaluate baseline stabilization. If peak ghosting, which is a<br />

characteristic of protein carryover, is present, perform a run with 100% Eluent<br />

B for 4 min, maintain elevated temperature, <strong>and</strong> repeat the blank injection. If<br />

ghosting is still present, yet minimized, repeat the run of 100% Eluent B. When<br />

not in use store column at room temperature in 25–75% (v/v) water-methanol<br />

with the column ends capped.<br />

References<br />

1. Anderson, N. L., Anderson, N. G., (2002) The human plasma proteome: history,<br />

character, <strong>and</strong> diagnostic prospects. Mol Cell Proteomics, 1, 845–67.<br />

2. Duan, X., Yarmush, D. M., Berthiaume, F., Jayaraman, A., <strong>and</strong> Yarmush, M. L.<br />

(2004) A moose serum two-dimensional gel map: application to profiling boon<br />

injury <strong>and</strong> infection. Electrophoresis, 25, 3055–65.<br />

3. Fujii, K., Nakano, T., Kawamura, T., Usui, F., B<strong>and</strong>o, Y., Wang, R., <strong>and</strong><br />

Nishimura, T. (2004) Multidimensional protein profiling technology <strong>and</strong> its application<br />

to human plasma proteome. J.Proteome Res., 3, 712–18.<br />

4. Zolotarjova, N., Martosella, J., Nicol, G., Bailey, J., Boyes, B.E., <strong>and</strong> Barrett,<br />

W.C. (2005) Differences among techniques for high-abundant protein depletion.<br />

Proteomics, 5, 3304–13.<br />

5. Harlow, E. <strong>and</strong> Lane, D., (1988) Antibodies, A Laboratory Manual. Cold Springs<br />

Harbor Laboratory: New York, pp. 726.<br />

6. Martosella, J., Zolotarjova, N., Liu, H., Nicol, G., <strong>and</strong> Boyes, B.E., (2005) Reversedphase<br />

high-performance liquid chromatographic prefractionation of immunodepleted<br />

human serum proteins to enhance mass spectrometry: identification of lowerabundant<br />

proteins. J. Proteome Res. 4, 1522–1537.


4<br />

Immunoaffinity <strong>Fractionation</strong> of Plasma Proteins<br />

by Chicken IgY Antibodies<br />

Lei Huang <strong>and</strong> Xiangming Fang<br />

Summary<br />

Separation of complex mixtures having a wide dynamic range of protein concentration,<br />

such as plasma or serum, presents a significant challenge for proteomic analysis.<br />

Immunoaffinity fractionation is one of the most effective methods used during sample<br />

preparation to improve the ability to detect low-abundant proteins (LAP), enhancing<br />

biomarker discovery. Avian IgY (Immunoglobulin Yolk) antibodies have unique <strong>and</strong><br />

advantageous features, which include strong avidity, high specificity, low nonspecific<br />

binding, <strong>and</strong> accumulative production. Polyclonal IgY antibodies covalently coupled to<br />

microbeads are particularly effective in specifically removing high-abundant proteins<br />

(HAP) from plasma, serum, CSF, urine, <strong>and</strong> other body fluid or cellular sources. IgY-12<br />

is a composition of IgY microbeads designed for one-step removal of the 12 most<br />

abundant proteins in human serum or plasma: albumin, IgG, transferrin, fibrinogen, 1antitrypsin,<br />

IgA, IgM, 2-macroglobulin, haptoglobin, apolipoproteins A-I <strong>and</strong> A-II, <strong>and</strong><br />

orosomucoid (1-acid glycoprotein). Removal of the 12 HAPs enables improved resolution<br />

<strong>and</strong> dynamic range for one-dimensional gel electrophoresis (1DGE), two-dimensional gel<br />

electrophoresis (<strong>2D</strong>GE), <strong>and</strong> liquid chromatography/mass spectrometry (LC/MS).<br />

Key Words: High abundance proteins; IgY antibodies; immunoaffinity fractionation;<br />

low abundant proteins; protein depletion; protein separation; proteomics; sample preparation.<br />

1. Introduction<br />

IgY antibody is immunoglobulin gamma isolated from egg yolks (so<br />

called IgY) of certain avian <strong>and</strong> reptilian vertebrates such as birds, reptiles,<br />

<strong>and</strong> amphibians (1–3). Chicken IgY antibodies have been developed <strong>and</strong><br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

41


42 Huang <strong>and</strong> Fang<br />

successfully applied for various types of immunoassays (4–8). An outst<strong>and</strong>ing<br />

advantage of chicken IgY antibodies is that they are secreted by hens into egg<br />

yolk, resulting in a high-yielding <strong>and</strong> easy to access reservoir of antibodies<br />

(9). Compared to drawing blood, collecting eggs is noninvasive, continuous,<br />

convenient, <strong>and</strong> scalable. One egg contains about 100 mg of total IgY. A laying<br />

hen can actively produce eggs for 2 years at an average of 20 eggs per<br />

month. Distinct from mammalian IgG antibodies in molecular structure <strong>and</strong><br />

biochemical features, IgY antibodies have been shown to have several advantages<br />

over IgG, particularly for their high avidity <strong>and</strong> less cross-reactivity<br />

to human proteins (10–12). This is because of the avian affinity maturation<br />

mechanism of gene conversion <strong>and</strong> the great evolutionary distance between<br />

chicken <strong>and</strong> mammals. When mammalian protein antigens are used to immunize<br />

chickens, more immunogenic epitopes are presented to the host, resulting in IgY<br />

antibodies with high affinity <strong>and</strong> broader recognition spectrum. In addition, the<br />

IgY Fc region does not bind human proteins such as complement, rheumatoid<br />

factor, Fc receptor, etc, thus significantly increasing IgY’s specificity of capture.<br />

Another unique feature of IgY antibodies is that they have a broader antigenbinding<br />

host range. This is also the result of greater evolutionary distance<br />

between chickens <strong>and</strong> mammals, <strong>and</strong> the sequence similarity among mammals.<br />

IgY antibodies raised against these high abundance proteins using human<br />

antigens also recognize the orthologous proteins from other mammalian species<br />

such as nonhuman primates, rat, mouse, pig, goat, cow, <strong>and</strong> dog.<br />

IgY microbeads are produced by covalently coupling IgY antibodies to<br />

60-μm polymeric beads via the oligosaccharides located on their Fc region. This<br />

orientated conjugation allows maximal capture of target proteins. Compared<br />

to other affinity reagents, including IgG microbead products, IgY microbeads<br />

have been shown to have distinct features <strong>and</strong> advantages (13,14). IgY-12 is a<br />

mixture of 12 types of IgY microbeads designed to collectively remove albumin,<br />

IgG, 1-antitrypsin, IgA, IgM, transferrin, haptoglobin, 1-acid glycoprotein<br />

(orosomucoid), 2-Macroglobulin, HDL (mainly apolipoproteins A-I <strong>and</strong> A-<br />

II), <strong>and</strong> fibrinogen from complex human body fluids such as serum, plasma,<br />

<strong>and</strong> cerebral spinal fluid (CSF) in a single step. IgY-12 spin column (0.6-mL<br />

bed size) can process 15–20 μL human plasma per loading, yielding 100–160<br />

μg of proteins partitioned of HAP. Larger samples can be processed by IgY-<br />

12 liquid chromatography (LC) columns. A 2-mL LC column can partition<br />

40–50 μL human plasma per injection <strong>and</strong> a 10-mL LC column allows a<br />

single loading of 200–250 μL. Through a simple procedure of sample loading,<br />

washing, eluting, <strong>and</strong> regenerating, approx 90–95% of total protein mass from<br />

human serum or plasma is removed. The LAP in the flow-through fractions<br />

can be further studied by <strong>2D</strong> polyacrylamide gel electrophoresis (<strong>PAGE</strong>) or<br />

LC/MS. The regenerated beads can be reused many times with minimal protein


Immunoaffinity <strong>Fractionation</strong> of Plasma Proteins by Chicken 43<br />

carry-over (15). The IgY microbeads can also be used in 96-well filter plate <strong>and</strong><br />

other formats for high-throughput partitioning of human serum/plasma samples<br />

or other body fluids.<br />

2. Materials<br />

2.1. IgY-12 High Capacity Spin Column Kit<br />

1. Prepacked IgY-12 spin column, containing 1.2-mL IgY microbeads slurry<br />

(Beckman Coulter, Fullerton, CA). Store at 2–8°C. Do not freeze.<br />

2. Dilution buffer (Tris Buffered Saline, TBS): 10 mM Tris-HCl, pH 7.4, 150 mM<br />

NaCl. For sample dilution, washing <strong>and</strong> equilibrating column, <strong>and</strong> rinsing pipet<br />

tips during resin transfer. Store at room temperature.<br />

3. Stripping buffer: 0.1M Glycine-HCl, pH 2.5. For stripping off bound proteins<br />

from column. Store at room temperature.<br />

4. Neutralization buffer: 1M Tris-HCl, pH 8.0. For neutralizing column <strong>and</strong> eluted<br />

proteins. Store at room temperature.<br />

5. 2-mL collection tubes. For collecting flow-through, washing, <strong>and</strong> eluted fractions.<br />

6. Empty spin columns with end caps.<br />

2.2. IgY-12 High Capacity LC2 or LC10 Column Kit<br />

1. Prepacked IgY-12 LC column, 2-mL or 10-mL packed bed (Beckman Coulter,<br />

Fullerton, CA). Store at 2–8°C. Do not freeze.<br />

2. Dilution, stripping, <strong>and</strong> neutralization buffers are same as for spin column.<br />

3. Spin filters. For sample clean up before loading column to remove sample particulates<br />

<strong>and</strong> extend column life.<br />

2.3. IgY-12 Microbeads for 96-Well Filter Plates<br />

1. IgY-12 microbeads, 50% slurry (GenWay Biotech, San Diego, CA). Store at<br />

2–8°C. Do not freeze.<br />

2. Dilution, stripping, <strong>and</strong> neutralization buffers are same as for spin column.<br />

3. 96-well filter plate, 400 μL, UHMW PE 25 μM, Long drip. (Innovative Microplate,<br />

Billerica, MA). Store at room temperature.<br />

4. Collection plate, Nunc 96-DeepWell Plates, 1.2-mL, Polypropylene (NUNC,<br />

Rochester, NY).<br />

2.4. SDS Gel Electrophoresis<br />

1. Tris-HCl SDS Gel, precast, 4–20% linear gradient (Rio-Rad, Hercules, CA). Store<br />

at 2–8°C.<br />

2. 5× SDS <strong>Sample</strong> Buffer: 10% (w/v) SDS, 20 mM dithiothreitol (DTT) or 25% (w/v)<br />

-mercaptoethanol (BME) (omitted under nonreducing condition), 20% (w/v)


44 Huang <strong>and</strong> Fang<br />

glycerol, 0.2M Tris-HCl, pH 6.8, 0.05% (w/v) bromophenol blue. Store at room<br />

temperature.<br />

3. Tris/glycine/SDS electrophoresis buffer: 25 mM Tris-base, 200 mM glycine, 0.1%<br />

(w/v) SDS.<br />

4. Coomassie Blue Staining Solution: 0.25% (w/v) Coomassie Brilliant Blue R250,<br />

40% (v/v) methanol, 10% (v/v) acetic acid. Store at room temperature.<br />

5. Destain Soluton: 40% (v/v) methanol, 10% acetic acid. Store at room temperature.<br />

3. Methods<br />

3.1. Spin Column (see Note 1)<br />

3.1.1. Immunocapture of 12 Abundant Serum/Plasma Proteins<br />

1. Dilute 15–20 μL serum or plasma sample in dilution buffer to obtain a final<br />

volume of 600 μL.<br />

2. Snap off the tip from the column <strong>and</strong> place the column in a 2-mL collection tube.<br />

3. Centrifuge the column for 30 sec at 400g in a microcentrifuge to obtain dried<br />

beads.<br />

4. Place the end cap to the column. Immediately add 0.5 mL diluted sample to the<br />

dried beads in the column. Seal the column with the top snap cap.<br />

5. Mix the beads <strong>and</strong> the sample completely by inverting <strong>and</strong> shaking the column,<br />

place it on an end-to-end rotator <strong>and</strong> incubate at room temperature for 15 min.<br />

6. Invert the column. Remove the end cap <strong>and</strong> place the column in a 2-mL collection<br />

tube. Centrifuge for 30 sec at 400g. Collect flow-through (IgY-12-depleted)<br />

sample for further analysis (see Note 2).<br />

3.1.2. Washing of Column<br />

1. Wash beads with 0.5 mL of dilution buffer, a total of three times. To obtain<br />

maximum yields of flow-through samples, the fraction from the first washing can<br />

be collected <strong>and</strong> combine with the flow-through sample from Section 3.1.1 step<br />

6 for further analysis.<br />

2. For each wash, always first insert the end cap, <strong>and</strong> then add 0.5 mL of dilution<br />

buffer <strong>and</strong> seal the column with top snap cap. Mix the beads <strong>and</strong> buffer completely<br />

by inverting <strong>and</strong> shaking the column, remove the end cap while inverting the<br />

column <strong>and</strong> place it in a 2-mL collection tube. Centrifuge for 30 sec at 400g <strong>and</strong><br />

save the flow-through for further analysis.<br />

3.1.3. Stripping of Bound Proteins<br />

1. Strip off bound proteins from beads using stripping buffer, a total of three to four<br />

times. For each elution, place the end cap to the column first after centrifugation,<br />

then add 0.5-mL stripping buffer <strong>and</strong> seal the column with top snap cap. Mix the<br />

beads <strong>and</strong> buffer completely (see Note 3) by inverting <strong>and</strong> shaking the column,<br />

incubate at room temperature for 3 min, remove the end cap while holding the


Immunoaffinity <strong>Fractionation</strong> of Plasma Proteins by Chicken 45<br />

column upside down <strong>and</strong> place it in a 2-mL collection tubes. Centrifuge for 30<br />

sec at 400g <strong>and</strong> collect the eluate. It is crucial for column stability to immediately<br />

neutralize the beads (see Section 3.1.4.).<br />

2. Pool eluted samples (total 1.5–2.0 mL) <strong>and</strong> neutralize with 150–200 μL of neutralization<br />

buffer. <strong>Sample</strong>s can be stored at -80°C if not analyzed immediately.<br />

3.1.4. Regeneration of IgY-12 Microbeads<br />

1. To regenerate IgY-12 microbeads after stripping bound serum proteins as<br />

described above, immediately neutralize beads with 0.6 mL of 1:10 diluted neutralization<br />

buffer. Mix beads <strong>and</strong> buffer completely by inverting <strong>and</strong> shaking the<br />

column. Incubate at room temperature for 5 min. Spin down beads in the column<br />

for 30 sec at 400g.<br />

2. Resuspend beads in 0.5-mL of dilution buffer. Beads are ready for next cycle or<br />

storage at 4°C. For storage of regenerated beads, add sodium azide (NaN3)to 0.02%. (w/v) in dilution buffer.<br />

3.2. LC Column (see Note 4)<br />

3.2.1. Protocol for 6.4 × 63 mm (2 mL) Column<br />

1. Set up the three Buffers as the only mobile phases.<br />

2. Purge lines with three Buffers at a flow rate of 1.0 mL/min for 10 min.<br />

3. Set up LC timetable (see Table 1 for details) <strong>and</strong> run two method blanks by<br />

injecting 125 μL of dilution buffer without a column.<br />

4. Attach column <strong>and</strong> equilibrate it in dilution buffer for 10 min at a flow rate of<br />

1.0 mL/min at room temperature.<br />

5. Dilute human serum five times (for example: 50 μL human serum with 200 μL<br />

of dilution buffer).<br />

6. Remove particulates with a 0.45-μm spin filter; 1 min at 9,000g.<br />

7. Inject 250 μL of the diluted <strong>and</strong> filtered plasma sample (Column capacity: 40–50<br />

μL of neat human serum/plasma per injection), start the method at a flow rate<br />

of 0.1 mL/min for 10 min, wash the column at a flow rate of 0.2 mL/min<br />

for 7 min, then change the flow rate to 1.0 mL/min to continue the wash for<br />

5 min, collect flow-through fraction <strong>and</strong> store collected fractions at –80°C if not<br />

analyzed immediately.<br />

8. Elute bound proteins from the column with stripping buffer at a flow rate of<br />

1.0 mL/min for 142 min, <strong>and</strong> neutralize the column with neutralizing buffer at<br />

a flow rate of 1.0 mL/min for 6 min.<br />

9. Regenerate column by equilibrating it with dilution buffer for an additional<br />

6 min at a flow rate of 1.0 mL/min.<br />

10. Store column after equilibrating with dilution buffer containing 0.02% (w/v)<br />

sodium azide (NaN3) at 2–8°C in a refrigerator.<br />

11. A st<strong>and</strong>ard chromatograph is illustrated in Fig. 1.


46 Huang <strong>and</strong> Fang<br />

Table 1<br />

LC Method for a 6.4 × 63 mm column<br />

Cycle Time<br />

(min)<br />

Dilution<br />

buffer<br />

Stripping<br />

buffer<br />

Neutralization<br />

buffer<br />

Flow rate<br />

(mL/min)<br />

Max<br />

pressure<br />

(psi)<br />

Injection<br />

Wash 0 100 0 0 0.1 100<br />

Wash 10.01 100 0 0 0.2 100<br />

Wash 17.01 100 0 0 1.0 100<br />

Elution 22.01 0 100 0 1.0 100<br />

Neutralization 36.01 0 0 100 1.0 100<br />

Re-equilibrium 42.01 100 0 0 1.0 100<br />

Stop 48.00<br />

Optimized for Beckman System Gold HPLC, Pump Module 1 Type: 118, Detector Model: 166<br />

3.2.2. Protocol for 12.7 × 79.0 mm (10 mL) Column<br />

1. Set up the three buffers as the only mobile phases.<br />

2. Purge lines with three buffers at a flow rate of 1.0 mL/min for 10 min.<br />

3. Set up LC timetable (see Table 2 for details) <strong>and</strong> run two method blanks by<br />

injecting 1.25 mL of dilution buffer without a column.<br />

Fig. 1. Chromatography of immunoaffinity separation of human plasma using IgY-<br />

12 high capacity LC2 column. Fifty microliters human plasma was fractionated on the<br />

column.


Immunoaffinity <strong>Fractionation</strong> of Plasma Proteins by Chicken 47<br />

4. Attach column <strong>and</strong> equilibrate it in dilution buffer for 10 min at a flow rate of<br />

2.0 mL/min at room temperature.<br />

5. Dilute human serum/plasma five times (for example: 250 μL human plasma with<br />

1.0 mL of dilution buffer).<br />

6. Remove particulates with a 0.45 μm spin filter; 1 min at 9,000g.<br />

7. Inject 1.5 mL of the diluted <strong>and</strong> filtered plasma sample (column capacity:<br />

200–250 μL of neat human serum/plasma), start the method at a flow rate of<br />

0.5 mL/min for 30 min, wash the column at a flow rate of 2.0 mL/min for<br />

5 min, collect flow-through fraction <strong>and</strong> store collected fractions at -80°C if not<br />

analyzed immediately.<br />

8. Elute bound proteins from the column with stripping buffer at a flow rate of<br />

2.0 mL/min for 15 min, <strong>and</strong> neutralize the column with neutralizing buffer at a<br />

flow rate of 2.0 mL/min for 10 min.<br />

9. Regenerate column by equilibrating it with dilution buffer for an additional<br />

10 min at a flow rate of 2.0 mL/min.<br />

10. Store column after equilibrating with dilution buffer containing 0.02% (w/v)<br />

sodium azide (NaN 3) at 2–8°C in a refrigerator.<br />

11. A st<strong>and</strong>ard chromatograph is illustrated in Fig. 2<br />

3.3. 96-Well Spin Filter Plate<br />

1. For each well, dilute 2–3 μL human serum or plasma in dilution buffer to obtain<br />

final volume of 100 μL.<br />

2. Aliquot 200 μL per well IgY-12 microbeads slurry into 96-well filter plate. Spin<br />

plate at 190g for 1 min in Eppendorf bench top centrifuge (see Note 5) with plate<br />

adapter to remove buffer.<br />

3. Add diluted sample to each well, mix with pipet tip. Incubate at room temperature<br />

on shaker for 15 min.<br />

Table 2<br />

LC method for a 12.7 × 79.0 mm column<br />

Cycle Time<br />

(min)<br />

Dilution<br />

buffer<br />

Stripping<br />

buffer<br />

Neutralization<br />

buffer<br />

Flow rate<br />

(mL/min)<br />

Max<br />

pressure<br />

(psi)<br />

Injection<br />

Wash 0 100 0 0 0.5 100<br />

Wash 30.01 100 0 0 2.0 100<br />

Elution 35.01 0 100 0 2.0 100<br />

Neutralization 50.01 0 0 100 2.0 100<br />

Re-equilibrium 60.01 100 0 0 2.0 100<br />

Stop 70.00<br />

Optimized for Beckman System Gold HPLC, Pump Module 1 Type: 118, Detector Model: 166


48 Huang <strong>and</strong> Fang<br />

Fig. 2. Chromatography of immunoaffinity separation of human plasma using IgY-<br />

12 LC10 column. Two hundred fifty microliters human plasma was fractionated on the<br />

column.<br />

4. Spin plate at 190g for 1 min. Collect flow-through fraction in collection plate,<br />

about 100 μL.<br />

5. Add 100 μL dilution buffer to each well. Gently shake the plate <strong>and</strong> spin at 190g<br />

for 1 min. Collect flow-through faction into the same collection plate from step 4.<br />

6. Wash beads with 100 μL dilution buffer, a total of 3 times. For each wash, add<br />

100 μL dilution buffer to each well, gently shake the plate <strong>and</strong> spin at 190g for<br />

1 min. Collect flow-through faction into collection plate for future analysis.<br />

7. Add 100 μL stripping buffer to each well. Gently shake the plate <strong>and</strong> incubate at<br />

room temperature on shaker for 2 min. Spin the plate at 190g for 1 min. Repeat<br />

for three to four times. Collect <strong>and</strong> combine flow-through factions into collection<br />

plate for future analysis.<br />

8. Immediately add 100 μL neutralization buffer to each well. Gently shake the plate<br />

<strong>and</strong> incubate at room temperature on shaker for 5 min.<br />

9. Spin the plate at 190g for 1 min. Add 100 μL dilution buffer to each well. Beads<br />

are ready for next cycle or storage at 4°C. For storage of regenerated beads, add<br />

sodium azide (NaN 3) to 0.02%. (w/v) in dilution buffer.<br />

3.4. Evaluation of <strong>Fractionation</strong> Efficiency by SDS-<strong>PAGE</strong> (see Note 6)<br />

1. Take a small aliquot of samples from neat plasma, flow-through <strong>and</strong> eluted<br />

fraction. Mix with 5× SDS sample buffer. Load approx 25–30 μL (see Note 7)


Immunoaffinity <strong>Fractionation</strong> of Plasma Proteins by Chicken 49<br />

Fig. 3. SDS-<strong>PAGE</strong> analysis of neat plasma, flow-through <strong>and</strong> eluted fractions using<br />

IgY-12 high capacity spin column (1.0 mL slurry). Twenty microliters of human plasma<br />

was partitioned on the column. Five cycles were repeated. Fifteen microliters of 1:70<br />

dilution of neat plasma, 15 μL of flow-through fraction, <strong>and</strong> 15 μL of eluted fraction<br />

were loaded to 4–20% SDS gradient gel. Coomassie blue staining. M: molecular weight<br />

marker, S: Neat plasma. F1-F5: Flow-through fractions from cycle 1 to 5, E1-E5:<br />

Eluted/bound fractions from cycle 1 to 5.<br />

of each sample on 4–20% SDS gel. The gel is run in Tris/Glycine/SDS<br />

electrophoresis buffer at 200 volts for 35 min.<br />

2. Remove gel from the gel cassette. Rinse the gel with deionized water. Stain gel<br />

in Coomassie Blue Staining solution for 30 min to 1honshaker.<br />

3. Rinse the gel with deionized water. Place gel in destaining solution <strong>and</strong> shake<br />

until the b<strong>and</strong>s emerge clearly from the background. Replace destaining solution<br />

with deionized water.<br />

4. A successful fractionation will result in distinct b<strong>and</strong>ing patterns on the gel as shown<br />

in Fig. 3. The major protein b<strong>and</strong>s of albumin, transferrin, IgG, <strong>and</strong> Apo-A1 that<br />

disappeared in the flow-through fraction will be shown in the eluted fraction.<br />

4. Notes<br />

1. Before loading plasma sample to the new IgY-12 Spin Column, perform the full<br />

procedure with buffers only for one or two cycles. The purpose is to remove any<br />

residual uncoupled IgY antibodies in the column.<br />

2. The flow-through fraction is now greatly diluted. The samples can be concentrated<br />

to desired concentration <strong>and</strong> volume for downstream analysis using molecular


50 Huang <strong>and</strong> Fang<br />

weight cutoff centrifugal concentrators, such as Vivaspin (Sartorius, Goettingen,<br />

Germany). An alternative method is TCA/Acetone precipitation.<br />

3. The beads tend to be packed tighter in acidic solution after centrifugation.<br />

Make sure beads are resuspended well for effective stripping. For more efficient<br />

stripping, 0.25 M Glycine-HCl, pH 2.5 can be used. In this case, 1 to 4 dilution<br />

of neutralization buffer should be used to regenerate beads <strong>and</strong> 250 μL of neutralization<br />

buffer should be added per 1 mL of eluted samples.<br />

4. The LC protocols are optimized for Beckman System Gold HPLC, Pump Module<br />

1 Type: 118, Detector Model: 166. If using other HPLC systems, some adjustment<br />

may be required.<br />

5. 96-well filter plate format can be adapted to automated liquid h<strong>and</strong>ling system.<br />

Some adjustments in procedure may be required.<br />

6. SDS-<strong>PAGE</strong> is a simple way to evaluate the efficiency of sample fractionation by<br />

IgY microbeads. The major plasma proteins, such as albumin, transferrin, IgG,<br />

<strong>and</strong> Apo-A1, can be easily visualized by Coomassie blue staining of SDS gel.<br />

The different protein b<strong>and</strong>ing patterns of neat plasma, flow-through fraction <strong>and</strong><br />

eluted fraction represent the protein composition in each sample. Under reducing<br />

condition (with DTT or BME in SDS sample buffer), the heavy <strong>and</strong> light chains<br />

of IgG are separated <strong>and</strong> migrate at different speed; while under nonreducing<br />

conditions (no DTT or BME in SDS sample buffer), the heavy <strong>and</strong> light chains of<br />

IgG are linked by disulphide bounds <strong>and</strong> migrate on gel as single b<strong>and</strong> at higher<br />

molecular weight.<br />

7. The total protein mass in plasma is about 60–80 mg/mL. Approximately 90% of<br />

proteins are captured by IgY-12 microbeads. The recovery rate is about 85–90%.<br />

The protein concentration in flow-through fraction is very low. To see protein<br />

b<strong>and</strong>ing pattern in SDS gel, load maximal volume of sample that a well of the gel<br />

can hold for flow-through <strong>and</strong> eluted/bound fractions, usually 25–30 μL(including<br />

5× <strong>Sample</strong> Buffer). As a control, load 15–20 μL of diluted neat plasma, usually<br />

at 1 to 70–80 dilutions. After flow-through fraction is concentrated <strong>and</strong> protein<br />

concentration is measured, an equal amount of protein (5–10 μG) from each<br />

fraction <strong>and</strong> neat plasma can be loaded for comparison on SDS gel. <strong>Fractionation</strong><br />

efficiency can be assessed.<br />

8. IgY microbeads can also be used to partition plasma/serum from other species,<br />

such as nonhuman primates, mouse, rat, cow, dog, etc. To ensure maximal<br />

separation efficiency, use 50% of human sample loading for other species.<br />

9. IgY microbeads can be recycled for at least 100 times under proper conditions.<br />

It is important to neutralize the beads immediately after stripping.<br />

Acknowledgments<br />

The author would like to thank Dr. Wei-Wei Zhang <strong>and</strong> Mr. Robert Gans<br />

for critical reading <strong>and</strong> editing of the manuscript. The IgY-12 microbeads<br />

products were developed <strong>and</strong> manufactured by GenWay Biotech <strong>and</strong> marketed<br />

by Beckman Coulter.


Immunoaffinity <strong>Fractionation</strong> of Plasma Proteins by Chicken 51<br />

References<br />

1. Leslie, G. A. <strong>and</strong> Clem, L. W. (1969) Phylogeny of immunoglobulin structure <strong>and</strong><br />

function. 3. Immunoglobulins of the chicken. J. Exp. Med. 130, 1337–52.<br />

2. Hadge, D. <strong>and</strong> Ambrosius, H. (1984) Evolution of low molecular weight immunoglobulins<br />

– IV. IgY-like immunoglobulins of birds, reptiles <strong>and</strong> amphibians,<br />

precursors of mammalian IgA. Mol. Immunol. 21, 699–707.<br />

3. Du Pasquier, L., Schwager, J., <strong>and</strong> Flajnik, M.F. (1989) The immune system of<br />

Xenopus. Annu. Rev. Immunol. 7, 251–75.<br />

4. Larsson, A. <strong>and</strong> Mellerstedt, H. (1992) Chicken antibodies: a tool to avoid interference<br />

by human anti-mouse antibodies in ELISA after in vivo treatment with<br />

murine monoclonal antibodies. Hybridoma 11, 33–9.<br />

5. Larsson, A., Balow, R. M., Lindahl, T. L., <strong>and</strong> Forsberg, P. O. (1993) Chicken<br />

antibodies: Taking advantage of evolution – A review. Poultry Science 72,<br />

1807–12.<br />

6. Warr, G. W., Magor, K. E., <strong>and</strong> Higgins, D. A.. (1995) IgY: clues to the origins<br />

of modern antibodies. Immunol. Today 16, 392–98.<br />

7. Schade, R. <strong>and</strong> Hlinak, A. (1996) Egg yolk antibodies, state of the art <strong>and</strong> future<br />

prospects. ALTEX. 13, 5–9.<br />

8. Zhang, W.-W. (2003). The use of gene-specific IgY antibodies for drug target<br />

discovery. Drug Discovery Today 8, 364–71.<br />

9. Patterson, R., Youngner, J. S., Weigle, W. O., <strong>and</strong> Dixon, F.J. (1962) Antibody<br />

production <strong>and</strong> transfer to egg yolk in chicken. J. Immunol. 89, 272–8.<br />

10. Stuart, C. A., Pietrzyk, R. A., Furlanetto, R. W., <strong>and</strong> Green, A. (1988) Highaffinity<br />

antibody from hens’ eggs directed against the human insulin receptor <strong>and</strong><br />

the human IGF1 receptor. Anal. Biochem. 173, 142–50.<br />

11. Gassmann, M., Thommes, P., Weiser, T., <strong>and</strong> Hubscher, U. (1990) Efficient<br />

production of chicken egg yolk antibodies against a conserved mammalian protein.<br />

FASEB J. 4, 2528–32.<br />

12. Larsson, A., A. Karlsson-Parra, <strong>and</strong> J. Sjoquist. (1991) Use of chicken antibodies<br />

in enzyme immunoassays to avoid interference by rheumatoid factors. Clin. Chem.<br />

37, 411–14.<br />

13. Fang, X., Curran, K. W., Huang, L., Xiao, W., Strauss, W., Harvie, G. Feitelson, J.,<br />

<strong>and</strong> Zhang, W.-W. (2003) Polyclonal gene-specific IgY antibodies for proteomics<br />

<strong>and</strong> abundant plasma protein depletion, in frontiers of biotechnology <strong>and</strong> pharmaceuticals,<br />

Vol. 4 (Reiner, J., Zhao, K., Chen, S.-H., <strong>and</strong> Guo, M., eds.)„ Science<br />

Press USA, Inc. Monmouth Junction, NJ, pp. 222–45.<br />

14. Fang, X., Huang, L., Feitelson, J. S., <strong>and</strong> Zhang, W.-W. (2004) Affinity separation:<br />

divide <strong>and</strong> conquer the proteome. Drug Discovery Today: Technology 1, 141–48<br />

15. Huang, L., Harvie, G., Feitelson, J. S., Gramatikoff, K., Herold, D.A., Allen, D. L.,<br />

Amunagama, R., Hagler, R. A., Pisano, M. R., Zhang, W.-W., <strong>and</strong> Fang, X. (2005)<br />

Immunoaffinity separation of plasma proteins by IgY microbeads: meeting the<br />

needs of proteomic sample preparation <strong>and</strong> analysis. Proteomics 5, 3314–28.


5<br />

Proteomics of Cerebrospinal Fluid:<br />

Methods for <strong>Sample</strong> Processing<br />

John E. Hale, Valentina Gelfanova, Jin-Sam You, Michael D. Knierman,<br />

<strong>and</strong> Robert A. Dean<br />

Summary<br />

Cerebrospinal fluid (CSF) provides an important source of potential biomarkers for<br />

brain disorders <strong>and</strong> therapeutic drug development. Applications of proteomic technology<br />

to the identification <strong>and</strong> quantification of proteins in CSF are increasing rapidly. Key to<br />

obtaining reproducible <strong>and</strong> reliable data about protein levels in CSF are st<strong>and</strong>ardization<br />

of methods for sample collection, storage, <strong>and</strong> subsequent sample processing. Methods<br />

are described here for all steps of sample processing for a number of different proteomic<br />

approaches.<br />

Key Words: Cerebrospinal fluid; mass spectrometry; proteomics; silver staining;<br />

two-dimensional gel electrophoresis.<br />

1. Introduction<br />

Cerebrospinal fluid is the interstitial fluid that bathes the ventricles of the<br />

brain. CSF is produced at a rate of approx 500 mL/d (1) <strong>and</strong> participates in<br />

maintenance of hydrodynamic pressure, transportation of nutrients, <strong>and</strong> removal<br />

of metabolites from the brain (2). Because of its proximity to the different<br />

regions of the brain, CSF has long been considered an important source for<br />

biomarkers of diseases of the brain. CSF is physically separated from plasma<br />

by the blood-brain barrier (bbb). This prevents the free flow of large molecules<br />

(such as proteins) from one space to the other. Smaller molecules may diffuse<br />

more freely however penetration of the bbb is dependent on the physical<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

53


54 Hale et al.<br />

properties of the individual molecule. The proteins of CSF have been studied<br />

by two-dimensional (2-D) gel electrophoresis for some time (3). Many of<br />

the proteins seen in CSF are abundant serum proteins, which has led to the<br />

mistaken impression that CSF is simply an ultrafiltrate of serum. There are<br />

many differences between the protein composition of CSF <strong>and</strong> serum however.<br />

Thus CSF is not a simple ultrafiltrate of serum (1). Several proteins have been<br />

identified that are present at much higher concentration in CSF than in serum<br />

(4). Proteins produced <strong>and</strong> secreted in the brain will be prevented from rapidly<br />

diffusing into the serum by the same blood brain barrier that limits diffusion<br />

of molecules into the brain. The use of CSF for biomarker discovery <strong>and</strong><br />

measurement is of obvious importance. However, differential levels of proteins<br />

may arise for many different reasons. Of major concern is blood contamination,<br />

which may occur during sample collection. The protein concentration of blood<br />

is 200–400 times higher than that for CSF so a very small percentage of blood<br />

can have a very dramatic effect on the protein profile of CSF of some proteins<br />

may be altered by differential h<strong>and</strong>ling of CSF samples <strong>and</strong> care should be<br />

taken to ensure that samples are collected <strong>and</strong> stored in as similar a fashion as<br />

possible. The success of any proteomic analysis of CSF is largely dependent<br />

on the quality of the sample analyzed. Although the samples are not collected<br />

by the analytical chemist directly, an underst<strong>and</strong>ing of the clinical methods<br />

used for collection <strong>and</strong> the criteria for acceptance of samples is important in<br />

downstream interpretation of the results. This section is intended as a guide<br />

to aid the analytical chemist in underst<strong>and</strong>ing the issues involved in sample<br />

collection <strong>and</strong> in assessing the suitability of individual samples for subsequent<br />

analysis.<br />

Lumbar puncture (LP) is routinely performed to collect cerebral spinal fluid<br />

(CSF) for confirmation of suspected meningeal infection <strong>and</strong> subarachnoid<br />

hemorrhage. CSF is also frequently examined to detect malignancies, neurodegenerative<br />

processes, <strong>and</strong> other pathology involving the central nervous system<br />

(CNS). LP also provides an opportunity for direct measurement of intracranial<br />

pressure (5,6). Although gross assessment of CSF provides clues about the<br />

presence of disease, physicians rely on a broad spectrum of laboratory<br />

techniques to evaluate patient specimens. The wealth of data available from<br />

st<strong>and</strong>ardized chemical, cytological, <strong>and</strong> microbiological tests on CSF from<br />

healthy individuals <strong>and</strong> patients with specific diseases provides the background<br />

against which clinical laboratories <strong>and</strong> physicians compare data for individual<br />

patients. This knowledge base allows physicians to narrow down the diagnostic<br />

possibilities suggested by a patient’s complaint <strong>and</strong> presentation (5,7).<br />

In clinical research, LP has increasingly been paired with varied analytical<br />

technologies to better characterize normal <strong>and</strong> disease biology <strong>and</strong> enhance<br />

diagnostic accuracy (8,9). The procedure also is used to characterize the central


Proteomics of Cerebrospinal Fluid 55<br />

disposition, pharmacodynamic <strong>and</strong> clinical responses produced by established<br />

<strong>and</strong> c<strong>and</strong>idate therapeutics. In drug development, analyses of CSF are used<br />

to determine if drugs penetrate the CNS <strong>and</strong> alter biochemical pathways<br />

<strong>and</strong> cellular responses. This approach aims to more thoroughly define the<br />

mechanism of action <strong>and</strong> the optimal dose for drugs designed to act on<br />

the CNS (10). Collection of CSF for clinical research generally employs LP<br />

techniques similar to those used in routine clinical practice. In attempting to<br />

identify central pharmacodynamic effects, LP is occasionally performed before<br />

<strong>and</strong> following multidose administration of drug designed to achieve circulating<br />

steady state concentrations (11). Placement of an indwelling catheter<br />

for continuous sampling of CSF from the thecal sac has also been reported in<br />

clinical research (12). This approach provides an opportunity to characterize<br />

diurnal changes in CSF. Continuous CSF sampling also creates an opportunity<br />

to evaluate acute pharmacokinetic <strong>and</strong> pharmacodynamic responses with<br />

various pharmacological interventions (10).<br />

Lumbar puncture for CSF collection in research is generally safe (13).<br />

Nevertheless, LP is an invasive procedure. Whether done for diagnostic or<br />

research purposes, the procedure carries a number of inherent risks. The most<br />

common complication from LP is postdural puncture headache. The headache is<br />

typically frontotemporal <strong>and</strong> may be accompanied by neck stiffness, dizziness,<br />

<strong>and</strong> nausea. The headache may result from added tension on anchoring structures<br />

of the brain because of removal of CSF (14). Leakage of CSF at the dural<br />

puncture site may be an important factor. The latter explanation is supported<br />

by a decreased incidence of headache when LP is performed using a small<br />

gauge spinal needle shown to reduce CSF leakage in an in vitro cadaveric dural<br />

model (15). Maintaining the patient in a prone, slightly head down position<br />

can help resolve the headache <strong>and</strong> associated symptoms. Persistent headache<br />

can be treated by intravenous administration of caffeine or epidural injection of<br />

autologous whole blood (blood patch) at the LP site (14). Other more serious,<br />

but rare risks include hemorrhage, infection, <strong>and</strong> herniation of the brain. As a<br />

result, LP is contraindicated in individuals with a bleeding diathesis, thrombocytopenia<br />

(platelet count


56 Hale et al.<br />

or L3–L4 interspaces through which the spinal needle will pass (5,16). If the<br />

impact of the position on the planned analyses is unknown, it may be useful<br />

to record or predetermine the position in the research protocol. Strict attention<br />

to maintenance of sterile conditions is required. Administration of a local<br />

anesthetic reduces the risk of pain <strong>and</strong> tends to reduce unwanted movement of<br />

the patient during the course of the procedure.<br />

The care taken to avoid risk to a patient also helps ensure access to a high<br />

quality CSF specimen. A traumatic LP increases the likelihood of blood in the<br />

CSF specimen. Excessive blood contamination is particularly problematic as<br />

it can distort chemical measurements <strong>and</strong> cell counts performed on CSF. If<br />

sufficiently severe, this issue can compromise clinical interpretation or research<br />

conclusions. The possibility of a traumatic tap producing a blood contaminated<br />

specimen can sometimes occur even with careful preparation of the patient <strong>and</strong><br />

close attention to procedural technique. Accordingly, CSF is typically collected<br />

in a series of sterile tubes that are submitted for laboratory analyses in a fashion<br />

that reduces the potential for such confounds. In diagnostic settings, the first<br />

tube is typically used for chemical <strong>and</strong> immunological analyses, a second for<br />

microbiological examination, <strong>and</strong> a third for total cell count <strong>and</strong> differential<br />

cell counts (17). Despite such precautions, clinical laboratories may refuse<br />

to perform selected analyses on grossly bloody CSF specimens. Similarly,<br />

research laboratories should pay close attention to this common, undesired<br />

source of preanalytical variability <strong>and</strong> define criteria that render a CSF specimen<br />

unacceptable for the intended use. Once collected, the CSF must be processed,<br />

preserved, <strong>and</strong> transported in a timely manner so as not to render a good<br />

specimen unacceptable. Preservation of specimen constituents consistent with<br />

the state at the time of collection requires attention to the potential impact of the<br />

collection device <strong>and</strong> vials, materials used for processing time <strong>and</strong> temperature.<br />

It is important to assure that “biology in the tube” does not confound attempts<br />

to characterize the biological state under investigation through degradation of<br />

chemical <strong>and</strong> cellular elements.<br />

Although we realize we cannot anticipate all possible proteomic methodologies<br />

that may be applied to CSF, the purpose of this review is to provide<br />

basic methodology for the collection, storage, qualification, <strong>and</strong> processing of<br />

CSF for proteomic applications.<br />

2. Materials<br />

2.1. <strong>Sample</strong> Simplification<br />

1. Montage equilibration buffer, wash buffer <strong>and</strong> columns are provide with the<br />

Montage Albumin Deplete Kit (Millipore).<br />

2. Protein G Sepharose (Amersham Biosciences).


Proteomics of Cerebrospinal Fluid 57<br />

2.2. Gel Electrophoresis<br />

1. Ammonium carbonate solution: 1M ammonium carbonate adjusted to pH 11 with<br />

30 % ammonium hydroxide.<br />

2. Reduction-alkylation cocktail: 97.5% ACN, 2% iodoethanol, <strong>and</strong> 0.5%<br />

triethylphosphine.<br />

3. Rehydration sample buffer: 11 cm IPG strips, (pH 3–10 nonlinear) <strong>and</strong> 8–16%<br />

linear gradient Protean II gels were from Bio-Rad, Hercules, CA, USA.<br />

4. IPG reduction buffer: 0.1M Tris-HC1, pH 6.8, 6M urea, 2M thiourea, 1% SDS,<br />

6.4 mM dithiothreitol (DTT), 30% glycerol, <strong>and</strong> a few grains of bromophenol<br />

blue.<br />

5. IPG alkylation buffer: 0.1M Tris-HC1, pH 6.8, 6M urea, 2M thiourea, 1% SDS,<br />

24 mM iodoacetamide , 30% glycerol, <strong>and</strong> a few grains of bromophenol blue.<br />

2.3. Silver Stain<br />

1. Fixer 1: 50% MeOH, 10% Acetic acid.<br />

2. Fixer 2: 5% MeOH, 10% acetic acid.<br />

3. DTT solution: 5 mg/mL dithiothreitol.<br />

4. Silver solution: 0.1% AgNO 3 <br />

5. Developer: 30 g sodium carbonate in 1LH 2O + 500 μL 37% formaldehyde.<br />

6. Stopping solution: 2.3M citric acid.<br />

2.4. In-Gel Digestion<br />

1. Farmers reagent: 30 mM potassium ferricyanide (solution 1) <strong>and</strong> 100 mM sodium<br />

thiosulphate (solution 2). The working reagent is 1 part solution 1 <strong>and</strong> 1 part<br />

solution 2 mixed together just before use.<br />

2. Gel reduction-alkylation reagent: 50% 0.1M ammonium carbonate solution,<br />

48.75% acetonitrile (by volume), 1% iodoethanol, <strong>and</strong> 0.25% triethylphosphine<br />

(final pH 10).<br />

3. Gel trypsin solution: 20 ng/μL analytical grade (Promega,Madison, WI). One vial<br />

containing 20 μg was reconstituted with 1 mL 50 mM sodium bicarbonate, pH 8.<br />

4. Extraction solution: 100 mM NH 4HCO 3, pH 8.5.<br />

5. Destain reagent: 50% acetonitrile, 25 mM NH 4HCO 3, pH 8.0.<br />

6. DTT solution 2: 50 mM DTT in 100 mM NH 4HCO 3, pH 8.0.<br />

7. Iodoacetamide solution: 100 mM iodoacetamide in 100 mM NH 4HCO 3, pH 8.0.<br />

2.5. Processing CSF for LC/MS/MS Analysis<br />

1. Chicken lysozyme (Sigma, St Louis, MO) is dissolved at 1 mg/mL in H2O <strong>and</strong><br />

stored in single use aliquots at –80°C. Working solutions are prepared by diluting<br />

at 12.5 μg/mL in urea solution before use.<br />

2. Urea solution: 8M urea in 100 mM ammonium carbonate, pH 11.0.<br />

3. LC/MS reduction-alkylation reagent: 97.5 % acetonitrile, 2% iodoethanol, 0.5%<br />

triethylphosphine (see Note 1).


58 Hale et al.<br />

4. LC/MS trypsin solution: TPCK treated bovine pancreatic trypsin (Worthington,<br />

Lakewood, NJ) is dissolved at 1 mg/mL in H 2O <strong>and</strong> stored in single use aliquots<br />

at –80°C. Working solutions are prepared by diluting to 5 μg/mL in 100 mM<br />

ammonium bicarbonate pH 8.0 before use.<br />

2.6. MALDI-TOF Mass Spectrometry<br />

1. O-methylisourea solution: 1M solution O-methylisourea-hemisulfate (Arcos) in<br />

100 mM sodium carbonate buffer adjusted to pH 10 with 5.0N NaOH (see Note 2).<br />

2. Matrix solution: Recrystallized -cyano-4-hydroxycinnamic acid (see Note 3) is<br />

reconstituted with 200 μL 50 % acetonitrile, 0.05 % TFA to make saturated matrix<br />

solution.<br />

2.7. LC-MS/MS Mass Spectrometry.<br />

1. The C-18 reversed phase column was a Zorbax SB300 1× 50 mm (Agilent).<br />

2. Solvent A: 0.1% formic acid (Aldrich) in water (Burdick <strong>and</strong> Jackson HPLC<br />

grade).<br />

3. Solvent B: 50% acetonitrile, 0.1% formic acid (Aldrich) in water (Burdick <strong>and</strong><br />

Jackson HPLC grade).<br />

4. Solvent C: 80% acetonitrile, 0.1% formic acid (Aldrich) in water (Burdick <strong>and</strong><br />

Jackson HPLC grade).<br />

3. Methods<br />

3.1. Simplification of CSF by Removal of Abundant Proteins<br />

Similar to serum, albumin <strong>and</strong> immunoglobulin comprise more than 50 % of<br />

the protein concentration of CSF. To visualize proteins of lower abundance, it<br />

is desirable to reduce the levels of these proteins before separation or analysis.<br />

One procedure for the reduction of these proteins is described here.<br />

1. Resuspend Protein G Sepharose beads in Montage equilibration buffer, centrifuge<br />

at 500g for 2 min, <strong>and</strong> discard supernatant. Repeat Montage equilibration buffer<br />

addition <strong>and</strong> centrifuging once. Resuspend beads pellet in equal volume of<br />

Montage equilibration buffer.<br />

2. Dilute aliquots (50 μg protein) of CSF with Montage equilibration buffer to a<br />

volume of 300 μL.<br />

3. To the CSF samples, add 20μL Protein G Sepharose bead suspension <strong>and</strong> agitate<br />

slowly for 1hatRT.<br />

4. While samples are incubating with Protein G Sepharose, rehydrate Montage<br />

columns via manufacturer specifications: add 400 μL of Montage equilibration<br />

buffer to column insert <strong>and</strong> centrifuge at 500g for 2 min. Discard eluate from the<br />

collection tube.<br />

5. Pellet Protein G Sepharose beads at 500g for 2 min.


Proteomics of Cerebrospinal Fluid 59<br />

6. Carefully remove (without disturbing beads) <strong>and</strong> transfer 280 μL of the effluent to<br />

a rehydrated Montage column. Centrifuge column at 500g for 2 min. Reapply the<br />

eluate to the column <strong>and</strong> centrifuge again. Add two consecutive 100-μL washes<br />

of Montage wash buffer over the column via 500g centrifugation for 2 min (final<br />

volume approx 500 μL). Discard the column insert.<br />

7. Speed vacuum samples to approx 30–50 μL. Monitor volumes during speed<br />

vacuuming to prevent evaporation to dryness.<br />

3.2. Processing CSF for 2-Dimensional Gel Electrophoresis<br />

Separation of proteins by 2-dimensional gel electrophoresis may be adversely<br />

affected by the presence of salts in the sample. Additionally, smearing of b<strong>and</strong>s<br />

has been noted in <strong>2D</strong> gel electrophoresis that is attributable to incomplete<br />

reduction of disulfides in proteins (18). We have addressed both of these issues<br />

in the procedure that follows.<br />

1. Dialyze CSF against deionized water using 3-kDa MWCO dialysis tubing.<br />

2. To reduce <strong>and</strong> alkylate the protein samples, place 50 μg of CSF protein (approx<br />

50 μL) in a tube. Add 5 μL of ammonium carbonate solution followed by 50 μL<br />

of reduction-alkylation cocktail ( see Note 1). Cap the tube <strong>and</strong> incubate for 60<br />

min at 37°C.<br />

3. The sample is then uncapped <strong>and</strong> evaporated in a speedvacuum with medium heat<br />

for 2 h. After the sample is dried, reconstitute it in Bio-Rad rehydration sample<br />

buffer.<br />

4. <strong>Sample</strong>s are isoelectrically focused using 11 cm IPG strips, (pH 3–10, nonlinear).<br />

Rehydrate the IPG strips overnight at room temperature. The IPG strips are run<br />

at 8,000 V for 60,000 Vh using an IEF cell.<br />

5. The second dimension separation is performed in 8–16% linear gradient Protean<br />

II gels at 120 V for 2 h.<br />

6. Stain the gels with silver (Section 3.3) <strong>and</strong> scan with a Bio- Rad FX-Imager at<br />

50-μm resolution.<br />

3.2.1. Alternative 2-Dimensional Gel Electrophoresis Procedure<br />

This procedure incorporates the alkylation of cysteines into the prepping of<br />

the IPG strips for the second dimension.<br />

1. After dialysis against water, dissolve samples in approx 0.4 mL of rehydration<br />

solution <strong>and</strong> apply to immobilized pH gradient strips. The rehydration of the IPG<br />

strips is performed overnight at room temperature. Run the rehydrated IPG strips<br />

at 8,000 V for 60,000 Vh using an IEF cell.<br />

2. Following focusing, reduce the proteins on the strips by immersing the strips in<br />

IPG reduction buffer for 15 min at room temperature.<br />

3. Alkylate the proteins by immersing the strips in IPG alkylation buffer for 15 min<br />

at room temperature.


60 Hale et al.<br />

4. The IPG strips are then overlayed on SDS <strong>PAGE</strong> gels (8–16 % linear gradient<br />

Protean II gels) <strong>and</strong> separated at 120 V for 2 h.<br />

3.3. Mass Spectrometry Compatible Silver Stain<br />

Some silver staining procedures use reagents, such as glutaraldehyde, which<br />

will interfere with proteomic analysis of proteins (19). Glutaraldehyde will react<br />

with lysines in proteins <strong>and</strong> cross-link them. This interferes with trypsin, which<br />

is the typical enzyme used for in-gel digestion procedures. Formaldehyde is<br />

capable of inducing the reduction of silver while not cross linking proteins. The<br />

following procedure has been used for staining of gels before in-gel digestion<br />

<strong>and</strong> mass spectral analysis.<br />

1. Following electrophoresis soak the gels in fixer 1 for 30 min (see Note 4). Then<br />

for an additional 30 min in fixer 2. Agitate on a rocker platform gently.<br />

2. Rinse gels with Milli-Q H 2O with gentle agitation for 20 min to 1 h (200 mL at<br />

a time).<br />

3. Soak gels in a DTT solution for 30 min (volume sufficient to cover gel i.e.,<br />

100 mL) with gentle rocking.<br />

4. Pour the DTT solution off <strong>and</strong> add 100 mL silver solution to cover gel which is<br />

soaked 30 min with gentle rocking.<br />

5. Pour off the silver solution <strong>and</strong> rinse the gel once with 100 mL H 2O (rapidly).<br />

Rinse the gel twice with 100 mL developer for ∼30 sec. Then soak in 100 mL<br />

developer until proteins reach the desired intensity (see Note 5).<br />

6. Stop the development by adding 5 mL stopping solution per 100 mL developer<br />

<strong>and</strong> agitating for 10 min.<br />

7. Rinse the gel with several changes of distilled H 2O over 30 min. Fig. 1 shows an<br />

image of a silver stained 2-D gel of albumin depleted CSF. Fig. 2 lists the spots<br />

that were identified after digestion <strong>and</strong> LC/MS/MS analysis.<br />

3.4. In-Gel Digestion Procedure<br />

3.4.1. St<strong>and</strong>ard Protocol<br />

Following gel separation <strong>and</strong> staining, a common procedure for identification<br />

of proteins is excising the gel b<strong>and</strong> or spot <strong>and</strong> digestion of the protein in the gel<br />

with a proteolytic enzyme (most commonly trypsin). The following procedure<br />

may be used with dye or silver stained gels.<br />

1. Place Coomassie blue or Sypro ruby-stained gel pieces in Eppendorf centrifuge<br />

tubes or in the wells of a 96-well plate.<br />

2. Silver stained gel pieces are first de-stained by immersing them in Farmers reagent<br />

for 5–10 min followed by 3 washes with Milli-Q water (see Note 6).


Proteomics of Cerebrospinal Fluid 61<br />

Fig. 1. Albumin was removed from a pooled CSF sample using the procedure in<br />

Section 3.1 without the protein G step. Proteins were separated by two-dimensional<br />

electrophoresis. The gel was silver stained.<br />

3. Immerse the gel pieces in gel reduction-alkylation reagent sufficient to completely<br />

cover them (usually ∼100 μL (see Note 1). Cap the tubes, or seal the plate, <strong>and</strong><br />

incubate the samples at 37°C for 60 min.<br />

4. After incubation, the reagent is drawn off <strong>and</strong> discarded. Dehydrate the gel pieces<br />

with 100 μL of pure acetonitrile for approx 5 min.<br />

5. Decant the excess acetonitrile, <strong>and</strong> dry the gel pieces on a speed vacuum for at<br />

least 15min with medium heat.<br />

6. Rehydrate the gel pieces with gel trypsin solution (usually 5–10 μL,) <strong>and</strong> incubate<br />

overnight at 37°C (see Note 7).<br />

7. For LC/MS/MS analysis, extract peptides with extraction solution for 60 min at<br />

37°C <strong>and</strong> desalt with a μC-18 Ziptip before injection onto the mass spectrometer.<br />

For MALDI analysis, extract peptides as described in Section 3.6.<br />

3.4.2. Alternative Procedure for Reduction <strong>and</strong> Alkylation of Gel Pieces<br />

For proteins separated in gels without prior reduction <strong>and</strong> alkylation this<br />

procedure can be substituted for the reduction/alkylation step described previously.


62 Hale et al.<br />

Fig. 2. Spots were cut out <strong>and</strong> digested with trypsin (see 3.4). LC-MS/MS was<br />

performed with the procedure listed in Section 3.7 <strong>and</strong> proteins identified searching<br />

the non-redundant database with SEQUEST.<br />

1. Crush excised Coomassie blue or Sypro stained gel pieces <strong>and</strong> soak in destain<br />

reagent for at least 5 min. Decant destain <strong>and</strong> add fresh destain reagent for another<br />

5 min. Repeat at least 3 times total.<br />

2. Silver stained gel pieces are destained as described previously.<br />

3. Reduce protein in the gel with 50 μL of DTT solution 2, for 30 min at 56°C.<br />

4. Remove the DTT solution <strong>and</strong> replace with 50 μL of iodoacetamide solution, for<br />

30 min at 45°C.<br />

5. Remove the iodoacetamide solution, wash the gel pieces with extraction solution,<br />

<strong>and</strong> then with acetonitrile <strong>and</strong> dry on a speedvacuum.<br />

6. Rehydrate the gel pieces with gel trypsin solution (usually 5–10 μL,) <strong>and</strong> incubate<br />

overnight at 37°C (see Note 7).<br />

7. For LC/MS/MS analysis, extract peptides with extraction solution for 60 min at<br />

37°C <strong>and</strong> desalt with a μC-18 Ziptip before injection onto the mass spectrum. For<br />

MALDI analysis, extract peptides as described in Section 3.6.


Proteomics of Cerebrospinal Fluid 63<br />

3.5. Processing CSF for LC/MS/MS Analysis<br />

More frequently, gel electrophoresis procedures are being eliminated <strong>and</strong><br />

protein mixtures are being analyzed by chromatographic separation of proteolytic<br />

digests of the mixtures. This procedure has been used for processing of<br />

unfractionated cerebrospinal fluid before LC/MS/MS analysis.<br />

1. Prepare the albumin <strong>and</strong> immunoglobulin depleted CSF as in Section 3.1.<br />

2. Add 40 μL of chicken lysozyme solution (see Note 8).<br />

3. Add 100 μL of LC/MS reduction/alkylation reagent (see Note 1). Close the tubes<br />

<strong>and</strong> incubate for 1hat37°C.<br />

4. Speedvacuum the solutions to dryness, at least 3 h with medium heat.<br />

5. Redissolve the pellet in 200 μL of LC/MS trypsin solution to produce a 1.6M<br />

urea solution <strong>and</strong> an enzyme: substrate ratio of 1:50 (w/w).<br />

6. Incubate at 37°C overnight.<br />

7. Typically, 100 μL of this digest is injected onto the mass spectrometer.<br />

3.6. MALDI-TOF Mass Spectrometry<br />

Peptides from in-gel digests of proteins may be identified from their peptide<br />

fingerprint. MALDI-TOF mass spectrometry can provide that fingerprint which<br />

consists of the singly protonated mass of each tryptic peptide in a given protein.<br />

These fingerprints may be used to search protein databases using software<br />

written for this purpose. Conversion of lysines into homoarginines increases<br />

the sensitivity of detection of lysine containing tryptic peptides (20–22). This<br />

procedure has been used for 1 <strong>and</strong> 2 D gel separated proteins.<br />

1. Following in-gel trypsin digestion (Section 3.4), extract peptides with Omethylisourea<br />

solution for 60 min at 37°C to convert lysines to homoarginine<br />

2. Desalt samples with a μC-18 Ziptip before MALDI mass spectral analysis.<br />

3. Spot the desalted peptides onto a MALDI target (1 μL) <strong>and</strong> add an equal volume<br />

of saturated matrix solution.<br />

4. After the spots dry, MALDI-TOF spectra are obtained on a Voyager DE-Pro mass<br />

spectrometer (see Note 9).<br />

5. Search peptide masses using the program Knexus. The mass of the homoarginine<br />

residue (170.23) <strong>and</strong> the S-ethanol-cysteine residue (147.03) are added to the<br />

list of user defined modifications. For peptides obtained from in-gel digests, the<br />

oxidized methionine modification is also selected.<br />

3.7. LC-MS/MS Mass Spectrometry<br />

For unfractionated protein digests or for gel digests that contain multiple<br />

proteins, LC-MS/MS analysis can separate <strong>and</strong> fragment peptides providing


64 Hale et al.<br />

spectra that can be used to search databases for peptide identification. The<br />

following procedure is capable of separating peptides from digests of unfractionated<br />

CSF.<br />

1. Inject tryptic digests (100–200 L from Section 3.5) onto a C-18 reversed<br />

phase column at a flow rate of 50 L/min on a Surveyor HPLC system<br />

(ThermoFinnigan).<br />

2. The gradient conditions are 90% solvent A, 10% solvent B to 5% solvent A, 95%<br />

solvent B over 120 min, followed by a 0.1 min ramp to 100% solvent C, <strong>and</strong> held<br />

at 100% solvent C for 5 min, followed by a 0.1 min ramp to 90 % solvent A,<br />

10% solvent B, <strong>and</strong> held for 17 min. Divert the effluent to waste for the first<br />

5 min to keep the source clean. The total column effluent is connected to the<br />

electrospray interface of an LTQ ion trap mass spectrometer (ThermoFinnigan).<br />

The source is operated in positive ion mode with 4.8 kV electrospray potential, a<br />

sheath gas flow of 20 arbitrary units, <strong>and</strong> a capillary temperature of 225°C. The<br />

source lenses are set by maximizing the ion current for the 2+ charge state of<br />

angiotensin.<br />

3. Collect data in the triple play mode with the following parameters: centroid parent<br />

scan set to 1 microscan <strong>and</strong> 50 ms maximum injection time, profile zoom scan<br />

set to 3 microscans <strong>and</strong> 500 ms maximum injection time, <strong>and</strong> a centroid MS/MS<br />

scan set to 2 microscans <strong>and</strong> 2000 ms maximum injection time. Set dynamic<br />

exclusion settings to a repeat count of one, exclusion list duration of two minutes,<br />

<strong>and</strong> rejection widths of –0.75 m/z <strong>and</strong> +2.0 m/z.<br />

4. Carry collisional activation out at a relative collision energy of 35% <strong>and</strong> an<br />

exclusion width of 3 m/z.<br />

5. Search MS/MS spectra against a nonredundant protein database with<br />

SEQUEST (23).<br />

4. Notes<br />

1. The reduction alkylation solution should be prepared just before use. Triethylphosphine<br />

is pyrophoric <strong>and</strong> should be h<strong>and</strong>led in a fume hood in accordance with the<br />

material safety data sheet.<br />

2. If the pH is not properly adjusted to 10.0 the reaction may not proceed to<br />

completion.<br />

3. -cyano-4-hydroxycinnamic acid (Sigma, St Louis, MO) is recrystalized with<br />

the following procedure. Place 50 mg -cyano-4-hydroxy-cinnamic acid in a 50<br />

mL tube <strong>and</strong> add 10 mL of DI water. Add 30 % ammonium hydroxide until<br />

completely dissolved. Centrifuge <strong>and</strong> pour into a new 50-mL tube. In a fume<br />

hood add neat trifluoroacetic acid until pH is below 3 to precipitate the -cyano-<br />

4-hydroxycinnamic acid (check with indicator paper), centrifuge, <strong>and</strong> decant the<br />

supernatant. Wash the pellet 3× with 0.1 % TFA. Dissolve the final pellet into<br />

enough 50% acetonitrile 0.1% TFA to just dissolve it, no more than 40 mL.<br />

Aliquot 200 μL into 1.0 mL tubes, speed vaccuum to dryness <strong>and</strong> store at –20°C.


Proteomics of Cerebrospinal Fluid 65<br />

4. To minimize background staining, clean glassware with which the gel will come<br />

into contact with 50% HNO 3. Wear gloves when h<strong>and</strong>ling the gel as fingerprints<br />

will produce background stain.<br />

5. Development should be executed with continuous agitation. If a precipitate should<br />

begin to form discard the developer <strong>and</strong> replace it with fresh developer.<br />

6. The gel pieces should be clear. If yellow color remains continue washes until it<br />

is gone.<br />

7. The volume of trypsin solution added should be just enough to rehydrate the gel.<br />

There should not be excess liquid immersing the gel.<br />

8. Lysozyme is added as an internal st<strong>and</strong>ard to monitor the efficiency of<br />

reduction/alkylation <strong>and</strong> digestion.<br />

9. The mass spectrometer was calibrated with des-Arg-bradykinin, angiotensin I,<br />

glu1-fibrinopeptide,<strong>and</strong> ACTH [1–17, 18–39, <strong>and</strong> 7–38], insulin (bovine), thioredoxin<br />

(Escherichia coli), <strong>and</strong> apomyoglobin (horse) (Sigma, St. Louis, MO).<br />

References<br />

1. Huang, T., Chung, H., Chen, M., Giiang, L., Chin, S., Lee, C., Chen, C., <strong>and</strong><br />

Liu, Y. (2004) Supratentorial cerebrospinal fluid production rate in healthy adults:<br />

quantification with two-dimensional cine phase-contrast MR imaging with high<br />

temporal <strong>and</strong> spatial resolution. Radiology 233:603–8.<br />

2. Nilsson C., Lindvall-Axelsson M., <strong>and</strong> Owman C. (1992) Neuroendocrine<br />

regulatory mechanisms in the choroid plexus-cerebrospinal fluid system. Brain<br />

Research - Brain Res. Rev. 17(2):109–38.<br />

3. Rohlff C. (2000) Proteomics in molecular medicine: applications in central nervous<br />

systems disorders. Electrophoresis 21(6):1227–34.<br />

4. Walsh M. J., Limos L., <strong>and</strong> Tourtellotte W. W. (1984) Two-dimensional<br />

electrophoresis of cerebrospinal fluid <strong>and</strong> ventricular fluid proteins, identification<br />

of enriched <strong>and</strong> unique proteins, <strong>and</strong> comparison with serum. J. Neurochem.<br />

43(5):1277–85.<br />

5. Ebers, G. Lumbar puncture <strong>and</strong> cerebrospinal fluid analysis. In: Kelley’s Textbook<br />

of Internal Medicine, 4th ed. Humes, H. D., ed. Lippincott Williams & Wilkins,<br />

Philadelphia. 2000, pp 2996–98.<br />

6. Griggs, R. C., Józefowicz, R. F., Aminoff, M. J. Approach to the patient with<br />

neurologic disease. In: Cecil Textbook of Medicine, 22nd Ed. Philadelphia, Eds:<br />

Goldman L, Ausiello D. Saunders, pp 2196–2205.<br />

7. Smith G. P., Kjeldsberg C. R. Cerebrospinal, synovial, <strong>and</strong> serous body fluids, In:<br />

Clinical Diagnosis <strong>and</strong> Management by Laboratory Methods, 19th Ed. Henry J. B.,<br />

ed. W.B. Saunders Company, Philadelphia. 1996, pp 457–82.<br />

8. Altemus M., Fong J., Yang R.,Damast S., Luine V., <strong>and</strong> Ferguson D. (2004)<br />

Changes in cerebrospinal fluid neurochemistry during pregnancy. Biol. Psychiatr.<br />

56(6):386–92.<br />

9. Baker D. G., West S. A., Nicholson W. E., et al. (1999)Serial CSF corticotropinreleasing<br />

hormone levels <strong>and</strong> adrenocortical activity in combat veterans with<br />

posttraumatic stress disorder. Am. J. Psychiatr. 156(4):585–8.


66 Hale et al.<br />

10. Bieck P. R.<strong>and</strong> Potter W. Z. (2005) Biomarkers in psychotropic drug development:<br />

integration of data across multiple domains. Ann. Rev. Pharmacol. Toxicol. 45,<br />

227–46.<br />

11. Boz C., Ozmenoglu M., Velioglu S., Kilinc K., Orem A., Alioglu Z.,<br />

Altunayoglu V.. (2006) Matrix metalloproteinase-9 (MMP-9) <strong>and</strong> tissue inhibitor<br />

of matrix metalloproteinase (TIMP-1) in patients with relapsing-remitting multiple<br />

sclerosis treated with interferon beta. Clin. Neurol. Neurosurg. 108:124–28,<br />

12. Geracioti T. D., Jr, Orth D. N., Ekhator N. N., Blumenkopf B., Loosen P. T.<br />

(1992) Serial cerebrospinal fluid corticotropin-releasing hormone concentrations<br />

in healthy <strong>and</strong> depressed humans. J. Clin. Endocrinol. Metab. 74:1325–30<br />

13. Jhee S. S., Zarotsky V. (2003) Safety <strong>and</strong> tolerability of serial cerebrospinal<br />

fluid (CSF) collections during pharmacokinetic/pharmacodynamic studies: 5 years<br />

experience. Clin. Res. Reg. Affairs 20(3):357–63.<br />

14. Raskin N. H. Headache. In: Harrison’s Principles of Internal medicine, 16th<br />

ed. Kasper D. L., Fauci A. S., Longo D. L., Brauwald E., Hauser S. L., <strong>and</strong><br />

Jameson J. L. eds. McGraw Hill, New York, pp 85–94<br />

15. Angle P. J., Kronberg J, E., Thompson D. E., et al. (2003) Dural tissue trauma<br />

<strong>and</strong> cerebrospinal fluid leak after epidural needle puncture: effect of needle design,<br />

angle, <strong>and</strong> bevel orientation. Anesthesiology 99(6):1376–82.<br />

16. S<strong>and</strong>oval M., Shestak W., Sturmann K., <strong>and</strong> Hsu C. (2004) Optimal patient<br />

position for lumbar puncture, measured by ultrasonography. Emergency Radiology<br />

10(4):179–81.<br />

17. Kjeldsberg C. R. <strong>and</strong> Knight J. A. (1986) Body Fluids, Laboratory Examination of<br />

Amniotic, Cerebrospinal, Seminal, Serous, <strong>and</strong> Synovial Fluids: A Textbook Atlas,<br />

2nd ed. American Society of Clinical Pathologists Press, Chicago, IL.<br />

18. Herbert, B., Galvani, M., Hamdan, M., et al (2001) Reduction <strong>and</strong> alkylation of<br />

proteins in preparation of two-dimensional map analysis: why, when, <strong>and</strong> how.<br />

Electrophoresis 22, 2046–57.<br />

19. Shevchenko, A., Wilm, M., Vorm, O., Mann, M.(1996) Mass spectrometric<br />

sequencing of proteins silver-stained polyacrylamide gels. Anal. Chem. 68, 850–58.<br />

20. Hale, J. E.,. Butler, J. P, Knierman, M. D., Becker, G. W. (2000) Increased sensitivity<br />

of tryptic peptide detection by MALDI-TOF mass spectrometry is achieved<br />

by conversion of lysine to homoarginine. Anal. Biochem. 287, 110–17.<br />

21. Brancia, F. L., Oliver, S. G., Gaskell, S. J. (2000) Improved matrix-assisted<br />

laser desorption/ionization mass spectrometric analysis of tryptic hydrolysates of<br />

proteins following guanidination of lysine-containing peptides.Rapid Commun.<br />

Mass Spectrom. 14, 2070–73.<br />

22. Beardsley, R. L., Karty, J. A., Reilly, J. P. (2000) Enhancing the intensities of<br />

lysine-terminated tryptic peptide ions in matrix-assisted laser desorption/ionization<br />

mass spectrometry. Rapid Commun. Mass Spectrom. 14, 2147–53.<br />

23. Eng, J. K., McCormack, A. L., Yates, J. R. (1999) An approach to correlate t<strong>and</strong>em<br />

mass spectral data of peptides with amino acid sequences in a protein database,<br />

Anal. Chem. 71, 4981–88.


6<br />

<strong>Sample</strong> <strong>Preparation</strong> of Bronchoalveolar Lavage Fluid<br />

Baptiste Leroy, Paul Falmagne, <strong>and</strong> Ruddy Wattiez<br />

Summary<br />

Respiratory diseases are important health problem throughout the world. The<br />

bronchoalveolar lavage (BAL) fluid obtained by fiber-optic bronchoscopy is a biofluid<br />

reflecting the expression of secreted pulmonary proteins <strong>and</strong> the products of activated<br />

cells. The characterization of the BALF proteome provides an opportunity to establish<br />

diagnostic <strong>and</strong> get prognostic indicators of airway diseases. The main part of the chapter<br />

is devoted to the description of the most effective <strong>and</strong> reliable method of BALF sample<br />

preparation <strong>and</strong> processing for proteomic studies. Principal BALF proteome characteristics<br />

<strong>and</strong> the difficulties to work with this fluid are also introduced. Finally, the best<br />

conditions for high resolution <strong>and</strong> high reliability 2-dimensional electrophoresis (DE) of<br />

BALF samples are given.<br />

Key Words: BALF sampling; bronchoalveolar lavage fluid; epithelial lining fluid;<br />

lung; mass spectrometry; proteome.<br />

1. Introduction<br />

The respiratory tract, <strong>and</strong> in particular the alveoli, are covered by a thin film<br />

called epithelium lining fluid (ELF). This fluid plays important functions of<br />

protection against external aggressions <strong>and</strong> preservation of the gas-exchange<br />

capability of the airways. ELF contains several categories of cells (mainly<br />

of the immune system) <strong>and</strong> a wide variety of soluble components (lipids,<br />

nucleic acids, <strong>and</strong> proteins/peptides) (1). Diverse techniques exist to sample<br />

this particular fluid, but the most commonly used remains the bronchoalveolar<br />

lavage (BAL) during fiber-optic bronchoscopy. This minimally invasive <strong>and</strong><br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

67


68 Leroy et al.<br />

reproducible procedure consists of liquid instillation through a fibrobronchoscope<br />

<strong>and</strong> recovery of the bronchoalveolar lavage fluid (BALF) by gentle<br />

aspiration (2,3). This biofluid allows the monitoring of the expression of<br />

secreted pulmonary proteins <strong>and</strong> the products of activated cells.<br />

In the last decade, the number of BALF focusing studies has grown exponentially.<br />

BALF proteome study has been initiated by some groups that have<br />

demonstrated that investigations of BALF proteome <strong>and</strong> the discovery of<br />

disease associated proteins should contribute to a better knowledge of the<br />

lung at the molecular level <strong>and</strong> to the study of the lung disorders at the<br />

clinical level (4–8). The most abundant proteins are plasma proteins that<br />

derive by diffusion across the blood barrier. A comparative analysis of serum<br />

<strong>and</strong> BALF proteomes revealed the presence of proteins specifically produced<br />

in the airways (9). These proteins are, therefore, good c<strong>and</strong>idates for lung<br />

specific disease biomarkers. The proteins produced locally are very heterogeneous<br />

<strong>and</strong> can be classified according to their function: proteins involved<br />

in defense mechanisms, tissue remodelling, lipid metabolism, inflammatory<br />

processes, cell growth, <strong>and</strong> oxidant-antioxidant systems. Differential-display<br />

proteomics studies showed that nonphysiological conditions cause significant<br />

modifications of the BALF proteome; these modifications can be used to<br />

better underst<strong>and</strong> pathogenesis mechanisms or to reveal diseases. Significant<br />

efforts have been devoted to large scale identification of BALF proteins<br />

or their degradation products <strong>and</strong> to the evaluation of potential markers<br />

for lung diseases, especially, for fibrosing interstitial lung diseases such as<br />

sarcoidosis <strong>and</strong> idiopathic pulmonary fibrosis (4,6,8,10,11). A majority of<br />

BALF proteomic studies use the two dimensional gel electrophoresis (2-DE)<br />

approach associated with mass spectrometry technologies. The technological<br />

progress encountered recently also allow scientists to reach the objective<br />

of quantitative analysis that is of crucial importance in differential-display<br />

proteomic. Reproducible two-dimensional BALF proteome patterns can be<br />

obtained using immobilized pH gradients (IPG) in the first dimension (Fig. 1)<br />

(12). After staining, the two dimensional patterns are compared to reveal<br />

up- or downregulated proteins. High throughput identification of proteins by<br />

mass spectrometry techniques also permits the creation of reference gels of<br />

the BALF proteome (9). These gels are now available on the World Wide<br />

Web (http://W3.umh.ac.be/biochim/proteomic.htm). The complexity of BALF<br />

proteome has been demonstrated by the great diversity of proteins present in<br />

this fluid.<br />

Although BALF proteome analysis is now largely facilitated by previous<br />

works, BALF sample preparation remains a critical step (13). Indeed, majors<br />

problems associated with BALF proteome analysis are ascribable to the low


<strong>Sample</strong> <strong>Preparation</strong> of Bronchoalveolar Lavage Fluid 69<br />

100 kDa<br />

MW<br />

5 kDa<br />

3.5<br />

α1-antitrypsin<br />

SP-A<br />

CC16<br />

Albumin<br />

Immunoglobulin light chain, κλ<br />

Transthyretin<br />

pI 10<br />

IgG heavy chain, γ<br />

Fig. 1. 2-D gel electrophoresis of human BALF proteins.<br />

protein content <strong>and</strong> the high salt (because of the phosphate-buffered saline used<br />

for the lavage procedure) <strong>and</strong> lipid concentrations of this sample. Moreover, as<br />

for other human fluid, the wide dynamic range of protein concentration with the<br />

occurrence of some major proteins gives rise to difficulties in the study of lowabundance<br />

proteins. In this context, several efforts have been made to optimize<br />

BALF sample preparation <strong>and</strong> to identify less abundant proteins (13). Recently,<br />

alternative or supplement methods to the 2-DE approach have been developed<br />

such as multidimensional liquid chromatography (multi-LC): proteins from<br />

BALF are separated by chromatography (ionic <strong>and</strong>/or reverse phase) coupled<br />

“on line” with mass spectrometry analysis (multi LC-MS) (10,14). This new<br />

promising approach allows the identification <strong>and</strong> the quantification of small,<br />

minor, <strong>and</strong> hydrophobic proteins present in the BALF samples. Nevertheless,<br />

at this moment, LC–MS technology dedicated to BALF proteome study lacks<br />

quantitative data.<br />

In this chapter, we describe BALF sample preparation for 2-DE, the most<br />

currently used proteomic tool. Clearly, this sample preparation dedicated to<br />

2-DE is also adequate to LC-MS approach. Finally, the conditions used in our<br />

laboratory to achieve 2-DE separation of BALF proteins are also detailed.


70 Leroy et al.<br />

2. Materials<br />

1. Sterile saline solution: 0.9% (w/v) NaCl, filtered through a 0.2-μm syringe filter<br />

unit (Minisart, Sartorius, Germany).<br />

2. Ultrapure water (double-distilled, deionized, >18) is used for all reagent<br />

preparations.<br />

3. Protease inhibitor cocktail tablets (Complete mini, Roche, Germany).<br />

4. Dialysis membrane: Spectra/Por membrane (MWCO : 3.5 kDa) (Spectrum).<br />

5. Dialysis buffer: 50 mM NH4HCO3. 6. Speed Vac.<br />

7. Immobilized pH gradient 3–10 NL 180mm (Pharmacia-Amersham).<br />

8. Rehydration solution: 7 M urea, 2 M thiourea, 4% CHAPS (w/v), 2% ampholytes<br />

3–10 (v/v), 65 mM DTE, <strong>and</strong> a trace of bromophenol blue. Prepare fresh each<br />

time.<br />

9. <strong>Sample</strong> buffer: 7 M urea, 2 M thiourea, 4% CHAPS (w/v), 2% ampholytes<br />

3–10 (v/v), 65 mM DTE <strong>and</strong> trace amounts of bromophenol blue. Prepare fresh<br />

each time.<br />

10. Equilibration buffer: 50 mM Tris-HCl, pH 6.8, 6 M urea, 30% glycerol (w/v),<br />

2% SDS (w/v). Prepare fresh each time.<br />

11. DryStrip Reswilling tray (Amersham biosciences or Bio-Rad).<br />

12. Multiphor II (Amersham biosciences) or Protean IEF Cell (Bio-Rad) device.<br />

13. St<strong>and</strong>ard vertical electrophoresis units for SDS-<strong>PAGE</strong>.<br />

14. Programmable power supply able to deliver >3000 V.<br />

15. Thermostatic circulator (Multitemp II, Amersham biosciences).<br />

3. Methods<br />

3.1. BALF Sampling<br />

1. Under medical controle, place a flexible fiberoptic bronchoscope through an<br />

endotracheal tube wedged into a subsegmental bronchus of an anesthetized patient<br />

(see Note 1 <strong>and</strong> 2).<br />

2. Instill, through the bronchoscope, 20 mL of sterile saline solution warmed to<br />

37°C.<br />

3. Collect the fluid by gentle aspiration <strong>and</strong> dispose in sterile siliconated bottles on<br />

ice (see Note 3). All subsequent manipulation of the samples should be realized<br />

on ice.<br />

4. Repeat procedure 1–3 four times <strong>and</strong> combine harvested fluids except the first<br />

sample (see Note 4).<br />

5. To elimine cells from the sample, centrifuge the combined fluids at 800g for 5min<br />

at 4°C (see Note 5, 6, 7).<br />

6. To avoid sample degradation, add an adequat amount (1 tablet for 60 mL of BALF<br />

sample) of protease inhibitor cocktail tablets (Complete mini, Roche, Germany)<br />

(see Note 8).<br />

7. Store BALF at –80°C until use (see Note 9).


<strong>Sample</strong> <strong>Preparation</strong> of Bronchoalveolar Lavage Fluid 71<br />

3.2. BALF <strong>Preparation</strong> for 2-DE<br />

Numerous BALF sample preparations have been used by different authors.<br />

Among these, precipitation (using trichloracetic acid or two-step combination<br />

of precipitant) is one of the most commonly used methodologies for protein<br />

analysis. However, limitations are observed with such a procedure because of a<br />

nonquantitative protein resolubilization after the precipitation step (13). Finally,<br />

the most efficient method of preparing a BALF sample before 2-DE analysis<br />

uses ultra filtration <strong>and</strong> dialysis-lyophilization (see Note 10).<br />

1. Rehydrate dialysis membranes (cut-off value 3.5 kDa) by soaking them in<br />

ultrapure water heated to about 90°C for 5 min or following manufacturer’s<br />

instructions. Length of the membrane must be adapted to the sample volume.<br />

Alternatively or if small volumes need to be dialyzed, ready to use dialysis<br />

devices are available commercially (Slide-A-Lyser 3.5K, Pierce).<br />

2. Rinse extensively the rehydrated membranes with ultrapure water. The<br />

membranes may not run dry.<br />

3. Close the dialysis tubes on one side <strong>and</strong> improve sealing with ultrapure water.<br />

4. Fill the membrane with BALF sample <strong>and</strong> close the second side of the dialysis<br />

tube. Check seal.<br />

5. Immerse the membrane tube in dialysis buffer (see Note 11) <strong>and</strong> place on a<br />

magnetic stirrer at 4°C for 3 h.<br />

6. Replace dialysis buffer twice.<br />

7. Harvest dialyzed BALF sample into an appropriate container.<br />

8. Reduce BALF sample volume to 20 μL in the Speed Vac (see Note 12).<br />

10. Suspend the sample in a minimum volume of sample buffer dedicated to 2-DE<br />

analysis or LC-MS method.<br />

11. Centrifuge at 18,000g for 15 min at 4°C to remove any unsolubilized material.<br />

12. <strong>Sample</strong> is now ready for protein assay <strong>and</strong> 2-DE or LC-MS analysis<br />

(see Note 13).<br />

Proteomic analysis of BALF is often hampered by the predominance of<br />

several highly abundant proteins including albumins <strong>and</strong> immunoglobulins.<br />

Depletion of these proteins is necessary before proteome analysis for detection<br />

of minor proteins. See other chapters in this book.<br />

3.3. BALF 2-DE analysis<br />

Here we propose the optimum conditions for obtaining high resolution <strong>and</strong><br />

reliable 2-DE of BALF samples (see Note 14). Use of different pH gradients in<br />

IEF or reticulations in SDS-<strong>PAGE</strong> should be needed in narrower study of BALF<br />

proteins. Indeed, the high number of protein spots in the pH range 4.5–6.7<br />

are increased or decreased in different lung pathologies such as sarcoidosis<br />

or fibrosis (6,8,9). In this context, the use of narrow range IPG strips for


72 Leroy et al.<br />

IEF improves the resolution of the separation <strong>and</strong> increases the probability of<br />

detecting less abundant proteins.<br />

1. Place the IPG strips in an adequate DryStrip Reswilling tray.<br />

2. Cover the IPG strips first with 500 μL of rehydration buffer <strong>and</strong> second with lowviscosity<br />

paraffin oil, <strong>and</strong> let the strips rehydrate overnight at room temperature.<br />

3. Remove the rehydrated IPG gels from the grooves, rinse them with ultrapure<br />

water, <strong>and</strong> place them, gel-side down, on water saturated filter paper. Filter<br />

papers are first blotted to remove excess water.<br />

4. Place rehydrated IPG strip (pH 3–10, 18 cm) in the Multiphor (Amersham<br />

Biosciences) or Protean IEF Cell (Bio-Rad) device set to 20°C. The correct<br />

settings of strips <strong>and</strong> cup loading can be achieved by following the manufacturer’s<br />

instructions.<br />

5. Apply 100 μg of protein/strip in cups at the anodic side of the IPG strip (see<br />

Note 15, 16).<br />

6. Increase voltage linearly from 300–5,000 V during the first 3 h <strong>and</strong> stabilize the<br />

voltage at 5,000 V for 20 h (see Note 17).<br />

7. After electrofocusing, place the IPG strips individually in capped glass tubes<br />

<strong>and</strong> equilibrate them for 20 min at room temperature in 10 mL of equilibration<br />

buffer containing 2% DTE (w/v) under gentle agitation.<br />

8. Replace buffer by 10 mL of fresh equilibration buffer containing 2% iodoacetamide<br />

(w/v) <strong>and</strong> incubate for 20 min at room temperature under gentle<br />

agitation.<br />

9. Run the second dimension on a 9–16% polyacrylamide linear gradient gel<br />

(18 × 20 × 1.5 cm) at 40 mA/gel constant current <strong>and</strong> 10°C.<br />

10. The gels are then ready to be stained using st<strong>and</strong>ard silver staining (see Note 18).<br />

4. Notes<br />

1. Human bronchoalveolar lavage is an invasive method that must be carried<br />

out under the informed consent of the concerned subjects <strong>and</strong> approved by a<br />

competent ethics committee.<br />

2. Human bronchoalveolar lavage requires topical lidocaine anaesthesia <strong>and</strong><br />

endotracheal tube manipulation <strong>and</strong> must thus be performed by a competent<br />

physician in an adequate environment.<br />

3. Typically, the mean recovery of BALF is 55 % of the instilled volume.<br />

4. The first sample is separated from the others to avoid bronchial contamination.<br />

5. With this procedure, typical protein concentration of BALF ranges from 0.05 to<br />

1.20 mg/mL.<br />

6. During the BAL process, the cellular elements can secrete a variety of components.<br />

Therefore, the cells should be removed immediately to provide optimal<br />

proteome stability.<br />

7. After centrifugation, the phenotype of cells (macrophages, lymphocytes) can<br />

be analyzed. The normal cellular pattern of BALF contains mainly alveolar


<strong>Sample</strong> <strong>Preparation</strong> of Bronchoalveolar Lavage Fluid 73<br />

macrophages, a small percentage of lymphocytes <strong>and</strong> less than 2% of<br />

polymorphs. This cellular pattern changes in many lung pathologies.<br />

8. Protease inhibitors are necessary but peptide inhibitors, e.g., high concentration<br />

of aprotinin, may interfere with mass spectrometry analysis.<br />

9. <strong>Sample</strong>s should be divided into small aliquots before storage to avoid repeated<br />

freezing <strong>and</strong> thawing of samples.<br />

10. Desalting by dialysis <strong>and</strong> ultra membrane centrifugation are very effective<br />

techniques for salt removal, leading to minimal sample loss compared to precipitation<br />

or filtration methods. However, during membrane ultracentrifugation,<br />

protein adsorption onto the membrane surface is a problem that can be, in part,<br />

circumvented by repeated washing steps with sample solution after centrifugation<br />

(13).<br />

11. Bath dialysis volume must be 100× sample volume.<br />

12. Speed Vac centrifugation is an easier method that generates less protein loss<br />

than freeze-drying (13).<br />

13. If electrophoresis is not to be run at this time, store the lyophilized BALF at<br />

–80°C until used. Protein concentration of BAL fluid must be determined using<br />

a suited protein assay.<br />

14. IEF running conditions depend on the pH gradient <strong>and</strong> the length of the IPG gel<br />

strip used. Conditions presented here assume the use of nonlinear wide-range<br />

immobilized pH gradient (3–10) 18 cm long IPG strips (optimized for body<br />

fluids) <strong>and</strong> the Multiphor II electrophoresis system of Amersham Biosciences.<br />

We also recommend the second dimension to be run on linear gradient polyacrylamide<br />

gels (9–16%) for best resolution.<br />

15. Different methods can be used to apply BALF sample on IPG strips such as<br />

the sample cup method or during the strip rehydration process. Classically, to<br />

increase the loading capacity, <strong>and</strong> enhance the resolution of 2-DE, the entire<br />

IPG gel can be used for sample application, with the proteins entering the gel<br />

during rehydration. Nevertheless, the best BALF 2-DE quality is obtained using<br />

the cup loading approach. A recent new type of in-gel sample application named<br />

“paper bridge sample application” has been optimized for the BALF proteome<br />

analysis. This procedure allows increasing the loading capacity of BALF sample<br />

without loss of the 2-DE resolution (8).<br />

16. After IEF but before equilibration, strips may be stored at –80°C until the second<br />

dimension.<br />

17. The quantity may be varied according to the sensitivity of the staining method.<br />

18. Quantification of proteins is a major problem of the 2-DE approach, especially<br />

after silver staining. However, a new fluorescent protein labeling protocol (<strong>2D</strong>-<br />

DIGE) before electrophoretic separation has been developed. (See other chapters<br />

in this book).<br />

Acknowledgments<br />

R. Wattiez is Research Associate of the Belgian FNRS. The authors thanks<br />

Catherine S’Heeren for her technical assitance.


74 Leroy et al.<br />

References<br />

1. Wattiez, R. <strong>and</strong> Falmagne, P. (2005) Proteomics of bronchoalveolar lavage fluid.<br />

J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 815, 169–78.<br />

2. Baughman, R. P. <strong>and</strong> Drent, M. (2001) Role of bronchoalveolar lavage in interstitial<br />

lung disease. Clin. in Chest Med. 22, 331–41.<br />

3. Reynolds, H.Y. (2000) Use of bronchoalveolar lavage in humans—past necessity<br />

<strong>and</strong> future imperative. Lung 25, 271–93.<br />

4. Wattiez, R., Hermans, C., Bernard, A., Lesur, O., <strong>and</strong> Falmagne, P. (1999)<br />

Human bronchoalveolar lavage fluid: two-dimensional gel electrophoresis, amino<br />

acid microsequencing <strong>and</strong> identification of major proteins. Electrophoresis 20,<br />

1634–45.<br />

5. Noel-Georis, I., Bernard, A., Falmagne, P., <strong>and</strong> Wattiez, R. (2002) Database of<br />

bronchoalveolar lavage fluid proteins. J. Chromatogr. B Analyt. Technol. Biomed.<br />

Life Sci. 771, 221–36.<br />

6. Magi, B., Bini, L., Perari, M.G., Fossi, A., Sanchez, J.C., Hochstrasser, D.,<br />

Paesano, S., Raggiaschi, R., Santucci, A., Pallini, V., <strong>and</strong> Rottoli, P. (2002)<br />

Bronchoalveolar lavage fluid protein composition in patients with sarcoidosis<br />

<strong>and</strong> idiopathic pulmonary fibrosis: a two-dimensional electrophoretic study.<br />

Electrophoresis 23, 3434–44.<br />

7. Lindahl, M., Stahlbom, B., <strong>and</strong> Tagesson, C. (1999) Newly identified proteins in<br />

human nasal <strong>and</strong> bronchoalveolar lavage fluids: potential biomedical <strong>and</strong> clinical<br />

applications. Electrophoresis 20, 3670–6.<br />

8. Sabounchi-Schutt, F., Astrom, J., Hellman, U., Eklund, A., <strong>and</strong> Grunewald, J.<br />

(2003) Changes in bronchoalveolar lavage fluid proteins in sarcoidosis: a<br />

proteomics approach. Eur. Respir. J. 21, 414–20.<br />

9. Noel-Georis, I., Bernard, A., Falmagne, P. <strong>and</strong> Wattiez, R. (2001) Proteomics as<br />

the tool to search for lung disease markers in bronchoalveolar lavage. Dis. Markers<br />

17, 271–8.<br />

10. Kriegova, E., Melle, C., Kolek, V., Hutyrova, B., Mrazek, F., Bleul, A., du<br />

Bois, R. M., von Eggeling, F. <strong>and</strong> Petrek, M. (2006) Protein Profiles of<br />

Bronchoalveolar Lavage Fluid from Patients with Pulmonary Sarcoidosis. Am. J.<br />

Resp. Crit. Care Med. 26, 1145–54.<br />

11. Lenz, A. G., Meyer, B., Costabel, U., <strong>and</strong> Maier, K. (1993) Bronchoalveolar lavage<br />

fluid proteins in human lung disease: analysis by two-dimensional electrophoresis.<br />

Electrophoresis 14, 242–4.<br />

12. Lenz, A. G., Meyer, B., Weber, H., <strong>and</strong> Maier, K. (1990) Two-dimensional<br />

electrophoresis of dog bronchoalveolar lavage fluid proteins. Electrophoresis 11,<br />

510–3.<br />

13. Plymoth, A., Lofdahl, C. G., Ekberg-Jansson, A., Dahlback, M., Lindberg, H.,<br />

Fehniger, T. E., <strong>and</strong> Marko-Varga, G. (2003) Human bronchoalveolar lavage:<br />

biofluid analysis with special emphasis on sample preparation. Proteomics<br />

3, 962–72.<br />

14. Wu, J., Kobayashi, M., Sousa, E., Lieu, W., Cai, J., Goldman, S. J., Dorner, A. J.,<br />

Projan, S. J., Kavuru, M. S., Qiu, Y., <strong>and</strong> Thomassen, M. J. (2005) Differential


<strong>Sample</strong> <strong>Preparation</strong> of Bronchoalveolar Lavage Fluid 75<br />

proteomic analysis of bronchoalveolar lavage fluid in asthmatics following<br />

segmental antigen challenge. Mol. Cell. Proteomics 4, 1251–64.<br />

15. Chromy, B., Gonzales, A., Perkins, J., Choi, M., Corzett, M., Chang, B. C.,<br />

Corzett, C. H., <strong>and</strong> McCutchen-Maloney, S. L. (2004) Proteomic analysis of<br />

human serum by two-dimensional differential gel electrophoresis after depletion<br />

of high-abundant proteins. J. Proteome Res. 6, 4–8.


7<br />

<strong>Preparation</strong> of Nasal Secretions for Proteome Analysis<br />

Begona Casado, Paolo Iadarola, <strong>and</strong> Lewis K. Pannell<br />

Summary<br />

The determination of protein patterns in nasal secretions of healthy subjects can help in<br />

the early diagnosis of diseases such as acute sinusitis. The comparison of nasal lavage fluid<br />

collected from subjects with acute sinusitis before <strong>and</strong> after pharmacological treatment<br />

gives information about the drug effects on gl<strong>and</strong>ular secretions. Nasal secretions were<br />

stimulated with 1× NS (0.9% Normal Saline) <strong>and</strong> 24× NS in healthy subjects <strong>and</strong> in<br />

sinusitis subjects before <strong>and</strong> after pharmacological treatment. The nasal lavage fluid<br />

(NLF) proteins are precipitated with a solution of “acid-ethanol.” Using this solution, the<br />

high molecular weight proteins precipitate <strong>and</strong> separate from the low molecular weight<br />

proteins. The proteins are digested <strong>and</strong> the peptides are separated using a capillary liquid<br />

chromatographic system. Eluted peptides are analyzed on ESI-Q-TOF mass spectrometry<br />

instrument.<br />

Key Words: CapLC-ESI-Q-ToF; liquid-liquid extraction; Nasal secretions; pharmacological<br />

treatment; proteomics; sample preparation; sinusitis.<br />

1. Introduction<br />

Nasal secretions (NS) are a barrier against pathogenic (e.g., bacteria <strong>and</strong><br />

viruses) <strong>and</strong> nonpathogenic (e.g., fine particles) antigens that are present in<br />

the air <strong>and</strong> are breathed in through the nose. NS serve to humidify, heat<br />

or cool, <strong>and</strong> clean inhaled air <strong>and</strong> contain proteins of the innate immune<br />

system. These proteins are from plasma, gl<strong>and</strong>ular mucous <strong>and</strong> serous cells (1,2)<br />

<strong>and</strong> their release is started during allergen exposure, rhinovirus, adenovirus,<br />

influenza, bacterial rhinosinusitis, cystic fibrosis, <strong>and</strong> occupational exposure.<br />

The hyperresponsiveness is a typical characteristic of inflamed mucosa <strong>and</strong><br />

airways (3–5) although different molecular mechanisms may be involved in<br />

allergic, infectious, <strong>and</strong> nonallergic disorders. The release of nasal secretions,<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

77


78 Casado et al.<br />

is generally induced by spraying a solution of normal (S) <strong>and</strong> hypertonic<br />

saline (HTS). The latter, in humans, is an airway irritant of nasal mucosa by<br />

stimulating nociceptive nerves <strong>and</strong> gl<strong>and</strong>ular secretion. For example, substance<br />

P is released in the mucosa after a neural depolarization with local axon<br />

responses caused by hypertonic saline stimulation (6,7). HTS nasal provocations<br />

have been employed also to underst<strong>and</strong> the different neural response between<br />

acute sinusitis, acute rhinitis, <strong>and</strong> the nonallergic rhinitis present in subjects<br />

with chronic fatigue syndrome (CFS) (8,9). In the last 4 years, the interest of<br />

the scientific community on nasal mucosa <strong>and</strong> nasal secretions has increased.<br />

The collection of nasal lavage, in fact, is a simple way for obtaining samples<br />

from the upper airways <strong>and</strong> may be performed using noninvasive procedures.<br />

For example, the agent responsible for the Creutzfeldt-Jakob disease can be<br />

identified “in vivo” in nasal mucosa (10), <strong>and</strong> an anthrax vaccine based on<br />

the use of an anthrax protective antigen (PA) protein carried by liposomeprotamine-DNA<br />

(LPD) is nasally dosed in mice (11).<br />

The complete knowledge of the nasal secretion constituents has not been<br />

achieved yet. It is apparent that the identification of specific mucous protein<br />

profiles may help elucidate the different mechanisms involved in host defense.<br />

To date the determination of mucus protein profiles can be achieved using<br />

a proteomic procedure. Different proteomic approaches have been used so<br />

far to analyze nasal lavage fluid (NLF). Lindahl et al. used two dimensional<br />

electrophoresis (2-DE) with matrix-assisted laser desorption/ionization<br />

(MALDI) mass spectrometry to analyze either the proteome of NLFs<br />

from subjects exposed to methyltetrahydrophthalic anhydride (MHHPA)<br />

or dimethylbenzylamine (DMBA), <strong>and</strong> that from healthy nonsmokers <strong>and</strong><br />

smokers (12–18). Another approach has been described by Casado et al. who<br />

applied liquid chromatography (LC) with electrospray ionization (ESI) mass<br />

spectrometry to analyze NLFs of subjects affected by acute sinusitis before<br />

<strong>and</strong> after pharmacological treatment, <strong>and</strong> for the comparison of NLFs of<br />

normal subjects before <strong>and</strong> after nasal provocation (19,20). Kristiansson et al.<br />

have used the same procedure for the analysis of HHPA-(hexahydrophthalic<br />

anhydride) adducted albumin tryptic peptides in nasal lavage fluid as<br />

biomarkers of exposure (21,22). These two complementary approaches<br />

provided new information on proteins involved in host protection <strong>and</strong> defence<br />

against microorganisms <strong>and</strong> occupational exposure.<br />

2. Materials<br />

2.1. Pharmacological Treatment<br />

1. Antibiotic: amoxicillin-clavulanic acid (Augmentin, GlaxoSmithKline, Research<br />

Triangle Park, NC, USA).


<strong>Preparation</strong> of Nasal Secretions for Proteome Analysis 79<br />

2. Steroid: fluticasone propionate nasal spray (Flonase, GlaxoSmithKline, Research<br />

Triangle Park, NC, USA).<br />

3. 2 adrenergic agonist vasoconstrictor: oxymetazoline nasal spray (Super G,<br />

L<strong>and</strong>over, MD, USA).<br />

4. United States Pharmacopea normal saline: saline nasal spray (0.9% NaCL) (Abbott<br />

Laboratories, IL, USA).<br />

2.2. Nasal Provocation<br />

1. United States Pharmacopea normal saline: saline nasal spray (0.9% NaCL) (Abbott<br />

Laboratories, IL, USA).<br />

2. Hypertonic saline 21.6%<br />

3. Beconase AQ pump aspirator spray device (23,24) (Glaxo-Wellcome, Triangle<br />

Park, NC, USA).<br />

4. Nasal secretion collection: 5-ounce Dixie wax-paper cup (James River Corp.,<br />

Norwalk, CT, USA) or polypropylene beakers (Fisher Scientific, Fair Lawn,<br />

NJ, USA).<br />

2.3. Nasal Lavage Fluid <strong>Preparation</strong> for Liquid Chromatography<br />

<strong>and</strong> Mass Spectrometry Analysis<br />

1. Protein assay on 96-well micro plates using MRX Microplate Reader Instrument<br />

(Dynex Technologies, Chantilly, VA, USA).<br />

2. Bovine albumin as st<strong>and</strong>ard protein for total protein assay (Sigma, MO, USA).<br />

3. Acid-ethanol solution: 50% 0.2N acetic acid, 50% ethanol, 0.02% sodium bisulfite<br />

(25) stored at 4°C. Ethanol <strong>and</strong> acetic acid obtained from Fisher Scientific<br />

(Fair Lawn, NJ, USA), <strong>and</strong> sodium bisulfite from Mallinckrodt Laboratory<br />

Chemicals (Phillipsburg, NJ, USA).<br />

4. Protein digestion: sequencing grade modified trypsin (Promega, Madison, WI,<br />

USA).<br />

5. Digestion buffer: 0.1M ammonium bicarbonate (pH 7.8) (Sigma, St. Louis,<br />

MO, USA).<br />

2.4. Liquid Chromatography <strong>and</strong> Mass Spectrometry Analysis<br />

of Nasal Lavage Fluid<br />

1. Desalt <strong>and</strong> concentration: 35 × 0.32 mm BioBasic C18 precolumn ( Thermo<br />

Hypersil-Keystone, Bellefonte, PA, USA).<br />

2. Peptide separation: Reverse-phase Zorbax C18 column (100 mm × 150 μm id)<br />

(Micro-Tech Scientific, Sunnyvale, CA, USA).<br />

3. Solvent A: HPLC grade H2O with 0.2% formic acid (Fisher Scientific,<br />

Fair Lawn, NJ, USA).<br />

4. Solvent B: HPLC grade acetonitrile with 0.2% formic acid (Fisher Scientific,<br />

Fair Lawn, NJ, USA).


80 Casado et al.<br />

5. Capillary LC instrument (Waters Inc, Milford, MA, USA).<br />

6. Electrospray-Quadrupole-Time of Flight mass spectrometer (Waters Inc, Milford,<br />

MA, USA).<br />

3. Methods<br />

3.1. Nasal Provocation<br />

1. Subjects’ nasal cavities need to be pre-washed with 24 sprays (100 μL each<br />

nostril) of sterile normal saline (1 × NS, 0.9% NaCl) using a Beconase AQ<br />

pump aspirator spray device.<br />

2. Subjects gently blow out through their noses, <strong>and</strong> the lavage fluid from both<br />

nostrils into a 5-ounce Dixie wax-paper cup. This material is discarded.<br />

3. 10 min later step 2 is repeated using 12 sprays of 1 × NS, <strong>and</strong> the lavage fluid<br />

discarded. Now the nasal provocation is performed.<br />

4. 100 μL of 0.9% normal saline is administered separately into each nostril.<br />

5. After 5 min, NLF is collected using 12 sprays of 0.9% NS (see Note 1).<br />

6. The NLF must gently blow out into a cup. NLF from left <strong>and</strong> right nostrils are<br />

mixed together (first series) (see Note 2).<br />

7. Immediately after this collection, the same subject is sprayed with hypertonic<br />

saline (HTS) (21.6% NaCl, 24 times the tonicity of NS (24 × NS). The pH of<br />

HTS (freshly prepared solution with double distilled deionized water) is 6.07<br />

(see Note 3).<br />

8. After 5 min, 12 puffs of NS are sprayed into each nostril.<br />

9. The NLF must gently blow out into a cup. NLFs from left <strong>and</strong> right nostrils are<br />

mixed together (second series).<br />

10. After pharmacological treatment on day 6, the first <strong>and</strong> second series for day 6<br />

are collected repeating the procedure indicated in steps 1–9.<br />

11. Lavage fluids are gently shaken to disperse mucous globules <strong>and</strong> pipetted into<br />

Eppendorf tubes. <strong>Sample</strong>s are frozen at –20°C until analysis is performed.<br />

3.2. Nasal Lavage Fluid <strong>Preparation</strong><br />

3.2.1. Total Protein Assay<br />

1. Measure the total protein concentration in each sample using modified Lowry’s<br />

method (see other chapters in this book).<br />

2. Place st<strong>and</strong>ard human albumin or real samples (10 μL) in triplicate in polystyrene<br />

microtiter plates, <strong>and</strong> add assay reagents.<br />

3. Measure the optical densities (650 nm) with a microtiter-plate reader.<br />

4. Interpolate the protein concentrations in the samples from the regression analysis<br />

of the st<strong>and</strong>ard curve (protein concentration in normal NLF: 1st series 878 μg/mL<br />

<strong>and</strong> 2nd series 1,700 μg/mL; protein concentration in sinusitis NLF: day 1 1st<br />

series 1,321 μg/mL <strong>and</strong> 2nd series 1,512 μg/mL, <strong>and</strong> day 6 1st series 638 μg/mL<br />

<strong>and</strong> 2nd series 725 μg/mL).


<strong>Preparation</strong> of Nasal Secretions for Proteome Analysis 81<br />

3.2.2. Acid-Ethanol Precipitation<br />

1. 30 μL of nasal lavage fluid are mixed with an equal volume of 50% ethanol, 50%<br />

0.2 N acetic acid, 0.02% sodium bisulfite.<br />

2. Leave the protein to precipitate at –20°C overnight (see Note 4). <strong>Sample</strong>s are<br />

stable in acid-ethanol solution for long time if stored at –20°C.<br />

3. Centrifuge the mixture for 30 min at 4°C. The supernatant containing endogenous<br />

peptides, lipids <strong>and</strong> sugars is discarded.<br />

3.2.3. Protein Digestion<br />

1. Prepare a fresh solution of 0.1 M ammonium bicarbonate pH 7.8.<br />

2. Dissolve the protein pellet in 10 μL of 0.1 M ammonium bicarbonate pH 7.8<br />

vortex until the protein pellet is dissolved.<br />

3. Dissolve 25 μg of trypsin in 25 μL of ammonium bicarbonate buffer (see Note 5).<br />

4. Trypsin is added to the samples in trypsin:protein ratio of 1:20 (w/w).<br />

5. Incubate the solution at 37°C overnight.<br />

6. Inactivate the trypsin by adding 1 μL of 0.1% formic acid.<br />

3.3. Analyis of Nasal Lavage Fluid <strong>Preparation</strong>s by Liquid<br />

Chromatography Coupled to Mass Spectrometry<br />

Electrospray has the advantage of ionizing macromolecules in a liquid. The<br />

ions observed are formed by addition of proton (hydrogen ion) to give the [M+H]<br />

ion in which M = analyte molecule, H = hydrogen ion. For large macromolecules<br />

(such as peptides) there will often be a distribution of many charge states.<br />

1. Same amounts of tryptic peptides are injected into a capillary liquid chromatography<br />

(CapLC) system after testing the LC system (see Note 6).<br />

2. Peptide mixture are concentrated <strong>and</strong> desalted on a BioBasic C18 precolumn<br />

applying an isocratic procedure (95% water in 0.2% formic acid (FA)) with a<br />

flow rate of 20 μL/min for 10 min (see Note 7).<br />

Table 1<br />

m/z, charge state, mean <strong>and</strong> SD of elution time, <strong>and</strong> CV of five chosen tryptic<br />

peptides from albumin from NLF.<br />

m/z Charge state Time (min) (X+±) CV (%)<br />

682.38 +3 47±0.9 1.8<br />

812.41 +2 61±0.9 1.5<br />

693.82 +2 43±1.2 2.8<br />

671.83 +2 47±1.3 2.8<br />

575.32 +2 34±0.9 2.6


82 Casado et al.<br />

Fig. 1. Total ion chromatogram (TIC) profile of nasal lavage fluid.<br />

Fig. 2. The histograms showing the proteins tabulated according biological function<br />

<strong>and</strong> origin, found in sinusitis NLFs pre- (Day1) <strong>and</strong> post- (Day6) pharmacological<br />

treatment.


<strong>Preparation</strong> of Nasal Secretions for Proteome Analysis 83<br />

3. The peptides are separated on a Zorbax C18 column (100 mm x 150 μm I.D.)<br />

using a gradient from 95% water in 0.2% FA to 95% acetonitrile in 0.2% FA<br />

over 100 min. The flow is set at 10 μL/min. A splitter is used to carry 1 μL/min<br />

on the analytical column (see Note 8).<br />

4. Separated peptides are analyzed using Electrospray-Quadrupole-Time of Flight<br />

mass spectrometer. The samples are run in duplicate. The reproducibility of elution<br />

times is determined by comparison with the retention time of five tryptic peptides<br />

of endogenous albumin (Table 1). Fig. 1 shows a total ion chromatogram (TIC)<br />

profile of nasal lavage fluid (see Note 9).<br />

Table 2<br />

Keratins from normal NLF are reported. We detect type I <strong>and</strong> type II reflecting<br />

the spectrum of cutaneous, transitional, <strong>and</strong> type I <strong>and</strong> II form heterodimers in<br />

intermediate fibers respiratory mucosal cells. Sinusitis had a more limited<br />

spectrum with k1, k5, k6a, k6f, k10, <strong>and</strong> k13<br />

Keratin Epithelium Location<br />

k1–k2 <strong>and</strong> k9–k10 epidermis anterior nares<br />

k6, k4, k16 terminally differentiated<br />

squamous cells<br />

anterior nares<br />

k1–k10 hairs nasal vestibule<br />

k25 inner root sheaths<br />

k6, k16 outer root sheath’s inner layer<br />

k5–k14 outer layer <strong>and</strong> sebaceous<br />

gl<strong>and</strong>s<br />

k8–k18, k7, k19 transitional cuboidal<br />

epithelium <strong>and</strong><br />

pseudoatratified respiratory<br />

epithelium<br />

nasopharynx<br />

k5–k14 basal cells (progenitors of<br />

respiratory epithelium<br />

from k5–k14 to k1–k10 differentiation of<br />

pseudostratified respiratory<br />

epithelial suprabasal cells<br />

k6, k16 or (k17) wet stratified squamous<br />

epithelial lining epithelial<br />

oral <strong>and</strong> esophageal<br />

invaginations of submucosal<br />

gl<strong>and</strong>s <strong>and</strong> ducts<br />

mucoseae<br />

k5–k14 myoepithelial cells surround<br />

submucosal gl<strong>and</strong>s


84 Casado et al.<br />

3.4. Data Interpretation<br />

1. The protein identification can be performed using MASCOT MS/MS ion search<br />

software (see Note 10).<br />

2. If the spectra are manually sequenced, the new peptide sequence can be matched<br />

to a protein using peptide match program in the protein identification resource<br />

(PIR, www.pir.georgetown.edu).<br />

3. PIR BLAST similarity search is used to search unknown protein query sequences.<br />

4. Protein sequence alignments are constructed using the CLUSTAW program<br />

in PIR.<br />

5. The identified proteins are compared on the basis of their origin <strong>and</strong> function<br />

(26). In Fig. 2, the proteins identified before <strong>and</strong> after pharmacological treatment<br />

were grouped according the biological function <strong>and</strong> origin. All the inflammatory<br />

proteins were identified only on Day 1 <strong>and</strong> not on Day 6. We can hypothesize that<br />

the treatment was successful <strong>and</strong> blocked the influx of inflammatory cells (e.g.,<br />

IL-16 <strong>and</strong> IL-17E), the generation of their mediators (e.g., TGF- 2 receptor),<br />

vascular permeability, <strong>and</strong> gl<strong>and</strong>ular hypersecretion. On Day 1, keratins associated<br />

with respiratory epithelium <strong>and</strong> the squamous metaplasia present in sinusitis were<br />

detected. Different actin protein (actin , 1, <strong>and</strong> 2) reflect the hyperplasia<br />

of the respiratory epithelium. Keratin proteome in normal NLF reflected the<br />

anticipated normal type of basal, pseudostratified, respiratory, gl<strong>and</strong>ular, <strong>and</strong><br />

stratified nonkeratinized <strong>and</strong> keratinized squamous epithelium that is present in<br />

the nose. A large number of keratins have been detected in normal NLF then in<br />

sinusitis NLF. Keratin profile in sinusitis NLF was consistent with desquamation<br />

of terminally differentiated cells <strong>and</strong> the presence of squamous metaplasia. The<br />

results demonstrate the changes that are taking place in respiratory epithelial cells<br />

during inflammation (Table 2).<br />

4. Notes<br />

1. The subjects pressed their left nostrils closed, <strong>and</strong> then spritzed 12 sprays of<br />

1× NS into their right nostrils.<br />

2. Because the amount of nasal secretions blown out from left <strong>and</strong> right nostrils<br />

are different, it is important to mix the two samples together. Because both<br />

nostrils are not always simultaneously closed or open, this situation may cause<br />

a different pattern of proteins. It is very important to mix the samples as,<br />

mixing the specimens from the two nostrils, the sample homogeneity is strongly<br />

improved. Big globules of mucus must be dissolved to liberate the proteins in<br />

the mucous net.<br />

3. Hypertonic saline stimulates the gl<strong>and</strong>ular secretions, local mucosal substance<br />

P release <strong>and</strong> pain. Because it is a provocation, the presence of a physician is<br />

recommended.<br />

4. Acid-ethanol solution precipitates high molecular weight proteins. Supernatant<br />

contains peptides that can be used for following determinations. Although


<strong>Preparation</strong> of Nasal Secretions for Proteome Analysis 85<br />

proteins precipitate in few hours, it is recommended to carry out the procedure<br />

overnight.<br />

5. If the trypsin stock is not entirely used, we suggest resuspending it in acetic acid<br />

to prevent autodigestion of trypsin.<br />

6. Before injecting the NLF samples, it is important to test the LC system. one<br />

microliter of nine peptide mixtures (neurotensin, angiotensin I, angiotensin 2,<br />

Glu-fibrinopetide B, somatostatin, bradykynin, bombesin, enkephalin, <strong>and</strong><br />

substance P; 1 pmol of each peptide) is injected on LC system. Resolution <strong>and</strong><br />

sensitivity can be checked.<br />

7. During peptide concentration <strong>and</strong> desalting, small (3–4 amino acids) <strong>and</strong> highly<br />

hydrophilic peptides are washed off from the precolumn. Those losses can<br />

prevent damage of the column.<br />

8. It is important to look carefully at the samples before injecting them on the<br />

CapLC to check if samples are contaminated with mucous globules. This material<br />

in fact can block the capillary causing the damage to the system. For LC-<br />

ESI-MS/MS formic acid replaces trifluoroacetic acid (TFA) in the LC mobile<br />

phase because an efficient ionization is prevented by the strong ion pairing<br />

characteristics of TFA.<br />

9. Duplicate <strong>and</strong> triplicate runs are necessary to examine the reproducibility of the<br />

elution. Two options are available for checking consistency of the chromatographic<br />

system. First, it is possible to spike the sample with one or more st<strong>and</strong>ard<br />

peptides. Second, it is possible to choose tryptic peptides from an endogenous<br />

protein present in the sample. You must know before analyzing your sample<br />

which abundant protein is present in the sample <strong>and</strong> if it is easily cleaved by<br />

the enzyme you are using. Those peptides need to be consistently present in<br />

your runs. We choose the second option <strong>and</strong> we looked at five peptides from<br />

albumin. The mean, st<strong>and</strong>ard deviation of retention time, <strong>and</strong> CV % of the five<br />

peptides are calculated to see how reproducible the experiments are.<br />

10. The program can be found in the web (www.matrixscience.com). There is a<br />

disadvantage in using MASCOT in the web. Only the first 300 peptides can be<br />

searched on the database. Using the nonrestricted MASCOT more information<br />

can be retrieved from the raw data. The following general search parameters<br />

were used: monoisotopic molecular masses, enzyme trypsin, peptide tolerance<br />

of ± 0.4 Da <strong>and</strong> MS/MS tolerance of ± 0.3 Da. The search is restricted to Homo<br />

sapiens species to make the search easier.<br />

References<br />

1. Baraniuk, J. N. (2000) Immunology <strong>and</strong> Allergy Clinics of North America<br />

(Lasley, M. <strong>and</strong> V. Altman, L. C. eds.), Saunders, Philadelphia, pp. 245–64.<br />

2. Baraniuk, J. N., Staevska, M. (2004) Current Therapy in Allergy Immunology <strong>and</strong><br />

Rheumatology (Lichtenstein, L. M., Busse, W. W. <strong>and</strong> Geha, R. S., eds.), Mosby,<br />

Philadelphia, pp. 17–24.


86 Casado et al.<br />

3. Meyer, P., Andersson, M., Persson, C.G., <strong>and</strong> Greiff, L. (2003) Steroid-sensitive<br />

indices of airway inflammation in children with seasonal allergic rhinitis. Pediatr.<br />

Allergy Immunol. 14, 60–65.<br />

4. Dahl, R., <strong>and</strong> Mygind, N. (1998) Mechanisms of airflow limitation in the nose <strong>and</strong><br />

lungs. Clin. Exp. Allergy 28, 17–25.<br />

5. Svensson, C., Andersson, M., Greiff, L., <strong>and</strong> Persson, C.G. (1998) Nasal mucosal<br />

end organ hyperresponsiveness. Am. J. Rhinol. 12, 37–43.<br />

6. Baraniuk, J. N., Ali, M., Yuta, A., Fang, S-Y., <strong>and</strong> Naranch, K. (1999) Hypertonic<br />

saline nasal provocation stimulates nociceptive nerves, substance P release, <strong>and</strong><br />

gl<strong>and</strong>ular mucus exocytosis in normal humans. Am. J. Respir. Crit. Care Med.<br />

160, 655–62.<br />

7. Sanico, A. M., Philip, G., Lai, G. K., <strong>and</strong> Togias, A. (1999) Hyperosmolar saline<br />

induces reflex nasal secretions, evincing neural hyperresponsiveness in allergic<br />

rhinitis. J. Appl. Physiol. 86, 1202–10.<br />

8. Baraniuk, J. N., Clauw, D. J., <strong>and</strong> Gaumond, E. (1998) Rhinitis symptoms in<br />

chronic fatigue syndrome. Ann. Allergy Asthma Immunol. 81, 359–65.<br />

9. Fukuda, K., Straus, S. E., Hickei, I., Sharpe, M. C., Dobbins, J. C., <strong>and</strong> Komaroff, A.<br />

(1994) The chronic fatigue syndrome: a comprehensive approach to its definition<br />

<strong>and</strong> study. Ann. Intern. Med. 121, 953–9.<br />

10. Tabaton, M., Monaco, S., Cordone, M. P., Colucci, M., Giaccone, G., Tagliavini, F.,<br />

<strong>and</strong> Zanusso, G. (2004) Prion deposition in olfactory biopsy of sporadic<br />

Creutzfeldt-Jakob disease. Ann Neurol. 55, 294–6.<br />

11. Sloat, B. R., <strong>and</strong> Cui, Z. (2005) Strong mucosal <strong>and</strong> systemic immunities<br />

induced by nasal immunization with anthrax protective antigen protein incorporated<br />

in liposome-protamine-dna particles. Pharm Res. Dec 6; [Epub ahead<br />

of print]<br />

12. Lindahl, M., Stahlbom, B., <strong>and</strong> Tagesson, C. (1995) Two-dimensional gel<br />

electrophoresis of nasal <strong>and</strong> bronchoalveolar lavage fluids after occupational<br />

exposure. Electrophoresis 16, 1199–1204.<br />

13. Lindahl, M., Stahlbom, B., Svartz, J., <strong>and</strong> Tagesson, C. (1998) Protein patterns of<br />

human nasal <strong>and</strong> bronchoalveolar lavage fluids analyzed with two-dimensional gel<br />

electrophoresis. Electrophoresis 19, 3222–29.<br />

14. Lindahl, M., Stahlbom, B., <strong>and</strong> Tagesson, C. (1999) Newly identified proteins in<br />

human nasal <strong>and</strong> bronchoalveolar lavage fluids: potential biomedical <strong>and</strong> clinical<br />

applications. Electrophoresis 20, 3670–76.<br />

15. Lindahl, M., Svartz, J., <strong>and</strong> Tagesson, C. (1999) Demonstration of different forms<br />

of the anti-inflammatory proteins lipocortin-1 <strong>and</strong> Clara cell protein-16 in human<br />

nasal <strong>and</strong> bronchoalveolar lavage fluids. Electrophoresis 20, 881–90.<br />

16. Ghafouri, B., Stahlbom, B., Tagesson, C., <strong>and</strong> Lindahl, M. (2002) Newly identified<br />

proteins in human nasal lavage fluid from non-smokers <strong>and</strong> smokers using twodimensional<br />

gel electrophoresis <strong>and</strong> peptide mass fingerprinting. Proteomics 2,<br />

112–20.<br />

17. Lindahl, M., Stahlbom, B., <strong>and</strong> Tagesson, C. (2001) Identification of a new<br />

potential airway irritation marker, palate lung nasal epithelial clone protein, in


<strong>Preparation</strong> of Nasal Secretions for Proteome Analysis 87<br />

human nasal lavage fluid with two-dimensional electrophoresis <strong>and</strong> matrix-assisted<br />

laser desorption/ionization-time of flight. Electrophoresis 22, 1795–1800.<br />

18. Lindahl, M., Ir<strong>and</strong>er, K., Tagesson, C., <strong>and</strong> Stahlbom, B. (2004) Nasal lavage fluid<br />

<strong>and</strong> proteomics as means to identify the effects of the irritating epoxy chemical<br />

dimethylbenzylamine. Biomarkers 9, 56–70.<br />

19. Casado, B., Pannell, L. K., Viglio, S., Iadarola, P. et al. (2004) Analysis of<br />

the sinusitis nasal lavage fluid proteome using capillary liquid chromatography<br />

interfaced to electrospray ionization-quadrupole time of flight- t<strong>and</strong>em mass<br />

spectrometry. Electrophoresis 25, 1386–93.<br />

20. Casado, B., Pannell, L. K., Iadarola, P., Baraniuk, J.N. (2005) Identification of<br />

human nasal mucous proteins using proteomics. Proteomics 5, 2949–59.<br />

21. Kristiansson, M. H., Lindh, C. H., <strong>and</strong> Jonsson, B. A. (2003) Determination of<br />

hexahydrophthalic anhydride adducts to human serum albumin. Biomarkers 8,<br />

343–59<br />

22. Kristiansson, M. H., Lindh, C. H., <strong>and</strong> Jonsson, B. A. (2004) Correlations between<br />

air levels of hexahydrophthalic anhydride (HHPA) <strong>and</strong> HHPA-adducted albumin<br />

tryptic peptides in nasal lavage fluid from experimentally exposed volunteers.<br />

Rapid Commun Mass Spectrom. 18, 1592–8.<br />

23. Ali, M., Maniscalco, J., <strong>and</strong> Baraniuk, J. N. (1996) Spontaneous release of submucosal<br />

gl<strong>and</strong> serous <strong>and</strong> mucous cell macromolecules from human nasal explants in<br />

vitro. Am. J. Physiol. 270, L595–L600.<br />

24. Baraniuk, J. N., Silver, P. B., Kaliner, M. A., <strong>and</strong> Barnes, P. J. (1994) Int. Arch.<br />

Allergy Immunol. 103, 202–8.<br />

25. Baraniuk, J. N., Okayama, M., Lundgren, J. D., Mullol, M. et al. (1990) Vasoactive<br />

intestinal peptide in human nasal mucosa. J. Clin. Invest. 86, 825–31.<br />

26. Wu, H. C. H., Huang, H., Yeh, Lai-Su, L., Barker, C. W. (2003) Protein family<br />

classification <strong>and</strong> functional annotation. Comput. Biol. Chem. 27, 37–47.


8<br />

<strong>Preparation</strong> of Urine <strong>Sample</strong>s for Proteomic Analysis<br />

Rembert Pieper<br />

Summary<br />

Reproducible procedures for the preparation of protein samples isolated from human<br />

urine are essential for meaningful proteomic analyses. Key applications are the discovery<br />

of novel proteins or their modifications in the human urine as well as protein biomarker<br />

discovery for diseases <strong>and</strong> drug treatments. The methodology presented here features<br />

experimental steps aimed at limiting protein losses because of organic solvent precipitation,<br />

effective separation of proteins from other compounds in the human urine<br />

<strong>and</strong> molecular weight-based enrichment of proteins in two distinct fractions. Urinary<br />

proteins are separated from cellular debris in the urine via centrifugation, concentrated<br />

with 5-kDa-cutoff membrane concentration devices <strong>and</strong> separated via size exclusion<br />

chromatography into fractions with a higher <strong>and</strong> a lower molecular weight than 30 kDa,<br />

respectively. A successive optional affinity removal step for highly abundant plasma<br />

proteins is described. Finally, buffer exchange steps useful for specific downstream<br />

proteomic analysis experiments of urinary proteins are presented, such as 2-dimensional<br />

gel electrophoresis, differential protein or peptide labeling <strong>and</strong> digestion with trypsin for<br />

LC-MS/MS analysis.<br />

Key Words: Biomarker discovery; gel electrophoresis; human urine; multidimensional<br />

liquid chromatography; proteomic sample preparation; urinary proteome.<br />

1. Introduction<br />

Human urine plays a central role in clinical diagnostics. The human urinary<br />

proteome has been investigated particularly in the context of renal <strong>and</strong> bladder<br />

malfunction <strong>and</strong> cancer (1–6). Under normal physiological conditions, small<br />

protein amounts are excreted with the urinary fluid (0.5–5 mg per voiding),<br />

because the kidney restricts passage of plasma proteins, particularly in the<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

89


90 Pieper<br />

M r range above 40 kDa during the filtration process in the glomeruli. More<br />

protein is lost in diseases, particularly those affecting the kidney, leading to<br />

proteinurea (7). Marshall <strong>and</strong> Williams as well as Anderson et al. pioneered the<br />

research in the characterization of the human urinary proteome in the eighties<br />

<strong>and</strong> nineties including sample preparation methods to isolate proteins from other<br />

matter in the urinary fluid (2,5,8–13). The most frequently used urine sample<br />

preparation methods for proteomic analysis are based on selective protein<br />

precipitation (12–17) or ultrafiltration <strong>and</strong> molecular weight-based enrichment<br />

steps (3,11,15,16,18). Urinary sample preparation should be performed at<br />

4°C <strong>and</strong> in the presence of protease inhibitors to avoid protein degradation.<br />

Cellular debris in urinary fluid should be removed before protein enrichment<br />

to avoid contamination with cellular proteins. Urine concentration is required<br />

to effectively separate proteins in size exclusion chromatography (SEC) experiments<br />

into protein fractions of distinct M r ranges. Immunoaffinity subtraction<br />

(IAS) permits the selective removal of highly abundant plasma proteins in<br />

urine concentrates <strong>and</strong> enrichment of lower abundance urinary proteins (3,19).<br />

Concentrated or lyophilized urinary protein samples are eventually prepared in<br />

buffers compatible with a variety of proteomic analysis techniques.<br />

2. Materials<br />

2.1. Urinary Protein Concentrate <strong>Preparation</strong><br />

1. 250-mL conical bottom polypropylene centrifugation tubes (Fisher Scientific).<br />

2. CompleteTM protease inhibitor cocktail tablets (Roche, Indianapolis, IN).<br />

3. Centricon ® Plus-80 (5,000 NMWL) centrifugal filter devices (Millipore,<br />

Billerica, MA).<br />

4. Amicon ® Ultra-4 (5,000 NMWL) centrifugal filter devices (Millipore,<br />

Billerica, MA).<br />

5. Swinging bucket rotor with 250-mL conical tube adaptors <strong>and</strong> centrifuge for<br />

velocities up to 4,000g (Beckman-Coulter, Fullerton, CA).<br />

6. Buffer A: 100 mM sodium phosphate, pH 7.0, 150 mM NaCl, 0.02% sodium<br />

azide.<br />

7. BCA assay reagents (Pierce Chemicals, Rockford, IL).<br />

2.2. Size Exclusion Chromatography of Urinary Protein Concentrates<br />

1. HiLoad 16/60 Superdex 75 prep grade column (GE Healthcare, Piscataway, NJ).<br />

2. Liquid chromatography system (FPLC) with fraction collector adjustable to 4°C.<br />

3. Centricon ® Plus-20 (5,000 NMWL) centrifugal filter devices (Millipore,<br />

Billerica, MA).<br />

4. Broad range gel filtration st<strong>and</strong>ard (Bio-Rad, Hercules, CA) (see Note 1).<br />

5. Buffer B: 25 mM ammonium bicarbonate, 1 mM benzamidine, 1 mM Na-EDTA.


<strong>Preparation</strong> of Urine <strong>Sample</strong>s for Proteomic Analysis 91<br />

2.3. Immunoaffinity Subtraction of Proteins<br />

1. Multiple Affinity Removal Spin Cartridge for the Depletion of High Abundance<br />

Proteins in Human Serum (Agilent Technologies) or Vivapure Anti-HSA/IgG<br />

Removal Kit (Sartorius AG) or Affinity Depletion Cartridge for Removal of HSA<br />

<strong>and</strong> Immunoglobulins from Human Serum (Applied Biosystems).<br />

2. Elution buffer: 0.5% glycine, 0.25% CHAPS, 150 mM NaCl, 2M urea, pH 2.5.<br />

3. Ultrafree ® -CL 0.45-μm centrifugal filter devices, (Millipore, Billerica, MA)<br />

2.4. Final <strong>Sample</strong> <strong>Preparation</strong> for Proteomic Analysis<br />

1. IPG rehydration solution: 8M urea, 2M thiourea, 4% CHAPS, 18 mM DTT<br />

<strong>and</strong> 0.5% Bio-Lyte ® pH 3–10 carrier ampholytes (Bio-Lyte ® is from Bio-Rad,<br />

Hercules, CA).<br />

2. Freeze-dry/lyophilization unit (vacuum pump, evacuable centrifuge, cold trap).<br />

3. Methods<br />

An overview of sample preparation <strong>and</strong> fractionation steps for urinary<br />

proteins is provided in Fig. 1. The schematic also shows downstream applications<br />

for urinary proteome analysis.<br />

3.1. Urinary Protein Concentrate <strong>Preparation</strong><br />

1. The urine sample is collected, e.g., from a patient in a clinical laboratory, <strong>and</strong><br />

transferred into a 250-mL tube with a conical bottom. It is cooled on ice <strong>and</strong><br />

two CompleteTM protease inhibitor cocktail tablets are added to minimize protein<br />

degradation. The sample tube is centrifuged at 3,000g for 60 min at 4°C (see<br />

Note 2). In order not to disturb the precipitate, the supernatant is pipeted carefully<br />

into a new polypropylene tube. It can be frozen <strong>and</strong> stored for days at –80°C<br />

or processed immediately. The precipitate containing cellular debris <strong>and</strong> other<br />

insoluble matter is discarded.<br />

2. The supernatant is transferred to a Centricon ® Plus-80 device <strong>and</strong> spun at 3,000g<br />

at 4°C, until the urine sample volume is reduced to approx 4–5 mL (see Note 3).<br />

This sample is collected <strong>and</strong> transferred into an Amicon ® Ultra-4 centrifugal filter<br />

device. It is concentrated to 1 mL by spinning at 4,000g at 4ºC, rediluted with<br />

buffer A to 4 mL <strong>and</strong> reconcentrated to approx 500 μL.<br />

3. The protein concentrate is transferred to a 1.5-mL microtube <strong>and</strong> usually has<br />

a brownish color. It can be frozen <strong>and</strong> stored for days at -80°C or processed<br />

immediately.<br />

4. The urinary protein concentrate is spun at 10,000g for 15 min at 4°C. The<br />

supernatant of the centrifugation step is recovered <strong>and</strong> the pellet discarded. The<br />

protein amount is measured <strong>and</strong> the sample is ready to be subjected to the size<br />

exclusion chromatography experiment.


92 Pieper<br />

Fig. 1. Overview of urinary protein sample preparation procedures. 1. Removal of<br />

precipitates via centrifugation at 3,000g; 2. Concentration of urine in Centricon ® Plus-<br />

80 centrifugal filter devices; further concentration in Amicon ® Ultra-4 centrifugal filter<br />

devices; 3. Size exclusion chromatography (Superdex 75) generating two fraction pools<br />

(SEC ≤30 kDa fraction <strong>and</strong> >30 kDa fraction); 4. Reconcentration of SEC samples;<br />

5. Immunoaffinity subtraction generating the IAS >30 kDa fraction; 6. Final sample<br />

concentration in Amicon ® Ultra-4 centrifugal filter devices. Downstream applications<br />

for proteomic analysis: 2-DE gel electrophoresis; digestion with trypsin (followed<br />

by LC-MS/MS analysis); LC separation of urinary proteins; covalent (isotope-coded)<br />

labeling of urinary proteins for differential quantitation using MS methods.<br />

5. For protein quantitation using the BCA assay, 1- or 2-μL aliquots are transferred<br />

to a microtiter plate, incubated for 10 min with 100 μL BCA solution at 37ºC <strong>and</strong><br />

measured in a spectrophotometer at =A 562. In parallel, a BCA assay st<strong>and</strong>ard<br />

curve with concentrations of 0.25 to 2 mg/mL bovine serum albumin is generated<br />

to calculate the total protein amount in the urinary sample.<br />

3.2. Size Exclusion Chromatography of Urinary Protein Concentrate<br />

1. The 16/60 Superdex 75 column is equilibrated in buffer A using an FPLC system<br />

at 4ºC in a cold cabinet (see Note 4). Once a stable baseline is observed monitoring<br />

UV light absorption at =A280, the initial experiment pertains to the molecular


<strong>Preparation</strong> of Urine <strong>Sample</strong>s for Proteomic Analysis 93<br />

weight (M r) column calibration. This experiment should be repeated on separate<br />

days to ascertain reproducibility.<br />

2. 100 μL (approx 3.5 mg protein) of the Bio-Rad gel filtration st<strong>and</strong>ard (M r range<br />

from 670 to 1.4 kDa) are loaded into the sample loop. The flow rate for the LC<br />

separation is 0.5 mL/min <strong>and</strong> fractions are eluted in volumes of approx 4–5 mL.<br />

As shown in the chromatogram of Fig. 2A, resolved LC peaks appear for the gel<br />

filtration st<strong>and</strong>ard proteins with the exception of the two high M r proteins (670<br />

<strong>and</strong> 150 kDa), which elute as a double peak.<br />

3. Using an X/Y scatter diagram with the M r units in logarithmic scale, the graphic<br />

display of M r values <strong>and</strong> elution volumes should yield a nearly linear fit <strong>and</strong><br />

enable the determination of the elution volume corresponding to the M r of 30<br />

Fig. 2. Size exclusion chromatography of urinary protein concentrates on a Superdex<br />

75 column. Chromatogram A: Bio-Rad gel filtration st<strong>and</strong>ard with thyroglobulin (670<br />

kDa) <strong>and</strong> Ig G (150 kDa) in double peak 1, ovalbumin (45 kDa) in peak 2, myoglobin<br />

(18 kDa) in peak 3 <strong>and</strong> vitamin B12 (1.4 kDa) in peak 4. B <strong>and</strong> C: two urinary protein<br />

concentrates, fractions 3–7: SEC >30 kDa sample pool; fractions 8–14: SEC ≤ 30 kDa<br />

sample pool. The UV 280 traces were monitored. This Figure has been reproduced with<br />

permission 3 .


94 Pieper<br />

kDa (see Note 5). The fraction number corresponding to this elution volume is<br />

determined.<br />

4. After the protein concentration measurement of urinary samples using the BCA<br />

assay, the sample volume equivalent to an amount of 4 mg protein is determined.<br />

If the 500 μL urinary concentrate contains more than 4 mg protein, the appropriate<br />

sample volume is aliquoted <strong>and</strong> re-diluted to 500 μL with buffer A, while freezing<br />

the remaining concentrate. If there is less than 4 mg protein, all of the urinary<br />

protein concentrate is applied to the LC experiment. This sample is kept on ice<br />

before application to the SEC experiment.<br />

5. Using the same LC column, LC method, <strong>and</strong> fraction collector settings, urinary<br />

protein concentrates are loaded onto the 16/60 Superdex 75 SEC column <strong>and</strong><br />

fractionated. As shown in the chromatograms of Fig. 2B <strong>and</strong> C for two different<br />

urinary protein samples, A 280 elution traces may vary from sample to sample.<br />

6. The fractions should be placed on ice after collection <strong>and</strong> combined into two<br />

fraction pools: (1) the fraction pool with proteins corresponding to a M r higher<br />

than 30 kDa (SEC >30 kDa) <strong>and</strong> (2) the fraction pool with proteins corresponding<br />

toaM r equal to <strong>and</strong> lower than 30 kDa (SEC ≤30 kDa). The 30 kDa M r fraction<br />

itself is added to the latter fraction pool. No fractions are collected in the baseline<br />

area (A 280 = 0), usually for fractions collected before the LC peak for the 670/150<br />

kDa gel filtration st<strong>and</strong>ards <strong>and</strong> after elution of the LC peak for the 1.4 kDa<br />

st<strong>and</strong>ard (peaks 1 <strong>and</strong> 4 in Fig. 2A, respectively).<br />

7. The two urinary protein sample pools are concentrated in Centricon ® Plus-20<br />

units to approx 1 mL. If fraction pool volumes are larger than 20 mL, concentrate<br />

in a stepwise process in the same Centricon ® tubes. Protein amounts in the SEC<br />

>30 kDa <strong>and</strong> SEC ≤30 kDa urinary protein concentrates are measured using<br />

the BCA assay as described under Section 8.3.1. step 5 <strong>and</strong> are frozen at –80°C.<br />

3.3. Immunoaffinity Subtraction of the SEC >30 kDa Fraction<br />

1. An optional fractionation step is the enrichment of less abundant urinary proteins<br />

depleting highly abundant plasma proteins via immunoaffinity subtraction (IAS).<br />

A detailed protocol for plasma protein depletion is provided in a different chapter<br />

in this book.<br />

2. Removal of highly abundant plasma proteins such as albumin <strong>and</strong> immunoglobulins<br />

(Ig) is desirable to increase the dynamic range for protein detection <strong>and</strong><br />

quantitation in downstream proteomic analyses. Most abundant plasma proteins<br />

have Mr values higher than 30 kDa <strong>and</strong> are present in the SEC >30 kDa urinary<br />

fraction. This is the fraction to be subjected to IAS (see Note 6).<br />

3. Different commercial products are available to deplete abundant plasma proteins<br />

from serum or urine <strong>and</strong> may be available in batch <strong>and</strong>/or as sealed cartridges<br />

(see Note 7).<br />

4. This short protocol describes depletion with IAS resin in batch form. Approx<br />

0.5–1 mL resin (available in batch or removed from a cartridge) is suspended<br />

in 1 mL buffer A <strong>and</strong> placed in a approx 5-mL microtube. The SEC >30 kDa


<strong>Preparation</strong> of Urine <strong>Sample</strong>s for Proteomic Analysis 95<br />

fraction (500 μL) is added to the suspended IAS resin <strong>and</strong> incubated for 15 min<br />

at room temperature. During the incubation step, the suspension is occasionally<br />

agitated by gently pipeting.<br />

5. The suspension is transferred to an Ultrafree-CL 0.45-μm filter device <strong>and</strong><br />

centrifuged at 1,000g for 1–2 min. The filtrate (bottom tube) is collected <strong>and</strong><br />

placed in an Amicon ® Ultra-4 centrifugal filter device. The resin is resuspended<br />

gently in 1.5 mL buffer A, briefly equilibrated, spun again at 1,000g for 1–2 min<br />

<strong>and</strong> the filtrate added to the Amicon ® Ultra-4 tube. This step is repeated <strong>and</strong><br />

the three combined filtrates are concentrated to approx 250 μL by spinning the<br />

Fig. 3. Protein spot profiles in 2-DE gels in a IAS >30 kDa fraction (top gel) <strong>and</strong><br />

a SEC ≤ 30 kDa fraction (bottom gel). <strong>Sample</strong>s with approx 200 μg protein were<br />

loaded in each IEF tube gel. In the first dimension, proteins were focused in the pI<br />

range between 4 <strong>and</strong> 7 applying 25,000 Volt-hours (Vh). The tube gel was stacked<br />

on an 8–15 %T second-dimension slab gel <strong>and</strong> proteins were resolved in the M r<br />

range between 8 <strong>and</strong> 200 kDa over 1,300 Vh. Protein spots in the gels were stained<br />

with Coomassie Brilliant Blue G. Spot trains denoted in the gels are: (1) albumin*;<br />

(2) (-1-acid glycoprotein*; (3) Ig light chains*; (4) prostagl<strong>and</strong>in H2 D-isomerase;<br />

(5) (-1-microglobulin. *These proteins were of very high abundance in the SEC >30<br />

kDa fraction <strong>and</strong> mostly removed via IAS. Spot trains for Ig light chains (3) <strong>and</strong><br />

prostagl<strong>and</strong>in H2 D-isomerase (4) overlap in the bottom gel.


96 Pieper<br />

Amicon ® Ultra-4 device at 4,000g at 4°C. This concentrate is the IAS >30 kDa<br />

(flow-through) fraction.<br />

6. To recycle the resin, it is resuspended in 2 mL elution buffer in the Ultrafree-CL<br />

device at room temperature, equilibrated for 2–3 min <strong>and</strong> spun at 1,000g for<br />

1–2 min. The resin re-suspension <strong>and</strong> elution step is repeated. The eluates are<br />

discarded <strong>and</strong> the resin is immediately neutralized with 4 mL buffer A <strong>and</strong> spun<br />

at 1,000g for 1–2 min. The resin is resuspended in a small volume of buffer A<br />

(0.5–1 mL) <strong>and</strong> stored in suspension at 4°C. The resin can be reused for IAS<br />

using another urinary protein SEC >30 kDa fraction (see Note 8).<br />

3.4. Final Urinary Protein <strong>Sample</strong> <strong>Preparation</strong><br />

for Proteomic Analysis<br />

1. The SEC ≤30 kDa fraction <strong>and</strong> the SEC >30 kDa fraction, if not processed via<br />

IAS, were stored at –80°C. After thawing, they are transferred to Amicon ® Ultra-4<br />

filter devices <strong>and</strong> concentrated to approx 250 μL at 4°C spinning at 4,000g, as<br />

described for the IAS >30 kDa fraction in Section 3.3. step 5.<br />

2. Final protein amounts are determined using the BCA assay as described in<br />

Section 3.1. step 5.<br />

3. A 40-fold buffer exchange in the Amicon ® Ultra-4 follows rediluting <strong>and</strong> reconcentrating.<br />

For 250 μL, the total exchange buffer volume is therefore 10 mL, which<br />

is added stepwise to the sample in the Amicon ® Ultra-4 tube (4 mL capacity).<br />

4. For the preparation of 2-DE gel samples <strong>and</strong> for further LC separation steps,<br />

buffer B is used as the exchange buffer. The 2-DE gels in Fig. 3 display the spot<br />

profiles for two urinary protein fractions. To prepare urinary protein samples for<br />

trypsin digestion, 25 mM ammonium bicarbonate is used as the exchange buffer.<br />

These protein samples are lyophilized for 24 h <strong>and</strong> the protein amounts of the<br />

freeze-dried samples are noted.<br />

5. For the preparation of samples in which proteins are to be covalently labeled, e.g.,<br />

with isotope-coded amine-reactive tags for differential MS analysis, the exchange<br />

buffer is 50 mM HEPES, pH 7.8 <strong>and</strong> the volume is reduced to obtain a final<br />

protein concentration of approx 5 mg/mL.<br />

4. Notes<br />

1. The gel filtration st<strong>and</strong>ard contains thyroglobulin (670 kDa), bovine -globulin<br />

(150 kDa), chicken ovalbumin (45 kDa), equine myoglobin (18 kDa), <strong>and</strong> vitamin<br />

B12 (1.4 kDa) <strong>and</strong>, once reconstituted in 500 μL water, has a protein concentration<br />

of 36 mg/mL.<br />

2. Under ideal circumstances, human urine specimens collected in clinical laboratories<br />

should be cooled on ice <strong>and</strong> supplemented with protease inhibitor tablets


<strong>Preparation</strong> of Urine <strong>Sample</strong>s for Proteomic Analysis 97<br />

(Complete TM ) immediately. Centrifugation at 3,000g to remove cellular debris<br />

should also occur in the clinical laboratory right after sample cooling. The resulting<br />

supernatant can be frozen at –80°C <strong>and</strong> shipped on ice.<br />

3. The 200–250 mL urinary fluid is added stepwise to the same Centricon ® Plus-80<br />

device (capacity of 80 mL). If precipitation occurs during the concentration step,<br />

the collected <strong>and</strong> concentrated sample is aliquoted into three 1.5-mL microtubes<br />

<strong>and</strong> spun for 15 min at 10,000g.<br />

4. It is ideal to equilibrate <strong>and</strong> run the Superdex 75 column at 4ºC. If a cold cabinet<br />

or a cooling system is not available, the LC experiments (gel filtration st<strong>and</strong>ard,<br />

urinary protein samples) can also be performed at room temperature. At room<br />

temperature, 1 mM benzamidine <strong>and</strong> 1 mM EDTA should be added to buffer A.<br />

It is important to equilibrate the reservoir with buffer A <strong>and</strong> the chromatography<br />

column to the same temperature for LC separation of the samples (either at 4ºC<br />

or at room temperature).<br />

5. A less ideal alternative is to approximate the 30 kDa elution volume as the<br />

fraction located equidistantly between the 45 kDa (ovalbumin) <strong>and</strong> the 18 kDa<br />

(myoglobin) LC peaks.<br />

6. Detailed protocols for the immunoaffinity subtraction (IAS) technology are<br />

described in a separate chapter in this book. In the human urine, several blood<br />

plasma proteins are frequently observed as highly abundant proteins (3). These<br />

proteins are human albumin (approx 67 kDa), IgG (approx 150 kDa) <strong>and</strong> -1-acid<br />

glycoprotein (approx 42 kDa). However, their concentrations are known to vary<br />

dramatically in human urine, sample- <strong>and</strong> donor-dependent fashion. Other proteins<br />

frequently of high abundance in human urine are prostagl<strong>and</strong>in H2 d-isomerase<br />

(approx 25 kDa) <strong>and</strong> -1-microglobulin (approx 31 kDa).<br />

7. Three simple-to-use IAS cartridges for the removal of plasma proteins are<br />

available (see Materials). Albumin <strong>and</strong> IgG are subtracted by all three<br />

immunoaffinity removal cartridges. For additional plasma protein subtraction<br />

specificities, further information should be obtained from the cartridge manufacturer.<br />

In a proteomic experiment comparing several urinary protein samples, only<br />

one of the IAS resin products should be used. The methods for the use of an IAS<br />

LC cartridge are different. Commercially available IAS LC resin cartridges (with<br />

a volume greater than 0.5 mL) are typically sufficient for high-abundance plasma<br />

protein removal from a SEC >30 kDa fraction with up to 3 mg total urinary protein.<br />

8. The described protocol works particularly well with protein A- or protein<br />

G-derivatized resins in which proteins A/G are cross-linked to plasma proteinspecific<br />

polyclonal antibodies via dimethylpimelimidate. In particular, this<br />

pertains to the elution <strong>and</strong> resin recycling steps which allow for effective elution<br />

of affinity-bound proteins, retain the cross-linkage <strong>and</strong> maintain the binding<br />

functions of the immobilized antibodies. Whether the elution buffer is compatible<br />

with the IAS resins of all depletion cartridges (based on cross-linkage chemistry<br />

between resin <strong>and</strong> antibodies) should be verified with the manufacturers. An<br />

alternative elution buffer provided by the manufacturer may be used for the<br />

recycling of the resin.


98 Pieper<br />

References<br />

1. Celis, J. E., Wolf, H., <strong>and</strong> Ostergaard, M. (2000) Bladder squamous cell carcinoma<br />

biomarkers derived from proteomics. Electrophoresis 21, 2115–21<br />

2. Edwards, J. J., Anderson, N. G., Tollaksen, S. L., von Eschenbach, A. C.,<br />

<strong>and</strong> Guevara, J., Jr. (1982) Proteins of human urine. II. Identification by twodimensional<br />

electrophoresis of a new c<strong>and</strong>idate marker for prostatic cancer. Clin<br />

Chem 28, 160–63<br />

3. Pieper, R., Gatlin, C. L., McGrath, A. M., et al. (2004) Characterization of the<br />

human urinary proteome: a method for high-resolution display of urinary proteins<br />

on two-dimensional electrophoresis gels with a yield of nearly 1400 distinct protein<br />

spots. Proteomics 4, 1159–74<br />

4. Saito, M., Kimoto, M., Araki, T., et al. (2005) Proteome analysis of gelatin-bound<br />

urinary proteins from patients with bladder cancers. Eur Urol 48, 865–71<br />

5. Williams, K. M., Williams, J., <strong>and</strong> Marshall, T. (1998) Analysis of Bence Jones<br />

proteinuria by high resolution two-dimensional electrophoresis. Electrophoresis<br />

19, 1828–35<br />

6. Decramer, S., Wittke, S., Mischak, H., et al. (2006) Predicting the clinical outcome<br />

of congenital unilateral ureteropelvic junction obstruction in newborn by urinary<br />

proteome analysis. Nat Med 12, 398–400<br />

7. Waller, K. V., Ward, K. M., Mahan, J. D., <strong>and</strong> Wismatt, D. K. (1989) Current<br />

concepts in proteinuria. Clin Chem 35, 755–765<br />

8. Anderson, N. G., Anderson, N. L., <strong>and</strong> Tollaksen, S. L. (1979) Proteins of human<br />

urine. I. Concentration <strong>and</strong> analysis by two-dimensional electrophoresis. Clin Chem<br />

25, 1199–1210<br />

9. Edwards, J. J., Tollaksen, S. L., <strong>and</strong> Anderson, N. G. (1982) Proteins of human<br />

urine. III. Identification <strong>and</strong> two-dimensional electrophoretic map positions of some<br />

major urinary proteins. Clin Chem 28, 941–48<br />

10. Marshall, R. J., Turner, R., Yu, H., <strong>and</strong> Cooper, E. H. (1984) Cluster analysis of<br />

chromatographic profiles of urine proteins. J Chromatogr 297, 235–44<br />

11. Marshall, T., <strong>and</strong> Williams, K. M. (1993) Centriprep ultrafiltration for fractionation<br />

of serum <strong>and</strong> urinary proteins before electrophoresis. Clin Chem 39, 1558<br />

12. Marshall, T., <strong>and</strong> Williams, K. (1996) Two-dimensional electrophoresis of human<br />

urinary proteins following concentration by dye precipitation. Electrophoresis 17,<br />

1265–72<br />

13. Marshall, T., <strong>and</strong> Williams, K. M. (1997) Two-dimensional electrophoresis of<br />

human urine <strong>and</strong> cerebrospinal fluid following protein concentration by dye precipitation.<br />

Biochem Soc Trans 25, S657<br />

14. Thongboonkerd, V., Chutipongtanate, S., <strong>and</strong> Kanlaya, R. (2006) Systematic evaluation<br />

of sample preparation methods for gel-based human urinary proteomics:<br />

quantity, quality, <strong>and</strong> variability. J Proteome Res 5, 183–91<br />

15. Thongboonkerd, V., McLeish, K. R., Arthur, J. M., <strong>and</strong> Klein, J. B. (2002)<br />

Proteomic analysis of normal human urinary proteins isolated by acetone precipitation<br />

or ultracentrifugation. Kidney Int 62, 1461–69


<strong>Preparation</strong> of Urine <strong>Sample</strong>s for Proteomic Analysis 99<br />

16. Tantipaiboonwong, P., Sinchaikul, S., Sriyam, S., Phutrakul, S., <strong>and</strong> Chen, S. T.<br />

(2005) Different techniques for urinary protein analysis of normal <strong>and</strong> lung cancer<br />

patients. Proteomics 5, 1140–49<br />

17. Sun, W., Li, F., Wu, S., et al. (2005) Human urine proteome analysis by three<br />

separation approaches. Proteomics 5, 4994–5001<br />

18. Oh, J., Pyo, J. H., Jo, E. H., (2004) Establishment of a near-st<strong>and</strong>ard twodimensional<br />

human urine proteomic map. Proteomics 4, 3485–97<br />

19. Pieper, R., Su, Q., Gatlin, C. L., Huang, S. T., Anderson, N. L., <strong>and</strong><br />

Steiner, S. (2003) Multi-component immunoaffinity subtraction chromatography:<br />

an innovative step towards a comprehensive survey of the human plasma proteome.<br />

Proteomics 3, 422–32


9<br />

Isolation of Cytoplasmatic Proteins from Cultured Cells<br />

for Two-Dimensional Gel Electrophoresis<br />

Ying Wang, Jen-Fu Chiu, <strong>and</strong> Qing-Yu He<br />

Summary<br />

Cytoplasma is the cell interior place between the cellular membrane <strong>and</strong> the nucleus,<br />

where various intracellular activities take place, including energy production, reactive<br />

oxygen species (ROS) detoxification, heme synthesis, nitrogen <strong>and</strong> lipid metabolism,<br />

phosphorylation in signal transduction, <strong>and</strong> cytoskeletal meshwork construction. The rich<br />

cytoplasmatic proteins carrying out these intracellular functions are interesting targets<br />

for biochemical <strong>and</strong> molecular biological studies. The relatively recent discipline of<br />

proteomics offers a chance to globally analyze the changes in cytoplasmic proteins corresponding<br />

to drug treatments or disease conditions, <strong>and</strong> thus provide target c<strong>and</strong>idates for<br />

further biological validation in drug development <strong>and</strong> biomarker discovery. Isolation of<br />

cytoplasmic proteins from cells is a necessary step for high resolution protein separation<br />

by two-dimensional gel electrophoresis (<strong>2D</strong>E) <strong>and</strong> specific proteomic analysis.<br />

Key Words: Cytoplasmatic proteins; protein isolation; proteomics; sample preparation;<br />

subcellular fractionation; two-dimensional gel electrophoresis.<br />

1. Introduction<br />

The cytoplasm is crowded <strong>and</strong> highly ordered with transport vesicles,<br />

mitochondria, chloroplasts, <strong>and</strong> other organelles. The endocytosis <strong>and</strong> exocytosis<br />

in cytoplasm provide paths between the cell interior <strong>and</strong> the surrounding<br />

medium, allowing for the uptake of extracellular components <strong>and</strong> the secretion<br />

of proteins <strong>and</strong> other components produced within the cell. The cytoplasm is<br />

the place for energy metabolism (1), reactive oxygen species (ROS) detoxification<br />

(2), heme synthesis (3), nitrogen <strong>and</strong> lipid metabolism (4,5), mixed<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

101


102 Wang et al.<br />

phospholipids biosynthesis (6), cytoskeleton rearrangements (7), <strong>and</strong> various<br />

protein-protein interactions (8). Mapping the altered expression in cytoplasmic<br />

proteins by proteomic analysis under given conditions can provide a network<br />

of responses to intracellular <strong>and</strong> extracellular signals (9).<br />

Proteomics is a research technique that can identify, characterize, <strong>and</strong><br />

quantitate proteins expressed in cells, tissues, or organisms under given conditions<br />

such as chemotherapeutic drug challenge (10,11). The altered proteins<br />

identified by proteomic analysis can be further characterized as potential drug<br />

targets; the global analysis of the protein alterations can result in valuable<br />

information for underst<strong>and</strong>ing the drug action mechanisms. By comparing the<br />

cytoplasmic protein profiles of HONE1 cells treated by gold(III) porphyrin<br />

1a to untreated control, we identified a number of differentially expressed<br />

proteins by peptide-mass-finger printing (PMF) (12). The identification of the<br />

altered proteins provided valuable clues to illustrate the underlying drug action<br />

mechanisms.<br />

2. Materials<br />

2.1. Cell Culture <strong>and</strong> Wash Buffer<br />

1. RPMI 1640 Medium or Dulbecco’s Modified Eagle’s Medium (DMEM) plus<br />

10% FBS, supplemented with 2 mM/L l-glutamine, 100 units/mL penicillin, <strong>and</strong><br />

100 μg/mL streptomycin.<br />

2. Solution of trypsin (0.25%) <strong>and</strong> ethylenediamine tetraacetic acid (EDTA) (1 mM).<br />

3. Cell washing buffer for <strong>2D</strong>E: 10 mM Tris-HCl, pH 7.0, 250mM sucrose.<br />

4. Teflon cell scrapers.<br />

5. Tissue grinder.<br />

2.2. Buffers <strong>and</strong> Reagents for Cytoplasmic Protein Precipitation<br />

1. Extraction buffer: 10 mM Tris-HCl, pH 7.6, 10 mM KCl, 5 mM MgCl2. 2. Nuclei isolation buffer: 10 mM Tris-HCl, pH 7.6, 10 mM KCl, 5 mM MgCl2, 0.35M sucrose.<br />

3. Radioimmunoprecipitation assay buffer (RIPA): 10 mM Tris-HCl, pH 7.5, 150<br />

mM NaCl, 1% NP 40 (w/v), 0.1% SDS (w/v), 0.5% sodium deoxycholate (w/v)<br />

(see Note 1),1mMDTT. 4. Protease inhibitors (see Note 2): 200 mM stock solution of phenylmethanesulfonyl<br />

fluoride (PMFS) in isopropanol (store at room temperature); 1 mg/mL leupeptin<br />

in water (store frozen in aliquots), 1 mg/mL aprotinin in water (store frozen in<br />

aliquots), 1 mg/mL pepstatin in methanol (store frozen in aliquots).<br />

5. Phosphatase inhibitors (see Note 3): activated 200 mM stock solution of sodium<br />

vanadate in water <strong>and</strong> 200 mM sodium fluoride stock solution, store at room<br />

temperature.


Isolation of Cytoplasmatic Proteins from Cultured Cells 103<br />

6. Methylene blue solution: 1.4% (w/v) methylene blue in 95% ethanol <strong>and</strong> filtered<br />

through 0.45 μm filter paper.<br />

7. 10% trichloroacetic acid (TCA).<br />

2.3. Buffers for Western Blot Analysis<br />

1. Transfer buffer: 25 mM Tris base, 192 mM glycine, 0.05% SDS (w/v), 20%<br />

methanol.<br />

2. TBS-T: 20 mM Tris-HCl, pH 7.6, 0.15M NaCl, 0.1% (w/v) Tween 20.<br />

3. Stripping buffer: 50 mM glycine, 1 % SDS (w/v), pH 2.0.<br />

4. ECL reagents (GE Healthcare).<br />

2.4. Buffers <strong>and</strong> Reagents for Two-Dimensional Gel<br />

Electrophoresis (<strong>2D</strong>E)<br />

1. <strong>2D</strong>E clean up kit (GE Healthcare).<br />

2. Rehydration solution: 8M urea, 2% CHAPS (stored frozen in aliquots). Add<br />

0.002% bromophenol blue, 0.005% IPG buffer, <strong>and</strong> 2.8 mg/mL DTT freshly<br />

before use.<br />

3. SDS equilibration buffer: 6M urea, 50 mM Tris-HCl, pH 8.8, 30% glycerol, 2%<br />

SDS.<br />

4. 1.5M Tris-HCl (pH 8.8): dissolve 181.7 g Tris base into 750 mL ddH 2O, add 12N<br />

HCl to adjust to pH 8.8 <strong>and</strong> then add ddH 2O to final volume of 1 L, after that,<br />

filtered by 0.45 μm filter paper.<br />

5. Thirty percent acrylamide gel stock solution: 30% acrylamide (w/v), 0.8% N’N’methylenebiasacrylamide<br />

(w/v), <strong>and</strong> filtered by 0.45 μm filter paper.<br />

6. SDS running buffer: 25 mM Tris base, 0.192 M glycine, 0.1% SDS (w/v).<br />

7. Agarose sealing solution: 0.5% agarose (normal or low-melting point), 0.002%<br />

bromophenol blue in SDS running buffer.<br />

8. Bromophenol blue stock solution: 1% (w/v) bromophenol blue powder, 50 mM<br />

Tris-Base, filtered through 0.45 μm filter paper.<br />

9. Silicon oil (e.g., DryStrip ® Cover Fluid from GE Healthcare).<br />

2.5. Silver Staining Solutions<br />

1. Fixation solution: 40% ethanol, 10% acetic acid.<br />

2. Incubation solution: 30% ethanol, 4.1% sodium acetate (anhydrous) (w/v), 0.2%<br />

sodium thiosulfate (anhydrous) (w/v).<br />

3. Silver nitrate solution: 0.1% silver nitrate (w/v), 0.02% formaldehyde (v/v).<br />

4. Development solution: 2.5% sodium-carbonate (w/v), 0.01% formaldehyde.<br />

5. Stop solution: 1.46% sodium-EDTA·2H2O (w/v).<br />

6. Preservation solution: 4.0% glycerol (w/v), 30% ethanol.


104 Wang et al.<br />

2.6. Coomassie Brilliant Blue Staining Solutions<br />

1. Fixation solution: 40% methanol (v/v) <strong>and</strong> 5% phosphoric acid (v/v).<br />

2. Coommasie blue G-250 staining solution: 0.08% Coomassie brilliant blue G-250<br />

(w/v) in 12% trichloroacetic acid, pH < 1.0.<br />

3. Coomassie blue R-250 staining solution: 0.1% Coomassie brilliant blue R-250<br />

(w/v), in 50% methanol (v/v), <strong>and</strong> 10 % acetic acid (v/v).<br />

4. Destaining solution: 15% methanol (v/v), 10% acetic acid (v/v), 7% acetic acid<br />

(optional) (stored up to 1 month at room temperature).<br />

3. Methods<br />

3.1. Cell Harvest (see Note 4)<br />

1. When the attached cells reach about 80% confluence, discard the media <strong>and</strong> wash<br />

two times with ice cold washing buffer (see Note 5), keep 1 mL washing buffer<br />

in the dish.<br />

2. Harvest cells by prechilled cell scrapper, then transfer 1 mL of cell suspension<br />

to a clean 2.0-mL Eppendorf tube, spin down at 3,000g for 5 min (4°C), <strong>and</strong><br />

wash two times with washing buffer, 1 mL each time. Cell pellet should be lysed<br />

immediately for extraction of cytoplasmatic proteins.<br />

3. When suspended cells reach about 80% confluence, spin down at 3,000g for 5<br />

min (4°C), <strong>and</strong> wash two times with ice cold washing buffer, 10 mL for each<br />

plate, <strong>and</strong> spin down to get cell pellet. Cell pellet should be lysed immediately<br />

for extraction of cytoplasmatic proteins.<br />

3.2. Precipitation of Cytoplasmatic Proteins (see Note 6)<br />

1. Re-suspend about 1×107 cells in 1 mL extraction buffer with protease inhibitors<br />

(0.2 mM PMSF, 5 μg/mL leupeptin, 2 μg/mL aprotinin, <strong>and</strong> 2 μg/mL pepstatin).<br />

Incubate the cells on ice for 10 min, <strong>and</strong> lysis by addition of Triton X-100 to the<br />

final concentration of about 0.3% (w/v).<br />

2. Homogenize cells in an ice-cold tissue grinder. 30–50 passes with the grinder are<br />

recommended; however, efficient homogenization may depend on the cell type.<br />

To check the efficiency of homogenization, pipet 2–3 μL of the homogenized<br />

suspension onto a cover slide, stained with methylene blue solution <strong>and</strong> observe<br />

under a microscope. Nuclei appear as dark blue <strong>and</strong> cytoplasm as light purple<br />

blue under white field lens. If about 80% of the nuclei are not surrounded by<br />

cytoplasm, proceed to step 3. Otherwise, perform 10–20 additional passes using<br />

the tissue grinder. Excessive homogenization should also be avoided, as it can<br />

cause damage to the heavy membranes, e.g., mitochondrial membrane, which<br />

triggers release of mitochondrial components (see Note 7).<br />

3. Slowly add 0.5 volume of nuclei isolation buffer to the bottom of extraction buffer,<br />

then remove nuclei by centrifugation at 500–700g for 10 min (swinging-bucket<br />

rotor) (4°C).


Isolation of Cytoplasmatic Proteins from Cultured Cells 105<br />

4. Further centrifuge the supernatant from nuclei isolation at 10,000g for 30 min<br />

(fixed-angle rotor) (4°C). Save the supernatant as cytoplasmatic fraction, <strong>and</strong> the<br />

pellet as heavy membrane fraction containing mitochondria.<br />

5. Add 0.25 volumes of 10% TCA to cytoplasmatic fraction to precipitate cytoplasmatic<br />

proteins; incubate on ice for 10 min. Spin down at 5,000g for 5 min<br />

(4°C). Remove supernatant, leaving protein pellet intact. Wash pellet with 200<br />

μL prechilled acetone. Spin down at 5,000g for 5 min (4°C) (see Note 8). Wash<br />

twice with acetone again. Air dry the pellet, <strong>and</strong> add rehydration solution for <strong>2D</strong>E<br />

analysis (see Note 9). Or add RIPA assay buffer for Western blot analysis to test<br />

the purity of the isolated subcellular fraction.<br />

3.3. Western Blot Analysis to Detect the Purity of the <strong>Sample</strong><br />

1. Determine the purity of the isolated subcellular fractions by Western blot<br />

analysis against specific protein markers (Table 1). Separate the samples by onedimensional<br />

SDS <strong>PAGE</strong>.<br />

Table 1<br />

Marker proteins of individual subcellular fractions.<br />

Subcellular<br />

localization<br />

Marker protein Database<br />

accession<br />

number<br />

References<br />

Mitochondria Cox II gi 142786 (13)<br />

Cox IV gi 142789 (14, 15)<br />

Cytochrome oxidase 1 gi 551699 (16)<br />

VDAC gi 340201 (14, 15, 17)<br />

MnSOD gi 34707 (18, 19)<br />

HSP60 gi 77702086 (12)<br />

mtHSP70 gi 292160 (20)<br />

Cytochrome P450 gi 5733409 (21)<br />

Cytoplasma Tubulin gi 4507729 (22)<br />

GAPDH gi 7669492 (16, 23)<br />

-actin gi 4501885 (12)<br />

LDH gi 9257228 (20)<br />

Calpain gi 791040 (24)<br />

Cytokeratin gi 1419564 (25)<br />

Vimentin gi 62414289 (26)<br />

Nuclei Histone gi 3649600 (18, 19)<br />

c-Jun gi 20986521 (27)<br />

c-Fos gi 29904 (28, 29)


106 Wang et al.<br />

2. Before transfer, soak PVDF membrane in 100% methanol for 1 min, then soak<br />

with transfer buffer.<br />

3. After SDS <strong>PAGE</strong>, transfer the gel to the membrane electrophoretically, using<br />

transfer buffer. Assemble the filter paper, SDS gel <strong>and</strong> membrane into a blotting<br />

s<strong>and</strong>wich as shown in Fig. 1 (see Note 10). The current for transfer should be 0.8<br />

times the area of the membrane in mA.<br />

4. After transfer, incubate the membrane in 5% nonfat milk in TBS-T buffer (4°C<br />

overnight or room temperature for 2 h).<br />

5. Wash the membrane three times with TBS-T buffer (10 min each, room temperature),<br />

followed by incubation with primary antibody, in 1% nonfat milk (4°C<br />

overnight or room temperature for 2 h).<br />

6. Wash the membrane three times by TBS-T buffer (10 min each, room temperature),<br />

then incubate with secondary antibody in 1% nonfat milk (4°C overnight<br />

or room temperature, 45 min to 1 h).<br />

7. Discard the secondary antibody <strong>and</strong> wash the membrane three times by TBS-T<br />

buffer (10 min each, room temperature). Develop the membrane using the ECL<br />

reagents.<br />

8. Once a satisfactory exposure for the results has been obtained, the membrane<br />

is stripped with stripping buffer <strong>and</strong> then reprobed with another antibody. Wash<br />

the membrane twice with TBS-T buffer (10 min each, room temperature), then<br />

incubate the membrane in stripping buffer for 20 min (room temperature), wash<br />

twice with TBS-T again (10 min each, room temperature), then go to steps<br />

4–7. Fig. 2 shows an example of Western blot analysis of specific protein<br />

markers of cytoplasmic <strong>and</strong> heavy membrane fraction from cisplatin treated<br />

HONE1 cells.<br />

Fig. 1. Western Blot assembly.


Isolation of Cytoplasmatic Proteins from Cultured Cells 107<br />

Fig. 2. Western blot analysis of cytoplasmatic fraction obtained from HONE1 cells<br />

treated with cisplatin for 6, 12, <strong>and</strong> 24 h.<br />

3.4. Two-Dimensional Electrophoresis<br />

1. Add 2.8 mg DTT, 5 μL carrier ampholytes or IPG buffer of the respective<br />

pH gradient, <strong>and</strong> 2 μL bromophenol blue to 1 mL rehydration stock solution<br />

(without DTT <strong>and</strong> IPG buffer) immediately before adding the protein samples.<br />

2. Add protein samples to the above mentioned rehydration solution (volume<br />

recommendations are given in Table 2), vortex <strong>and</strong> spin down the samples. The<br />

recommended protein loading is given in Table 3.<br />

3. IPG-strip sample loading by in-gel-rehydration. Pipet the sample solution into<br />

an IPG-strip holding device. Remove the protective cover from the IPG strip,<br />

position the IPG strip with the gel side down. To help coat the entire IPG strip,<br />

gently lift <strong>and</strong> lower the strip <strong>and</strong> slide it back <strong>and</strong> forth along the surface of the<br />

solution. Be careful not to trap bubbles under the IPG strip. Overlay the strips by<br />

Immobiline DryStrip® Cover Fluid (GE Healthcare) to ensure that rehydrated<br />

Table 2<br />

Rehydration solution volume per Immobiline DryStrip<br />

IPG Strip Length (cm) Total volume per strip (μL)<br />

7 125<br />

13 250<br />

18 340


108 Wang et al.<br />

Table 3<br />

Protein loads for silver <strong>and</strong> Coomassie blue staining.<br />

IPG Strip Length (cm) pH-range<br />

Recommended protein load (μg)<br />

Silver stain Coomassie stain<br />

7 4–7 25–50 50–100<br />

3–10, 3–10 NL 50–75 50–150<br />

13 3–10, 3–10 NL 50–100 100–200<br />

18 4–7 200–400 400–1000<br />

3–10, 3–10 NL 100–400 200–1000<br />

Immobiline DryStrip gels do not dry out during electrophoresis. Finally, place<br />

the cover on the strip holder.<br />

4. IEF is performed according to a step-wise voltage increase procedure:<br />

rehydration at 30 V for 10–16 h, followed by 500 V <strong>and</strong> 1000 V for 1 h<br />

each, <strong>and</strong> 5,000–8,000 V for about 10 h with a total of 64,000 V hours<br />

(56,000 V hours is acceptable for 7-cm strips). After IEF, the strips can be<br />

subjected to second dimension immediately or can be kept at –70 ° C for several<br />

weeks.<br />

5. Prepare the 1.5-mm thick polyacrylamide gels. Seal the gel surface by 1-butanol,<br />

allow the gel to be polymerized for at least 30 min at room temperature or<br />

polymerize the gel overnight (see Notes 11 <strong>and</strong> 12).<br />

6. The strips after IEF are subjected to two-step equilibration in equilibration<br />

buffers with 1% DTT (w/v) for the first step, <strong>and</strong> 2.5% (w/v) iodoacetamide for<br />

the second step. Wash strips by SDS running buffer for three times before load<br />

onto the acrylamide gel (see Note 13).<br />

7. Electrophoresis conditions: set buffer circulation temperature to 10ºC <strong>and</strong> start<br />

the run at 15 mA per gel. After 30 min increase current to 30 mA per gel.<br />

8. After SDS page, visualize proteins with silver staining or Coomassie brilliant<br />

blue staining. For silver staining, fix the gels in fixation solution overnight,<br />

<strong>and</strong> then change to incubation solution for 30 min. After washing three times<br />

in water for 10 min each, stain the gels in silver nitrate solution for 40<br />

min. Perform development for 15 min in development solution. Stop staining<br />

by stop solution <strong>and</strong> then wash the stained gels three times in water for<br />

5 min each.<br />

9. Alternatively, visualize the gels by Coomassie brilliant blue stain. Fix the gels<br />

in fixation solution for Coomassie brilliant blue stain overnight, followed by<br />

Coommasie blue G-250 or R-250 stain for more than 12 h. Destain by destaining<br />

solution until background is acceptable.<br />

10. Acquire images by a suitable Image Scanner <strong>and</strong> preserve gels in preservation<br />

solution for further analysis. Fig. 3 shows <strong>2D</strong>E pattern of cytoplasmatic proteins<br />

compared with the pattern of a whole cell protein extract.


Isolation of Cytoplasmatic Proteins from Cultured Cells 109<br />

Fig. 3. <strong>2D</strong>E pattern of cytoplasmatic proteins compared with the pattern of whole<br />

cell protein. One hundred μg of cytoplasmatic protein or whole cell protein from human<br />

nasopharyngeal carcinoma HONE1 cells was separated on 13-cm IPG-strips, pH 3–10,<br />

followed by SDS-<strong>PAGE</strong> (12% SDS gel), <strong>and</strong> visualized by silver staining.<br />

4. Notes<br />

1. Sodium deoxycholate is an ionic detergent to extract proteins. Prepare 10 %<br />

stock solution in water, protect the solution from light. Do not add sodiumdeoxycholate<br />

when preparing lysates for kinase assays. Ionic detergents can<br />

denature enzymes, causing them to lose activity.<br />

2. Commercially available protease inhibitor cocktails can be used instead, for<br />

example, protease inhibitor cocktail (Catalog number P8340) from Sigma-<br />

Aldrich, protease inhibitor cocktail set I (Catalog number 539131) from<br />

Calbiochem.<br />

3. Do not add phosphatase inhibitors when preparing lysates for phosphatase assays.<br />

4. Wash cells intensively to avoid contamination of serum proteins in the culture<br />

medium.<br />

5. Do not wash the cells with PBS in the last washing step, because PBS contains<br />

150 mM sodium chloride, which will interfere with isoelectric focusing. Use<br />

instead 250 mM sucrose, 10 mM Tris-HCl, pH 7.5.<br />

6. Always perform the isolation of cytoplasmic proteins on ice. Chill all buffers<br />

before use. Perform the isolation steps on ice to ensure the purity of subcellular<br />

fractions, because activities of most cellular proteases are inhibited at low<br />

temperature.<br />

7. Under ideal homogenization conditions, particulate organelles such as nuclei,<br />

mitochondria, lysosomes, <strong>and</strong> peroxisomes remain intact. Golgi complexes,<br />

plasma membranes, <strong>and</strong> reticular organelles will fragment <strong>and</strong> can, at least to<br />

some extent, vesiculate. It is not possible to outline a general protocol suitable<br />

for the production of a reasonable homogenate for all kinds of cultured cells,


110 Wang et al.<br />

because different conditions are required for different cell types <strong>and</strong> experimental<br />

purposes.<br />

8. Long centrifuge at high speed will cause difficulties in dissolving protein pellet<br />

in rehydration solution or RIPA assay buffer.<br />

9. <strong>2D</strong>E clean-up kit can be used to improve the quality of <strong>2D</strong>E results by removing<br />

interfering contaminants.<br />

10. Bubbles between the SDS gel <strong>and</strong> the membrane will lead to inefficient transfer.<br />

11. It is critical that all the glass plates for <strong>2D</strong>E have been cleaned <strong>and</strong> rinsed<br />

extensively with distilled water. Clean the glass plates with distilled water<br />

followed by 95% ethanol to remove the acid <strong>and</strong> air-dry prior use.<br />

12. Prepare 12.5% second dimensional SDS gels for st<strong>and</strong>ard separations; prepare<br />

8–10% gels to better separate large molecular weight proteins <strong>and</strong> 15% gels<br />

for low molecular weight proteins. For the equilibration step, add DTT <strong>and</strong><br />

iodoacetamide to equilibration buffer just prior use.<br />

Acknowledgments<br />

This investigation was partially supported by grants from Hong Kong<br />

University funding (No. 200511159099 to Q.Y.H.) <strong>and</strong> the Area of Excellence<br />

Scheme of the Hong Kong University Grants Committee.<br />

Reference<br />

1. Mesecke N., Terziyska N., Kozany C., Baumann F., Neupert W., Hell K.,<br />

Herrmann J. M. (2005) A disulfide relay system in the intermembrane space of<br />

mitochondria that mediates protein import, Cell 121, 1059–69.<br />

2. Ferret P. J., Hammoud R., Tulliez M., et al. (2001) Detoxification of<br />

reactive oxygen species by a nonpeptidyl mimic of superoxide dismutase<br />

cures acetaminophen-induced acute liver failure in the mouse, Hepatology 33,<br />

1173–80.<br />

3. Szaszi K., Jones J. J., Nathens A. B., et al. (2005) Glutathione depletion<br />

inhibits lipopolysaccharide-induced intercellular adhesion molecule 1 synthesis,<br />

Free Radic.Biol.Med. 38, 1333–43.<br />

4. Zou M. H., Kirkpatrick S. S., Davis B. J., et al. (2004) Activation of the AMPactivated<br />

protein kinase by the anti-diabetic drug metformin in vivo. Role of<br />

mitochondrial reactive nitrogen species, J.Biol.Chem. 279, 43940–51.<br />

5. Osawa Y., Uchinami H., Bielawski J., Schwabe R. F., Hannun Y. A., Brenner D. A.<br />

(2005) Roles for C16-ceramide <strong>and</strong> sphingosine 1-phosphate in regulating<br />

hepatocyte apoptosis in response to tumor necrosis factor-alpha, J.Biol.Chem. 280,<br />

27879–87.<br />

6. Nakamura Y., Awai K., Masuda T., Yoshioka Y., Takamiya K., Ohta H. (2005)<br />

A novel phosphatidylcholine-hydrolyzing phospholipase C induced by phosphate<br />

starvation in Arabidopsis, J.Biol.Chem. 280, 7469–76.


Isolation of Cytoplasmatic Proteins from Cultured Cells 111<br />

7. Favoreel H. W., Van Minnebruggen G., Adriaensen D., Nauwynck H. J. (2005)<br />

Cytoskeletal rearrangements <strong>and</strong> cell extensions induced by the US3 kinase of an<br />

alphaherpesvirus are associated with enhanced spread, Proc.Natl.Acad.Sci.U.S.A<br />

102, 8990–5.<br />

8. Yang T., Chaudhuri S., Yang L., Chen Y., Poovaiah B. W. (2004)<br />

Calcium/calmodulin up-regulates a cytoplasmic receptor-like kinase in plants,<br />

J.Biol.Chem. 279, 42552–559.<br />

9. Jiang X. S., Dai J., Sheng Q. H., et al. (2005) A comparative proteomic strategy<br />

for subcellular proteome research: ICAT approach coupled with bioinformatics<br />

prediction to ascertain rat liver mitochondrial proteins <strong>and</strong> indication of mitochondrial<br />

localization for catalase, Mol.Cell Proteomics 4, 12–34.<br />

10. He Q. Y. <strong>and</strong> Chiu J. F. (2003) Proteomics in biomarker discovery <strong>and</strong> drug<br />

development, J.Cell Biochem. 89, 868–86.<br />

11. Wang Y., Chiu J. F., <strong>and</strong> He Q. Y. (2005) Proteomics in computer-aided drug<br />

design, Current Computer-Aided Drug Design 1, 43–52.<br />

12. Wang Y., He Q. Y., Sun R. W., Che C. M., Chiu J. F. (2005) Gold(III) Porphyrin<br />

1a induced apoptosis by mitochondrial death pathways related to reactive oxygen<br />

species, Cancer Res. 65, 11553–64.<br />

13. Bidere N., Lorenzo H. K., Carmona S., Laforge M., Harper F., Dumont C., Senik A.<br />

(2003) Cathepsin D triggers Bax activation, resulting in selective apoptosisinducing<br />

factor (AIF) relocation in T lymphocytes entering the early commitment<br />

phase to apoptosis, J.Biol.Chem. 278, 31401–11.<br />

14. Arnoult D., Gaume B., Karbowski M., Sharpe J. C., Cecconi F., Youle R. J. (2003)<br />

Mitochondrial release of AIF <strong>and</strong> EndoG requires caspase activation downstream<br />

of Bax/Bak-mediated permeabilization, EMBO J. 22, 4385–99.<br />

15. Arnoult D., Parone P., Martinou J. C., Antonsson B., Estaquier J., Ameisen J. C.<br />

(2002) Mitochondrial release of apoptosis-inducing factor occurs downstream of<br />

cytochrome c release in response to several proapoptotic stimuli, J.Cell Biol. 159,<br />

923–9.<br />

16. Sanchez-Alcazar J. A., Bradbury D. A., Pang L., Knox A. J. (2003) Cyclooxygenase<br />

(COX) inhibitors induce apoptosis in non-small cell lung cancer through<br />

cyclooxygenase independent pathways, Lung Cancer 40, 33–44.<br />

17. Vahsen N., C<strong>and</strong>e C., Briere J. J., et al. (2004) AIF deficiency compromises<br />

oxidative phosphorylation, EMBO J. 23, 4679–89.<br />

18. Wang H., Yu S. W., Koh D. W., et al. (2004) Apoptosis-inducing factor substitutes<br />

for caspase executioners in NMDA-triggered excitotoxic neuronal death,<br />

J.Neurosci. 24, 10963–73.<br />

19. Yu S. W., Wang H., Poitras M. F., et al. (2002) Mediation of poly(ADP-ribose)<br />

polymerase-1-dependent cell death by apoptosis-inducing factor, Science 297,<br />

259–63.<br />

20. Otera H., Ohsakaya S., Nagaura Z., Ishihara N., Mihara K. (2005) Export of<br />

mitochondrial AIF in response to proapoptotic stimuli depends on processing at<br />

the intermembrane space, EMBO J. 24, 1375–86.


112 Wang et al.<br />

21. Leung Y. K., Lau K. M., Mobley J., Jiang Z., Ho S. M. (2005) Overexpression<br />

of cytochrome P450 1A1 <strong>and</strong> its novel spliced variant in ovarian cancer cells:<br />

alternative subcellular enzyme compartmentation may contribute to carcinogenesis,<br />

Cancer Res. 65, 3726–34.<br />

22. Jayanthi S., Deng X., Noailles P. A., Ladenheim B., Cadet J. L. (2004)<br />

Methamphetamine induces neuronal apoptosis via cross-talks between endoplasmic<br />

reticulum <strong>and</strong> mitochondria-dependent death cascades, FASEB J. 18, 238–51.<br />

23. Gallego M. A., Joseph B., Hemstrom T. H., et al. (2004) Apoptosis-inducing factor<br />

determines the chemoresistance of non-small-cell lung carcinomas, Oncogene 23,<br />

6282–91.<br />

24. Richard I., Broux O., Allam<strong>and</strong> V., et al. (1995) Mutations in the proteolytic<br />

enzyme calpain 3 cause limb-girdle muscular dystrophy type 2A, Cell 81, 27–40.<br />

25. Marceau N. <strong>and</strong> Loranger A. (1995) Cytokeratin expression, fibrillar organization,<br />

<strong>and</strong> subtle function in liver cells, Biochem.Cell Biol. 73, 619–25.<br />

26. Parry D. A. (1995) Hard alpha-keratin IF: a structural model lacking a head-totail<br />

molecular overlap but having hybrid features characteristic of both epidermal<br />

keratin <strong>and</strong> vimentin IF, Proteins 22, 267–72.<br />

27. Watanabe M., Sasaki M., Itoh K., et al. (2005) JunB induced by constitutive CD30extracellular<br />

signal-regulated kinase 1/2 mitogen-activated protein kinase signaling<br />

activates the CD30 promoter in anaplastic large cell lymphoma <strong>and</strong> reed-sternberg<br />

cells of Hodgkin lymphoma, Cancer Res. 65, 7628–34.<br />

28. Moreno-Manzano V., Ishikawa Y., Lucio-Cazana J., <strong>and</strong> Kitamura M. (1999)<br />

Suppression of apoptosis by all-trans-retinoic acid. Dual intervention in the c-Jun<br />

n-terminal kinase-AP-1 pathway, J.Biol.Chem. 274, 20251–58.<br />

29. Ozaki T., Katsumoto E., Mui K., Furutsuka D., Yamagami S. (1998) Distribution<br />

of Fos- <strong>and</strong> Jun-related proteins <strong>and</strong> activator protein-1 composite factors in mouse<br />

brain induced by neuroleptics, Neuroscience 84, 1187–96.


10<br />

<strong>Sample</strong> <strong>Preparation</strong> of Culture Medium from<br />

Madin-Darby Canine Kidney Cells<br />

Daniel Ambort, Daniel Lottaz, <strong>and</strong> Erwin Sterchi<br />

Summary<br />

A reproducible, st<strong>and</strong>ardized <strong>and</strong> simple sample preparation methodology is the key<br />

to successful two-dimensional gel electrophoresis (2-DE). This chapter describes step-bystep<br />

the sample preparation of culture medium from Madin-Darby canine kidney (MDCK)<br />

cells. Tips <strong>and</strong> tricks are given to circumvent possible pitfalls.<br />

Key Words: Bicinchoninic acid (BCA) assay; culture medium (CM); isoelectric<br />

focusing (IEF); Madin-Darby canine kidney (MDCK); rehydration loading;<br />

two-dimensional gel electrophoresis (2-DE) ultracentrifugation; ultrafiltration .<br />

1. Introduction<br />

Two-dimensional gel electrophoresis (2-DE), introduced by O’Farrell <strong>and</strong><br />

Klose in 1975 (1,2), enabled separation of complex protein mixtures into<br />

individual protein species according to their net charge (pI) in the first<br />

dimension by isoelectric focusing (IEF) <strong>and</strong> in the second dimension according<br />

to their molecular mass (Mr) by sodium dodecyl sulfate-polyacrylamide gel<br />

electrophoresis (SDS-<strong>PAGE</strong>) (3). In the conventional approach, IEF was<br />

performed in carrier ampholyte-generated pH gradients, which moved towards<br />

the cathode on prolonged focusing time. This “cathodic drift” phenomenon<br />

was thereafter remedied by nonequilibrium pH gradient gel electrophoresis<br />

(NEPHGE) (4) <strong>and</strong> finally eliminated with the invention of fixed immobilized<br />

pH gradients (IPG) (5–8). The development of microanalytical techniques,<br />

namely Edman sequencing (9–11) <strong>and</strong> mass spectrometry (12–14) enabled<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

113


114 Ambort et al.<br />

identification of proteins at amounts available from a single 2-D gel. 2-DE<br />

advanced to the core technology of proteome analysis (7,8,15) <strong>and</strong> was brought<br />

from art to craft in an industrial st<strong>and</strong>ard.<br />

Appropriate sample treatment is the key to good results. The ideal sample<br />

solubilization procedure should result in the disruption of all noncovalently<br />

bound protein complexes <strong>and</strong> aggregates into a solution of individual<br />

polypeptides (15). Denaturation <strong>and</strong> reduction of proteins is achieved in the<br />

st<strong>and</strong>ard lysis buffer (O’Farrell 1975) (1) which is composed of 8–9 M urea,<br />

2–4% CHAPS, 1% DTT or DTE <strong>and</strong> 0.8–2% carrier ampholytes. Hydrophobic<br />

proteins are better dissolved in 2 M thiourea <strong>and</strong> 7 M urea instead of 9 M urea<br />

(16). Optimized procedures for different sample types do exist (17). However,<br />

a general “Prepares them all” procedure is not available (18). Another highpriority<br />

issue is the removal <strong>and</strong> inactivation of all interfering substances:<br />

nucleic acids, lipids, salts, small ionic compounds, polysaccharides, proteases,<br />

<strong>and</strong> insoluble particles. <strong>Sample</strong> preparation for 2-DE is a very cumbersome <strong>and</strong><br />

time-consuming task that is subject to trial <strong>and</strong> error.<br />

Because of the complex biological architecture of eukaryotic cells into<br />

organelles <strong>and</strong> large cellular structures, fractionation techniques are applied<br />

before comprehensively studying the subproteomes. In such reductionist<br />

approaches classic biochemical techniques, namely centrifugation <strong>and</strong> affinitymediated<br />

isolation using antibodies against molecular tags, are applied to enrich<br />

for subcellular fractions as reviewed by Yates 3rd (19). Beside analysis of<br />

cytosolic, organelle-specific <strong>and</strong> transmembrane proteins several investigations<br />

were aimed at identifying those proteins secreted by various cell types into the<br />

extracellular milieu or medium (20–24).<br />

This chapter describes the sample preparation of culture medium from<br />

Madin-Darby canine kidney (MDCK) cells for 2-DE (Fig. 1). The methodology<br />

is subsectioned into four parts with basic introductory information on each topic:<br />

cell culture (see Subheading 3.1.), ultracentrifugation <strong>and</strong> ultrafiltration (see<br />

Subheading 3.2.), protein quantitation by BCA assay (see Subheading 3.3.)<br />

<strong>and</strong> finally, rehydration loading <strong>and</strong> isoelectric focusing (see Subheading 3.4.).<br />

2. Materials<br />

2.1. Cell Culture<br />

2.1.1. Equipment<br />

1. BD Falcon TM bulk packaged serological pipets (10 mL) (BD Biosciences, Franklin<br />

Lakes, NY, USA)<br />

2. BD Falcon TM individually wrapped serological pipets (25 mL) (BD Biosciences,<br />

Franklin Lakes, NY, USA)


<strong>Sample</strong> <strong>Preparation</strong> of Culture Medium 115<br />

Fig. 1. Two-dimensional gel electrophoresis of Madin-Darby canine kidney cell<br />

culture supernatant (80 μg protein). First dimension: isoelectric focusing in an immobilized<br />

pH gradient (IPG) pH 3–10 nonlinear in a 24-cm-long gel strip. Second dimension:<br />

SDS-<strong>PAGE</strong> in a 12.5% gel. Silver stained.<br />

3. BD Falcon TM st<strong>and</strong>ard cell culture dish, st<strong>and</strong>ard tissue-culture treated<br />

(100 × 20 mm) (BD Biosciences, Franklin Lakes, NY, USA)<br />

4. CELLSTAR ® PP-test tubes (50 mL, sterile) (Greiner Bio-One Inc., Longwood,<br />

FL, USA)<br />

5. Erlenmeyer flasks (250 mL)<br />

6. Laminar air flow cabinet (Brouwer AG, Luzern, Switzerl<strong>and</strong>)<br />

7. Neubauer improved counting chamber (Assistent, Sondheim, Germany)<br />

8. NUAIRE TM US autoflow CO 2 water-jacketed incubator NU-4750 (Vitaris AG,<br />

Baar, Switzerl<strong>and</strong>)<br />

9. Water bath<br />

2.1.2. Solutions <strong>and</strong> Reagents<br />

1. Dulbecco’s Phosphate Buffered Saline (D-PBS) (1X) (500 mL) (GIBCO Invitrogen<br />

corporation, Gr<strong>and</strong> Isl<strong>and</strong>, NY, USA)<br />

2. Foetal bovine serum (FBS) (E. U. approved South American origin, virus <strong>and</strong><br />

mycoplasma tested) (500 mL) (GIBCO Invitrogen corporation, Gr<strong>and</strong> Isl<strong>and</strong>, NY,<br />

USA)<br />

3. Minimum essential medium (MEM) (1X) (with Earle´s salts, without l-glutamine)<br />

(500 mL) (GIBCO Invitrogen corporation, Gr<strong>and</strong> Isl<strong>and</strong>, NY, USA)<br />

4. Penicillin-streptomycin-glutamine (100X) (100 mL) (GIBCO Invitrogen corporation,<br />

Gr<strong>and</strong> Isl<strong>and</strong>, NY, USA)<br />

5. Trypsin-EDTA (1X) (100 mL) (GIBCO Invitrogen corporation, Gr<strong>and</strong> Isl<strong>and</strong>,<br />

NY, USA)


116 Ambort et al.<br />

6. Culture medium: 1X MEM (see Note 1), 5% (v/v) FBS, 100 U/mL penicillin,<br />

100 μg/mL streptomycin <strong>and</strong> 292 μg/mL l-glutamine. To 500 mL of MEM (one<br />

bottle) aseptically add 25 mL of FBS (see Note 2) <strong>and</strong> 5 mL of 100X penicillinstreptomycin-glutamine<br />

stock solution. Store at 4°C. Before use warm up to 37°C<br />

in a water bath.<br />

7. Serum-free medium: 1X MEM (see Note 1), 100 units/mL penicillin, 100 μg/mL<br />

streptomycin, <strong>and</strong> 292 μg/mL l-glutamine. To 500 mL of MEM (one bottle)<br />

aseptically add 5 mL of 100X penicillin-streptomycin-glutamine stock solution.<br />

Store at 4°C. Before use warm up to 37°C in a water bath.<br />

2.2. Ultracentrifugation <strong>and</strong> Ultrafiltration<br />

2.2.1. Equipment<br />

1. Centricon ® Plus-70 centrifugal filter devices (Millipore corporation, Billerica,<br />

MA, USA)<br />

2. Centrifuge filter system (50 mL, 0.2 μm) (Costarcorporation, Cambridge, MA,<br />

USA)<br />

3. Eppendorf centrifuge 5415R (Eppendorf AG, Hamburg, Germany)<br />

4. Eppendorf tubes (1.5 mL, 2 mL)<br />

5. KONTRON CENTRIKON TFT 70.38 fixed-angle rotor (KONTRON Instruments<br />

AG, Zürich, Switzerl<strong>and</strong>)<br />

6. KONTRON CENTRIKON T-2060 ultracentrifuge (KONTRON Instruments<br />

AG, Zürich, Switzerl<strong>and</strong>)<br />

7. KONTRON CENTRIKON ultracentrifuge tubes (32.5 mL) (KONTRON Instruments<br />

AG, Zürich, Switzerl<strong>and</strong>)<br />

8. Mettler AC 100 analytical balance (Mettler Instrumente AG, Greifensee, Zürich,<br />

Switzerl<strong>and</strong>)<br />

9. Sorvall RT6000D centrifuge (Kendro Laboratory Products AG, Zürich,<br />

Switzerl<strong>and</strong>)<br />

10. Sorvall H1000B swinging bucket rotor (Kendro Laboratory Products AG, Zürich,<br />

Switzerl<strong>and</strong>)<br />

11. Water bath<br />

2.2.2. Solutions <strong>and</strong> Reagents<br />

1. Ethylenedinitrilo tetraacetic acid disodium salt dihydrate (Na 2-EDTA 2H 2O,<br />

Titriplex ® III) (GR for analysis)<br />

2. Phenylmethylsulfonyl fluoride (PMSF) (Sigma, St. Louis, MO, USA)<br />

3. 2-Propanol (GR for analysis)<br />

4. Sodium hydroxide (NaOH) (pellets GR for analysis)<br />

5. Tris(hydroxymethyl)aminomethane (Tris) (GR for analysis buffer substance)


<strong>Sample</strong> <strong>Preparation</strong> of Culture Medium 117<br />

6. 0.5 M EDTA pH 8.0 stock solution: To make 100 mL of stock solution, dissolve<br />

2 g of NaOH pellets in 80 mL of ddH 2O. Add 18.6 g of Na 2-EDTA-2H 2O under<br />

constant stirring at RT (see Note 3). Titrate solution to pH 8 with 5 N NaOH<br />

(liquid). Adjust to a final volume of 100 mL with ddH 2O <strong>and</strong> check pH again.<br />

Filter solution with a 0.2 μm bottle top filter. This solution can be stored at RT.<br />

7. 0.1 M PMSF stock solution: To prepare 25 mL, dissolve 0.44 g of PMSF in 25<br />

mL of 2-propanol (isopropanol). Warm up to 37°C in a water bath (see Note 4).<br />

Portion solution into 2 mL aliquots in 2 mL Eppendorf tubes <strong>and</strong> store at –20°C.<br />

8. 0.2 M Tris pH 10.5 stock solution: To make 0.5 L, dissolve 12.12 g of Tris in<br />

0.5 L of ddH 2O(see Note 5). Filter solution with a 0.2 μm bottle top filter. This<br />

solution can be stored at RT.<br />

9. <strong>Sample</strong> solubilization buffer I: 20 mM Tris-HCl pH 9.0, 1 mM EDTA, 1 mM<br />

PMSF. To prepare 150 mL, dilute 15 mL of 0.2 M Tris pH 10.5 stock solution to<br />

a final volume of 150 mL in ddH 2O. Add 0.3 mL of 0.5 M EDTA pH 8.0 stock<br />

solution <strong>and</strong> 1.5 mL of 0.1 M PMSF stock solution under constant stirring at RT<br />

(see Note 6). Filter solution with a 0.2 μm bottle top filter. This solution should<br />

be prepared freshly just before use!<br />

2.3. Protein Quantitation by BCA Assay<br />

2.3.1. Equipment<br />

1. Beaker (25 mL)<br />

2. CELLSTAR ® micro-plate (TC, sterile) (Greiner Bio-One Inc., Longwood, FL,<br />

USA)<br />

3. Incubator<br />

4. Vortex mixer<br />

5. Vmax ® microplate reader (Molecular Devices Corporation, Sunnyvale, CA, USA)<br />

2.3.2. Solutions <strong>and</strong> Reagents<br />

1. Albumin St<strong>and</strong>ard Ampules (2 mg/mL, 10 x 1 mL) (Pierce, Rockford, IL, USA)<br />

2. BCATM Protein Assay Kit (Pierce, Rockford, IL, USA)<br />

3. BCATM Protein Assay Reagent A (500 mL) (Pierce, Rockford, IL, USA)<br />

4. BCATM Protein Assay Reagent B (25 mL) (Pierce, Rockford, IL, USA)<br />

2.4. Rehydration Loading <strong>and</strong> Isoelectric Focusing<br />

2.4.1. Equipment<br />

1. Beaker (50 mL)<br />

2. Centrifuge filter system (50 mL, 0.2 μm) (Costarcorporation, Cambridge, MA,<br />

USA)<br />

3. Eppendorf centrifuge 5415R (Eppendorf AG, Hamburg, Germany)<br />

4. EPS 3501 XL power supply (Amersham Biosciences, Uppsala, Sweden)


118 Ambort et al.<br />

5. IEF electrode strips (Amersham Biosciences, Uppsala, Sweden)<br />

6. Immobiline TM DryStrip Kit (Amersham Biosciences, Uppsala, Sweden)<br />

7. Immobiline TM DryStrip Reswelling Tray (7–24 cm) (Amersham Biosciences,<br />

Uppsala, Sweden)<br />

8. Vortex mixer<br />

9. Multiphor TM II horizontal electrophoresis apparatus (Amersham Biosciences,<br />

Uppsala, Sweden)<br />

10. Multitemp TM III thermostatic circulator (Amersham Biosciences, Uppsala,<br />

Sweden)<br />

11. Multi-purpose rotator (Scientific Industries Inc., Queens Village, NY, USA)<br />

12. Parafilm (50 × 15 m) (American National Can Company, Chicago, IL, USA)<br />

13. Petri dishes<br />

14. Tweezers<br />

2.4.2. Solutions <strong>and</strong> Reagents<br />

1. Bromophenol blue (BPB) (Bio-Rad Laboratories, Richmond, CA, USA)<br />

2. 3-[(3-cholamidopropyl)-dimethylammonio]-1-propane sulfonate (CHAPS)<br />

(ultrapure) (USB corporation, Clevel<strong>and</strong>, OH, USA)<br />

3. 1,4-dithioerythritol (DTE) (for biochemistry) (Merck, Darmstadt, Germany)<br />

4. ImmobilineTM DryStrip pH 3–10 NL (IPG) (240 × 3 × 0.5 mm) (Amersham<br />

Biosciences, Uppsala, Sweden)<br />

5. Mixed bed ion exchanger resin<br />

6. Paraffin (Merck, Darmstadt, Germany)<br />

7. PharmalyteTM 3–10 (for IEF) (Amersham Biosciences, Uppsala, Sweden)<br />

8. Thiourea (puriss. p. a. ACS; ≥99% [RT]) (Fluka, Buchs, Switzerl<strong>and</strong>)<br />

9. ZOOM ® urea (Invitrogen life technologies, Carlsbad, CA, USA)<br />

10. DTE aliquots: 65 mM in 1.5 mL of sample solubilization buffer II (working<br />

solution). Portion 0.015 g (15 mg) of DTE in 1.5 mL Eppendorf tube <strong>and</strong> store<br />

at 4°C until use (see Note 7).<br />

11. <strong>Sample</strong> solubilization buffer II (stock solution): 7 M urea, 2 M thiourea, 4%<br />

(w/v) CHAPS. To prepare 25 mL, dissolve 10.5 g of urea, 3.8 g of thiourea in<br />

10 mL of ddH2O under constant stirring at RT. Fill up to a final volume of 30<br />

mL with ddH2O(see Note 8).Add5gofmixed bed ion exchanger resin <strong>and</strong><br />

stir for 10 min. Remove beads by filtration through a 0.2 μm bottle top filter.<br />

Dissolve 1gofCHAPS, add a trace of BPB <strong>and</strong> portion solution into 1.5 mL<br />

aliquots in 1.5 mL Eppendorf tubes. Store at –20°C until use.<br />

12. <strong>Sample</strong> solubilization buffer II (working solution): 7 M urea, 2 M thiourea, 4%<br />

(w/v) CHAPS, 1% (w/v) (65 mM) DTE, 2% (v/v) (0.8% (w/v)) Pharmalyte<br />

3–10. To make up 1.5 mL, add 1.5 mL of sample solubilization buffer II<br />

(stock solution) to one DTE aliquot in 1.5-mL Eppendorf tube. Add 30 μL of<br />

Pharmalyte 3–10 <strong>and</strong> incubate for 15 minutes at RT with occasional vortexing<br />

until DTE is completely dissolved. This solution is prepared just before use (see<br />

Note 9).


<strong>Sample</strong> <strong>Preparation</strong> of Culture Medium 119<br />

3. Methods<br />

3.1. Cell Culture<br />

Mammalian renal tubular epithelium consists of at least seven different<br />

segments, complicating biochemical investigation of this heterogeneous tissue.<br />

Cultured monolayers of dog kidney (Madin-Darby canine kidney (MDCK))<br />

cells display many typical features of renal tubular epithelia, such as brush<br />

border membrane, tight junctions <strong>and</strong> adherent junctions. MDCK strain II (26)<br />

was used to prepare samples of cell culture supernatants for 2-DE. High-quality<br />

protein samples for 2-DE are only obtained from serum-free media. Serumderived<br />

proteins heavily contaminate cell-derived secreted proteins in culture<br />

media (see Note 10). Hence it is essential to wash the cells thoroughly (twice<br />

in PBS or serum-free medium) when changing from complete culture media to<br />

serum-free conditions.<br />

1. Harvest confluent MDCK cells by trypsinization. To five confluent 100-mm cell<br />

culture dishes add 1.5 mL of prewarmed (37°C) Trypsin-EDTA solution per dish<br />

<strong>and</strong> incubate at 37°C in a humidified incubator in an atmosphere of 5% CO 2 until<br />

cells detach (see Note 11).<br />

2. Resuspend trypsinized cells in culture medium. To each dish add 6.5–7 mL of<br />

prewarmed (37°C) culture medium <strong>and</strong> pool resuspended cells from the five<br />

dishes into one 50-mL Greiner tube. Fill up to a total volume of 40 mL with<br />

culture medium (see Note 12).<br />

3. Seed 1.15 × 10 6 cells onto 100-mm cell culture dishes. Adjust final volume with<br />

prewarmed (37°C) culture medium to 9 mL per dish. Prepare a total of 18 dishes<br />

per condition (see Note 13). Incubate the cultures for about three days at 37°C<br />

in a humidified incubator in an atmosphere of 5% CO 2 until cells are confluent<br />

(see Note 14).<br />

4. Aspirate medium <strong>and</strong> wash cells twice in 4 mL of prewarmed (37°C) serum-free<br />

medium per dish (see Note 15). Add 4 mL of serum-free medium to each dish<br />

<strong>and</strong> incubate for 22 h at 37°C in a humidified incubator in an atmosphere of<br />

5% CO 2.<br />

5. Harvest cell culture supernatants. Pool media from 18 dishes into one 250-mL<br />

Erlenmeyer flask to give a final volume of 70–72 mL (see Note 13). Immediately<br />

proceed to ultracentrifugation <strong>and</strong> ultrafiltration (see Subheading 3.2.).<br />

3.2. Ultracentrifugation <strong>and</strong> Ultrafiltration<br />

Beside body fluids, such as human plasma, urine, <strong>and</strong> cerebrospinal fluid,<br />

cell culture supernatants are among the most difficult samples to be prepared<br />

for 2-D <strong>PAGE</strong>. Culture media contain the complete set of interfering substances<br />

that are incompatible with the first dimensional isoelectric focusing: insoluble<br />

particles (dead cells, cell debris), nucleic acids (from dead cells), lipids<br />

(membranes <strong>and</strong> exosomes (22)), salts (from medium see Note 1), small ionic


120 Ambort et al.<br />

compounds (amino acids from medium), phenolic compounds (Phenol red<br />

from medium) <strong>and</strong> proteases (secreted during cultivation). Therefore these<br />

contaminants are removed in a two-step purification strategy: 1) insoluble<br />

particles, nucleic acids <strong>and</strong> lipids by high-speed centrifugation; 2) salts, small<br />

ionic <strong>and</strong> phenolic compounds by ultrafiltration. Proteases are inhibited by<br />

addition of inhibitors <strong>and</strong> basic pH conditions. Alternative methods such as<br />

TCA/acetone precipitation (27) <strong>and</strong> dialysis must not be applied. Unspecific<br />

salt precipitation <strong>and</strong> loss of proteins are associated with these methods (see<br />

Note 16).<br />

1. Add 140 μL of 0.5 M EDTA pH 8.0 <strong>and</strong> 700 μL of 0.1 M PMSF to 70 mL of<br />

culture medium in 250 mL Erlenmeyer flask <strong>and</strong> put on ice (see Note 17).<br />

2. Transfer 4 × 17.5 mL of medium to four prechilled KONTRON CENTRIKON<br />

ultracentrifuge tubes. Adjust precise volumes with ddH 2O on an analytical<br />

balance <strong>and</strong> put tubes on ice.<br />

3. Place precooled (4°C) KONTRON CENTRIKON TFT 70.38 fixed-angle rotor<br />

into KONTRON CENTRIKON T-2060 ultracentrifuge. Place tubes into the rotor<br />

in appropriate positions <strong>and</strong> close the lid. Ultracentrifuge for 60 min at 31,200<br />

rpm (100,000g) at 4°C.<br />

4. Carefully remove tubes from ultracentrifuge <strong>and</strong> put on ice.<br />

5. Rinse Centricon ® Plus-70 components consisting of cap, concentrate/retentate<br />

cup, sample filter cup <strong>and</strong> filtrate collection cup with ddH 2O to remove dust<br />

particles (see Note 18).<br />

6. Place sample filter cup into filtrate collection cup <strong>and</strong> leave on ice.<br />

7. Pool the medium by inverting tubes into the same sample filter cup <strong>and</strong> close.<br />

Then place assembled Centricon ® Plus-70 centrifugal filter device into precooled<br />

(4°C) Sorvall H1000B swinging bucket rotor fixed in a Sorvall RT6000D<br />

centrifuge. Centrifuge for 60 min at 3,200 rpm (2,190g) at 4°C.<br />

8. Discard the flow-through in the filtrate collection cup. Add 70 mL of prechilled<br />

sample solubilization buffer I (20 mM Tris pH 9.0, 1 mM EDTA, 1 mM PMSF)<br />

into sample filter cup (see Note 19). Centrifuge for 60 min at 3,200 rpm (2,190g)<br />

at 4°C.<br />

9. Repeat step 8 twice.<br />

10. After the last washing step place concentrate/retentate cup upside down onto the<br />

filtrate collection cup. Invert assembly <strong>and</strong> place back into centrifuge. Recover<br />

sample concentrate for 5 min at 2,200 rpm (1,000g) at 4°C.<br />

11. Determine volume with a pipet. Typical final concentrate volumes are between<br />

250 μL <strong>and</strong> 350 μL.<br />

12. Transfer protein concentrate to a 1.5 mL Eppendorf tube <strong>and</strong> spin for 5 min<br />

at 13,200 rpm (16,100g) at 4°C to remove precipitates (see Note 20). Put<br />

samples on ice <strong>and</strong> proceed to protein quantitation (see Subheading 3.3.) or store<br />

at –20°C until use.


<strong>Sample</strong> <strong>Preparation</strong> of Culture Medium 121<br />

3.3. Protein Quantitation by BCA Assay<br />

Quantitative determination of protein solubilized in modified lysis buffer<br />

(16) (7 M urea, 2 M thiourea, 4% CHAPS, 65 mM DTE <strong>and</strong> 2% Pharmalyte<br />

3–10) is not possible. The Bradford assay (29) cannot be used for two reasons:<br />

1) Coomassie Brilliant Blue G-250 binds to detergents (CHAPS) <strong>and</strong> carrier<br />

ampholytes (Pharmalyte 3–10); 2) Coomassie Brilliant Blue G-250 may not<br />

bind to protein at all under basic pH conditions (urea). The second problem<br />

may be remedied by acidification of sample with 0.1 N HCl before quantitation<br />

(30). St<strong>and</strong>ard Lowry (31), Biuret (32), <strong>and</strong> BCA (bicinchoninic acid) (33)<br />

assays based on the reduction of Cu 2+ to Cu + for development of color interfere<br />

with thiol reducing agents (DTE) <strong>and</strong> thiourea. Thiourea forms complexes with<br />

copper ions. TCA/acetone precipitation (27) must not be applied in combination<br />

with any of these techniques. Protein may be lost on precipitation leading<br />

to underestimation of solubilized protein. The best solution to all problems<br />

mentioned above is quantitation of protein before solubilization in lysis buffer.<br />

In this section the BCA assay from Pierce is used to accurately quantitate<br />

protein concentration in sample solubilization buffer I (20 mM Tris pH 9.0,<br />

1mM EDTA, 1 mM PMSF). Although the BCA assay interferes with chelating<br />

agents (EDTA) concentrations below 10 mM are tolerated (34).<br />

1. Prepare a set of albumin (BSA) st<strong>and</strong>ards in 1.5-mL Eppendorf tubes (see<br />

Table 1 for details). Use ddH 2O as diluent. Gently vortex tubes. There will be<br />

sufficient volume for two replications of each diluted st<strong>and</strong>ard.<br />

2. Prepare protein samples in 1.5-mL Eppendorf tubes. Dilute 5 μL of each protein<br />

concentrate to a final volume of 100 μL in ddH 2O (1:20 dilution). Gently vortex<br />

tubes. While performing BCA assay put undiluted protein concentrates on ice<br />

(see Subheading 3.2. step 12).<br />

Table 1<br />

<strong>Preparation</strong> of diluted albumin (BSA) st<strong>and</strong>ards<br />

Vial Volume of diluent a Volume <strong>and</strong> source of BSA Final BSA concentration<br />

A 150 μL 150 μL of stock b 1000 μg/mL<br />

B 25 μL 75 μL of A 750 μg/mL<br />

C 50 μL 50 μL of A 500 μg/mL<br />

D 75 μL 25 μL of A 250 μg/mL<br />

E 88 μL 12.5 μL of A 125 μg/mL<br />

F 98 μL 2.5 μL of A 25 μg/mL<br />

a Use ddH2O to prepare albumin (BSA) st<strong>and</strong>ards.<br />

b The concentration of albumin st<strong>and</strong>ard stock solution is 2 mg/mL.


122 Ambort et al.<br />

3. Pipet 25 μL of each st<strong>and</strong>ard or unknown sample replicate into a microplate<br />

well. St<strong>and</strong>ards are applied in duplicates, protein samples in triplicates. Use<br />

ddH 2O for blanks.<br />

4. Prepare BCA working reagent by mixing 50 parts of BCA TM Reagent A with 1<br />

part of BCA TM Reagent B in a 25 mL beaker. Mix thoroughly (see Note 21).<br />

For each st<strong>and</strong>ard, unknown sample or blank 200 μL of BCA working reagent<br />

is required. Include two extra replicates in your calculation.<br />

5. Add 200 μL of BCA working reagent to each well. Gently shake microplate by<br />

h<strong>and</strong> for a few seconds.<br />

6. Incubate microplate for 30 min at 37°C in an incubator.<br />

7. Cool plate to RT <strong>and</strong> measure absorbance at 550 nm on a microplate reader (see<br />

Note 22).<br />

8. Subtract the average 550 nm absorbance measurement of the blank replicates<br />

from the 550 nm absorbance measurements of all other individual st<strong>and</strong>ards <strong>and</strong><br />

unknown sample replicates.<br />

9. Prepare a st<strong>and</strong>ard curve by plotting the average blank-corrected 550 nm<br />

absorbance measurement (A 550, y-axis) for each albumin st<strong>and</strong>ard versus its<br />

amount (in μg, x-axis) in increasing order (see Note 23).<br />

10. Use the st<strong>and</strong>ard curve to determine the protein amount of each unknown<br />

sample (Fig. 2). The protein concentration in unknown sample is calculated as<br />

follows: (protein amount of unknown in μg)/(volume of diluted sample in μL) ×<br />

(dilution factor)= protein concentration in mg/mL (see Note 24). Typical protein<br />

concentrations are between 5 mg/mL <strong>and</strong> 7 mg/mL.<br />

11. Portion undiluted protein concentrates into appropriate aliquots (80 μg for<br />

analytical load) in 1.5 mL Eppendorf tubes <strong>and</strong> store at –20°C until use (see<br />

Subheading 3.4.1).<br />

3.4. Rehydration Loading <strong>and</strong> Isoelectric Focusing<br />

Traditionally, protein samples prepared in st<strong>and</strong>ard lysis buffer (O’Farrell<br />

1975) (1) were loaded onto the basic end (cathode) of an isoelectric focusing<br />

(IEF) tube gel. Before sample loading the gel rods were prerun to establish a<br />

carrier ampholyte-derived pH gradient. On development of fixed pH gradients<br />

samples were applied with rubber frames or sample cups to rehydrated immobilized<br />

pH gradient (IPG) strips at the acidic or basic end (5–8) or simultaneously<br />

at both ends (35). The problem with cup-loading sample application<br />

techniques is that proteins may precipitate during the sample entry phase, which<br />

leads to horizontal streaking at the sample application point. This problem is<br />

remedied by in-gel sample rehydration where protein solubilized in lysis buffer<br />

is directly diluted with the rehydration solution used for IPG strip reswelling<br />

(36,37). Unfortunately, some proteins that are soluble in lysis buffer may be<br />

lost on dilution into rehydration solution because of lower concentrations of<br />

chaotropic agents <strong>and</strong> detergents. For simplicity protein sample preparation


<strong>Sample</strong> <strong>Preparation</strong> of Culture Medium 123<br />

Fig. 2. Typical color response curve for albumin (BSA) st<strong>and</strong>ards using the BCA<br />

assay. Each point represents the mean of two replications.<br />

<strong>and</strong> rehydration can be done all-in-one in modified lysis buffer (16) (7 M<br />

urea, 2 M thiourea, 4% CHAPS, 65 mM DTE <strong>and</strong> 2% Pharmalyte 3–10). This<br />

strategy works very well in combination with the Multiphor II horizontal flatbed<br />

isoelectric focusing system with final maximum voltage limited to 3,500 V<br />

for steady-state IEF (38). Higher voltage settings may become problematic,<br />

because zwitterionic detergent (CHAPS), reducing agent (DTE) <strong>and</strong> carrier<br />

ampholytes (Pharmalyte 3–10) heavily contribute to the current in the<br />

strip.<br />

1. Thaw protein samples on ice (see Subheading 3.3.11). Dilute each aliquot of<br />

protein solution (80 μg for analytical load) to a final volume of 450 μL in sample<br />

solubilization buffer II (see Note 25).<br />

2. Gently vortex tubes <strong>and</strong> solubilize protein for 60 min on a rotary shaker at RT.<br />

3. Centrifuge for 30 min at 13,200 rpm (16,100g) at 22°C in a tabletop centrifuge<br />

to remove insoluble particles.<br />

4. Slide the protective lid completely off the Immobiline TM DryStrip Reswelling<br />

Tray <strong>and</strong> level the tray by turning the leveling feet until the bubble in the spirit<br />

level is centered.<br />

5. Remove IPG strips (240 mm long, 3 mm wide ready-made Immobiline TM<br />

DryStrips pH 3–10 NL cast on GelBond PAGfilm) from the freezer <strong>and</strong> warm<br />

up to RT.


124 Ambort et al.<br />

6. Evenly apply the entire sample-containing solution into the groove of the<br />

reswelling tray (see Note 26).<br />

7. Peel off the protective cover sheet from the ready-made IPG strip starting at the<br />

acidic (+) end <strong>and</strong> grip the strip with tweezers at the overlapping basic plastic<br />

end (see Note 27).<br />

8. Slowly lower the IPG strip (gel side down) onto the solution with the<br />

acidic (+) end oriented towards the number labels of the reswelling tray (see<br />

Note 26).<br />

9. Cover the strip with 3 mL of paraffin oil (see Note 28). Repeat steps 6–9 for<br />

each sample.<br />

10. Slide the lid onto the reswelling tray <strong>and</strong> rehydrate the IPG strips overnight<br />

at RT.<br />

11. To remove rehydrated IPG strips sequentially from the reswelling tray, open<br />

the lid, slide the tip of tweezers along the sloped end of the slot <strong>and</strong> into the<br />

slight depression under the IPG strip. Grab the acidic (+) end of the strip with<br />

tweezers <strong>and</strong> lift the strip out of the tray. While still holding the strip, rinse it<br />

briefly with ddH 2O. Place it on a piece of damp filter paper at one edge to drain<br />

off excess liquid. Repeat procedure for each strip.<br />

12. Set the temperature on the MultiTemp TM III thermostatic circulator to 20°C.<br />

13. Place the ceramic cooling plate in Multiphor II unit <strong>and</strong> make sure the surface<br />

is level.<br />

14. Starting at the top of the plate near the cooling tubes pipet 1 mL of paraffin oil<br />

in a straight line onto the middle of the plate. Use the grid of the cooling plate<br />

as a guide. Then pipet 1 mL of paraffin oil on each side of the paraffin oil line<br />

(a total of 3 mL) onto the bottom of the plate. The additional 1 mL of paraffin<br />

oil on each side is evenly spread on the cooling plate to form a triangle which<br />

begins with the base line at the bottom <strong>and</strong> extends to the middle of the paraffin<br />

oil line (see Note 29).<br />

15. Slowly lower the Immobiline TM DryStrip tray onto the bottom of the paraffin<br />

oil triangle with the red (anodic, positively charged) electrode connection of the<br />

tray positioned at the top of the plate near the cooling tubes.<br />

16. Connect the red <strong>and</strong> black electrode leads on the tray to the Multiphor II unit.<br />

17. Pour 10 mL of paraffin oil into the tray at the bottom of the cooling plate.<br />

18. Slowly lower the Immobiline TM DryStrip aligner, 12 grooves side up, onto the<br />

bottom of the paraffin oil layer next to the black electrode.<br />

19. Transfer the rehydrated IPG strips with tweezers to adjacent grooves of the<br />

aligner in the tray. Place the strips gel side up with the acidic (+) end at the top<br />

of the tray near the red electrode.<br />

20. Cut one IEF electrode strip into two pieces each to a length of 110 mm. Moisten<br />

the two IEF electrode strips with deionized water (see Note 30).<br />

21. Place the damp electrode strips across the acidic <strong>and</strong> basic ends of the aligned<br />

IPG strips.


<strong>Sample</strong> <strong>Preparation</strong> of Culture Medium 125<br />

22. Align each electrode over an electrode strip, ensuring the marked side corresponds<br />

to the side of the tray giving electric contact (see Note 31). When the<br />

electrodes are properly aligned, press them down to contact the electrode strips.<br />

23. Pour 100 mL of paraffin oil into the tray to cover the IPG <strong>and</strong> electrode strips.<br />

24. Close the lid of the Multiphor II unit. Connect the leads on the lid to the EPS<br />

3501 XL power supply <strong>and</strong> start IEF according to programmed parameters (see<br />

Note 32).<br />

25. After IEF remove electrodes <strong>and</strong> IEF electrode strips.<br />

26. Grip each IPG strip with tweezers at the overlapping basic plastic end, carefully<br />

remove it from the tray <strong>and</strong> rinse it with ddH 2O.<br />

27. Place each IPG strip into a petri dish with the plastic side of the strip facing the<br />

inner wall of the petri dish <strong>and</strong> cover. Seal it with a piece of parafilm <strong>and</strong> store<br />

at –20° C until use.<br />

28. IPG strip equilibration, SDS-<strong>PAGE</strong> <strong>and</strong> postseparation visualization techniques<br />

applied following sample preparation <strong>and</strong> IEF are not topic of this chapter.<br />

Useful tips <strong>and</strong> tricks concerning these methods can be found in the Amersham<br />

2-D electrophoresis h<strong>and</strong>book (40). As an example the final 2-D map of Madin-<br />

Darby canine kidney (MDCK) cell culture supernatant is shown in Fig. 1.<br />

4. Notes<br />

1. Composition of Minimum Essential Medium (MEM) (25): 264 mg/mL CaCl2 2H2O, 400 mg/mL KCl, 200 mg/mL MgSO4 7H2O, 6800 mg/mL NaCl,<br />

2200 mg/mL NaHCO3, 158 mg/mL NaH2PO4 2H2O, 1000 mg/mL<br />

d-glucose, 10 mg/mL Phenol red, 126 mg/mL l-arginine HCl, 24 mg/mL<br />

l-cystine, 42 mg/mL l-histidine HCl H2O, 52 mg/mL l-isoleucine, 52<br />

mg/mL l-leucine, 73 mg/mL l-lysine HCl, 15 mg/mL l-methionine, 32 mg/mL<br />

l-phenylalanine, 48 mg/mL l-threonine, 10 mg/mL l-tryptophan, 36 mg/mL<br />

l-tyrosine, 46 mg/mL l-valine, 1 mg/mL d-Ca pantothenate, 1 mg/mL choline<br />

chloride, 1 mg/mL folic acid, 2 mg/mL i-inositol, 1 mg/mL niacinamide, 1<br />

mg/mL pyridoxine HCl, 0.1 mg/mL riboflavin, <strong>and</strong> 1 mg/mL thiamine HCl.<br />

2. Before use fetal bovine serum (FBS) is heat-inactivated! Incubate one bottle<br />

(500 mL) of FBS for 30 min at 56°C in a water bath. Portion solution into<br />

25 mL aliquots <strong>and</strong> store at –20°C.<br />

3. The EDTA may not completely dissolve in 0.5 M stock solution below pH 8.0.<br />

Therefore titration of EDTA stock solution with a few drops of 5 N NaOH<br />

(liquid) is necessary. The solution is ready when the pale white color turns into<br />

a crystal clear solution.<br />

4. PMSF is very toxic! Protect your eyes <strong>and</strong> skin. PMSF is not very stable in<br />

water <strong>and</strong> has a half-life of about 30 min. Hence the solution is prepared in<br />

isopropanol. PMSF is difficult to dissolve at RT; therefore the solution is warmed<br />

up to 37°C.<br />

5. The 0.2 M Tris stock solution has a pH of 10.5–10.6. Do not titrate with HCl!<br />

The chloride ions extremely contribute to the current (heat production) during


126 Ambort et al.<br />

isoelectric focusing (IEF). Any heat produced during IEF will cause protein<br />

precipitation <strong>and</strong> produce horizontal streaks in the final 2-D gel.<br />

6. The sample solubilization buffer I has a pH of 9.0–9.1. Do not titrate with HCl!<br />

The Tris serves as a positively charged ion that helps in solubilization of proteins<br />

<strong>and</strong> not to maintain constant pH (see Note 19). This solution should be prepared<br />

shortly before use because of the poor stability of PMSF in water (see Note 4).<br />

The 0.1 M PMSF stock solution should be warmed up to 37°C in a water bath.<br />

Otherwise the PMSF may precipitate!<br />

7. DTE is not very stable in solution. Hence it is stored as solid in small aliquots<br />

at 4°C.<br />

8. The final volume of 30 mL compensates for the dead volume of the magnetic<br />

stir bar in a 50-mL beaker <strong>and</strong> equals to a total volume of 25 mL. Add urea<br />

<strong>and</strong> thiourea in small portions with the help of a spatula under constant stirring<br />

at RT. Urea <strong>and</strong> thiourea will cool down the solution <strong>and</strong> decrease solubility!<br />

Therefore in between additions wait for several minutes until each small portion<br />

is fully dissolved. Do not heat urea-containing solutions above 37°C to avoid<br />

carbamylation of proteins!<br />

9. The sample solubilization buffer II (working solution) should be prepared<br />

freshly. Never reuse remaining buffer, better discard it!<br />

10. It is essential to wash the cells thoroughly (twice in PBS or serum-free medium)<br />

to remove serum proteins. Culture media heavily contaminated with fetal bovine<br />

serum (FBS) resemble human plasma!<br />

11. Before trypsinization confluent cells may be thoroughly washed (twice in 2–3 mL<br />

of PBS or serum-free medium) (see Note 10). Prewarm PBS <strong>and</strong> Trypsin-EDTA<br />

solution to 37°C in a water bath!<br />

12. To each dish treated with 1.5 mL of Trypsin-EDTA solution 6.5–7 mL of culture<br />

medium is added to give a total volume of 40 mL. Prewarm culture medium to<br />

37°C in a water bath!<br />

13. In total 18 dishes per condition are prepared. The final volume of culture medium<br />

used per dish is 9 mL. Once MDCK cells reach confluence serum-free medium is<br />

added. The final volume of serum-free medium used per dish is 4 mL. This gives<br />

a total volume of 70–72 mL per condition <strong>and</strong> corresponds to the appropriate<br />

processing volume for the Centricon ® Plus-70 centrifugal filter devices (see<br />

Note 18).<br />

14. The doubling time of MDCK strain II cells is one day. Confluence is reached<br />

after 3–4 days.<br />

15. Alternatively, use PBS instead of serum-free medium if costs are a major<br />

concern.<br />

16. TCA/acetone (27) may precipitate calcium-phosphate <strong>and</strong> small amino acids<br />

from the culture media. Very high contaminant concentrations may be achieved<br />

that extremely interfere with isoelectric focusing. Dialysis must not be used!<br />

Very high solute volumes are needed to remove salts <strong>and</strong> unspecific protein<br />

binding to the dialysis membrane may occur.


<strong>Sample</strong> <strong>Preparation</strong> of Culture Medium 127<br />

17. The 0.1 M PMSF stock solution is warmed up to 37°C in a water bath. PMSF<br />

is added before putting medium on ice to avoid precipitation. EDTA inhibits<br />

metalloproteases by chelation of free metal ions. PMSF inhibits serine proteases<br />

<strong>and</strong> some cysteine proteases. Inhibitor cocktails (for example Complete Mini,<br />

EDTA-free from Roche) must not be used! These cocktails contain protein- <strong>and</strong><br />

peptide-based inhibitors that can reach very high concentrations on ultrafiltration<br />

(up to 300X) <strong>and</strong> hence abundantly mask the secreted proteins present in the<br />

medium.<br />

18. Usage guidelines for Centricon ® Plus-70 centrifugal filter devices are given in<br />

the user guide (28). The filter material is made of a polyethersulfone Biomax<br />

membrane with a 5 kDa cut-off.<br />

19. The pH of 9.0 from Tris serves two functions: 1) it maximizes protein extraction<br />

at basic pH conditions (almost any protein is in deprotonated state); 2) minimizes<br />

protease activity. The Tris itself serves as a positively charged ion that helps in<br />

solubilization of proteins (see Note 6). Very basic proteins may be lost!<br />

20. On concentration protein precipitation may occur!<br />

21. When Reagent B is first added to Reagent A, turbidity is observed that quickly<br />

disappears on mixing to yield a clear, green color.<br />

22. Alternatively, wavelengths from 540–590 nm may be used with this<br />

method (34).<br />

23. Amount of albumin st<strong>and</strong>ards (see Table 1) used: F, 0.625 μg; E, 3.125 μg; D,<br />

6.25 μg; C, 12.5 μg; B, 18.75 μg, <strong>and</strong> A, 25 μg. The st<strong>and</strong>ard amount is referred<br />

to a volume of 25 μL. Average blank-corrected 550 nm absorbance values above<br />

0.8 must not be used for st<strong>and</strong>ard curve preparation!<br />

24. For example: (protein amount of unknown is 7 μg)/(volume of diluted sample<br />

is 25 μL) × (dilution factor is 20) = 5.6 mg/mL.<br />

25. A final volume of 450 μL is recommended by the supplier (Amersham<br />

Biosciences) for rehydration of one 24 cm Immobiline TM DryStrip. For first trial<br />

prepare a duplicate! The upper limit is twelve samples per run.<br />

26. Avoid trapping of air bubbles.<br />

27. Do not wear gloves during removal of protective cover sheet! The rubber material<br />

tends to stick to the “naked” gel <strong>and</strong> hence will damage it.<br />

28. Overlaying of IPG strips with paraffin oil reduces risk of urea crystallization<br />

during rehydration.<br />

29. In this case the paraffin oil evenly distributes the heat produced during IEF<br />

between the tray <strong>and</strong> the cooling plate.<br />

30. Do not use ddH 2O or tap water! The former leads to very low conductivity<br />

between electrode <strong>and</strong> IPG strip, the latter to very high.<br />

31. Each electrode has a side marked red or black.<br />

32. Program for 24 cm IPG pH 3–10 NL strips using the EPS 3501 XL power<br />

supply (adapted from Hoving (40)): Phase 1, 300 V, 1 Vh (0.006 h); Phase 2,<br />

300 V, 900 Vh (3 h); Phase 3, 3500 V, 9500 Vh (5 h), <strong>and</strong> Phase 4, 3500 V,<br />

52500 Vh (15 h). Current <strong>and</strong> power are set non-limiting (2 mA, 5 W). Phases<br />

1–4 are programmed in the gradient mode. The voltage will be ramping up to


128 Ambort et al.<br />

the maximum set in the phase, starting from zero in the first phase <strong>and</strong> in phases<br />

to follow from the end point of the phase before. Therefore in phases 1 <strong>and</strong> 3<br />

voltage is linearly increased <strong>and</strong> in phases 2 <strong>and</strong> 4 held constant. The current<br />

check option must be switched off!<br />

Acknowledgments<br />

The authors wish to acknowledge <strong>and</strong> thank Ursula Luginbühl for excellent<br />

technical assistance. This work was funded by the Swiss National Science<br />

Foundation (SNSF) (grant 3100A0-100772 to E.E.S.) <strong>and</strong> the European Science<br />

Foundation (ESF) Integrated Approaches for Functional Genomics (grant 0341<br />

to D. A.).<br />

References<br />

1. O’Farrell, P. H. (1975) High resolution two-dimensional electrophoresis of<br />

proteins. J. Biol. Chem. 250, 4007–021.<br />

2. Klose, J. (1975) Protein mapping by combined isoelectric focusing <strong>and</strong><br />

electrophoresis in mouse tissues. A novel approach to testing for induced point<br />

mutations in mammals. Humangenetik 26, 231–43.<br />

3. Lämmli, U. K. (1970) Cleavage of structural proteins during the assembly of the<br />

head of bacteriophage T4. Nature 227, 680–85.<br />

4. O’Farrell, P. Z., Goodman, H. M., O’Farrell, P. H. (1970) High-resolution twodimensional<br />

electrophoresis of basic as well as acidic proteins. Cell 12, 1133–42.<br />

5. Bjellqvist, B., Ek, K., Righetti, P. G., Gianazza, E., et al. (1982) Isoelectric focusing<br />

in immobilized pH gradients: principle, methodology <strong>and</strong> some applications. J.<br />

Biochem. Biophys. Methods 6, 317–39.<br />

6. Görg, A., Postel, W., Günther, S. (1988) The current state of two-dimensional<br />

electrophoresis with immobilized pH gradients. Electrophoresis 9, 531–46.<br />

7. Görg, A., Obermaier, C., Boguth, G., et al. (2000) The current state of twodimensional<br />

electrophoresis with immobilized pH gradients. Electrophoresis 21,<br />

1037–53.<br />

8. Görg, A., Weiss, W., Dunn, M. J. (2004) Current two-dimensional electrophoresis<br />

technology for proteomics. Proteomics 4, 3665–85.<br />

9. Matsudaira, P. (1987) Sequence from picomole quantities of proteins electroblotted<br />

onto polyvinylidene difluoride membranes. J. Biol. Chem. 262, 10035–38.<br />

10. Aebersold, R. H., Leavitt, J., Saavedra, R. A., Hood, L. E., Kent, S. B. (1987)<br />

Internal amino acid sequence analysis of proteins separated by one- or twodimensional<br />

gel electrophoresis after in situ protease digestion on nitrocellulose.<br />

Proc. Natl. Acad. Sci. 84, 6970–74.<br />

11. Rosenfeld, J., Capdevielle, J., Guillemot, J. C., Ferrara, P. (1992) In-gel digestion<br />

of proteins for internal sequence analysis after one- or two-dimensional gel<br />

electrophoresis. Anal. Biochem. 203, 173–79.


<strong>Sample</strong> <strong>Preparation</strong> of Culture Medium 129<br />

12. Yates 3rd, J. R., Speicher, S., Griffin, P. R., Hunkapiller, T. (1993) Peptide mass<br />

maps: a highly informative approach to protein identification. Anal. Biochem. 214,<br />

397–408.<br />

13. James, P., Quadroni, M., Carafoli, E., Gonnet, G. (1994) Protein identification in<br />

DNA databases by peptide mass fingerprinting. Protein Sci. 3, 1347–50.<br />

14. Cottrell, J. S. (1994) Protein identification by peptide mass fingerprinting. Pept.<br />

Res. 7, 115–24.<br />

15. Herbert, B. R., Sanchez, J.C., Bini, L. (1997) Two-dimensional electrophoresis:<br />

The state of the art <strong>and</strong> future directions, in Proteome Research: New Frontiers<br />

in Functional Genomics (Wilkins, M. R., Williams, K. L., Appel, R. D.,<br />

Hochstrasser, D. F., eds.), Springer, Berlin, pp. 13–33.<br />

16. Rabilloud, T., Adessi, C., Giraudel, A., Lunardi, J. (1997) Improvement of the<br />

solubilization of proteins in two-dimensional electrophoresis with immobilized pH<br />

gradients. Electrophoresis 18, 307–16.<br />

17. Link, A. J. (ed.) (1999) 2-D Proteome Analysis Protocols. Humana, Totowa, NJ.<br />

18. Westermeier, R. (2001) Electrophoresis in Practice, 3rd Edition, Wiley-VCH,<br />

Weinheim.<br />

19. Yates 3rd, J. R., Gilchrist, A., Howell, K. E., Bergeron, J. J. (2005) Proteomics of<br />

organelles <strong>and</strong> large cellular structures. Nature 6, 702–14.<br />

20. Lim, J. W. E., Bodnar, A. (2002) Proteome analysis of conditioned medium from<br />

mouse embryonic fibroblast feeder layers which support the growth of human<br />

embryonic stem cells. Proteomics 2, 1187–1203.<br />

21. Boraldi, F., Bini, L., Liberatory, S., Armini, A., et al. (2003) Normal human dermal<br />

fibroblasts: Proteomic analysis of cell layer <strong>and</strong> culture medium. Electrophoresis<br />

24, 1292–1310.<br />

22. Mears, R., Craven, R. A., Hanrahan, S., Totty, N., et al. (2004) Proteomic<br />

analysis of melanoma-derived exosomes by two-dimensional polyacrylamide gel<br />

electrophoresis <strong>and</strong> mass spectrometry. Proteomics 4, 4019–31.<br />

23. Prowse, A. B. J., McQuade, L. R., Bryant, K. J., Van Dyk, D. D., et al. (2005)<br />

A proteome analysis of conditioned media from human neonatal fibroblasts used<br />

in the maintenance of human embryonic stem cells. Proteomics 5, 978–89.<br />

24. Volmer, M. W., Stühler, K., Zapatka, M., Schöneck, A., et al. (2005) Differential<br />

proteome analysis of conditioned media to detect Smad4 regulated secreted<br />

biomarkers in colon cancer. Proteomics 5, 2587–2601.<br />

25. Eagle, H. (1959) Amino acid metabolism in mammalian cell cultures. Science 130,<br />

432–7.<br />

26. Richardson, J. C., Scalera, V., Simmons, N. L. (1981) Identification of two<br />

strains of MDCK cells which resemble separate nephron tubule segments. Biochim.<br />

Biophys. Acta 673, 26–36.<br />

27. Damerval, C., DeVienne, D., Zivy, M., Thiellement, H. (1986) Technical improvements<br />

in two-dimensional electrophoresis increase the level of genetic variation<br />

detected in wheat-seedling protein. Electrophoresis 7, 53, 54.<br />

28. http://www.millipore.com/userguides.nsf/dda0cb48c91c0fb6852567430063b5d6/6<br />

03b133b9b2a919c85256b3e0050b862/$FILE/P36006.pdf (User guide for<br />

Centricon ® Plus-70 centrifugal filter devices from Millipore)


130 Ambort et al.<br />

29. Bradford, M. M. (1976) A rapid <strong>and</strong> sensitive method for the quantitation of<br />

microgram quantities of protein utilizing the principle of protein-dye binding. Anal.<br />

Biochem. 72, 248–54.<br />

30. Ramagli, L. S., Rodriguez, L. V. (1985) Quantitation of microgram amounts<br />

of protein in two-dimensional polyacrylamide gel electrophoresis sample buffer.<br />

Electrophoresis 6, 559–63.<br />

31. Lowry, O. H., Rosebrough, N. J., Farr, A. L., R<strong>and</strong>all, R. J. (1951) Protein<br />

measurement with the Folin phenol reagent. J. Biol. Chem. 193, 265–95.<br />

32. Mokrasch, L. C., McGilvery, R. W. (1956) Purification <strong>and</strong> properties of fructose-<br />

1, 6-diphosphatase. J. Biol. Chem. 221, 909–17.<br />

33. Smith, R. K., Krohn, R. I., Hermanson, G. T., et al. (1985) Measurement of protein<br />

using bicinchoninic adic. Anal. Biochem. 150, 76–85.<br />

34. http://www.piercenet.com/files/1296dh4.pdf (Instructions for BCA TM Protein<br />

Assay Kit from Pierce)<br />

35. Langen, H., Roder, D., Juranville, J. F., Fountoulakis, M. (1997) Effect of protein<br />

application mode <strong>and</strong> acrylamide concentration on the resolution of protein spots<br />

separated by two-dimensional gel electrophoresis. Electrophoresis 18, 2085–90.<br />

36. Rabilloud, T., Valette, C., Lawrence, J. J. (1994) <strong>Sample</strong> application by ingel<br />

rehydration improves the resolution of two-dimensional electrophoresis with<br />

immobilized pH gradients in the first dimension. Electrophoresis 15, 1552–58.<br />

37. Sanchez, J. C., Rouge, V., Pisteur, M., Ravier, F., et al. (1997) Improved <strong>and</strong><br />

simplified in-gel sample application using reswelling of dry immobilized pH<br />

gradients. Electrophoresis 18, 324–27.<br />

38. Hoving, S., Voshol, H., van Oostrum, J. (2000) Towards high perfomance twodimensional<br />

gel electrophoresis using ultrazoom gels. Electrophoresis 21, 2617–21.<br />

39. Hoving, S., Gerrits, B., Voshol, H., Muller, D., et al. (2002) Preparative twodimensional<br />

gel electrophoresis at alkaline pH using narrow range immobilized<br />

pH gradients. Proteomics 2, 127–34.<br />

40. http://www1.amershambiosciences.com/applic/upp00738.nsf/vLookupDoc/319<br />

798244-C534/$file/80642960.pdf (Amersham 2-D electrophoresis h<strong>and</strong>book)


11<br />

<strong>Sample</strong> <strong>Preparation</strong> for Mass Spectrometry Analysis<br />

of Formalin-Fixed Paraffin-Embedded Tissue<br />

Proteomic Analysis of Formalin-Fixed Tissue<br />

Nicolas A. Stewart <strong>and</strong> Timothy D. Veenstra<br />

Summary<br />

One of the great hopes in biomedical research is that proteomic technology can be used<br />

to identify novel biomarkers for diseases such as cancer. The challenge to discovering<br />

biomarkers starts with sample collection <strong>and</strong> continues right through data acquisition <strong>and</strong><br />

bioinformatic analysis. Because the ultimate goal is to find indicators of human disease it<br />

is ideal to be able to study clinical samples. Unfortunately clinical samples such as serum,<br />

plasma, urine, <strong>and</strong> especially tissue biopsies are precious <strong>and</strong> are often difficult to obtain<br />

in sufficient quantities or numbers to conduct proteomic discovery studies. There exists,<br />

however, a vast archive of pathologically characterized clinical samples in the form of<br />

formalin fixed paraffin embedded tissue blocks. This chapter describes methods that have<br />

been developed to allow the proteins from these tissue samples to be excised in a form<br />

that is amenable for proteomic analysis by mass spectrometry.<br />

Key Words: Cancer biomarkers; formalin-fixed paraffin embedded tissue; mass<br />

spectrometry; proteomics; tissue microdissection.<br />

1. Introduction<br />

One of the greatest determinants on the survival rate from any cancer is<br />

the stage at which it is detected. The survival rate from cancers detected at an<br />

early stage is generally higher but decreases as the stage of detection increases.<br />

Therefore a major impetus in proteomics research is to identify biomarkers<br />

of early stage diseases. Many of these biomarker discovery efforts are aimed<br />

towards the use of biofluids such as serum <strong>and</strong> plasma. These sample types,<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

131


132 Stewart <strong>and</strong> Veenstra<br />

however, are inherently difficult to analyze <strong>and</strong> are often too precious to be<br />

given over to proteomic-type discovery studies that are continuing to evolve in<br />

both efficacy <strong>and</strong> success. Another complication is that although biomarkers<br />

that originate from the site of a tumor may have a high local concentration,<br />

by the time they become diluted within the circulatory system, their effective<br />

concentration in the acquired clinical sample may be vanishingly small. Ideally,<br />

it would be beneficial to identify a biomarker at the tumor level <strong>and</strong> determine<br />

if it could be translated to a marker detectable in serum or plasma. Tissue<br />

or tumor biopsies therefore, are ideal sample c<strong>and</strong>idates for the discovery of<br />

potential biomarkers. However, the feasibility of their use for such studies is<br />

hampered because of the fact that they are relatively difficult to obtain <strong>and</strong><br />

require careful storage <strong>and</strong> h<strong>and</strong>ling.<br />

Formalin-fixed, paraffin-embedded (FFPE) tissues, on the other h<strong>and</strong>,<br />

represent a vast resource for retrospective protein biomarker investigation.<br />

Formalin fixation <strong>and</strong> paraffin embedding of tissue is practiced in pathology<br />

labs worldwide as a st<strong>and</strong>ard processing method by which tissues can be<br />

stored <strong>and</strong> catalogued as stable entries. Unfortunately, the storage stability<br />

of these tissues partially arises from the high degree of covalently crosslinked<br />

proteins. Currently, immunohistochemistry (IHC) is the only published<br />

technology capable of providing protein information from these samples (1,2).<br />

In addition, because IHC requires a priori knowledge of individual proteins<br />

being analyzed, it is not a discovery-based technology.<br />

As with tissue biopsies, FFPE sections are heterogeneous in that they<br />

contain many different types of cells. To acquire a homogeneous population<br />

of cells, laser-capture microdissection (LCM) is used. This technique has the<br />

ability to directly isolate a user-defined population of cells from their tissue<br />

microenvironment (3). Although LCM of fresh tissue is widely practiced,<br />

outside of the interest in conducting tissue microdissection of FFPE tissues for<br />

mRNA extraction, microdissection of FFPE tissues is not widely practiced for<br />

extraction <strong>and</strong> analysis of soluble protein (4).<br />

Mass spectrometry (MS) is arguably the driving technology in discoverydriven<br />

proteomics. Dramatic technical improvements in MS instrumentation<br />

combined with the rapid growth of genomic <strong>and</strong> proteomic databases have<br />

enabled development of approaches to identify <strong>and</strong> quantify large numbers of<br />

proteins from complex samples such as serum <strong>and</strong> tissues (5). Combining LCM<br />

of FFPE tissues <strong>and</strong> MS has the potential for generating protein biomarker data<br />

necessary for discovering proteins that are key determinants or indicators of<br />

diseases such as cancer.<br />

A common misnomer is that MS identifies proteins in proteomic studies.<br />

Actually in its present form, MS is best suited to the identification of peptides<br />

that are produced from the enzymatic digestion of proteins. It is through


<strong>Sample</strong> <strong>Preparation</strong> for Mass Spectrometry Analysis 133<br />

the analysis of peptide surrogates that proteins are identified. Because of<br />

their size, peptides are very amenable to t<strong>and</strong>em MS (MS/MS) methods that<br />

produce partial amino acid sequence information that leads to their identification.<br />

Besides the advances made in MS instrumentation, its coupling with<br />

nanoflow reversed-phase liquid chromatography (nanoRPLC) has made the<br />

analysis of complex peptide mixtures possible. To compare the tryptic peptide<br />

abundances obtained from two different FFPE tissues, stable-isotope labeling<br />

using trypsin-mediated 16 O/ 18 O incorporation may also be employed.<br />

2. Materials<br />

2.1. Tissue Processing<br />

1. Mayer’s hematoxylin stain.<br />

2. Eosin stain.<br />

3. Graded ethanol solutions.<br />

4. SubX organic solvent (Surgipath Medical Industries, Richmond, IL).<br />

2.2. Protein Extraction<br />

1. 50% glycerol.<br />

2. LCM instrument.<br />

3. Low-binding microcentrifuge tubes.<br />

4. The Liquid Tissue-MS protein prep kit (Expression Pathology, Inc.,<br />

Gaithersburg, MD).<br />

2.3. Trypsin Digest<br />

1. Porcine sequencing grade modified trypsin (Promega, Madison, WI).<br />

2. Dithiothreitol (DTT, Sigma, St. Louis, MO).<br />

3. Trifluoroacetic acid (TFA) (≥ 98.0% pure).<br />

4. Incubator for the digest at 37°C in microcentrifuge tubes.<br />

5. High performance liquid chromatography (HPLC)-grade acetonitrile (ACN)<br />

(EMD Chemicals Inc., Gibbstown, NJ).<br />

6. C-18 reverse phase microcolumns (e.g. ZipTip®, Millipore, Billerica, MA).<br />

7. Conditioning, loading <strong>and</strong> eluting buffers for reverse phase desalting microcolumns:<br />

ACN, 0.1% TFA in ddH2O, <strong>and</strong> 60% ACN, 0.1% TFA solution, respectively.<br />

2.4. LC-MS/MS analysis of tryptic peptides<br />

1. HPLC system capable of NanoRPLC (e.g., Agilent 1100 capillary LC system,<br />

Agilent Technologies, Palo Alto, CA).<br />

2. Fused silica capillary column: 75 μm inner diameter × 360 outer diameter × 10<br />

cm long, slurry packed with C-18 silica-bonded stationary phase; 3 μm, 300 Å<br />

pore size.


134 Stewart <strong>and</strong> Veenstra<br />

3. Linear ion trap MS coupled to the NanoRPLC (e.g., LTQ, Thermo Electron, San<br />

Jose, CA).<br />

4. High resolution MS coupled to the NanoRPLC for 18 O isotope labeling analysis<br />

(e.g., LTQ-FTICR, Thermo Electron, San Jose, CA).<br />

5. Trifluroroacetic acid is diluted to 0.1% (v/v) <strong>and</strong> is used to resolubilize tryptic<br />

peptides before LC-MS/MS.<br />

6. A solution of 0.1 % (v/v) formic acid (FA) in ddH 2O (NANOPure Diamond water<br />

system, Barnstead International, Dubuque, IA) (Mobile Phase A) <strong>and</strong> a solution<br />

of 0.1% FA in HPLC-grade ACN (Mobile Phase B) are prepared for the gradient<br />

used in the reverse phase liquid chromatographic separation of tryptic peptides.<br />

2.5. Mass Spectrometry Analysis <strong>and</strong> Bioinformatic Analysis<br />

1. Accompanying software for the collection of the raw MS/MS data generated.<br />

2. Software to search raw data files to a database (e.g., SEQUEST).<br />

3. Methods<br />

3.1. Tissue Processing<br />

1. For tissue microdissection, 10 μm thick tissue sections are cut from FFPE whole<br />

mount tissue blocks <strong>and</strong> placed on coated slides.<br />

2. The section is then heated for 60 min at 58°C. To remove paraffin, treat the<br />

section with SubX organic solvent (Surgipath Medical Industries, Richmond, IL)<br />

twice for 5 min.<br />

3. The tissue is then rehydrated through multiple, graded ethanol solutions <strong>and</strong><br />

distilled water. Tissue is then counterstained with Mayer’s hematoxylin, <strong>and</strong><br />

dehydrated through graded ethanol solutions, <strong>and</strong> air-dried.<br />

3.2. Laser-capture Microdissection<br />

1. Rehydrate the tissue with 50% glycerol in water for 5 min before laser capture<br />

microdissection (LCM).<br />

2. Place the slide containing the tissue upside-down below the objective lens <strong>and</strong><br />

locate cellular regions with specific histological features of interest (see Note 1).<br />

3. Capture cells with the LCM instrument utilizing an excimer laser (MPB<br />

Technologies PSX-100) operating at the following conditions: 248 nm<br />

wavelength, 2.5 ns pulse, Emax= 5 mJ, repetition rate = 0.1–100 Hz (see Note 2).<br />

4. Captured cells within the selected area are then transferred into a 1.5 mL<br />

low-binding microcentrifuge receiving tube. Approx 100,000–200,000 cells are<br />

required for subsequent proteomic analysis (see Note 3).<br />

3.3. Protein Extraction <strong>and</strong> Trypsin digest<br />

1. Cells collected by microdissection for nano-reversed-phase liquid<br />

chromatography-t<strong>and</strong>em mass spectrometry (RPLC-MS/MS) analysis were


<strong>Sample</strong> <strong>Preparation</strong> for Mass Spectrometry Analysis 135<br />

processed using proprietary reagents according to the manufacturer’s recommendations<br />

(Liquid Tissue, Expression Pathology Inc., Gaithersburg, MD).<br />

2. The material for nanoRPLC-MS/MS analysis was suspended in 20 μL of Liquid<br />

Tissue reaction buffer, <strong>and</strong> incubated for 90 min at 95°C, followed by cooling<br />

on ice for 3 min.<br />

3. DTT is added to a final concentration of 10 mM.<br />

4. Heat samples for 5 min at 95°C, <strong>and</strong> let cool to room temperature.<br />

5. Trypsin (15–18 units) is added <strong>and</strong> the sample is incubated at 37°C for 18 h.<br />

6. The resulting proteome digestates may be stored at –20°C until analysis.<br />

7. Tryptic peptides generated from the comparative FFPE samples are desalted<br />

using C-18 microcolumns. Microcolumns are conditioned by aspirating <strong>and</strong><br />

dispensing with conditioning buffer (10 μL, 3×), followed by loading buffer<br />

(10 μL 3×).<br />

8. Peptide samples are re-solubilized in loading buffer (10 μL) <strong>and</strong> washed with<br />

loading buffer (10 μL 3×).<br />

9. Peptides are eluted from the microcolumn with the elution buffer (10 μL) into<br />

microcentrifuge tubes, <strong>and</strong> lyophilized.<br />

10. Peptide samples are re-solubilized in loading buffer (10 μL) <strong>and</strong> transferred to<br />

vials for autosampler.<br />

3.4. Trypsin-mediated 18O-Labeling 1. For isotope labeling, the protein extracts from equivalent numbers of cells<br />

16 18 microdissected from FFPE tissues are reconstituted separately in H2 O <strong>and</strong> H2 O<br />

each containing 20% methanol (v/v).<br />

2. Sequencing grade trypsin is resuspended in the appropriately labeled water (i.e.<br />

16 18 H2 OorH2 O), <strong>and</strong> added to the related sample at an enzyme to protein ratio<br />

of 1:20.<br />

3. Incubate at 37°C for 16 h. After this time, an additional equivalent aliquot of the<br />

trypsin is added <strong>and</strong> the samples incubated at 37°C for an additional 6h(see<br />

Note 7).<br />

4. TFA is added to a final concentration of 0.4% (v/v) to stop the reaction (see Note 8).<br />

5. The differentially labeled proteome samples are pooled <strong>and</strong> lyophilized to dryness.<br />

6. <strong>Sample</strong>s are resolubilized in loading buffer (10 μL) <strong>and</strong> transferred to vials for<br />

autosampler.<br />

3.5. Nanoflow RPLC-MS/MS Analysis<br />

1. Nanoflow RPLC is performed on a capillary LC system coupled online to a ion<br />

trap MS or an MS capable of high resolution (i.e., 100,000) for quantitation of<br />

16 18 O/ O-labeled samples (see Note 9).<br />

2. Wash column for 30 min with 98% mobile phase A at a flow rate of 0.5 μL/min<br />

prior to sample injection.<br />

3. Inject sample (typically 1–6 μL) onto the column.


136 Stewart <strong>and</strong> Veenstra<br />

4. Elute peptides using a linear gradient of 2–40% mobile phase B in 110 min, then<br />

to 98% mobile phase B in an additional 30 min, all at a constant flow rate of 0.25<br />

μL/min.<br />

3.6. Mass Spectrometry Analysis <strong>and</strong> Bioinformatic Analysis<br />

1. The MS is operated in a data dependent MS/MS mode in which each full MS scan is<br />

followed by five MS/MS scans of the five most abundant peptide molecular ions.<br />

2. Subject peptides to collision-induced dissociation (CID) using a normalized<br />

collision energy of 35% (see Note 10). The heated capillary temperature <strong>and</strong><br />

electrospray voltage of the mass spectrometer are typically set at 160°C <strong>and</strong><br />

1.5 kV, respectively.<br />

3. Although data collection may be dictated based on the available mass to charge<br />

(m/z) range of the instrument, it is typically collected over a broad range of<br />

400–2,000.<br />

4. Because the starting amount of protein obtained from the LCM cells is low,<br />

multidimensional fractionation prior to MS analysis is inefficient owing to sample<br />

h<strong>and</strong>ling losses associated with chromatography.<br />

5. To maximize the number of peptides identified, segmented precursor selection<br />

scan ranges (i.e., gas phase fractionation in the m/z dimension, GPFm/z) are<br />

used. The following overlapping m/z intervals may be used as a guideline:<br />

m/z 400–605, 595–805, 795–1005, 995–1,205, 1,195–1,405, 1,395–1,605,<br />

1,595–1,805, 1,795–2,000, 400–805, <strong>and</strong> 795–1,200. Data for the 16O/ 18O-labeled experiments are acquired using FTICR detection in centroid mode for the full MS<br />

scan (m/z 400–2000) at 100,000 resolution followed by MS/MS of the top five<br />

most abundant molecular ions detected.<br />

6. The t<strong>and</strong>em mass spectra are searched against the UniProt human<br />

proteomic database from the European Bioinformatics Institute (http://<br />

www.ebi.ac.uk/integr8) using a combination of database <strong>and</strong> searching algorithm<br />

software (e.g. SEQUEST). Peptides are searched using fully tryptic cleavage<br />

constraints <strong>and</strong> a dynamic 4.008 amu modification on the C-terminus for when<br />

18O isotope labeling analysis. When using SEQUEST, a legitimately identified<br />

peptide should have cross correlation (Xcorr) scores of 1.9 for [M+H] 1+ , 2.2 for<br />

[M+2H] 2+ <strong>and</strong> 3.5 for [M+3H] 3+ <strong>and</strong> a minimum delta correlation score (Cn) of 0.08<br />

4. Notes<br />

1. Microdissection is best performed using software-directed laser pulses to strike at<br />

a constant velocity <strong>and</strong> rate throughput over the previously defined <strong>and</strong> mapped<br />

cellular regions.<br />

2. Target slides are optically transparent quartz coated with an energy transfer<br />

coating with the exact dimensions of a st<strong>and</strong>ard histology glass slide. The slide<br />

stage is a computer-controlled, XY translation stage. A 1/8 beam-splitter is


<strong>Sample</strong> <strong>Preparation</strong> for Mass Spectrometry Analysis 137<br />

used to split the laser to an energy meter with the remaining beam traveling<br />

to an ultraviolet reflective mirror <strong>and</strong> directed down (-Z) to a ×10 microscope<br />

objective (LMU-10X-UVR, OFR). The objective focuses the laser onto the slide<br />

<strong>and</strong> the LCM process is observable using a confocally aligned CCD camera.<br />

3. Because many FFPE tissues have been in storage for years, if not decades, an<br />

obvious concern when conducting proteomic investigations of these samples is<br />

the effect of long-term storage. Presently there are not enough studies on this<br />

topic to make any solid conclusions, however, no obvious detrimental effect has<br />

been observed in those studies that have examined FFPE tissues using MS (8,9).<br />

One of the studies showed that formalin-fixation <strong>and</strong> storage does not result in<br />

an inordinate degree of oxidation to methionyl <strong>and</strong> cysteinyl residues nor were<br />

any adverse effects on the tryptic digestion efficiency observed (8).<br />

4. Unfortunately the maximum number of cells that can typically be microdissected<br />

from FFPE tissues is on the order of 200,000. Although this number will<br />

vary depending on the tissue <strong>and</strong> its heterogeneity, it typically does not yield<br />

enough protein to employ multidimensional fractionation before MS analysis.<br />

The protein yield for a typical experiment is on the order of 10–20 μg, whereas<br />

typically 200 μg is required for a multidimensional fractionation consisting of<br />

strong cation exchange prior to RPLC to be employed.<br />

5. There are experimental <strong>and</strong> data analysis issues related to quantitative proteomics<br />

whether it is done using O 16 /O 18 labeling or subtractively (i.e., when the number<br />

of peptides identified from one proteome sample is compared to that identified<br />

in another). The trypsin-mediated incorporation of O 18 is not always absolutely<br />

complete because of the incomplete exchange at the peptide’s carboxy-terminus.<br />

Although no absolute reason for this effect has been established, it may be<br />

because of the low abundance of the peptide within the complex mixture. Manual<br />

analysis of the MS spectrum should always be conducted when a potential<br />

interesting abundance change is suggested. When using a subtractive proteomics<br />

approach, the precision level is such that abundance changes below three-fold<br />

are not considered reliable.<br />

6. A constant issue when dealing with the identification of large numbers of<br />

peptides from complex mixtures is the false positive rate. It is impossible to<br />

orthogonally validate all of the peptides/proteins identified in these studies,<br />

therefore acceptable filtering criteria are necessary when evaluating correct<br />

identifications. SEQUEST is a commonly used software program used for identifying<br />

peptides based on t<strong>and</strong>em MS spectra. The filtering criteria based on X corr<br />

<strong>and</strong>C n scores are provided as a guideline only. In bioinformatic analyses, they<br />

have been shown to provide a false positive rate of approx 1.5% (5).<br />

7. Because the proteins are extracted from the FFPE tissues as tryptic peptides, the<br />

role of trypsin during this step is to enzymatically exchange the C-terminal carboxyl<br />

oxygen atoms with the appropriate oxygen (i.e., 16 Oor 18 O) isotope (6).<br />

8. An alternative means to stop the trypsin-mediated incorporation of 18 O is to boil<br />

the sample at the end of the digest. This is acceptable, but the sample should


138 Stewart <strong>and</strong> Veenstra<br />

not be boiled after addition of acid as deamidation of asparagine or glutamine<br />

residues may occur.<br />

9. An MS capable of high resolution (e.g., FTICR-MS) is necessary for accurately<br />

quantifying the different isotopically-labeled counterparts from the two proteome<br />

samples to be compared.<br />

10. To minimize the selection of peptides that have already been subjected to CID,<br />

dynamic exclusion is used. While this is a user defined value, 90 s. is typical.<br />

Acknowledgments<br />

This project has been funded in whole or in part with federal funds from the<br />

National Cancer Institute, National Institutes of Health, under Contract N01-<br />

CO-12400. The content of this publication does not necessarily reflect the views<br />

or policies of the Department of Health <strong>and</strong> Human Services, nor does mention<br />

of trade names, commercial products, or organization imply endorsement by<br />

the United States Government.<br />

References<br />

1. MacIntyre, N. (2001) Unmasking antigens for immunohistochemistry. Br. J. Biomed.<br />

Sci. 58, 190–96.<br />

2. Shi, S. R., Cote, R. J. <strong>and</strong> Taylor, C. R. (2001) Antigen retrieval techniques: current<br />

perspectives. J. Histochem. Cytochem. 49, 931–37.<br />

3. Emmert-Buck, M. R., Bonner, R. F., Smith, P. D., et al. (1996) Laser capture<br />

microdissection. Science 274, 998–1001.<br />

4. Gillespie, J. W., Best, C. J., Bichsel, V. E., et al. (2002) Evaluation of non-formalin<br />

tissue fixation for molecular profiling. Am. J. Pathol. 160, 449–57.<br />

5. Peng, J. <strong>and</strong> Gygi, S. P. (2001) Proteomics: the move to mixtures. J. Mass Spectrom.<br />

36, 1083–91.<br />

6. Yao, X., Freas, A., Ramirez, J., Demirev, P. A., <strong>and</strong> Fenselau, C. (2001) Proteolytic<br />

18O labeling for comparative proteomics: model studies with two serotypes of<br />

adenovirus. Anal. Chem. 73, 2836–42.<br />

7. Yates, J. R. III, Eng, J. K., McCormack, A. L., <strong>and</strong> Schieltz, D. (1995) Method to<br />

correlate t<strong>and</strong>em mass spectra of modified peptides to amino acid sequences in the<br />

protein database. Anal. Chem. 67, 1426–36.<br />

8. Hood, B. L., Darfler, M. M., Guiel, T. G., et al. (2005) Proteomic analysis of<br />

formalin-fixed prostate cancer tissue. Mol. Cell. Proteomics 4, 1741–53.<br />

9. Crockett, D. K., Lin, Z., Vaughn, C. P., Lim, M. S., Elenitoba-Johnson, K. S.<br />

(2005) Identification of proteins from formalin-fixed paraffin-embedded cells by<br />

LC-MS/MS. Lab. Invest. 85, 1405–15.


12<br />

Metalloproteomics in the Molecular Study of Cell<br />

Physiology <strong>and</strong> Disease<br />

Hermann-Josef Thierse, Stefanie Helm, <strong>and</strong> Patrick Pankert<br />

Summary<br />

Physical <strong>and</strong> chemical stresses as well as metal-related diseases can disrupt the<br />

normal trafficking of metal ions. Moreover, homeostatic imbalance of such metal ions<br />

may modulate essential cellular functions (including signal transduction pathways), may<br />

catalyze oxidative damage, <strong>and</strong> may affect the folding of nascent proteins. Here we<br />

describe a new qualitative subproteomic method for the detection, isolation, <strong>and</strong> identification<br />

of metal-interacting proteins. Combining both classical immobilized metal ion<br />

affinity chromatography (IMAC) <strong>and</strong> modern proteomic techniques (e.g., two dimensional<br />

gel electrophoresis [2-DE]), metal-specific proteins have been successfully isolated <strong>and</strong><br />

identified to define a metalloproteome. These metal-specific proteomes may give new<br />

insights into metal-related pathophysiological processes, such as the allergic reaction to<br />

nickel, which represents the most common form of human contact hypersensitivity.<br />

Key Words: 2-DE; 2-dimensional gel electrophoresis; disease proteomics; IMAC;<br />

immobilized metal ion affinity chromatography; metal affinity; metalloproteome; nickel;<br />

nickel allergy; nitrilotriacetic acid.<br />

1. Introduction<br />

Because immobilized metal ion affinity chromatography (IMAC) was first<br />

developed for metal-specific protein isolation by Porath et al. (1,2), a large<br />

number of IMAC protocols have been developed. All IMAC protocols share<br />

the same principal that proteins with exposed histidine <strong>and</strong> cysteine side chains<br />

have a distinct affinity for certain metals, like Ni2+ ,Co2+ ,Zn2+ ,Cu2+ ,Fe3+ ,<br />

or Mn2+ (3,4). Depending on the chosen metal-chelating group <strong>and</strong> metal ion<br />

more or fewer coordination sites are accessible for potential protein binding.<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

139


140 Thierse et al.<br />

In general, binding affinity of proteins to metal ions is dependent on the<br />

amino acid composition of a given protein, with histidine showing the highest<br />

metal affinity followed by tryptophan, phenylalanine, tyrosine, <strong>and</strong> cysteine<br />

(5). However, the affinity between a metal ion <strong>and</strong> an amino acid is also highly<br />

dependent on the specific metal ion itself, with Fe 3+ , for example, having the<br />

highest affinity to carboxyl- <strong>and</strong> phosphate-groups. Besides the amino acid<br />

sequence <strong>and</strong> the distribution of a certain amino acid, surface characteristics<br />

<strong>and</strong> protein folding (3-dimensional structure) are additional important parameters<br />

for binding affinity. In an optimal column or bead-based performance<br />

the selected metal is strongly held by the matrix bound metal-chelating group,<br />

e.g., nitrilotriacetic acid (NTA) or iminodiacetic acid (IDA), still leaving metal<br />

coordination sites available to interact with the metal-specific protein lig<strong>and</strong>.<br />

Tetradentate NTA, for example, was found to bind Ni 2+ with three carboxyl<br />

groups <strong>and</strong> one nitrogen (6), leaving two other lig<strong>and</strong> positions accessible to<br />

Ni 2+ -specific proteins or recombinant 6His-tagged proteins (7). Interestingly,<br />

when compared to the dissociation constant (K d) of most antibody bindings (K d<br />

from10 −5 M to 10 −12 M), 6His-tagged protein to Ni 2+ K d has been shown to be<br />

relatively high, 10 −13 M at pH 8 (8,9). Moreover, Ni-NTA itself has been shown<br />

to be stable over a pH range of 2.5–13 <strong>and</strong> withst<strong>and</strong>s extreme conditions like<br />

2% SDS <strong>and</strong> 100% ethanol.<br />

Today, IMAC is not only commonly used for purification of the mentioned<br />

recombinant 6His-tagged proteins or the evaluation of protein folding <strong>and</strong><br />

endotoxin removal from protein preparations, but also advantageous in the<br />

enrichment of phosphopeptides (10). Reversible protein phosphorylation (e.g.,<br />

at Ser, Thr, <strong>and</strong> Tyr residues) is one of the most important post-translational<br />

modifications, controlling signal transduction pathways that direct cellular<br />

activation, differentiation, <strong>and</strong> proliferation, as well as apoptosis (11). Because<br />

phosphopeptides are acidic <strong>and</strong> show strong binding characteristics to some<br />

metal ions (Fe 3+ ,Ga 3+ , <strong>and</strong> Al 3+ ) different IMAC protocols have been introduced<br />

for isolation of phosphopeptides (10,12). Among the most recently<br />

described phosphopeptide IMAC methods are titanium dioxide (TiO 2) phosphopeptide<br />

enrichment (13,14), <strong>and</strong> metal oxide affinity chromatography (MOAC)<br />

where the affinity of the phosphate group for Al(OH) 3 is exploited (15). For<br />

detailed information on IMAC phosphopeptide enrichment see Corthals et al.,<br />

2005 <strong>and</strong> Ueda et al., 2003 (5,10).<br />

Affecting up to 15% of the women in industrialized countries, human<br />

nickel (Ni) allergy represents one of the major metal-related diseases in the<br />

human population. However, according to several studies transition metal Ni<br />

is two-faced. Nickel is considered as a beneficial physiological agent used for<br />

essential functions as an ultra trace element. Conversely, Nickel can act as a<br />

pathological agent by interacting directly with DNA or DNA-binding proteins


Metalloproteomics in the Molecular Study of Cell Physiology 141<br />

or metal-specific T cells, thus causing cellular toxicity or a metal-specific<br />

allergic contact dermatitis (ACD). To elucidate such physiological <strong>and</strong> diseaserelated<br />

molecular processes, a new qualitative subproteomic method, combining<br />

IMAC <strong>and</strong> 2-dimensional gel electrophoresis (2-DE), has been developed to<br />

enrich <strong>and</strong> identify Ni 2+ -interacting proteins from different human cell types<br />

(e.g., human antigen presenting cells, keratinocytes). After affinity binding of<br />

metal-interacting proteins to Ni-NTA beads, followed by stringent washing<br />

steps <strong>and</strong> elution with imidazole, proteins are applied to modern 2-DE using<br />

immobilized pH-gradients (16–19). In a typical proteomic workflow, proteins<br />

are identified by mass spectrometry to define the metalloproteome. Complementary<br />

methods such as the use of recombinant proteins <strong>and</strong> metal detection<br />

by atomic absorption spectrometry allow confirmation of direct metal-protein<br />

interactions (20). Thus, with a clear differential pattern to Cu-binding proteins<br />

in human liver cells, Ni-NTA affinity enrichment of proteins from human<br />

B-cell lysates resulted in the subproteomic identification of so far unknown<br />

Ni-interacting proteins (Fig. 1) (21–23). Quite unexpectedly, 16 of these Niinteracting<br />

proteins were found to belong to the group of stress-inducible<br />

heatshock proteins or chaperonins, including HSP-60, HSP-70, BIP, <strong>and</strong> the<br />

high-molecular heterooligomeric complex of TriC/CCT (21). Thus Ni 2+ ,in<br />

addition to the induced formation of T cell epitopes recognizable by the<br />

A B<br />

Protein solution<br />

cell lysate<br />

0.1% Triton X-100<br />

Ni-NTA-beads<br />

1h incubation, 4°C<br />

Washing steps<br />

500 mM NaCl (high salt)<br />

137 mM NaCl (low salt)<br />

Elution of Ni-interacting<br />

proteins<br />

250 mM imidazole<br />

2-DE, staining, spot<br />

picking, trypsinization<br />

MW<br />

pH 4 pH 7<br />

Fig. 1. Ni-affinity enriched proteins were isolated from human antigen presenting<br />

cells (2* 10 7 cells) as demonstrated in the subproteomic workflow (A) <strong>and</strong> analyzed<br />

by 2-DE (B, silver staining), <strong>and</strong> subsequently identified by mass spectrometry (for<br />

details of mass spectrometric protein identification see (21)).


142 Thierse et al.<br />

acquired immune system, intimately interacts with essential constituents of the<br />

innate defense system, thereby linking both arms of the immune system. In<br />

summary, the development <strong>and</strong> combined usage of new metal-specific <strong>and</strong><br />

proteomic techniques gives new insights into metal-related metabolic pathways<br />

<strong>and</strong> frequent metal-specific disorders, like human nickel ACD, leading to<br />

improved strategies in diagnosis <strong>and</strong> therapy of such environment-induced<br />

disease (21,24,25).<br />

2. Materials<br />

2.1. Cell Culture <strong>and</strong> Lysis<br />

1. RPMI 1640 medium for human antigen presenting cells containing 10% fetal calf<br />

serum, 2 mM l-glutamine, 1 mM sodium pyruvate, nonessential amino acids <strong>and</strong><br />

25 mM HEPES buffer (all from Gibco BRL Life Technologies, Paisley, UK).<br />

For primary keratinocytes Keratinocyte Basal Medium 2 was supplemented with<br />

SupplementPack/Keratinocyte Growth Medium 2 (KGM) (Promocell, Heidelberg,<br />

Germany).<br />

2. A solution of trypsin (0.1%) <strong>and</strong> ethylenediamine tetraacetic acid (EDTA) (0.02%)<br />

(Biochrom, Berlin, Germany).<br />

3. Phosphate buffered saline (D-PBS) (Invitrogen, Karlsruhe, Germany).<br />

4. Fetal calf serum (FCS) (Biochrom, Berlin, Germany).<br />

5. Lysis buffer (0.1% Triton): 137 mM NaCl, 20 mM Tris-HCl, 10% (v/v) glycerol,<br />

0.1% (v/v) Triton X-100, pH 8.2. Add protease inhibitor cocktail tablets,<br />

“complete mini, EDTA free” (Roche, Mannheim, Germany) before use. Store in<br />

aliquots at –20°C (see Note 1).<br />

2.2. Isolation of Nickel-Binding Proteins<br />

1. Nickel-nitrilotriacetic acid (Ni-NTA) Magnetic Agarose Beads 5% suspension,<br />

with binding capacity: 300 μg/mL (Qiagen, Hilden, Germany).<br />

2. High salt lysis buffer: 500 mM NaCl, 20 mM Tris-HCl, pH 8.2, 10% (v/v)<br />

Glycerol, 0.1% Triton X-100.<br />

3. Low salt lysis buffer: 137 mM NaCl, 20 mM Tris-HCl, pH 8.2, 10% (v/v)<br />

Glycerol, 0.1% Triton X-100.<br />

4. Imidazole solution: 250 mM imidazole (Sigma, Taufkirchen, Germany) in distilled<br />

water.<br />

5. Magnetic device for 1.5 mL Eppendorf tubes (Qiagen, Hilden, Germany).<br />

6. St<strong>and</strong>ard 2-D electrophoresis equipment.<br />

2.3. Silver Staining of Metal-Affinity Enriched Proteins Compatible<br />

for Mass Spectrometry (MS)<br />

Several protocols of 2-dimensional gel electrophoresis (2-DE) are useful<br />

for detection of IMAC separated proteins, without negatively affecting the


Metalloproteomics in the Molecular Study of Cell Physiology 143<br />

principal method described here (16,17,21). Nevertheless, after separating<br />

affinity-enriched proteins by 2-DE a protein staining method compatible for<br />

mass spectrometric analysis has to be applied. Following protein gel scanning,<br />

e.g., with the LabScan Image Scanner (GE Healthcare, München, Germany) or<br />

the new laser scanner FLA 5100 (FUJIFILM Life Science, Düsseldorf), spot<br />

picking can be performed by h<strong>and</strong> or automatically using the PROTEINEER<br />

spII system (Bruker Daltonics, Bremen, Germany). Subsequent MALDI mass<br />

spectra can be recorded e.g., with a Ultraflex MALDI-TOF spectrometer<br />

(Bruker Daltronics, Bremen, Germany) equipped with a 337 nm nitrogen laser<br />

(for details see (21)).<br />

1. Thiosulfate solution: 0.02% (w/v) sodium thiosulfate pentahydrate (Merck,<br />

Darmstadt, Germany).<br />

2. Silver staining solution: 0.2% (w/v) silver nitrate (Merck, Darmstadt, Germany),<br />

0.02% formaldehyde solution (34%) (J.T. Baker, Deventer, NL).<br />

3. Developer: 3% (w/v) sodium carbonate (J.T. Baker, Deventer, NL), 0.05%<br />

formaldehyde solution (37%) (J.T. Baker, Deventer, NL), 0.0005% thiosulfate<br />

solution.<br />

4. Stopping solution (suitable for mass spectrometry) (see Note 2): 50% methanol<br />

(Merck, Darmstadt, Germany), 12% acetic acid (glacial) (Merck, Darmstadt,<br />

Germany).<br />

5. Stopping solution (not suitable for mass spectrometry): 0.5% glycine (Roth,<br />

Karlsruhe, Germany).<br />

6. Ethanol.<br />

3. Methods<br />

3.1. Preparing <strong>Sample</strong>s for Isolation of Nickel-Binding Proteins<br />

1. Culture of immune cells in RPMI medium <strong>and</strong>/or primary keratinocytes in KGM<br />

in 10 mm tissue culture dish until passage 3 or earlier.<br />

2. When e.g., keratinocytes are confluent, wash with PBS <strong>and</strong> incubate with 3 mL<br />

of trypsin/EDTA for 5 min at 37°C. Rinse cells with a pasteur pipet <strong>and</strong> transfer<br />

into a tube supplied with 6 mL 10% FCS in PBS. Rinse the dish with additional<br />

3 mL of 10% FCS <strong>and</strong> transfer it into the tube.<br />

3. Wash cells 3× with cold PBS <strong>and</strong> count the cells.<br />

4. Resuspend cells in lysis buffer (2* 107 /mL), incubate at 4°C for 1 h with gentle<br />

shaking.<br />

5. Clarify the lysate by centrifugation (20,000g, 10 min, 4°C). (see Note 3)<br />

6. The supernatant can be stored in aliquots at –20°C or –80°C.<br />

3.2. Isolation of Nickel-Binding Proteins<br />

1. Resuspend the Ni-NTA Magnetic Agarose Beads by vortexing or pipeting.<br />

2. Incubate 150 μL Ni-NTA Beads with 1 mL lysate (recommended ratio) by rotation<br />

for2hat4°Cina1.5-mL Eppendorf tube (see Note 4).


144 Thierse et al.<br />

3. Insert the tube into a magnetic device for 1.5-mL Eppendorf tubes <strong>and</strong> remove<br />

supernatant (see Note 5).<br />

4. Wash pellet 2× with high salt lysis buffer, once with low salt lysis buffer. The<br />

pellet has to be resuspended thoroughly during each wash. Discard supernatants<br />

(see Note 6).<br />

5. For elution add the imidazole solution to the pellet <strong>and</strong> incubate for 10 min at<br />

room temperature. Use a volume equal to the original volume of Ni-NTA beads.<br />

6. Insert the tube into a magnetic device for 1.5-mL Eppendorf tubes <strong>and</strong> carefully<br />

transfer the eluate into a fresh tube. Store aliquots at –80°C.<br />

7. For first-dimension isoelectric focussing sample (e.g., 25 μg) can be applied by<br />

including it in the rehydration solution of the IPG strip, followed by st<strong>and</strong>ard<br />

protocols of 2-D electrophoresis.<br />

3.3. Silver Staining of Metal-Affinity Enriched Proteins Compatible<br />

for Mass Spectrometry (MS)<br />

After 2-DE of metal-affinity enriched proteins spots have to be stained with<br />

staining protocols adapted to mass spectrometric analysis (for details see (21)).<br />

Silver staining has to be performed in glass dishes <strong>and</strong> see also Notes 7–11.<br />

1. Fix the gel in 40% ethanol/10% (v/v) acetic acid for at least 1 h.<br />

2. Wash 3× in 30% (v/v) ethanol for 20 min.<br />

3. Reduce the gel in thiosulfate solution for 1 min.<br />

4. Wash 3× in distilled water for 20 sec.<br />

5. Stain the gel in staining solution for 20 min.<br />

6. Wash 3× in distilled water for 20 sec.<br />

7. Develop in developer for 3–5 min.<br />

8. Wash 3x in distilled water for 30 sec.<br />

9. Stop the reaction in stopping solution for 5 min.<br />

10. Wash 2× in distilled water for 30 sec <strong>and</strong> an additional time for 15 min.<br />

11. Gel can be stored in 1% (v/v) acetic acid or shrink-wrap. An example result is<br />

demonstrated in Fig. 1.<br />

Alternatively, gels (e.g., loaded with 100 μg protein concentration) may<br />

be stained by MS-compatible Coomassie staining, e.g., according to Jungblut<br />

et al. (26).<br />

4. Notes<br />

1. Freshly prepared lysis buffer can be stored at 4°C for several months under<br />

sterile conditions.<br />

2. The color of the gel deepens after a while.<br />

3. At this point a determination of the protein concentration is often useful.<br />

4. Incubation of the cell-lysate with Ni-NTA Magnatic Agarose Beads can be<br />

extended to overnight incubation.


Metalloproteomics in the Molecular Study of Cell Physiology 145<br />

5. The supernatant contains all proteins that do not bind to Nickel <strong>and</strong> maybe<br />

additional Nickel-binding Proteins. Therefore, store the supernatant at –20°C or<br />

–80°C just in case.<br />

6. Supernatants of the washing steps can be stored at –20°C or –80°C just in case.<br />

7. All steps should be performed at room temperature with gentle shaking.<br />

8. Because the silver stain is very sensitive, nitrile gloves are recommended. Latex<br />

gloves may leave fingerprints on the gel.<br />

9. The amount of fluid per gel depends on the size of the gel <strong>and</strong> the staining<br />

dishes. Approximately 250 mL for 20 × 20 cm gels is sufficient.<br />

10. The gel becomes less flexible through the staining procedure <strong>and</strong> tears very<br />

easily. To transport the gel safely onto the scanning device, very careful h<strong>and</strong>ling<br />

is required.<br />

11. All solutions, containing methanol or silver nitrate have to be disposed of<br />

according to the directions given by the local authorities.<br />

Acknowledgments<br />

We thank Doris Wild <strong>and</strong> Stefanie Eikelmeier for excellent technical assistance,<br />

<strong>and</strong> Dr. Ian Haidl, Depts. of Pediatrics, Microbiology <strong>and</strong> Immunology,<br />

Halifax, Canada, for very careful reading of the manuscript. This work<br />

was supported in part by the L<strong>and</strong>esstiftung Baden-Wüerttemberg, Germany,<br />

Forschungsprogramm “Allergologie” by grant P-LS-AL/26 (to HJT), <strong>and</strong> the<br />

European Union, as part of the project Novel Testing Strategies for In Vitro<br />

Assessment of Allergens (Sens-it-iv), LSHB-CT-2005 – 018681, (www.sensit-iv.eu).<br />

References<br />

1. Porath, J., Carlsson, J., Olsson, I., <strong>and</strong> Belfrage, G. (1975) Metal chelate affinity<br />

chromatography, a new approach to protein fractionation. Nature. 258, 598–9.<br />

2. Porath, J. (1992) Immobilized metal ion affinity chromatography. Protein Expr.<br />

Purif. 3, 263–81.<br />

3. Mondal, K. <strong>and</strong> Gupta, M. N. (2006) The affinity concept in bioseparation: evolving<br />

paradigms <strong>and</strong> exp<strong>and</strong>ing range of applications. Biomol. Eng. 23, 59–76.<br />

4. Sun, X., Chiu, J. F., <strong>and</strong> He, Q. Y. (2005) Application of immobilized metal<br />

affinity chromatography in proteomics. Expert Rev. Proteomics. 2, 649–57.<br />

5. Ueda, E. K., Gout, P. W., <strong>and</strong> Morganti, L. (2003) Current <strong>and</strong> prospective applications<br />

of metal ion-protein binding. J. Chromatogr. A. 988, 1–23.<br />

6. Hochuli, E., Dobeli, H., <strong>and</strong> Schacher, A. (1987) New metal chelate adsorbent<br />

selective for proteins <strong>and</strong> peptides containing neighbouring histidine residues. J.<br />

Chromatogr. 411, 177–84.<br />

7. Terpe, K. (2003) Overview of tag protein fusions: from molecular <strong>and</strong> biochemical<br />

fundamentals to commercial systems. Appl. Microbiol. Biotechnol. 60, 523–33.


146 Thierse et al.<br />

8. Harlow, E., <strong>and</strong> Lane, D. (1988) Antibodies: A Laboratory Manual, Cold Spring<br />

Harbor Laboratory, Cold Spring Harbor, NY.<br />

9. Schmitt, J., Hess, H., <strong>and</strong> Stunnenberg, H. G. (1993) Affinity purification of<br />

histidine-tagged proteins. Mol. Biol. Rep. 18, 223–30.<br />

10. Corthals, G. L., Aebersold, R., <strong>and</strong> Goodlett, D. R. (2005) Identification of<br />

phosphorylation sites using microimmobilized metal affinity chromatography.<br />

Methods Enzymol. 405, 66–81.<br />

11. Bollen, M. <strong>and</strong> Beullens, M. (2002) Signaling by protein phosphatases in the<br />

nucleus. Trends Cell Biol. 12, 138–45.<br />

12. Zhou, W., Merrick, B. A., Khaledi, M. G., <strong>and</strong> Tomer, K. B. (2000) Detection <strong>and</strong><br />

sequencing of phosphopeptides affinity bound to immobilized metal ion beads by<br />

matrix-assisted laser desorption/ionization mass spectrometry. J. Am. Soc. Mass<br />

Spectrom. 11, 273–82.<br />

13. Hata, K., Morisaka, H., Hara, K., et al. (2006) Two-dimensional HPLC on-line<br />

analysis of phosphopeptides using titania <strong>and</strong> monolithic columns. Anal. Biochem.<br />

350, 292–7.<br />

14. Larsen, M. R., Thingholm, T. E., Jensen, O. N., Roepstorff, P., <strong>and</strong> Jorgensen,<br />

T. J. (2005) Highly selective enrichment of phosphorylated peptides from peptide<br />

mixtures using titanium dioxide microcolumns. Mol. Cell Proteomics. 4, 873–86.<br />

15. Wolschin, F., Wienkoop, S., <strong>and</strong> Weckwerth, W. (2005) Enrichment of phosphorylated<br />

proteins <strong>and</strong> peptides from complex mixtures using metal oxide/hydroxide<br />

affinity chromatography (MOAC). Proteomics. 5, 4389–97.<br />

16. Gorg, A., Obermaier, C., Boguth, G., et al. (2000) The current state of twodimensional<br />

electrophoresis with immobilized pH gradients. Electrophoresis. 21,<br />

1037–53.<br />

17. Gorg, A., Weiss, W., <strong>and</strong> Dunn, M. J. (2004) Current two-dimensional<br />

electrophoresis technology for proteomics. Proteomics. 4, 3665–85.<br />

18. Klose, J. (1975) Protein mapping by combined isoelectric focusing <strong>and</strong><br />

electrophoresis of mouse tissues. A novel approach to testing for induced point<br />

mutations in mammals. Humangenetik. 26, 231–43.<br />

19. O’Farrell, P. H. (1975) High resolution two-dimensional electrophoresis of<br />

proteins. J. Biol. Chem. 250, 4007–21.<br />

20. Thierse, H. J., Moulon, C., Allespach, Y., et al. (2004) Metal-protein complexmediated<br />

transport <strong>and</strong> delivery of Ni2+ to TCR/MHC contact sites in nickelspecific<br />

human T cell activation. J. Immunol. 172, 1926–34.<br />

21. Heiss, K., Junkes, C., Guerreiro, N., et al. (2005) Subproteomic analysis of metalinteracting<br />

proteins in human B cells. Proteomics. 5, 3614–22.<br />

22. She, Y. M., Narindrasorasak, S., Yang, S., Spitale, N., Roberts, E. A., <strong>and</strong><br />

Sarkar, B. (2003) Identification of metal-binding proteins in human hepatoma lines<br />

by immobilized metal affinity chromatography <strong>and</strong> mass spectrometry. Mol. Cell.<br />

Proteomics. 2, 1306–18.<br />

23. Smith, S. D., She, Y. M., Roberts, E. A., <strong>and</strong> Sarkar, B. (2004) Using immobilized<br />

metal affinity chromatography, two-dimensional electrophoresis <strong>and</strong> mass<br />

spectrometry to identify hepatocellular proteins with copper-binding ability. J.<br />

Proteome Res. 3, 834–40.


Metalloproteomics in the Molecular Study of Cell Physiology 147<br />

24. Kulkarni, P. P., She, Y. M., Smith, S. D., Roberts, E. A., <strong>and</strong> Sarkar, B.<br />

(2006) Proteomics of metal transport <strong>and</strong> metal-associated diseases. Chemistry. 12,<br />

2410–22.<br />

25. Martin, S. F., Merfort, I., <strong>and</strong> Thierse, H. J. (2006) Interactions of chemicals <strong>and</strong><br />

metal ions with proteins <strong>and</strong> role for immune responses. Mini Rev. Med. Chem. 6,<br />

247–55.<br />

26. Jungblut, P., Baumeister, H., <strong>and</strong> Klose, J. (1993) Classification of mouse liver<br />

proteins by immobilized metal affinity chromatography <strong>and</strong> two-dimensional<br />

electrophoresis. Electrophoresis. 14, 638–43.


13<br />

Protein Extraction from Green Plant Tissue<br />

Ragnar Flengsrud<br />

Summary<br />

A method for preparation of protein from green plant tissue for two-dimensional<br />

electrophoresis is described. The method is demonstrated on barley leaves, potato leaves<br />

<strong>and</strong> spruce needles <strong>and</strong> appears to overcome the obstacles inherent in green plants to<br />

proteomic analysis. The yield <strong>and</strong> the representation of proteins are discussed.<br />

Key Words: Barley leaves; green plant tissue; potato leaves; protein extraction;<br />

spruce needles; two-dimensional electrophoresis.<br />

1. Introduction<br />

<strong>Preparation</strong> of proteins for two-dimensional (2-D) electrophoresis is an<br />

important, <strong>and</strong> sometimes crucial, part of this central proteomics technique, a<br />

fact that may be overlooked or underestimated.<br />

Plants <strong>and</strong> especially green plant tissues constitute considerable challenges<br />

here, because of low protein concentration <strong>and</strong> the presence of deleterious<br />

compounds in the cell.<br />

Plant proteases may contribute to the challenge because remarkable stability<br />

has been reported relevant to temperature (1–3), pH(2,3) <strong>and</strong> even urea or<br />

guanidine hydrochloride (2,4).<br />

During the work with green plant tissues several principles were applied<br />

that seem to overcome these problems, resulting in good, reproducible 2-D<br />

separation of green tissue proteins from three different species (5). These<br />

principles are: (a) Cell disruption <strong>and</strong> homogenization in liquid N2 <strong>and</strong> insoluble<br />

polyvinylpyrrolidone; (b) an extraction buffer including thiourea <strong>and</strong> SDS with<br />

a pH lower than 5.5; (c) protein precipitation in 90% (v/v) acetone at –20°C;<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

149


150 Flengsrud<br />

(d) dialysis against a buffer containing 9.5M urea, nonionic detergent <strong>and</strong><br />

lysine. The sample preparation method described in this chapter is well suited<br />

for barley leaves (Fig. 1), potato leaves <strong>and</strong> spruce needles. Electropherograms,<br />

both in the isoelectric focusing (Fig. 1) <strong>and</strong> the nonequilibrium pH gradient<br />

mode, is presented. There is no indication of protease activity. Total recovery<br />

of protein is 6.7–16.5% (5). Yet, the possibility exists that all or most proteins<br />

are represented in the extract.<br />

A study on the effect of different extraction solutions on the solubilization<br />

of endosperm proteins showed that the same proteins were extracted, but to<br />

different degree. SDS/urea was one of the extraction solutions with best overall<br />

results in this work. Even with 22% protein recovery, the extract was shown<br />

to contain hordein proteins (6). The sample preparation method was utilized in<br />

a stress diagnosis study on spruce needles (7,8). This study used both soluble<br />

<strong>and</strong> immobilized pH gradients <strong>and</strong> about 1,500 spots were detected by image<br />

analysis. Here, the changes in needle protein pattern were studied by image<br />

analysis of 300–350 spots. Protease activity was found to be negligible in this<br />

study.<br />

Fig. 1. Two-dimensional electrophoresis in the isoelectric focusing mode of proteins<br />

extracted from leaves of a mutant line (H354-33-7-5). of the barley variety cv. Carlsberg<br />

II. The loading was 33 μg protein <strong>and</strong> silver staining was used for detection.


Protein Extraction from Green Plant Tissue 151<br />

2. Materials<br />

1. Extraction buffer: 50 mM pyridine, 10 mM thiourea, 1% (w/v) SDS, adjusted to<br />

pH to 5.0 by HCl. The purity of pyridine (see Note 1), the pH-range (see Note 2)<br />

<strong>and</strong> the use of SDS (see Note 3) is important.<br />

2. Lysis solution: 9.5M urea, 2% (v/v) nonionic detergent (Igepal CA-630), 1.6%<br />

(v/v) ampholytes pH 5–7, 0.4% ampholytes pH 3–10, 2.5% (w/v) dithiothreitol<br />

(DTT).<br />

3. Modified lysis solution for dialysis: 2% (w/v) lysine substitutes the ampholytes.<br />

3. Methods<br />

1. Cut the leaves into 0.5 to 1.0 cm parts <strong>and</strong> needles into 2–4 mm parts. Typically,<br />

start with 0.4 g green tissue <strong>and</strong> grind it twice in a mortar with liquid N2 to give<br />

a fine powder. The storage of plant material before homogenization should be<br />

considered (see Note 4).<br />

2. Add an amount of insoluble polyvinylpyrrolidone (Polyclar AT) twice the weight<br />

of plant material (see Note 5) <strong>and</strong> mix.<br />

3. Add extraction buffer (9.5 mL) to the mixture in the mortar, stir for a few minutes<br />

<strong>and</strong> centrifuge at +5°C for 40min. at 8,000g.<br />

4. Transfer the supernatant to a thick-walled glass centrifuge-tube, add ice cold<br />

acetone (see Note 6) to give a final concentration of 90% (v/v) <strong>and</strong> mix well.<br />

Allow proteins to precipitate at –20°C for 2 h. The yield of the protein depends<br />

on the concentration of acetone (see Note 7).<br />

5. Collect proteins by centrifugation for 20 min at +5°C <strong>and</strong> 5,000g. Discard the<br />

supernatant <strong>and</strong> wash the precipitate once with ice cold acetone <strong>and</strong> centrifuge as<br />

above.<br />

6. Carefully dry the resulting precipitate in a stream of N2, add 400 μL of the lysis<br />

solution <strong>and</strong> mix well.<br />

7. Dialyse the mixture of precipitate <strong>and</strong> lysis solution overnight against 25 mL of<br />

the modified lysis solution.<br />

8. Centrifuge the dialysed sample at 8,000g for 10 min. Add the appropriate<br />

ampholytes to the clear supernatant to give a final 2% (v/v) concentration <strong>and</strong><br />

store at –20°C in suitable aliquots. The suitability depend on the detection method<br />

to be used following the 2-D electrophoresis (see Note 8).<br />

4. Notes<br />

1. The pyridine in the extraction buffer should be distilled over ninhydrin <strong>and</strong><br />

stored under nitrogen. Alternatively, at least HPLC-grade is used <strong>and</strong> stored under<br />

nitrogen.<br />

2. The study of thiourea as phenoloxidase inhibitor concluded that the pH in the<br />

extraction buffer should not be above 5.5 (9).<br />

3. Extraction with <strong>and</strong> without SDS in the extraction buffer showed that SDS was<br />

necessary for the solubilisation of membrane-bound proteins (5).


152 Flengsrud<br />

4. Storage of barley leaves at –20°C or –80°C up to 3 mo <strong>and</strong> at +5°C for 1 wk,<br />

before homogenization does not seem to affect the protein pattern.<br />

5. The original study (5) used twice the amount (g/g) of insoluble polyvinylpyrrolidone<br />

(Polyclar AT) to plant tissue for its homogenization. A later work (7)<br />

used routinely equal amounts <strong>and</strong> observed that less polyvinylpyrrolidone resulted<br />

in poor electropherograms.<br />

6. Acetone is kept at –20°C before its use in protein precipitation.<br />

7. A final concentration of 90% acetone at –20°C for 1 h will totally precipitate the<br />

proteins (10).<br />

8. The following aliquots of the extract are suitable for 2-D electrophoresis: for<br />

silver staining, 40–60 μL, corresponding to 30–46 μg protein; for Coomassie Blue<br />

staining, 150–260 μL. The aliquots should not be refrozen.<br />

References<br />

1. Fahmy, A. S., Ali, A. A., <strong>and</strong> Mohammed, S. A. (2004) Characterization of a<br />

cysteine protease from wheat Triticum aestivum (cv. Giza 164). Bioresour. Technol.<br />

91, 297–304.<br />

2. Kaneda, M., Yonezawa, H., <strong>and</strong> Uchikoba, T. (1995) Improved isolation, stability<br />

<strong>and</strong> substrate specificity of cucumisin, a plant serine endopeptidase. Biotechnol.<br />

Appl. Biochem. 22, 215–22.<br />

3. Patel, B. K. <strong>and</strong> Jagannadham, M. V. (2003) A high cysteine containing thiol<br />

proteinase from the latex of Ervatamia heyneana: purification <strong>and</strong> comparison with<br />

ervatamin B <strong>and</strong> C from Ervatamia coronaria. J. Agric. Food Chem. 51, 6326–34.<br />

4. Uchikoba, T., Niidome, T., Sata, I., <strong>and</strong> Kaneda, M. (1993) Protease D from the<br />

sarcocarp of honeydew melon fruit. Phytochemistry 33, 1005–08.<br />

5. Flengsrud, R. <strong>and</strong> Kobro, G. (1989) A method for two-dimensional electrophoresis<br />

of proteins from green plant tissues. Anal. Biochem. 177, 33–6.<br />

6. Flengsrud, R. (1993) Separation of acidic endosperm proteins by two-dimensional<br />

electrophoresis. Electrophoresis 14, 1060–66.<br />

7. Davidsen, N. B. (1995) Two-dimensional electrophoresis of acidic proteins isolated<br />

from ozone-stressed Norway spruce needles (Picea abies L. Karst): Separation<br />

method <strong>and</strong> image prosessing. Electrophoresis 16, 1305–11.<br />

8. Davidsen, N. B. (1996) Improved two-dimensional electrophoretic separation of<br />

acidic proteins extracted from Norway spruce needles by using immobilized pH<br />

gradients. Electrophoresis 17, 1280–81.<br />

9. Van Driessche, E., Beeckmans, S., Dejaegere. R., <strong>and</strong> Kanarek, L. (1984) Thiourea:<br />

the antioxidant of choice for the purification of proteins from phenol-rich plant<br />

tissues. Anal. Biochem. 141, 184–88.<br />

10. Neuhoff, V. (1973) Micromethods in Molecular Biology. Springer-Verlag<br />

(Kleinzeller, A., Springer, G.F., <strong>and</strong> Wittmann, H.G., eds.) 14, p133.


14<br />

The Terminator: A Device for High-Throughput<br />

Extraction of Plant Material<br />

B. M. van den Berg<br />

Summary<br />

The Terminator is a device for cost-efficient high-throughput homogenization of plant<br />

material <strong>and</strong> sample preparation. Protein <strong>and</strong> DNA samples can easily be prepared from<br />

large numbers of crude material for further analysis such as protein electrophoresis or<br />

polymerase chain reaction (PCR) followed by DNA electrophoresis. The functioning of the<br />

device is based on vibration of 96 stainless steel pegs in wells of a st<strong>and</strong>ard 96-well micro<br />

plate. Using the Terminator all types of plant tissue, including seeds, can be homogenized<br />

in st<strong>and</strong>ard micro plates in 3 min.<br />

Key Words: DNA extraction; electrophoresis; high throughput analysis; plant<br />

material; protein extraction; seeds.<br />

1. Introduction<br />

High-throughput homogenization <strong>and</strong> sample preparation is often a timelimiting<br />

step in large-scale analysis of biological material. This certainly holds<br />

for the analysis of seed or other plant tissue in several agricultural businesses—<br />

for example the seed business (1)—where many hundreds or even thous<strong>and</strong>s<br />

of individual samples are daily analyzed on a routine basis.<br />

In the seed business, gel isoelectric focusing of protein (but also other<br />

electrophoretic techniques) is widely used to determine the genetic purity of<br />

seed lots or the percentage of inbreds that may occur in hybrid seed lots. With<br />

the advance of isoelectric focusing in the 1980s, the need for high-throughput<br />

sample preparation techniques <strong>and</strong> equipment became important (1). During the<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

153


154 van den Berg<br />

years 1988 <strong>and</strong> 1989 a device was developed—later named the Terminator—<br />

that is perfectly suited to address the needs of high-throughput sample preparation<br />

for isoelectric focusing of protein (2). In more recent years, the Terminator<br />

has proven to be an efficient tool also in high-throughput extraction of<br />

DNA from seed <strong>and</strong> other plant tissue (1,3). In 2004, the Terminator was<br />

improved. The device was restyled, another power supply was added, <strong>and</strong> the<br />

design of the stainless steel pegs was improved to increase the efficiency of<br />

homogenization. Here, the functioning of the Terminator is presented in detail.<br />

2. Materials<br />

All chemicals were purchased from Sigma (St. Louis, OH, USA), unless<br />

otherwise stated.<br />

2.1. <strong>Sample</strong> <strong>Preparation</strong><br />

1. The Terminator used is the restyled <strong>and</strong> improved Terminator as produced <strong>and</strong><br />

sold by Elexa (Enkhuizen, the Netherl<strong>and</strong>s). This device consists of three parts: the<br />

Terminator base plate (Fig. 1A), the Terminator head consisting of a vibromotor<br />

<strong>and</strong> an aluminum plate with 96 stainless steel pegs (Fig. 1B), <strong>and</strong> a variable power<br />

supply (Fig. 1C). The base plate is a4cmthick stainless steel plate equipped<br />

with a micro plate holder consisting of an aluminum base, plastic holders, <strong>and</strong><br />

micro plate clips. St<strong>and</strong>ard 96-wells micro plates can be placed on the micro<br />

plate holder <strong>and</strong> fixed with the special clips. The heavy weight of the steel plate<br />

<strong>and</strong> the rubber feet assure that the Terminator remains at a fixed position during<br />

operation.<br />

Fig. 1. Image showing the three parts of the Terminator. A, the Terminator base<br />

plate; B, the Terminator head consisting of the vibrating motor <strong>and</strong> the 96-peg plate;<br />

C, the variable AC power supply (0–220 V).


A Device for High-Throughput Extraction of Plant Material 155<br />

The Terminator head consists of a special design vibromotor that operates at a<br />

vibration frequency of 50 Hz, <strong>and</strong> an aluminum plate attached to it which bears<br />

96 stainless steel pegs that fit perfectly in the wells of a st<strong>and</strong>ard micro plate.<br />

The variable power supply is a st<strong>and</strong>ard AC 220 V transformer that can deliver<br />

0–220 V (see Notes 1–3).<br />

2. 96-well flat-bottom micro plates (see Note 4) from Costar (Cambridge, USA).<br />

3. Seeds from Seminis Vegetable Seeds (Oxnard, CA, USA) <strong>and</strong> Syngenta (Nerac,<br />

France).<br />

4. Sunflower <strong>and</strong> corn seed protein extraction buffer for isoelectric focusing (IEF):<br />

10 mM Tris-HCl, pH 7.0, 2% (v/v) ampholytes of same pH range as that of the<br />

gel. Make fresh just before use.<br />

5. Tomato seed ADH (alcohol dehydrogenase) extraction buffer: 2% ampholytes<br />

(v/v) pH range 3–10, 0.25% (w/v) dithiothreitol. Make fresh just before use.<br />

6. Ampholytes: SinuLytes 3-7 <strong>and</strong> 3-10 from Sinus (Heidelberg, Germany). Store<br />

at 4°C (see Note 5).<br />

7. DNA seed extraction buffer (XB): 0.2M Tris-HCl pH 8.0, 0.5% (w/v) sodium<br />

dodecyl sulphate (SDS), 0.3M NaCl, 25 mM ethylenediamine tetraacetic acid<br />

(EDTA). Store at room temperature.<br />

8. Protein precipitation (PP) buffer: 2.5M potassium acetate pH 6.5. Add 245.0 g<br />

potassium acetate to 800 mL water. Stir until fully dissolved. Adjust the pH to<br />

6.5 with acetic acid. Bring the final volume to 1 L. Store at room temperature.<br />

9. Tris-EDTA (TE) buffer: dissolve 1.21 g Tris <strong>and</strong> 37.2 mg EDTA in 1 L water.<br />

Store at 4°C.<br />

2.2. Isoelectric Focusing (IEF)<br />

These instructions assume knowledge of making thin horizontal isoelectric<br />

focusing gels between glass plates <strong>and</strong> the use of equipment for horizontal<br />

isoelectric focusing (4–7).<br />

1. 16% (w/v) glycerol. Store at 4°C.<br />

2. 30% acrylamide/bis-acrylamide solution (29:1). Store at 4°C.<br />

3. SinuLytes 3-7 <strong>and</strong> 3-10 (Sinus, Heidelberg, Germany). Store at 4°C.<br />

4. 10% (w/v) ammonium persulfate. Store at 4°C, but no longer than 1 wk.<br />

5. Electrode paper: Whatman #17<br />

6. Electrode solution: 2% (v/v) ampholytes with pH-range identical to that of<br />

the gel.<br />

7. 96-well sample applicator strips from Elexa (Enkhuizen, The Netherl<strong>and</strong>s).<br />

8. Gel backing: GelGrip from Sinus (Heidelberg, Germany).<br />

9. Coomassie gel staining solution: 0.2% (w/v) coomassie brilliant blue (CBB),<br />

50% (v/v) water, 40% (v/v) ethanol, 10% (v/v) acetic acid. Add 0.1 g coomassie<br />

brilliant blue to 20 mL ethanol. Stir until fully dissolved. Then add 25 mL water<br />

<strong>and</strong> 5 mL acetic acid. Prepare fresh before use.<br />

10. CBB destaining solution: 50% (v/v) water, 40% (v/v) ethanol, 10% (v/v) acetic<br />

acid. Store at room temperature.


156 van den Berg<br />

11. ADH staining solution: 0.1M Tris-HCl, pH 7.5, 5% (v/v) ethanol, 0.2%<br />

-nicotinamide adenine dinucleotide (NAD), 0.2% (w/v) 1-(4,5-dimethylthiazol-<br />

2-yl)-3,5-diphenylformazan (MTT), 0.05% (w/v) phenazine methosulfate (PMS).<br />

Prepare fresh.<br />

12. 20% (w/v) trichloroacetic acid (TCA). Prepare fresh.<br />

2.3. Inter Simple Sequence Repeat PCR (ISSR-PCR)<br />

ISSR-PCR is a technique based on amplification of DNA between two simple<br />

sequence repeat (SSR; head-to-tail t<strong>and</strong>em arrays of short DNA repeat motifs)<br />

regions, that uses 5 ′ or 3 ′ anchored SSR PCR primers (8).<br />

1. Anchored primer: 5’-DVDTCTCTCTCTCTCTC (D = A,G,T ;V = A,C,G) from<br />

Invitrogen (Carlsbad, CA, USA).<br />

2. PCR mix: 9.06 μL water, 1.50 μL PCR buffer (10x), 0.60 μL 50 mM magnesium<br />

chloride, 0.6 μL dNTP mixture (2.5 mM each), 0.12 μL 25 μM primer, 0.12<br />

μL DNA polymerase, 3 μL pepper DNA solution. The PCR buffer (10×) <strong>and</strong><br />

the magnesium chloride (MgCl 2) come at the appropriate concentration with the<br />

DNA polymerase. The dNTP’s <strong>and</strong> primer must be diluted to the appropriate<br />

concentration with water.<br />

3. DNA polymerase from Invitrogen (Carlsbad, CA, USA).<br />

4. PCR reaction plates from Invitrogen (Carlsbad, CA, USA).<br />

2.4. DNA Electrophoresis<br />

These instructions assume knowledge of making thin horizontal gels between<br />

glass plates <strong>and</strong> the use of equipment for horizontal electrophoresis (3).<br />

1. Gel buffer (2×): 120 mM Tris-formic acid, pH 9.0.<br />

2. 10% (w/v) ammonium persulfate. Store at 4ºC, but no longer than 1 wk.<br />

3. 20% (w/v) glycerol. Store at 4ºC.<br />

4. 30% acrylamide/bis-acrylmide solution (29:1). Store at 4ºC.<br />

5. Gel backing: GelGrip from Sinus (Heidelberg, Germany).<br />

6. <strong>Sample</strong> buffer for DNA electrophoresis (SB): 2.5% gel buffer (2×), 0.02%<br />

Bromophenol Blue.<br />

7. In-gel well template <strong>and</strong> spacers (0.2 mm thick) from Elexa (Enkhuizen, The<br />

Netherl<strong>and</strong>s).<br />

8. Electrode paper: Whatman #17.<br />

9. Electrode solution: 10% (w/v) Tris-Base, 1.73% (w/v) boric Acid, 0.02%<br />

bromophenol blue.<br />

10. Fixing solution: 2% (v/v) nitric acid.<br />

11. Staining solution: 0.2% (w/v) silver nitrate.<br />

12. Developer solution: 0.05% (v/v) formaldehyde, 3% (w/v) sodium carbonate.<br />

Prepare fresh just before use.<br />

13. Stop solution: 5% (v/v) acetic acid.


A Device for High-Throughput Extraction of Plant Material 157<br />

3. Methods<br />

3.1. <strong>Sample</strong> <strong>Preparation</strong><br />

The Terminator homogenizes tissue in the wells of the micro plate by the<br />

vibration in all directions of the pegs in the wells. Fig. 2A illustrates a diagrammatic<br />

view of how plant tissue is squeezed <strong>and</strong> homogenized between the<br />

vibrating pegs <strong>and</strong> the wall <strong>and</strong> bottom of the micro plate wells. Optimal<br />

voltage for operation of the Terminator lies between 90 <strong>and</strong> 130 V depending<br />

on the type of tissue to be homogenized. For locations with 110 net Voltage, a<br />

pretransformer that gives 220V output can best be used.<br />

1. Small parts of the corn <strong>and</strong> sunflower seeds are punched using a home-made<br />

device to get samples that fit in the wells of a 96-well plate (see Note 7).<br />

Alternatively, parts can be cut using a scalpel. To cut the seed parts easier, the<br />

seeds may be soaked in water overnight. The seed parts are put in the wells of<br />

a 96-well micro plate, 200 μL seed extraction buffer is added, <strong>and</strong> the tissue is<br />

disrupted <strong>and</strong> fully homogenized using the Terminator. After 3 min operation of<br />

the Terminator the homogenates are centrifuged in a micro plate centrifuge <strong>and</strong><br />

the supernatant samples are used for IEF.<br />

2. Tomato seeds are put in the wells of a 96-well micro plate, 200 μL extraction buffer<br />

is added, <strong>and</strong> the seeds are disrupted <strong>and</strong> fully homogenized using the Terminator.<br />

Fig. 2. Composite image showing a close view of the pegs of the Terminator in<br />

micro plate wells. A drawing of a peg in a well of a micro plate illustrating how tissue<br />

is squeezed between the pegs <strong>and</strong> the walls <strong>and</strong> bottom of the plate. B <strong>and</strong> C show a<br />

photograph at close range of the pegs in the micro plate.


158 van den Berg<br />

After 3 min operation of the Terminator the homogenates are centrifuged in a<br />

micro plate centrifuge <strong>and</strong> the supernatant samples are used for IEF.<br />

3. Pepper seeds are put in the wells of a 96-well micro plate, 200 μL DNA<br />

extraction buffer is added, <strong>and</strong> the seeds are disrupted <strong>and</strong> fully homogenized<br />

using the Terminator. After 3 min operation of the Terminator the homogenates<br />

are centrifuged in a micro plate centrifuge <strong>and</strong> 90 μL of each of the individual<br />

supernatants are brought (taking care not to disturb the pellets) in another micro<br />

plate. 90 μL PP buffer is added to the supernatants <strong>and</strong> the micro plate is mixed<br />

using an orbital shaker for 3 min at moderate speed. Then the plate is centrifuged<br />

at 4°C, 1,300g, for 10 min with moderate deceleration.<br />

Of the resulting supernatants 90 μL is brought to a new micro plate <strong>and</strong> 90<br />

μL ice-cold isopropanol (–20°C) is added to the supernatants. The micro plate<br />

is briefly shaken using the orbital shaker at medium speed <strong>and</strong> then centrifuged<br />

at 4°C, 1,300g for 10 min with moderate deceleration. The resulting supernatant<br />

is discarded <strong>and</strong> the pellet dried at 65°C for 20 min. To the pellet 100 μL<br />

TE-buffer is added, <strong>and</strong> the resulting DNA solution is stored at 4ºC. For long<br />

term storage –20°C is recommended.<br />

3.2. Isoelectric Focusing (IEF)<br />

1. For IEF of corn <strong>and</strong> sunflower seed extracts, gels of size 260 × 188 mm, <strong>and</strong><br />

thickness of 0.2 mm are made fresh by combining the following solutions: 9.5<br />

mL 16% glycerol, 2.0 mL acrylamide/bis acrylamide solution, 1.0 mL SinuLytes<br />

3-7, 12 μL TEMED, 35 μL 10% ammonium persulfate. After swirling the<br />

solution the gel was poured.<br />

2. The gel is divided in two fields using electrode paper wicks on the long sides<br />

<strong>and</strong> in the middle of the gel. In this way 96 samples can be run with a running<br />

distance of 5 cm.<br />

3. Prefocusing is carried out at settings 600 Volts, 60 mA, 12 Watts for 75 Volthours<br />

4. The corn <strong>and</strong> sunflower seed extracts (8 μL per sample) are applied to the<br />

horizontal IEF gel in the range 3–7 using the 96-well applicator strip.<br />

5. Focusing is carried out in two runs. Run 1 with 200 Volts, 60 mA, 12 Watts<br />

for 50 Volthours <strong>and</strong> run 2 at settings 1000 Volts, 60 mA, 12 Watts for 1,000<br />

Volthours.<br />

6. After focusing, proteins are fixed in TCA solution for 10 min, stained with CBB<br />

solution for 10 min, <strong>and</strong> then the gel is destained several times for 10 min until<br />

the background is clear.<br />

7. For ADH IEF of tomato seed extracts gels of size 260 × 188 mm, <strong>and</strong> thickness<br />

of 0.2 mm are made fresh by combining the following solutions: 9.5 mL 16%<br />

glycerol, 2.0 mL acrylamide/bis acrylamide , 1.0 mL SinuLytes 3-10, 12 μL<br />

TEMED, 35 μL l10% ammonium persulfate. After swirling the solution the gel<br />

was poured.


A Device for High-Throughput Extraction of Plant Material 159<br />

8. The gel is divided in two fields using electrode paper wicks on the long sides<br />

<strong>and</strong> in the middle of the gel. In this way 96 samples can be run with a running<br />

distance of 5 cm.<br />

9. Prefocusing is carried out at settings 600 Volts, 60 mA, 12 Watts for 75 Volthours<br />

10. The tomato seed extracts (8 μl per sample) are applied to the horizontal IEF gel<br />

using the 96-well applicator strip.<br />

11. Focusing is carried out in two runs. Run 1 with 200 Volts, 60 mA, 12 Watts<br />

for 50 Volthours <strong>and</strong> run 2 at settings 1000 Volts, 60 mA, 12 Watts for 700<br />

Volthours.<br />

12. The ADH staining solution is prepared just before the end of the focusing.<br />

Tris-HCl buffer is brought to 37°C. 5 mL ethanol, 0.05 g NAD, 0.05 g MTT<br />

<strong>and</strong> 0.01 g PMS are added, <strong>and</strong> dissolved by mixing. The gel is stained at 37°C<br />

until the b<strong>and</strong>s can be clearly visualized (5–10 min). After removal of the stain,<br />

the gel is destained 10 min in 2% (v/v) acetic acid <strong>and</strong> then rinsed 10 min with<br />

water.<br />

3.3. Inter-SSR PCR<br />

1. The PCR mix is prepared just before PCR by adding the reaction mixture components<br />

together, including 3 μL of the DNA solution gained by extraction under<br />

Section 3.2.<br />

2. The PCR mix is added to the PCR plates <strong>and</strong> PCR is carried out with the following<br />

program steps: an initial 5 min step at 94ºC, the 40 cycles of 0.3 min at 94°C,<br />

0.45 min at 55°C, <strong>and</strong> 2 min at 72°C, which is followed by a final step of 5 min<br />

at 72°C.<br />

3.4. DNA Electrophoresis<br />

1. For horizontal electrophoresis of pepper ISSR PCR fragments, gels of size 260 ×<br />

188 mm <strong>and</strong> thickness of 0.2 mm are used. The following gel solution is used to<br />

pour the gels: 6.25 mL gel buffer, 9.5 mL 20% glycerol, 2.08 mL acrylamide/bis<br />

acrylamide 29: 1 solution, 0.02 mL TEMED, 0.14 mL 10% ammonium persulfate.<br />

The gel is poured <strong>and</strong> used the same day or stored at 4°C (for maximally 5 d).<br />

2. To the PCR mix 4 μL of SB is added <strong>and</strong> of these samples, 4 μL is pipetted into<br />

the in-gel wells of the gel.<br />

3. The gel is divided in two fields using electrode paper wicks on the long sides<br />

<strong>and</strong> in the middle of the gel. In this way 96 samples can be run with a running<br />

distance of 5 cm.<br />

4. Electrophoresis is carried out at 15°C at power settings of 600 V, 40 mA <strong>and</strong><br />

24 W for 75 min<br />

5. After electrophoresis the gel is incubated in fix solution for at least 3 min <strong>and</strong> then<br />

rinsed with water for 30 s. There after the gel is incubated in staining solution for<br />

20 min, washed in water for 1 min, <strong>and</strong> then the DNA fragments are visualized<br />

by adding developer solution. The developer solution is refreshed after 1 min.<br />

The time for development is 3–5 min depending on the amount of DNA present


160 van den Berg<br />

in the gel. Staining reaction is stopped by discarding the developer solution <strong>and</strong><br />

adding stop solution. After 5 min in the stop solution the gel is washed with water<br />

for at least 30 min <strong>and</strong> dried on the air.<br />

4. Notes<br />

1. Before operation of the Terminator, the head with the 96 pegs must be placed.<br />

It must be assured to place the head carefully, by lowering the Terminator head<br />

slowly above the Terminator base plate. Then it is slowly lowered to assure that<br />

the pegs fit into the wells of the micro plate. After operation, the head must be<br />

removed carefully to avoid sample going from one well to another.<br />

2. The Voltage to be applied for operation of the Terminator must be determined<br />

empirically. The knob of the variable power supply must be turned slowly to<br />

increase the Voltage until the head starts clearly vibrating <strong>and</strong> macerating the<br />

tissue. This generates quite some noise by the pegs that hit the bottom of the<br />

plate. If the tissue is disrupted—this may take only a few seconds—the noise<br />

decreases, <strong>and</strong> homogenization can continue at a constant voltage somewhere<br />

between 90 <strong>and</strong> 130 V. The time needed for complete homogenization may also<br />

be determined empirically but is no more than 3 min.<br />

3. Cleaning <strong>and</strong> maintenance of the Terminator is very simple. After operation, the<br />

Terminator head can be cleaned by spraying the pegs with water using a siphon<br />

<strong>and</strong> then the pegs can be dried on the air. The head can be placed with the<br />

pegs on a tissue. When kept clean, the Terminator needs no maintenance. Several<br />

Terminators operate now for more than 10 yr in several labs without maintenance.<br />

For thorough cleaning <strong>and</strong> decontamination the 96-peg plate can be detached<br />

from the vibrator head by turning the vibrator head counter clockwise.<br />

4. Micro plates from Costar are used. However, as st<strong>and</strong>ard 96-well flat-bottom<br />

micro plates are produced worldwide using the same format, the Terminator is<br />

compatible with 96-well flat-bottom micro plates of all major suppliers.<br />

5. SinuLytes are used as ampholytes in IEF gels. Alternatively, other sources of<br />

ampholytes are available. But SinuLytes are superior in our h<strong>and</strong>s because of<br />

a low molecular weight (average molecular weight is between 400 <strong>and</strong> 700<br />

Dalton) of the many amphoteric compounds, which means fast fixing, staining,<br />

<strong>and</strong> destaining of gels. In addition, high buffer capacity <strong>and</strong> solubility at pI, even<br />

conductivity along the gel <strong>and</strong> linear pH results in superior performance.<br />

6. If one views the operation of the Terminator, one may easily suspect (at first sight)<br />

that cross-contamination (sample of one well contaminates sample of another well)<br />

is likely. However, extensive experiments to study possible cross-contamination<br />

were carried out. One may also view Fig. 3B. The single-b<strong>and</strong>ed pattern does not<br />

contain a trace of other b<strong>and</strong>s that are present in other lanes. Further, for making<br />

the image of Fig. 4, pepper seeds were placed in such a way in the micro plates<br />

that each variety lacking the intense b<strong>and</strong> (see Fig. 4) is surrounded by seeds<br />

of a variety that has the intense b<strong>and</strong>. In this way cross-contamination would<br />

become easily visible from the resulting DNA b<strong>and</strong>ing pattern. This shows that


A Device for High-Throughput Extraction of Plant Material 161<br />

Fig. 3. Images of isoelectric focusing gels showing analysis of homogenates prepared<br />

using the Terminator. A CBB stained b<strong>and</strong>ing pattern of corn seed extract focused<br />

in the pH range 3–7. The image shows genetic variability of the corn seed storage<br />

proteins. B CBB stained b<strong>and</strong>ing pattern of sunflower seed extract focused in the pH<br />

range 3–7. The image shows genetic variability of the sunflower seed storage proteins<br />

C ADH stained b<strong>and</strong>ing pattern of tomato seed extract in the pH range 3–10. The<br />

image shows the two known variants of the dimeric enzyme alcohol dehydrogenase<br />

from tomato seeds.<br />

Fig. 4. B<strong>and</strong>ing pattern resulting from ultra-thin layer electrophoresis of PCR<br />

samples using pepper seed DNA as template. One anchored primer is used for PCR.<br />

Pepper varieties were taken that differ in presence of an intense PCR b<strong>and</strong>. Prior to<br />

homogenization using the Terminator, the pepper seeds were put in the micro plate<br />

in such a way that each seed having the b<strong>and</strong> was surrounded by a seed lacking the<br />

b<strong>and</strong>. The variable b<strong>and</strong> is easily visible in the rectangle on the left in the image. An<br />

exploded view is made in the right at the bottom. The white square indicates the area<br />

of the gel that was eluted for further PCR (see Note 6).


162 van den Berg<br />

no cross-contamination occurred. To exclude even traces of cross-contamination,<br />

the gel area indicated with a white square in Fig. 4 was eluted <strong>and</strong> the sample was<br />

used for PCR. DNA electrophoresis showed no trace of DNA at that particular<br />

place in the gel.<br />

7. Several types of cork borers are commercially available to cut round parts from<br />

large seeds such as corn, sunflower, <strong>and</strong> squash. Also leaf samples can best be<br />

taken using a cork borer or a puncher that cuts small leaf discs. It works best to<br />

cut discs of similar size as the bottom of the micro plate wells. First put the discs<br />

in the wells <strong>and</strong> then add the extraction fluid. In this way the leaf tissue can be<br />

optimally homogenized.<br />

References<br />

1. van den Berg, B.M. (1998) Isoelectric focusing in the vegetable seed industry.<br />

Electrophoresis 19, 1780–87.<br />

2. van den Berg, B.M. <strong>and</strong> Tamboer, J.H.A. (1992) The terminator, an apparatus for<br />

simultaneous homogenization of 96 small seeds individually. Electrophoresis 13,<br />

9, 10.<br />

3. van den Berg, B.M. (1997) Horizontal ultrathin-layer multi-zonal electrophoresis of<br />

DNA: an efficient tool in analysis of PCR fragments. Electrophoresis, 18, 2861–64.<br />

4. van den Berg, B.M., Burg, H.C.J., Tamboer, J.H.A., <strong>and</strong> Grapendaal, B. (1992)<br />

Equipment for rapid homogenization of high numbers of plant tissue for<br />

electrophoretic analysis of proteins. Electrophoresis 13, 76–81.<br />

5. van den Berg, B.M. <strong>and</strong> Gabillard, D. (1994) Isoelectric focusing in immobilized pH<br />

gradient of melon (Cucumis melo L.) seed protein: methodical <strong>and</strong> genetic aspects<br />

<strong>and</strong> application in breeding. Electrophoresis 15, 1541–51.<br />

6. van den Berg, B.M. (1990) Inbred testing of tomato (Lycopersicon esculentum) F1<br />

varieties by ultrathin-layer isoelectric focusing of seed protein. Electrophoresis 11,<br />

824–29.<br />

7. van den Berg, B.M. (1991) A rapid <strong>and</strong> economical method for hybrid purity testing<br />

of tomato (Lycopersicon esculentum L.). F1 hybrids using ultrathin-layer isoelectric<br />

focusing of alcohol dehydrogenase variants from seeds. Electrophoresis 12, 64–9.<br />

8. Zietkiewicz, E., Rafalski, A. <strong>and</strong> Labuda, D. (1994) Genome fingerprinting by<br />

simple sequence repeat (SSR)-anchored polymerase chain reaction amplification.<br />

Genomics 20, 176–83.


15<br />

Isolation of Mitochondria from Plant Cell Culture<br />

Etienne H. Meyer <strong>and</strong> A. Harvey Millar<br />

Summary<br />

Mitochondria carry out a variety of important processes in plants. Their major role<br />

is the synthesis of ATP through the coupling of a membrane potential to the transfer of<br />

electrons from NADH to O 2 via the electron transport chain. The NADH is generated by<br />

the oxidation of organic acids via the tricarboxylic acid cycle. However, mitochondria also<br />

perform many important secondary functions such as synthesis of nucleotides, amino acids,<br />

lipids, <strong>and</strong> vitamins. Mitochondria contain their own genome <strong>and</strong> undertake transcription<br />

<strong>and</strong> translation by some unique mechanisms; they actively import proteins <strong>and</strong> metabolites<br />

from the cytosol, are involved in programmed cell death processes in plants, <strong>and</strong> respond<br />

to cellular stress conditions. To underst<strong>and</strong> the extent <strong>and</strong> mechanisms of mitochondrial<br />

functions in plants <strong>and</strong> the way in which their functions are perceived by the nucleus<br />

requires detailed information about the protein components of these organelles. Isolation<br />

of mitochondria to identify their proteomes <strong>and</strong> the changes in these proteomes during<br />

development <strong>and</strong> environmental stresses is growing area of research. In this chapter we<br />

provide a useful method for the isolation of mitochondria from plant cell culture using<br />

a gentle method of cell disruption based on protoplasts isolation that provides relatively<br />

high mitochondrial yields.<br />

Key Words: Cell fractionation; mitochondria; percoll density gradients; protoplasts<br />

isolation.<br />

1. Introduction<br />

Plant mitochondria play an important role in plant metabolism. They provide<br />

energy in the form of ATP, but also they provide a large number of metabolic<br />

precursors in the form of tricarboxylic acids to the rest of the cell for nitrogen<br />

assimilation <strong>and</strong> biosynthesis of amino acids. They play key roles in plant<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

163


164 Meyer <strong>and</strong> Millar<br />

development, fertility, <strong>and</strong> susceptibility to disease. Between 1,000 <strong>and</strong> 1,500<br />

proteins are expected to be found in this cellular compartment <strong>and</strong> currently<br />

many hundreds of these proteins have been identified by proteomics. Studies of<br />

the plant mitochondria proteome require isolation methods that avoid rupture<br />

of mitochondrial membranes. These methods should be able to eliminate<br />

most of the cellular contaminant released after disruption of the plant cell.<br />

Several extensive methodology reviews (1–3) <strong>and</strong> more specific methodology<br />

papers (4–6) are already available on plant mitochondrial purification. All these<br />

methods are based on a cell disruption by grinding. Here we describe a gentle<br />

method based on protoplasts isolation. Protoplasts are plant cells after removal<br />

of the cell wall. This cell wall is made of fibrils of cellulose embedded in a<br />

matrix of several other kinds of polymers such as pectin <strong>and</strong> lignin. This rigid<br />

structure can be digested using two enzymatic activities, cellulase will digest the<br />

cellulose <strong>and</strong> pectolyase will break the intercellular pectin bonds. The resulting<br />

protoplasts are highly fragile <strong>and</strong> can easily be broken by filtration or homogenization.<br />

Protoplasts can be isolated from virtually any plant tissue. Plant organs<br />

give low yields of protoplasts, whereas cell cultures are an excellent starting<br />

material for protoplast isolation. The method we describe here was optimized<br />

for the purification of mitochondria from plant cell culture.<br />

2. Materials<br />

1. Plant cell material for protoplasts: Depending on the growth rate of the plant<br />

culture, 5–7 day old cell culture should be used. This culture should be sterile to<br />

avoid fungal <strong>and</strong> bacterial contamination.<br />

2. Enzyme buffer: 0.4M mannitol, 0.7 g/L MES-KOH pH 5.7. Just before use 0.4%<br />

(w/v) of cellulase <strong>and</strong> 0.05% (w/v) of pectolyase are added (see Note 1). This<br />

buffer has to be prepared just before use.<br />

3. Disruption buffer: 0.4M sucrose, 3 mM EDTA, 50 mM Tris-HCl, pH 7.5, 0.1%<br />

BSA, 2 mM DTT (Dithiothreitol) which is added just prior the disruption. The<br />

osmoticum (sucrose) maintains the mitochondria structure <strong>and</strong> prevents physical<br />

swelling <strong>and</strong> rupture of membranes, the buffer (Tris) prevents acidification from<br />

the contents of ruptured vacuoles, the EDTA inhibits the function of phospholipases<br />

<strong>and</strong> various proteases, the BSA will remove free fatty acids <strong>and</strong> the<br />

reductant (DTT) prevents damage from oxidants present in the tissue or produced<br />

on homogenization. This media can be freshly prepared, stored overnight at 4°C<br />

or frozen at –20°C <strong>and</strong> stored for many weeks (see Note 2).<br />

4. Wash buffer: 0.3 M sucrose, 1 mM EDTA, 10 mM MOPS-KOH, pH 7.2. This<br />

buffer is then used for resuspension of organelle pellets, as the base media for<br />

Percoll gradients <strong>and</strong> for washing purified organelle pellets. This media can be<br />

freshly prepared, stored overnight at 4°C or frozen at –20°C <strong>and</strong> stored for many<br />

weeks (see Note 2).


Isolation of Mitochondria from Plant Cell Culture 165<br />

5. Percoll gradient solutions: Gradients of Percoll (Pharmacia, Uppsala, Sweden)<br />

(see Note 3) are prepared in the wash buffer on the day of use. This is aided by<br />

making a 2× wash buffer <strong>and</strong> adding Percoll <strong>and</strong> distilled water to make 1× wash<br />

buffer with the appropriate percentage of Percoll required. The discontinuous step<br />

gradient was cast using a simple inverted syringe bodies (see Note 4).<br />

3. Methods<br />

3.1. Protoplasts Isolation<br />

The cell culture (5–7 day-old cells) is filtered through two layers of muslin.<br />

The cells are resuspended in the enzyme buffer (a maximum of 500 g of cells<br />

per L of buffer) <strong>and</strong> incubated for 3hinthedark under low agitation (45<br />

rpm) at 25°C. Then the protoplasts are washed twice to remove all traces of<br />

the digestion enzymes. The suspension is centrifuged (for 10 min at 800g) <strong>and</strong><br />

the pelleted protoplasts are resuspended in enzyme buffer without enzyme (see<br />

Note 5). After the second wash, the protoplast pellet is resuspended in cold<br />

disruption buffer.<br />

The following steps should be done either in a cold room or on ice using<br />

4ºC cooled glassware <strong>and</strong> centrifuge tubes (see Note 2).<br />

3.2. Protoplasts Disruption<br />

The protoplasts can be disrupted by filtration through Nylon meshes. The<br />

suspension is filtrated successively through three different Nylon meshes<br />

(100 μm, 75 μm, <strong>and</strong> 30 μm holes). Alternatively, the protoplasts can be broken<br />

by homogenization in a Dounce homogeniser (or potter) (see Note 6).<br />

We recommend checking the digestion as well as the disruption by optical<br />

microscopy (see Note 7).<br />

3.3. Differential Centrifugation to Obtain a Crude Organelle Pellet<br />

1. Transfer filtered homogenate into 50, 250, or 500 mL centrifuge tubes, depending<br />

on the volume of the preparation, <strong>and</strong> centrifuge in a precooled rotor for 5 min<br />

at ∼3,000g in a fixed angle rotor in a preparative centrifuge at 4°C.<br />

2. Decant supernatant gently into another set of centrifuge tubes taking care not<br />

to transfer the pellet material which contains plastids, nuclei <strong>and</strong> cell debris.<br />

Centrifuge supernatant for 15 min at ∼18,000g <strong>and</strong> the resulting high speed<br />

supernatant is discarded. The tan, yellow, or green coloured pellet in each tube<br />

contains an unwashed crude organelle pellet.<br />

3. Resuspend the pellet in 2–10 mL of wash medium with the aid of a clean, soft<br />

bristle paint brush.


166 Meyer <strong>and</strong> Millar<br />

Fig. 1. Percoll gradient purification of plant mitochondria. (A) Three-step Percoll<br />

gradient for the purification of Arabidopsis mitochondria made from 5 mL of 50%<br />

Percoll, 25 mL of 25% Percoll, <strong>and</strong> 5 mL of 18% Percoll solution from bottom to top.<br />

This gradient was centrifuged at 40,000g in a fixed angle rotor for 45 min. Amyloplast<br />

envelopes are concentrated in the 18 to 25% interphase (“a”), the fraction containing<br />

mitochondria in the 25–40% interphase (“m”). (B) One-step Percoll gradient made<br />

with 35 mL of 28% Percoll. The fraction containing mitochondria from the three-step<br />

Percoll gradient was loaded on this gradient which was then centrifuged at 40,000g in<br />

a fixed angle rotor for 45 min. Mitochondria (“m”) are present on top of the gradient<br />

whereas peroxisomes <strong>and</strong> other contaminants (“p”) are located in the bottom part of<br />

the gradient.


Isolation of Mitochondria from Plant Cell Culture 167<br />

3.4. Density Gradient Purification of Mitochondria<br />

The crude mitochondrial preparation described above is contaminated by<br />

thylakoid or amyloplast membranes, peroxisomes <strong>and</strong> endoplasmic reticulum.<br />

Further purification is carried out using Percoll (Pharmacia, Uppsala, Sweden)<br />

density gradients.<br />

1. Layer washed mitochondria, from up to 80 g of etiolated plant tissue or up to 40<br />

g green plant tissue, over 35 mL of a 18–25–40% step gradient (from bottom to<br />

top:5mLof50%, 25 mL of 25%, 5 mL of 18% Percoll in wash buffer) in a 50<br />

mL centrifuge tube (see Note 2).<br />

2. Centrifuge at ∼40,000g for 45 min in a fixed angle rotor of a preparative centrifuge<br />

without braking on the deceleration.<br />

3. After centrifugation, mitochondria form a white-brown b<strong>and</strong> in the bottom part<br />

of the gradient (Fig. 1). Aspirate the mitochondria with a Pasteur pipet avoiding<br />

collection of the yellow or green plastid fractions. Dilute suspension with at least<br />

four volumes of st<strong>and</strong>ard wash medium <strong>and</strong> centrifuge at ∼18,000g for 15 min<br />

in 50 mL tubes.<br />

4. The resultant loose pellets is resuspended in a small amount of wash medium <strong>and</strong><br />

loaded on top of a 28% Percoll continuous gradient (35 mL of 28% Percoll in<br />

wash buffer in a 50-mL centrifuge tube).<br />

5. Centrifuged at ∼40,000g for 45 min in an angle rotor of a preparative centrifuge<br />

without braking on the deceleration.<br />

6. After centrifugation the mitochondria form a white b<strong>and</strong> in the top part of the<br />

gradient whereas contaminants such as peroxisomes are located in the bottom part<br />

of the gradient (Fig. 1). Aspirate the mitochondria with a Pasteur pipet <strong>and</strong> dilute<br />

them with at least four volumes of wash buffer <strong>and</strong> centrifuge again at ∼18,000g<br />

for 15 min in 50-mL tubes.<br />

7. Remove the supernatant <strong>and</strong> resuspend the pellet in wash buffer. Centrifuge again<br />

at ∼18,000g for 15 min. Resuspend the mitochondrial pellet in wash medium at<br />

a concentration of 5–20 mg mitochondrial protein/mL. This can be determined<br />

using a Bradford or Lowry assay.<br />

8. In our h<strong>and</strong>s 100 g of cell culture yields 95 g of protoplasts <strong>and</strong> 15 mg of purified<br />

mitochondria.<br />

9. Once isolated by density gradient purification, plant mitochondria can be kept<br />

on ice for 5–6 h without significant losses in membrane integrity <strong>and</strong> respiratory<br />

function. Longer-term storage of mitochondria can be achieved by rapid-freezing<br />

of mitochondrial samples in liquid N 2. Frozen samples can be then kept at –80°C.<br />

4. Notes<br />

1. At these concentrations of enzymes, more than 95% of the cells will be converted<br />

in protoplasts after 3 h. Increasing the concentrations may result in a faster


168 Meyer <strong>and</strong> Millar<br />

digestion but also a lot of protoplasts could break due to a longer incubation in<br />

the buffer.<br />

2. <strong>Preparation</strong> of mitochondria should be undertaken as quickly as is possible <strong>and</strong><br />

without samples warming above 4°C or storage for extended periods between<br />

centrifugation runs. So we highly recommend using chilled buffers. The time<br />

between homogenization <strong>and</strong> preparation of the washed crude pellet is the most<br />

critical for ensuring integrity <strong>and</strong> high yield.<br />

3. The colloidal silica sol, Percoll, allows the formation of iso-osmotic gradients<br />

<strong>and</strong> through isopycnic centrifugation facilitates a range of methods for the density<br />

purification of mitochondria. The most common method is the sigmoidal, selfgenerating<br />

gradient obtained by centrifugation of a Percoll solution in a fixedangle<br />

rotor. The density gradient is formed during centrifugation at >10,000g<br />

due to the sedimentation of the poly-dispersed colloid (average particle size 29<br />

nm diameter, average density = 2.2 g/mL). The concentration of Percoll in<br />

the starting solution <strong>and</strong> the time of centrifugation can be varied to optimise a<br />

particular separation<br />

4. Step gradients of Percoll are often used as these aids the concentration of<br />

mitochondria fractions on a gradient at an interface between Percoll concentrations.<br />

Step gradients can easily be formed by setting up a series of inverted 20-mL<br />

syringes (fitted with 19-gauge needles) strapped to a flat block of wood, clamped<br />

to a retort st<strong>and</strong> over a rack at an angle of 45° containing the centrifuge tubes<br />

(Fig. 2). The needles are lowered to touch the bevels against the inside, lower<br />

edge of the tubes. The step gradient solutions are then added (from bottom to<br />

top) to the empty inverted syringe bodies <strong>and</strong> each allowed to drain through in<br />

turn before the addition of the next step solution.<br />

Fig. 2. Making density gradient. Home-made apparatus for discontinuous gradients,<br />

see explanation in Note 4


Isolation of Mitochondria from Plant Cell Culture 169<br />

5. The protoplasts are very fragile. Thus the agitation should be very gentle (45 rpm)<br />

on an orbital shaker. Also, the resuspension of the pelleted protoplasts should be<br />

as soft as possible to avoid disruption of protoplasts. We recommend resuspending<br />

the pellet by slowly swirling the tube.<br />

6. These methods of disruption are dependant on the size of the cells. Small cells<br />

(diameter below 10 μm) will not be disrupted. Mesh with smaller holes should<br />

then be used. The disruption of large cells will release a lot of membrane fragment<br />

which will block the holes of the 10-μm mesh. Then the filtration through the<br />

10-μm mesh will be replaced by a second filtration through the 30-μm mesh.<br />

7. The digestion has to be checked by optical microscopy after three hours. A drop<br />

(approx 50 μL) is largely sufficient. If a lot of nondigested cells remain (nonround<br />

cells), incubate the suspension a longer time in the enzyme medium. The<br />

disruption should also be checked to ensure that all the protoplasts are broken.<br />

Unbroken protoplasts will be pelleted during the low-speed centrifugation <strong>and</strong><br />

lost. If some unbroken protoplasts remain, repeat the filtration or homogenisation<br />

step.<br />

References<br />

1. Neuburger, M. (1985) Higher-Plant Cell Respiration, vol. 18 (Douce, R., Day, DA,<br />

ed.), pp. 7–24, Springer-Verlag, Berlin.<br />

2. Douce, R. (1985) Mitochondria in higher plants: Structure, function <strong>and</strong> biogenesis,<br />

American Society of Plant Physiologists, Academic Press, Orl<strong>and</strong>o, Florida.<br />

3. Millar, A. H., Liddell, A., <strong>and</strong> Leaver, C. J. (2001) Isolation <strong>and</strong> subfractionation<br />

of mitochondria from plants. Meth. Cell Biol., 65, 53–74.<br />

4. Neuburger, M., Journet, E. P., Bligny, R., Carde, J. P., <strong>and</strong> Douce, R. (1982)<br />

Purification of plant-mitochondria by isopycnic centrifugation in density gradients<br />

of percoll. Arch. Biochem. Biophys. 217, 312–23.<br />

5. Leaver, C. J., Hack, E., <strong>and</strong> Forde, B. G. (1983) Protein-synthesis by isolated<br />

plant-mitochondria. Meth. Enzymol. 97, 476–84.<br />

6. Day, D. A., Neuburger, M., <strong>and</strong> Douce, R. (1985) Biochemical-characterization of<br />

chlorophyll-free mitochondria from pea leaves. Aust. J. Plant Physiol. 12, 219–228.


16<br />

Isolation <strong>and</strong> <strong>Preparation</strong> of Chloroplasts<br />

from Arabidopsis thaliana Plants<br />

Sybille E. Kubis, Kathryn S. Lilley, <strong>and</strong> Paul Jarvis<br />

Summary<br />

A major area of research in the postgenomic era has been the proteomic analysis<br />

of various subcellular <strong>and</strong> suborganellar compartments. The success of these studies is<br />

to a large extent dependent upon efficient protocols for the preparation of highly pure<br />

organelles or suborganellar components. Here we describe a simple, rapid, <strong>and</strong> low-cost<br />

method for isolating a high yield of Arabidopsis chloroplasts. The method can readily<br />

be applied to wild-type plants <strong>and</strong> different mutants, <strong>and</strong> at different developmental<br />

stages ranging from 10-day-old seedlings to rosette leaves from older plants. The isolated<br />

chloroplast fraction is highly pure, with immunologically undetectable contamination from<br />

other cellular organelles. Chloroplasts isolated using the method described here have been<br />

successfully used for proteomic analysis, as well as in studies on chloroplast protein import<br />

<strong>and</strong> other aspects of chloroplast biology.<br />

Key Words: Arabidopsis thaliana; chloroplast isolation; chloroplast proteomics;<br />

organelle isolation; Percoll gradient; plastids; polytron homogenizer.<br />

1. Introduction<br />

Chloroplasts belong to a diverse group of organelles called plastids (1,2).<br />

Plastids are ubiquitous in plants <strong>and</strong> algae, <strong>and</strong> perform numerous essential<br />

functions including important steps in the biosynthesis of amino acids,<br />

lipids, nucleotides, hormones, vitamins, <strong>and</strong> secondary metabolites, as well<br />

as oxygenic photosynthesis (2,3). In the latter process, energy from sunlight<br />

is converted into usable chemical bond energy, <strong>and</strong> the associated redox<br />

reactions lead to the generation of oxygen from water. Chloroplasts are therefore<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

171


172 Kubis et al.<br />

important sites for the production of organic matter <strong>and</strong> oxygen, <strong>and</strong> so provide<br />

the fuels essential for all higher forms of life (4).<br />

Completion of the genome sequencing projects for Arabidopsis, rice<br />

<strong>and</strong> other species, <strong>and</strong> the development of efficient methods for routine<br />

protein identification by mass spectrometry, have enabled numerous largescale<br />

proteomic studies. Because of the dynamic range limitations associated<br />

with analyses on highly complex mixtures (i.e., the tendency of abundant<br />

proteins to mask the presence of less abundant proteins), these studies have<br />

tended to focus on isolated subcellular components. In plants, chloroplasts have<br />

received considerable attention in this regard (5–8). The uses of such proteomic<br />

studies are several-fold: they can confirm the expression <strong>and</strong> structure of genes<br />

predicted based on genome sequence analysis in silico; they can provide information<br />

on subcellular <strong>and</strong> suborganellar protein localization; <strong>and</strong> they can even<br />

be used to estimate the relative abundance of different proteins. Information of<br />

this nature is particularly important, because it has been estimated that up to<br />

50% of the proteins encoded by the >26,000 genes in the Arabidopsis genome<br />

are of unknown function (9). Proteomics is one of the tools being used to<br />

address this deficiency.<br />

Most proteins targeted to chloroplasts possess a cleavable, amino-terminal<br />

targeting signal called a transit peptide (10,11). Using computer programs (e.g.,<br />

TargetP) to detect the presence of a transit peptide, it has been estimated that<br />

∼4,000 proteins are targeted to chloroplasts in Arabidopsis (9). Unfortunately,<br />

these in silico methods are not 100% reliable (12), <strong>and</strong> so the only truly<br />

dependable method for the determination of protein localization is laboratory<br />

experimentation. However, there are presently less than 700 entries in a database<br />

of experimentally determined chloroplast proteins (13), clearly indicating a<br />

need for further studies. Furthermore, several recent reports have indicated<br />

that protein targeting to chloroplasts is not as simple as was once thought.<br />

In a large-scale study of the Arabidopsis chloroplast proteome, only ∼60%<br />

of the proteins identified were predicted to have a transit peptide (6,14). Of<br />

the remainder, many appeared to have a signal peptide for ER translocation,<br />

or no cleavable targeting sequence at all. Intriguingly, direct evidence for<br />

a protein transport pathway to chloroplasts through the ER <strong>and</strong> Golgi has<br />

now been presented (15), <strong>and</strong> the targeting of proteins lacking a cleavable<br />

peptide has been described in some detail (16,17). These data demonstrate that<br />

transit peptide prediction in silico cannot provide a complete picture of the<br />

chloroplast proteome. The existence of dual-targeted proteins (e.g., proteins<br />

targeted to mitochondria or the ER as well as chloroplasts) adds an additional<br />

level of complexity (18,19), further emphasizing the need for experimental<br />

determination of protein localization.


Isolation <strong>and</strong> <strong>Preparation</strong> of Chloroplasts 173<br />

A number of chloroplast proteomic studies have already been described. In<br />

addition to the whole chloroplast study mentioned earlier (14), several reports<br />

have described the proteomes of individual suborganellar compartments of the<br />

chloroplast: e.g., the thylakoid lumen (20,21), the envelope membrane (22,23),<br />

the stroma (24), <strong>and</strong> the lipid-containing structures called plastoglobuli (25).<br />

In addition, an analysis of plastids in dark-grown plants, called etioplasts,<br />

revealed a proteome consistent with what one would expect of plastids in<br />

heterotrophic tissue, along with some novel functions (26). As well as studies<br />

that simply catalogue the proteins present in a particular subcellular or suborganellar<br />

compartment, comparative proteomics has been employed with considerable<br />

success. For example, the chloroplasts from mesophyll cells <strong>and</strong> bundlesheath<br />

cells of maize, a C4 plant, were recently compared (27). The data<br />

not only revealed differential accumulation of carbon metabolism enzymes<br />

consistent with the C4 photosynthetic mechanism, but also shed light on how<br />

other plastidic functions are distributed between the two cell types. In another<br />

example, chloroplasts isolated from Arabidopsis mutants lacking different<br />

protein import receptor isoforms were compared with wild-type chloroplasts<br />

(28,29). Different groups of chloroplast proteins were found to be selectively<br />

deficient in different receptor mutants, indicating that the different receptor<br />

isoforms likely possess a degree of preprotein recognition specificity (10,11).<br />

These various studies have demonstrated the utility <strong>and</strong> value of chloroplast<br />

proteome analysis, <strong>and</strong> it is anticipated that proteomics will continue to form<br />

an essential component of chloroplast research in the future. The chloroplast<br />

isolation procedure described in technical detail here, <strong>and</strong> previously (30), has<br />

been successfully used in proteomic studies (28,29), as well as in other research<br />

on chloroplast biology (31–33).<br />

2. Materials<br />

2.1. Growth of Arabidopsis Seedlings<br />

1. Seeds, stored in a 1.5-mL microfuge tubes (with a small hole in the lid to allow<br />

evaporation of any residual moisture) at room temperature.<br />

2. 70% (v/v) ethanol containing 0.05% (v/v) Triton X-100 (Sigma-Aldrich Ltd.,<br />

Poole, UK), in a Duran bottle at room temperature. For 200 mL, mix 140 mL<br />

100% ethanol, 60 mL sterile deionized water, <strong>and</strong> 100 μL Triton X-100.<br />

3. 100% ethanol, in Duran bottle at room temperature.<br />

4. Laminar flow hood (e.g., Model P5HB, Bassaire Ltd., Southampton, UK).<br />

5. Circular filter papers, 9 cm in diameter (Whatman, Banbury, UK; or Fisher<br />

Scientific, Loughborough, UK).<br />

6. Industrial methylated spirit (IMS).<br />

7. Orbital shaker (e.g., Model S01, Stuart Scientific, Stone, UK).


174 Kubis et al.<br />

8. Petri dishes, 9 cm in diameter (e.g., Bibby Sterilin Ltd., Stone, UK).<br />

9. Murashige <strong>and</strong> Skoog (MS) medium: MS salt <strong>and</strong> vitamin mixture (Sigma),<br />

0.5% (w/v) sucrose, <strong>and</strong> 0.6% (w/v) agar. Sterilize the medium in an autoclave<br />

(15 min, 121°C, 15 psi), cool to 50°C in a water bath, <strong>and</strong> then pour into Petri<br />

plates to a depth of ∼3–4 mm in a laminar flow hood (400 mL medium is<br />

sufficient for ∼15–20 plates). Allow the plates to dry for 1hinthehood before<br />

replacing the lids. Prepoured plates can be stored at 4°C, up-side down, sealed<br />

in a plastic bag for up to 1 month.<br />

10. Leukopor tape (Beiersdorf AG, Hamburg, Germany) or equivalent (e.g.,<br />

Micropore tape, 3M, Bracknell, UK).<br />

11. Refrigerator or cold-room (4°C).<br />

12. Plant tissue culture chamber (e.g., Model CU-36L5, Percival Scientific Inc.,<br />

Perry, Iowa) set at 20°C, providing 100–120 μmol/m 2 /s white light with a longday<br />

cycle (16-h-light/8-h-dark).<br />

2.2. Chloroplast Isolation<br />

These materials are sufficient for one isolation on a single plant sample.<br />

If multiple samples (e.g., different genotypes) are to be analyzed, additional<br />

materials will be required (i.e., in points 1, 3, 6, 7, <strong>and</strong> 9 below).<br />

1. For one isolation procedure, 25–40 Petri plates of 10-day-old plants, each plate<br />

containing ∼150–200 seedlings as shown in Fig. 1B (see Notes 1, 2).<br />

2. Two ice buckets, containing ice.<br />

3. Two 1-L beakers, one 50-mL beaker, measuring cylinders, <strong>and</strong> one funnel.<br />

4. Polytron; e.g., Kinematica Model PT10-35 (Kinematica AG, Littau,<br />

Switzerl<strong>and</strong>), with a large rotor (PTA 20 S) <strong>and</strong> speed set to 4 on scale of 11<br />

(see Note 3).<br />

5. Cold-room (4°C).<br />

6. Miracloth (Calbiochem Ltd., Nottingham, UK); two squares of about 15 × 15 cm.<br />

7. Two 30-mL Nalgene tubes <strong>and</strong> one 250-mL Nalgene tube (Fisher).<br />

8. Percoll (Amersham Biosciences, Little Chalfont, UK); an opened bottle can be<br />

stored at 4°C for several months.<br />

9. Continuous Percoll gradient. Before use, make up 26 mL of gradient mixture as<br />

follows: 13 mL Percoll, 13 mL 2× chloroplast isolation buffer (see below), <strong>and</strong> 5<br />

mg glutathione (roughly, the tip of a small spatula). Mix the components together<br />

in a 30-mL Nalgene tube, ensuring that the glutathione is completely dissolved.<br />

Precentrifuge in a fixed angle rotor at 43,000g max for 30 min (brake off) at 4°C; this<br />

is equivalent to 19,000 rpm in an SS-34 rotor in a Sorvall RC6 centrifuge (Kendro,<br />

Asheville, North Carolina), with acceleration set to 7 <strong>and</strong> deceleration set to 2.<br />

Gradients can be prepared the day before, <strong>and</strong> then stored overnight at 4°C.<br />

10. Chloroplast isolation buffer (CIB): 0.3M sorbitol, 5 mM MgCl 2,5mM EGTA,<br />

5mM EDTA, 20 mM HEPES/KOH pH 8.0, 10 mM NaHCO 3. This is prepared<br />

as a 2× CIB stock (see Notes 4, 5). The final pH of the solution should be 8.0.


Isolation <strong>and</strong> <strong>Preparation</strong> of Chloroplasts 175<br />

Fig. 1. Different steps of the chloroplast isolation procedure. (A) An MS agar plate<br />

carrying Arabidopsis seeds sown at an appropriate density (∼150 seeds per plate) for<br />

use in chloroplast isolation after growth for 10–14 d. (B) A plate similar to that shown<br />

in A, after the plants have been allowed to grow for 14 d in a tissue culture cabinet.<br />

(C, D) The Arabidopsis seedlings are harvested from the plates by h<strong>and</strong>, taking care<br />

not carry over any of the agar medium. (E) The Arabidopsis tissue is transferred to a<br />

50-mL beaker containing cold chloroplast isolation medium, <strong>and</strong> then disrupted using<br />

five consecutive rounds of homogenization using a polytron blender. (F) Following<br />

filtration through Miracloth, the homogenate is loaded onto a preformed continuous<br />

Percoll gradient. (G) After centrifugation of the loaded Percoll gradient, two green<br />

b<strong>and</strong>s are apparent: the upper b<strong>and</strong> contains broken material, whereas the lower b<strong>and</strong><br />

contains intact chloroplasts.


176 Kubis et al.<br />

11. HEPES-MgSO 4-sorbitol (HMS) buffer: 50 mM HEPES/NaOH pH 8.0, 3 mM<br />

MgSO 4, 0.3M sorbitol (see Note 6). The final pH of the solution should be 8.0.<br />

2.3. Establishing Yield <strong>and</strong> Intactness of Chloroplasts<br />

1. Hemocytometer with a 0.1 mm deep counting chamber <strong>and</strong> a ruling pattern of 1/400<br />

mm 2 (e.g., Improved Neubauer BS748, Hawksley Technology, Lancing, UK).<br />

2. Cover glass (e.g., 22 × 22 mm, No.1, Chance Propper Ltd., Warley, UK).<br />

3. Phase-contrast microscope (e.g., Carl Zeiss AG, Oberkochen, Germany).<br />

4. HMS buffer (see Section 2.2.).<br />

5. Tissue paper (e.g., Kimcare, Kimberly-Clarke Europe Ltd., Reigate, UK).<br />

2.4. <strong>Preparation</strong> for Proteomics<br />

1. Ice bucket containing ice, with lid.<br />

2. Lysis buffer (see Table 1). Recommended detergent components: ASB-<br />

14 (amidosulfobetaine-14) (Calbiochem Ltd.); CHAPS (3-[(3-Cholamidopropyl)dimethylammonio]-1-propanesulfonate)<br />

(Sigma-Aldrich Ltd.); SB3-10<br />

Table 1<br />

Lysis buffers<br />

Buffer Component Concentration<br />

1. St<strong>and</strong>ard buffer CHAPS 4% (w/v)<br />

Urea 8M<br />

Tris-HCl, pH 168–169 10–30 mM<br />

Magnesium acetate 5 mM<br />

2. Thiourea buffer CHAPS 4% (w/v)<br />

Urea 7M<br />

Thiourea 2M<br />

Tris-HCl, pH 8.0–9.0 10–30 mM<br />

Magnesium acetate 5 mM<br />

3. ASB-14 buffer a ASB-14 2% (w/v)<br />

Urea 7M<br />

Thiourea 2M<br />

Tris-HCl, pH 8.0–9.0 10-30 mM<br />

Magnesium acetate 5 mM<br />

4. SDS buffer b SDS 2% (w/v)<br />

Tris-HCl, pH 8.0–9.0 10-30 mM<br />

Magnesium acetate 5 mM<br />

a ASB-14 can be substituted with NP40, SB3-10 or various other sulfobetaine-derived detergents.<br />

b SDS-containing solutions must be diluted to a final concentration of 0.2% or less before<br />

successful isoelectric focusing can take place.


Isolation <strong>and</strong> <strong>Preparation</strong> of Chloroplasts 177<br />

(3-(decyldimethylammonio)propanesulfonate inner salt) (Sigma-Aldrich Ltd.);<br />

Nonidet P40 (NP40) substitute ([octylphenoxy]polyethoxyethanol) (USB Corp.,<br />

Clevel<strong>and</strong>, Ohio).<br />

3. H<strong>and</strong>-held, plastic pestles for use with 1.5-mL microfuge tubes (e.g., pellet pestle,<br />

blue polypropylene, Sigma-Aldrich Ltd.).<br />

4. Bench-top microcentrifuge (e.g., Eppendorf 5415D, Eppendorf UK Ltd.,<br />

Cambridge, UK).<br />

5. Protein concentration estimation kit (e.g., DC Protein Assay, Bio-Rad Laboratories<br />

Ltd., Hemel Hempstead, UK; or PlusOne 2-D Quant, Amersham Biosciences).<br />

6. Plastic cuvets, 1 mL (e.g., Sarstedt Ltd., Leicester, UK).<br />

7. Spectrophotometer (e.g., Spectronic; Thermo Electron Corp., Waltham,<br />

Massachusetts).<br />

3. Methods<br />

3.1. Growth of Arabidopsis Seedlings<br />

1. Transfer the appropriate amount of seeds (e.g., for 40 Petri plates each carrying<br />

∼150–200 seeds, an amount equivalent to ∼240–320 μL will be required) into a<br />

sterile 1.5-mL microfuge tube, <strong>and</strong> add 1 mL of 70% (v/v) ethanol, 0.05% (v/v)<br />

Triton X-100 (see Note 7).<br />

2. Shake the tube by h<strong>and</strong> to ensure that all seeds are suspended in the solution, <strong>and</strong><br />

then place the tube, oriented horizontally, onto an orbital shaker. Shake at 250<br />

rpm for 5 min.<br />

3. Allow the seeds to settle at bottom of tube, remove the supernatant with a pipet,<br />

<strong>and</strong> then add 1 mL of 100% ethanol. Shake the tube by h<strong>and</strong> first of all, <strong>and</strong> then<br />

on the orbital shaker at 250 rpm for 10 min.<br />

4. Meanwhile, switch on the laminar flow hood <strong>and</strong> sterilize the interior surfaces<br />

with IMS. Take an appropriate number of round filter papers (at least one per<br />

seed sample), <strong>and</strong> fold them in half to create a crease (this will facilitate seed<br />

sowing later). In the hood, soak (<strong>and</strong> sterilize) the filter paper(s) with IMS. Allow<br />

the filter paper(s) to dry.<br />

5. Using a cut 1-mL Gilson pipet tip (lacking ∼5 mm from the fine end, to increase<br />

the aperture size), pipet the seeds onto the sterilized filter paper(s) <strong>and</strong> leave to<br />

dry. This takes about 15 min.<br />

6. Sow seeds onto Petri plates containing MS medium. For 10- to 14-day-old<br />

plants, an appropriate density is ∼150–200 seeds/plate, as shown in Fig. 1A. (see<br />

Notes 7, 8).<br />

7. Seal each plate with Leukopor tape.<br />

8. Incubate plates upside down (to prevent condensation accumulating on the surface<br />

of the agar) at 4°C for at least 2d(upto4dispossible) to break seed dormancy<br />

<strong>and</strong> synchronize germination.<br />

9. Grow plants for 10–14 d in a plant tissue culture chamber. Plants grown for 14 d<br />

are shown in Fig. 1B.


178 Kubis et al.<br />

3.2. Isolation of Chloroplasts<br />

The isolation procedure should be started as early as possible in the morning.<br />

During the isolation procedure, plant material should kept at 4°C. The first<br />

steps (stages 2–4 below) can be carried out on the bench in the laboratory, but<br />

the isolation itself should be carried out at 4°C in the cold-room. The method<br />

below describes an isolation on a single plant sample. If multiple samples (e.g.,<br />

different genotypes) are to be used, additional materials will be required (see<br />

Section 2.2).<br />

1. <strong>Preparation</strong>, on the day before the isolation: Place a 200-mL aliquot of 2× CIB, a<br />

50-mL aliquot of 2× CIB, a 50-mL aliquot of HMS buffer, <strong>and</strong> 200 mL deionized<br />

H 2O into the refrigerator or cold-room; in the morning, the frozen solutions<br />

should have thawed. Place all rotors in the cold-room to precool overnight.<br />

2. <strong>Preparation</strong>, on the day of the isolation: Prepare CIB by adding 200 mL chilled<br />

sterile deionized H 2O to 200 mL thawed 2× CIB; mix well <strong>and</strong> keep on ice. Put<br />

100 mL of the CIB into a 1-L plastic beaker <strong>and</strong> keep on ice. Place a second<br />

1-L plastic beaker on ice, with funnel containing two layers of Miracloth. The<br />

250-mL <strong>and</strong> 30-mL Nalgene tubes should also be placed on ice to precool.<br />

3. Prepare a continuous Percoll gradient as described in Section 2.2 (point 9), <strong>and</strong><br />

keep the tube on ice after precentrifugation (see Note 9).<br />

4. Take plates out of plant tissue culture chamber <strong>and</strong> remove the Leukopor tape.<br />

Remove the seedlings from the medium by gently scraping them off with a<br />

gloved h<strong>and</strong> (see Fig. 1C, D), avoiding carry over of medium because this<br />

interferes with the isolation, <strong>and</strong> place them into the 100 mL CIB in the 1-L<br />

beaker on ice.<br />

5. During homogenization, a total of 100 mL CIB is used per sample; this is used<br />

in five, consecutive rounds of homogenization, each one using 20 mL CIB (see<br />

Note 10).<br />

6. Place 20 mL fresh CIB into the 50-mL plastic beaker, <strong>and</strong> then transfer the<br />

seedlings into the beaker.<br />

7. Place the plant material under the rotor of polytron, <strong>and</strong> homogenize for 1-2 s (see<br />

Fig. 1E). The optimal conditions for the homogenization have to be established<br />

empirically (see Note 11).<br />

8. Filter the homogenate through two layers of Miracloth into the 1-L beaker on<br />

ice. Gently squeeze the Miracloth around the plant material.<br />

9. Place a second 20-mL aliquot of fresh CIB into 50-mL beaker, <strong>and</strong> return the<br />

plant material to the beaker.<br />

10. Repeat points 7–9 until all five 20-mL aliquots of CIB have been used, <strong>and</strong><br />

five rounds of homogenization <strong>and</strong> filtration have been completed. The plant<br />

material will gradually become disrupted during the procedure.<br />

11. Transfer the pooled, filtered homogenate into the 250-mL Nalgene tube on ice,<br />

<strong>and</strong> centrifuge at 1,000g max for 5 min (brake on) at 4°C; this is equivalent<br />

to 3,000 rpm in an SLA-1500 rotor in a Sorvall RC6 centrifuge, with both<br />

acceleration <strong>and</strong> deceleration set to 7).


Isolation <strong>and</strong> <strong>Preparation</strong> of Chloroplasts 179<br />

12. Pour off most of the supernatant, <strong>and</strong> resuspend the pellet in the residual ∼500<br />

μL supernatant by rotating the tube on ice; do not resuspend by pipeting.<br />

13. Transfer the resuspended homogenate onto the top of the preformed Percoll<br />

gradient, using a cut 1-mL Gilson pipet tip (lacking ∼5 mm from the fine end,<br />

to increase the aperture size). Pipet very slowly so as not to disturb the gradient<br />

(see Fig. 1F).<br />

14. To separate the intact chloroplasts from broken chloroplasts <strong>and</strong> other debris,<br />

centrifuge in a swing-out rotor at 7,800g max for 10 min (brake off) at 4°C; this<br />

is equivalent to 7,000 rpm in an HB-6 rotor in a Sorvall RC6 centrifuge, with<br />

acceleration set to 7 <strong>and</strong> deceleration set to 2.<br />

15. After centrifugation, remove the tube carefully <strong>and</strong> place it on ice. The lower<br />

green b<strong>and</strong> in the gradient contains intact chloroplasts, whereas the upper b<strong>and</strong><br />

contains broken chloroplasts (see Fig. 1G). Broken chloroplasts are removed<br />

<strong>and</strong> discarded by pipeting, <strong>and</strong> then the intact chloroplasts are recovered using<br />

a 1-mL Gilson pipet tip (cut at the end), <strong>and</strong> transferred into a precooled 30<br />

mL Nalgene tube. The volume of recovered intact chloroplasts can range from<br />

2mLto6mL.<br />

16. Add 25 mL HMS buffer to the chloroplasts <strong>and</strong> invert the tube carefully 2–3<br />

times to wash off the Percoll.<br />

17. Centrifuge the chloroplasts in a swing-out rotor at 1,000g max for 5 min (brake<br />

on) at 4°C; this is equivalent to 2,000 rpm in an HB-6 rotor in a Sorvall RC6<br />

centrifuge, with both acceleration <strong>and</strong> deceleration set to 7.<br />

18. Gently pour off the supernatant, <strong>and</strong> then resuspend the chloroplasts in<br />

∼150–400 μL fresh HMS buffer by rotating the tube on ice; do not resuspend<br />

by pipeting.<br />

3.3. Establishing the Yield, Intactness <strong>and</strong> Purity of the Isolated<br />

Chloroplasts<br />

If necessary, the yield of chloroplasts <strong>and</strong> their intactness can be assessed as<br />

follows. Alternatively, for some applications it may be appropriate to proceed<br />

directly to downstream procedures (e.g., Section 3.4).<br />

1. Add 5 μL of isolated chloroplasts to 495 μL of HMS buffer in a 1.5-mL microfuge<br />

tube, <strong>and</strong> then mix gently by flicking the tube to obtain a 1:100 dilution.<br />

2. Pipet ∼60–80 μL of the diluted suspension onto the counting chamber of the<br />

hemocytometer, <strong>and</strong> place a cover glass on top.<br />

3. Drain the excess suspension with tissue paper.<br />

4. Count the number of chloroplasts in 10 different 1/400 mm 2 squares (e.g., those<br />

on each diagonal line), using a phase contrast microscope with a 16× objective.<br />

The number of chloroplasts per square should average between 10 <strong>and</strong> 20. If too<br />

few or too many chloroplasts are present, adjust the dilution factor (point 1 above)<br />

accordingly <strong>and</strong> repeat the procedure. Intact chloroplasts appear round <strong>and</strong> bright<br />

green (see Fig. 2A), <strong>and</strong> under phase-contrast are surrounded by a bright halo of<br />

light.


180 Kubis et al.<br />

5. The concentration (number of chloroplasts per mL) is calculated as follows: n (the<br />

average number of chloroplasts per 1/400 mm 2 square) × 25 (the total number<br />

of squares in the grid) × 100 (the dilution factor employed) × 10 4 (scaling factor<br />

needed to express the data per mL, because the volume above the 25 squares is<br />

only 0.1 μL).<br />

6. Calculate the actual yield of chloroplasts by multiplying the concentration (number<br />

of chloroplasts per mL) by the volume of chloroplast suspension used in Section<br />

3.2, point 18.<br />

<strong>Sample</strong>s prepared using the methodology described here are mostly intact,<br />

<strong>and</strong> exhibit minimal contamination from other cellular compartments. To illustrate<br />

these points, we analyzed typical chloroplast preparations by phasecontrast<br />

light microscopy (see Fig. 2A,B), by transmission electron microscopy<br />

(see Fig. 2C), <strong>and</strong> by immunoblotting using high-titre antibodies against components<br />

of various cellular organelles (see Fig. 3). Microscopic analysis did not<br />

reveal any evidence of significant contamination from other cellular organelles.<br />

Fig. 2. Light <strong>and</strong> electron micrographs of isolated chloroplasts. (A, B) Chloroplasts<br />

isolated from 14-day-old Arabidopsis seedlings were analysed by phase-contrast<br />

light microscopy, at both low (A) <strong>and</strong> high (B) magnification, <strong>and</strong> the majority were<br />

adjudged to be intact. Size bars indicate 100 μm. (C) The integrity of the chloroplasts<br />

was confirmed by transmission electron microscopy, as described previously<br />

(30). No evidence of significant contamination of the chloroplast preparation with other<br />

organelles was observed using either method. Size bar indicates 2 μm.


Isolation <strong>and</strong> <strong>Preparation</strong> of Chloroplasts 181<br />

Immunologically, the chloroplasts contained undetectable contamination from<br />

all organelles tested:


182 Kubis et al.<br />

3.4. <strong>Preparation</strong> of the Chloroplasts for Proteomics<br />

1. Chloroplast lysis <strong>and</strong> solubilization: Resuspend the intact chloroplast sample in<br />

a small volume of the appropriate lysis buffer (see Table 1). To ensure optimal<br />

lysis, a plastic h<strong>and</strong>-held pestle should be employed in conjunction with a 1.5-mL<br />

microfuge tube. Solubilization should be allowed to proceed on ice for 30 min,<br />

with frequent grinding of the chloroplast material using the pestle (see Note 12).<br />

After this period, centrifuge at 1,400gmax (∼3,900 rpm in an Eppendorf 5415D<br />

microfuge using the st<strong>and</strong>ard 24-place rotor) for 10 min at 4°C, <strong>and</strong> then remove<br />

<strong>and</strong> retain the supernatant.<br />

2. Protein Precipitation: It is often necessary to precipitate proteins derived from<br />

plant material lysis, to ensure the removal of phytophenols, lipids <strong>and</strong> other<br />

cellular components that may interfere with downstream processing steps. There<br />

are several commercial kits available for this purpose; e.g., PlusOne 2-D Clean-Up<br />

Fig. 4. Analysis of chloroplast protein samples by CyDyeDIGE <strong>2D</strong>-<strong>PAGE</strong>. Chloroplasts<br />

were isolated from 10-day-old Arabidopsis seedlings using the described protocol,<br />

<strong>and</strong> then analysed using CyDyeDIGE technology as described previously (28,29). An<br />

image of a typical pH 3–10 nonlinear <strong>2D</strong>-DIGE gel is shown (the pH range is indicated<br />

at the top). Two different chloroplast protein samples (one from wild-type Arabidopsis,<br />

<strong>and</strong> another from a mutant) were labeled with minimal CyDyeDIGE fluors, <strong>and</strong> then<br />

analysed simultaneously. The images from the wild-type sample (Cy5) <strong>and</strong> the mutant<br />

sample (Cy3) have been overlaid. If the Cy5 signal were colored red <strong>and</strong> the Cy3 signal<br />

were colored green, spots corresponding to proteins spots corresponding to proteins<br />

deficient in the mutant would appear red, whereas proteins enriched in the mutant would<br />

appear green. The large (LSU) <strong>and</strong> small (SSU) subunits of Rubisco are indicated.


Isolation <strong>and</strong> <strong>Preparation</strong> of Chloroplasts 183<br />

kit (Amersham Biosciences), or PerfectFOCUS (Genotech, St. Louis, Missouri).<br />

Alternatively, refer to other chapters in this book.<br />

3. Protein estimation: The protein concentration of the sample(s) in lysis buffer<br />

must be determined before proceeding with any proteomics methodologies. There<br />

are several commercial kits available for this purpose, but it is imperative that<br />

the method used is compatible with samples containing detergents; e.g., RC/DC<br />

Protein Assay (Bio-Rad Laboratories Ltd.), or PlusOne 2-D Quant (Amersham<br />

Biosciences).<br />

4. Once these various preparative procedures have been completed, the solubilized<br />

chloroplast protein samples can be subjected to a variety of different methodologies<br />

for proteome analysis, such as difference gel electrophoresis (DIGE) (see<br />

Fig. 4).<br />

4. Notes<br />

1. For 10-day-old plants, this is roughly equivalent to ∼6,000–8,000 individuals<br />

or ∼20 g tissue. For older plants, few individuals are needed, <strong>and</strong> the density of<br />

plants per plate should be reduced.<br />

2. It is likely that more plates will be needed when working with sick, mutant<br />

plants.<br />

3. The method works just as well using a Kinematica PT20 polytron with a small<br />

rotor (13 mm diameter) at ∼40% maximum speed, as described previously (30).<br />

4. Prepare 2× CIB <strong>and</strong> store in 200-mL aliquots in 400 mL Duran bottles at –20°C.<br />

Before use, thaw overnight at 4°C, then make CIB by adding 200 mL sterile<br />

deionized H2O <strong>and</strong> mixing well.<br />

5. Small amounts of 2× CIB are needed for preparation of Percoll gradients, so<br />

aliquots of 2× CIB in 50-mL tubes are recommended for storage at –20°C. The<br />

buffer can be thawed <strong>and</strong> frozen several times (up to 5 times). Mix well after<br />

thawing.<br />

6. Prepare 50-mL aliquots <strong>and</strong> store at –20°C. The buffer can be thawed <strong>and</strong> frozen<br />

several times (up to 5 times). Mix well after thawing.<br />

7. Do not wear gloves when h<strong>and</strong>ling seeds. The seeds will stick to the gloves<br />

because of static electricity. Instead, sterilize h<strong>and</strong>s with IMS <strong>and</strong> avoid touching<br />

the seeds directly.<br />

8. When working with certain, particularly sick mutants, it may be beneficial to<br />

use MS medium supplemented with 100 mM (∼3%) sucrose.<br />

9. When using newly thawed aliquots of 2× CIB, mix well before use to obtain a<br />

homogeneous solution.<br />

10. Sometimes, yields can be improved by increasing the volume of CIB <strong>and</strong><br />

increasing the number of rounds of homogenization; this may be particularly<br />

advantageous when using large amounts of tissue.<br />

11. When using small rotors (e.g., PTA10S with the Kinematica PT10-35, or the<br />

13-mm rotor with the Kinematica PT20) good homogenization is achieved by<br />

moving the small beaker up <strong>and</strong> down 8–10 times quickly (taking ∼3–4sin


184 Kubis et al.<br />

total), in each one of the five rounds of homogenization. When working with<br />

chlorotic or sick mutants, homogenization times should be reduced according to<br />

the severity of the mutant phenotype. For example, when isolating chloroplasts<br />

from the ppi1 mutant (28), homogenization time was reduced to ∼1–2 s for each<br />

of the five rounds of homogenization. When using larger rotors (e.g., PTA20S<br />

with the Kinematica PT10-35), the up <strong>and</strong> down movement of the beaker is not<br />

possible because of the strong suction generated.<br />

12. Buffers containing ASB-14 will solidify at 4°C. Therefore, lysis <strong>and</strong> solubilization<br />

using such buffers should be carried out at room temperature, <strong>and</strong> the<br />

lysis buffer should include a protease inhibitor cocktail (e.g., Complete Protease<br />

Inhibitor Cocktail Tablets, Roche Diagnostics Ltd., Lewes, UK).<br />

Acknowledgments<br />

We thank Ramesh Patel for technical assistance, <strong>and</strong> Natalie Allcock<br />

<strong>and</strong> Stefan Hyman (Electron Microscope Laboratory, Faculty of Medicine<br />

<strong>and</strong> Biological Sciences, University of Leicester) for transmission electron<br />

microscopy. We are grateful to Sabina Kovacheva <strong>and</strong> Ramesh Patel for<br />

their helpful comments on the manuscript. We thank Marc Boutry (PMA2<br />

H + -ATPase), Karl-Josef Dietz (PrxII F), Richard Trelease (catalase), Peter<br />

Shaw (histone H3), Henrik Scheller (PSI-D), <strong>and</strong> Kenton Ko (SSU) for generously<br />

providing antibodies. This work was supported by the Biotechnology<br />

<strong>and</strong> Biological Sciences Research Council (BBSRC) Genomic Arabidopsis<br />

Resource Network (GARNet), by the Royal Society Rosenheim Research<br />

Fellowship (to P.J.), <strong>and</strong> by BBSRC Grants 91/P12928 <strong>and</strong> BBS/B/03629<br />

(to P.J.).<br />

References<br />

1. Whatley, J. M. (1978) A suggested cycle of plastid developmental interrelationships.<br />

New Phytol. 80, 489–502.<br />

2. Lopez-Juez, E. <strong>and</strong> Pyke, K. A. (2005) Plastids unleashed: their development <strong>and</strong><br />

their integration in plant development. Int. J. Dev. Biol. 49, 557–77.<br />

3. Leister, D. (2003) Chloroplast research in the genomic age. Trends Genet. 19,<br />

47–56.<br />

4. Nelson, N. <strong>and</strong> Ben-Shem, A. (2004) The complex architecture of oxygenic photosynthesis.<br />

Nat. Rev. Mol. Cell Biol. 5, 971–82.<br />

5. Baginsky, S. <strong>and</strong> Gruissem, W. (2004) Chloroplast proteomics: potentials <strong>and</strong><br />

challenges. J. Exp. Bot. 55, 1213–20.<br />

6. Jarvis, P. (2004) Organellar proteomics: chloroplasts in the spotlight. Curr. Biol.<br />

14, R317–R319.<br />

7. van Wijk, K. J. (2004) Plastid proteomics. Plant Physiol. Biochem. 42, 963–77.


Isolation <strong>and</strong> <strong>Preparation</strong> of Chloroplasts 185<br />

8. Pan, S., Carter, C. J. <strong>and</strong> Raikhel, N. V. (2005) Underst<strong>and</strong>ing protein trafficking<br />

in plant cells through proteomics. Expert Rev. Proteomics 2, 781–92.<br />

9. Haas, B. J. et al. (2005) Complete reannotation of the Arabidopsis genome:<br />

methods, tools, protocols <strong>and</strong> the final release. BMC Biol. 3, 7.<br />

10. Jarvis, P. <strong>and</strong> Robinson, C. (2004) Mechanisms of protein import <strong>and</strong> routing in<br />

chloroplasts. Curr. Biol. 14, R1064–R1077.<br />

11. Bédard, J. <strong>and</strong> Jarvis, P. (2005) Recognition <strong>and</strong> envelope translocation of chloroplast<br />

preproteins. J. Exp. Bot. 56, 2287–2320.<br />

12. Richly, E. <strong>and</strong> Leister, D. (2004) An improved prediction of chloroplast proteins<br />

reveals diversities <strong>and</strong> commonalities in the chloroplast proteomes of Arabidopsis<br />

<strong>and</strong> rice. Gene 329, 11–6.<br />

13. Friso, G., Giacomelli, L., Ytterberg, A. J., et al. (2004) In-depth analysis of the<br />

thylakoid membrane proteome of Arabidopsis thaliana chloroplasts: new proteins,<br />

new functions, <strong>and</strong> a plastid proteome database. Plant Cell 16, 478–99.<br />

14. Kleffmann, T., Russenberger, D., von Zychlinski, A., et al. (2004) The Arabidopsis<br />

thaliana chloroplast proteome reveals pathway abundance <strong>and</strong> novel protein<br />

functions. Curr. Biol. 14, 354–62.<br />

15. Villarejo, A. et al. (2005) Evidence for a protein transported through the secretory<br />

pathway en route to the higher plant chloroplast. Nat. Cell Biol. 7, 1124–31.<br />

16. Miras, S., Salvi, D., Ferro, M., et al. (2002) Non-canonical transit peptide for<br />

import into the chloroplast. J. Biol. Chem. 277, 47770–8.<br />

17. Nada, A. <strong>and</strong> Soll, J. (2004) Inner envelope protein 32 is imported into chloroplasts<br />

by a novel pathway. J. Cell Sci. 117, 3975–82.<br />

18. Duchêne, A. M. et al. (2005) Dual targeting is the rule for organellar aminoacyl-tRNA<br />

synthetases in Arabidopsis thaliana. Proc. Natl. Acad. Sci. USA 102, 16484–89.<br />

19. Levitan, A., Trebitsh, T., Kiss, V., Pereg, Y., Dangoor, I. <strong>and</strong> Danon, A. (2005)<br />

Dual targeting of the protein disulfide isomerase RB60 to the chloroplast <strong>and</strong> the<br />

endoplasmic reticulum. Proc. Natl. Acad. Sci. USA 102, 6225–30.<br />

20. Peltier, J. B. et al. (2002) Central functions of the lumenal <strong>and</strong> peripheral<br />

thylakoid proteome of Arabidopsis determined by experimentation <strong>and</strong> genomewide<br />

prediction. Plant Cell 14, 211–236.<br />

21. Schubert, M., Petersson, U.A., Haas, B.J., Funk, C., Schroder, W.P. <strong>and</strong><br />

Kieselbach, T. (2002) Proteome map of the chloroplast lumen of Arabidopsis<br />

thaliana. J. Biol. Chem. 277, 8354–8365.<br />

22. Ferro, M. et al. (2003) Proteomics of the chloroplast envelope membranes from<br />

Arabidopsis thaliana. Mol. Cell. Proteomics.<br />

23. Froehlich, J. E., Wilkerson, C. G., Ray, W. K., et al. (2003) Proteomic study of<br />

the Arabidopsis thaliana chloroplastic envelope membrane utilizing alternatives to<br />

traditional two-dimensional electrophoresis. J. Proteome Res. 2, 413–25.<br />

24. Peltier, J. B. et al. (2006) The oligomeric stromal proteome of Arabidopsis thaliana<br />

chloroplasts. Mol. Cell. Proteomics 5, 114–33.<br />

25. Ytterberg, A. J., Peltier, J. B. <strong>and</strong> van Wijk, K. J. (2006) Protein profiling of<br />

plastoglobules in chloroplasts <strong>and</strong> chromoplasts. A surprising site for differential<br />

accumulation of metabolic enzymes. Plant Physiol. 140, 984–997.


186 Kubis et al.<br />

26. von Zychlinski, A., Kleffmann, T., Krishnamurthy, N., Sjol<strong>and</strong>er, K., Baginsky, S.<br />

<strong>and</strong> Gruissem, W. (2005) Proteome analysis of the rice etioplast: metabolic<br />

<strong>and</strong> regulatory networks <strong>and</strong> novel protein functions. Mol. Cell. Proteomics 4,<br />

1072–84.<br />

27. Majeran, W., Cai, Y., Sun, Q. <strong>and</strong> van Wijk, K.J. (2005) Functional differentiation<br />

of bundle sheath <strong>and</strong> mesophyll maize chloroplasts determined by comparative<br />

proteomics. Plant Cell 17, 3111–40.<br />

28. Kubis, S. et al. (2003) The Arabidopsis ppi1 mutant is specifically defective in the<br />

expression, chloroplast import, <strong>and</strong> accumulation of photosynthetic proteins. Plant<br />

Cell 15, 1859–71.<br />

29. Kubis, S. et al. (2004) Functional specialization amongst the Arabidopsis Toc159<br />

family of chloroplast protein import receptors. Plant Cell 16, 2059–77.<br />

30. Aronsson, H. <strong>and</strong> Jarvis, P. (2002) A simple method for isolating import-competent<br />

Arabidopsis chloroplasts. FEBS Lett. 529, 215–20.<br />

31. Park, S. <strong>and</strong> Rodermel, S. R. (2004) Mutations in ClpC2/Hsp100 suppress the<br />

requirement for FtsH in thylakoid membrane biogenesis. Proc. Natl. Acad. Sci.<br />

USA 101, 12765–70.<br />

32. Sinvany-Villalobo, G., Davydov, O., Ben-Ari, G., Zaltsman, A., Raskind, A. <strong>and</strong><br />

Adam, Z. (2004) Expression in multigene families. Analysis of chloroplast <strong>and</strong><br />

mitochondrial proteases. Plant Physiol. 135, 1336–45.<br />

33. Koo, A. J., Fulda, M., Browse, J. <strong>and</strong> Ohlrogge, J. B. (2005) Identification of<br />

a plastid acyl-acyl carrier protein synthetase in Arabidopsis <strong>and</strong> its role in the<br />

activation <strong>and</strong> elongation of exogenous fatty acids. Plant J. 44, 620–32.<br />

34. Finkemeier, I., Goodman, M., Lamkemeyer, P., K<strong>and</strong>lbinder, A., Sweetlove, L. J.<br />

<strong>and</strong> Dietz, K. J. (2005) The mitochondrial type II peroxiredoxin F is essential for<br />

redox homeostasis <strong>and</strong> root growth of Arabidopsis thaliana under stress. J. Biol.<br />

Chem. 280, 12168–12180.<br />

35. Kunce, C. M., Trelease, R. N. <strong>and</strong> Turley, R. B. (1988) Purification <strong>and</strong> biosynthesis<br />

of cottonseed (Gossypium hirsutum L.) catalase. Biochem. J. 251, 147–155.<br />

36. Morsomme, P., Dambly, S., Maudoux, O. <strong>and</strong> Boutry, M. (1998) Single point<br />

mutations distributed in 10 soluble <strong>and</strong> membrane regions of the Nicotiana<br />

plumbaginifolia plasma membrane PMA2 H+-ATPase activate the enzyme <strong>and</strong><br />

modify the structure of the C-terminal region. J. Biol. Chem. 273, 34837–42.


17<br />

Isolation of Plant Cell Wall Proteins<br />

Elisabeth Jamet, Georges Boudart, Gisèle Borderies,<br />

Stephane Charmont, Claude Lafitte, Michel Rossignol, Herve Canut,<br />

<strong>and</strong> Rafael Pont-Lezica<br />

Summary<br />

The quality of a proteomic analysis of a cell compartment strongly depends on the<br />

reliability of the isolation procedure for the cell compartment of interest. Plant cell walls<br />

possess specific drawbacks: (1) the lack of a surrounding membrane may result in the loss<br />

of cell wall proteins (CWP) during the isolation procedure; (2) polysaccharide networks<br />

of cellulose, hemicelluloses, <strong>and</strong> pectins form potential traps for contaminants such as<br />

intracellular proteins; (3) the presence of proteins interacting in many different ways<br />

with the polysaccharide matrix require different procedures to elute them from the cell<br />

wall. Three categories of CWP are distinguished: labile proteins that have little or no<br />

interactions with cell wall components, weakly bound proteins extractable with salts, <strong>and</strong><br />

strongly bound proteins. Two alternative protocols are decribed for cell wall proteomics:<br />

(1) nondestructive techniques allowing the extraction of labile or weakly bound CWP<br />

without damaging the plasma membrane; (2) destructive techniques to isolate cell walls<br />

from which weakly or strongly bound CWP can be extracted. These protocols give very<br />

low levels of contamination by intracellular proteins. Their application should lead to<br />

a realistic view of the cell wall proteome at least for labile <strong>and</strong> weakly bound CWP<br />

extractable by salts.<br />

Key Words: Arabidopsis thaliana; bioinformatics; cell fractionation; cell wall; cell<br />

wall protein; plant; proteomics.<br />

1. Introduction<br />

Plant cell wall proteins (CWP) present specific complexities in addition to the<br />

difficulties usually encountered in proteome analysis, such as protein separation<br />

<strong>and</strong> detection of scarce proteins (1). They are embedded in an insoluble<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

187


188 Jamet et al.<br />

polysaccharide matrix <strong>and</strong> interact with other cell wall components, making<br />

their extraction challenging. Current models of cell wall structure describe the<br />

arrangement of their components into two structurally independent <strong>and</strong> interacting<br />

networks, embedded in a pectin matrix (2,3). Cellulose microfibrils <strong>and</strong><br />

hemicelluloses constitute the first network; the second one is formed by structural<br />

proteins. Three types of CWP can be distinguished, according to their<br />

interactions with cell wall components (4). CWP can have little or no interactions<br />

with cell wall components <strong>and</strong> thus move freely in the extracellular<br />

space. Such proteins can be found in liquid culture media of cell suspensions<br />

<strong>and</strong> seedlings or can be extracted with low ionic strength buffers. We call this<br />

fraction “labile proteins,” most of them have acidic pI ranging from 2 to 6<br />

(Fig. 1A). Alternatively, CWP might be weakly bound to the matrix by Van<br />

der Waals interactions, hydrogen bonds, <strong>and</strong> hydrophobic or ionic interactions.<br />

Such proteins may be extracted by salts <strong>and</strong> most of them have basic pI ranging<br />

from8to11(Fig. 1B) so that they are positively charged at the acidic pH of cell<br />

walls. Even though most of the cell wall polysaccharides are neutral, negatively<br />

Fig. 1. pIs of labile <strong>and</strong> weakly-bound CWP. pIs of CWP identified in<br />

several proteomic studies (4) were calculated (www.iut-arles.up.univ-mrs.fr/w3bb/d_<br />

abim/compo-p.html) after removal of their predicted signal peptides (http://<br />

psort.nibb.ac.jp/form.html). Three groups of proteins were considered: (A) labile<br />

proteins; (B) salt-extracted proteins, i.e. proteins extracted with salt solutions or<br />

chelating agent; (C) all proteins. Reprinted from (4), Copyright (2005), with permission<br />

from Elsevier.


Isolation of Plant Cell Wall Proteins 189<br />

charged pectins contain polygalacturonic acid that provides negative charges<br />

for interactions with basic proteins. Such interactions would be modulated by<br />

pH, degree of pectin esterification, Ca 2+ concentration, <strong>and</strong> by the mobility<br />

<strong>and</strong> diffusion coefficients of these macromolecules (3,5). Finally, CWP can<br />

be strongly bound to cell wall components so that they are still resistant to<br />

salt-extraction. As examples, extensins are cross-linked by covalent links (6,7)<br />

<strong>and</strong> peroxidases can have a high affinity for Ca 2+ -pectate (8).<br />

The available techniques described in this chapter allow the extraction of<br />

labile <strong>and</strong> weakly bound CWP. Because labile proteins can be lost during<br />

the preparation of cell walls, they must be extracted from tissues by nondestructive<br />

techniques such as vacuum infiltration (9), or recovered from liquid<br />

culture media from cell suspension cultures or seedlings (10,11). Weakly bound<br />

CWP can be extracted with salts or chelating agents from living cells with<br />

nondestructive techniques (9,10) or from purified cell walls with destructive<br />

techniques. At present, there is no efficient procedure to release CWP strongly<br />

bound to the extracellular matrix. Structural proteins, for instance extensins or<br />

PRP, can be cross-linked via di-isodityrosine bonds (6,12). Purified cell walls<br />

appear as the most suitable material to isolate such proteins. However, until<br />

now, extensins have only been eluted with salts before their insolubilization<br />

from cell suspension cultures (13).<br />

2. Materials<br />

A major problem in proteomics is the occurrence of keratins that can contaminate<br />

materials <strong>and</strong> working solutions. The presence of keratins can prevent the<br />

identification of proteins of interest by mass spectrometry. It is necessary to pay<br />

attention to all possible sources of contamination at all steps of the following<br />

protocols. Powder-free gloves should be permanently worn <strong>and</strong> washed with<br />

soap before their first use. Chemicals should be reserved for proteomic studies<br />

<strong>and</strong> should not be manipulated with spatula. Buffers should be filtered on 0.22<br />

μm pore size filters. Glass plates for electrophoresis should be cleaned with<br />

alcohol before use.<br />

2.1. Extraction of Labile or Weakly Bound CWP by Nondestructive<br />

Techniques<br />

2.1.1. CWP extraction <strong>and</strong> Analysis from Liquid Culture Medium<br />

of Seedlings<br />

1. Murashige <strong>and</strong> Skoog (MS) culture medium: Murashige <strong>and</strong> Skoog (14) liquid<br />

medium (Sigma Chemical, St Louis, MO, USA) is supplemented with 10 g/L<br />

sucrose <strong>and</strong> adjusted to pH 5.8 with KOH.<br />

2. PVPP (Sigma, St. Louis, MO, USA) is treated with acid to increase polymerization<br />

<strong>and</strong> to remove metal ions <strong>and</strong> contaminants. One g PVPP in 10 mL 10%


190 Jamet et al.<br />

HCl is boiled for 10 min, filtered through a G4 filter, <strong>and</strong> rinsed until neutral<br />

pH is reached. The residue is dehydrated with acetone <strong>and</strong> grinded in a mortar<br />

to obtain a fine powder (15).<br />

3. Low binding 12 kDa cutoff Spectra/Por ® cellulose ester (CE) dialysis bags<br />

(Merck Eurolab Poly Labo, Strasbourg, France).<br />

4. Centriprep ® device (MWCO: 10 kDa) (Millipore Corporation, Bedford, MA,<br />

USA).<br />

5. Bradford protein assay (Coomassie ® Protein assay Reagent Kit, Pierce, Rockford,<br />

IL, USA) (16).<br />

6. Immobilized pH gradient (IPG) buffers <strong>and</strong> 13 cm-strips pH 4 –7 or 6–11 (GE<br />

Healthcare Europe GmbH, Orsay, France).<br />

7. 2-DE (2-dimensional electrophoresis) sample buffer: 7M urea, 2M thiourea,<br />

4% (w/v) CHAPS, 65 mM DTE, 0.5% (v/v) IPG buffer (pH 4-7 or 6-11),<br />

bromophenol blue trace.<br />

2.1.2. CWP Extraction <strong>and</strong> Analysis from Cell Suspension Cultures<br />

1. Gamborg liquid medium: Gamborg B5 medium supplemented with 20 g/L<br />

sucrose, 2.5 μM naphthalene acetic acid (Sigma Chemical, St Louis, MO, USA)<br />

<strong>and</strong> adjusted to pH 5.7 with KOH (17).<br />

2. Cell washing buffer: 50 mM sodium acetate buffer pH 6.5, 10 mM DTT, 1 mM<br />

PMSF, 1% ethanol, 50% glycerol.<br />

3. C1 protein extraction buffers: 0.15M NaCl in cell washing buffer.<br />

4. C2 protein extraction buffer: 1M NaCl in cell washing buffer.<br />

5. C3 protein extraction buffer: 0.2M CaCl2 dihydrate in cell washing buffer.<br />

6. C4 protein extraction buffer: 2M LiCl in cell washing buffer.<br />

7. C5 protein extraction buffer: 50 mM 1,2-cyclohexanediamine tetracetic acid<br />

(CDTA) in cell washing buffer.<br />

8. Low binding 2 kDa cutoff Spectra/Por ® CE dialysis bags (Merck Eurolab Poly<br />

Labo, Strasbourg, France).<br />

9. One mL Hi-Trap SP Sepharose cation exchanger (GE Healthcare Europe GmbH,<br />

Orsay, France).<br />

10. Hi-Trap SP equilibration buffer: 10 mM MES-KOH, pH 5.2.<br />

11. Hi-Trap SP elution buffer: 10 mM MES-KOH, pH 5.2, 2M NaCl.<br />

12. Econo-Pac ® 10DG desalting column (Bio-Rad, Hercules, CA,USA).<br />

13. Desalting column equilibration buffer: 50 mM ammonium formate.<br />

14. Bradford protein assay (Coomassie ® Protein assay Reagent Kit, Pierce,<br />

Rockford, IL, USA) (16).<br />

15. 1-DE (1-dimensional electrophoresis) sample buffer: 62 mM Tris-HCl, pH 6.8,<br />

2% SDS, 10% glycerol, 5% mercapto-ethanol.<br />

16. Resuspending solution: 1M thiourea, 10 mM DTT, 1% (v/v) protease inhibitor<br />

cocktail for plant (Sigma, St. Louis, MO, USA) in UHQ water (see Note 1).<br />

Prepare as required.<br />

17. Immobilized pH gradient (IPG) buffers <strong>and</strong> 13-cm strips pH 4–7 or 6–11 (GE<br />

Healthcare Europe GmbH, Orsay, France).


Isolation of Plant Cell Wall Proteins 191<br />

18. 2-DE sample buffer: 7M urea, 2M thiourea, 4% (w/v) CHAPS, 65 mM DTE,<br />

0.5% (v/v) IPG buffer (pH 4–7 or 6–11), bromophenol blue trace.<br />

2.1.3. Extraction <strong>and</strong> Analysis of CWP from Rosette Leaves<br />

2.1.3.1. Extraction of Proteins<br />

1. Recovering solution: 0.3M mannitol, 66 mM DTT, 330 mM thiourea, 3.3% (v/v)<br />

protease inhibitor cocktail for plant (Sigma, St. Louis, MO, USA) (see Note 1).<br />

Prepare as required.<br />

2. R1 protein extraction buffer: 1M NaCl, in recovering solution. Adjust pH to 6.9<br />

with 0.5N NaOH. Prepare as required<br />

3. R2 protein extraction buffer: 0.2M CaCl2 dihydrate, in recovering solution. Adjust<br />

pH to 6.9 with 0.5N NaOH. Prepare as required.<br />

4. R3 protein extraction buffer: 2M LiCl, 0.3 in recovering solution. Adjust pH to<br />

6.9 with 1N NaOH. Prepare as required.<br />

5. R4 protein extraction buffer: 50 mM CDTA, in recovering solution. Adjust pH to<br />

6.9 with 5N NaOH. Prepare as required.<br />

6. Malate dehydrogenase (MDH) assay mixture: 50 mM Tris-HCl, pH 7.8 (2.15<br />

mL), 50 mM MgCl2 hexa-hydrate (300 μL), 150 mM DTT (100 μL), 10 mM<br />

NADP (150 μL), 30 mM malic acid (300 μL). Store DTT <strong>and</strong> NADP solutions in<br />

single use aliquots at –20°C. Store malic acid solution at –20°C no longer than<br />

one month. Prepare the MDH assay mixture as required<br />

2.1.3.2. Analysis of Labile CWP<br />

1. Low binding 2 kDa cutoff Spectra/Por ® CE dialysis bags (Merck Eurolab Poly<br />

Labo, Strasbourg, France). Store at –20°C.<br />

2. Resuspending solution: 1M thiourea, 10 mM DTT, protease inhibitor cocktail (1%<br />

v/v) in UHQ water. Prepare as required.<br />

3. Immobilized pH gradient (IPG) buffers <strong>and</strong> 7-cm strips pH 4–7 (GE Healthcare<br />

Europe GmbH, Orsay, France).<br />

4. 2 DE-sample buffer: 7M urea, 2M thiourea, 4% (w/v) CHAPS, 65 mM DTE,<br />

0.5% (v/v) IPG buffer (pH 4–7 or 6–11), bromophenol blue trace.<br />

2.1.3.3. Analysis of Weakly Bound CWP<br />

1. Low binding 2 kDa cutoff Spectra/Por ® CE dialysis bags (Merck Eurolab Poly<br />

Labo, Strasbourg, France). Store at –20°C.<br />

2. One mL Hi-Trap SP Sepharose cation exchanger (GE Healthcare Europe GmbH,<br />

Orsay, France).<br />

3. Hi-Trap SP equilibration buffer: 10 mM MES-KOH, pH 5.2.<br />

4. Hi-trap SP elution buffer: 10 mM MES-KOH, pH 5.2, 2M NaCl.<br />

5. Econo-Pac ® 10DG desalting column (Bio-Rad, Hercules, CA, USA).<br />

6. Desalting column equilibration buffer: 50 mM ammonium formate.<br />

7. 1-DE sample buffer: 62 mM Tris-HCl, pH 6.8, 2% SDS, 10% glycerol, 5%<br />

mercapto-ethanol.


192 Jamet et al.<br />

8. Resuspending solution: 1M thiourea, 10 mM DTT, 1% (v/v) protease inhibitor<br />

cocktail in UHQ water. Prepare as required.<br />

9. Immobilized pH gradient (IPG) buffers <strong>and</strong> 7-cm strips pH 4-7 (GE Healthcare<br />

Europe GmbH, Orsay, France).<br />

10. 2 DE-sample buffer: 7M urea, 2M thiourea, 4% (w/v) CHAPS, 65 mM DTE,<br />

0.5% (v/v) IPG buffer (pH 4–7 or 6–11), bromophenol blue trace.<br />

2.2. Extraction of Weakly Bound CWP by Destructive Techniques<br />

2.2.1. Cell wall preparation<br />

1. MS solid medium: Murashige <strong>and</strong> Skoog (14) liquid medium (Sigma Chemical,<br />

St Louis, MO, USA) is supplemented with 20 g/L sucrose <strong>and</strong> 12 g/L agar, <strong>and</strong><br />

adjusted to pH 5.8 with KOH.<br />

2. PVPP (Sigma, St. Louis, MO, USA) is treated with acid to increase polymerization<br />

<strong>and</strong> to remove metal ions <strong>and</strong> contaminants. One g PVPP in 10 mL 10% HCl<br />

is boiled for 10 min, filtered through a G4 filter, <strong>and</strong> rinsed until neutral pH is<br />

reached. The residue is dehydrated with acetone <strong>and</strong> grinded in a mortar to obtain<br />

a fine powder (15).<br />

3. Nylon nets (1.5-mm pore size <strong>and</strong> 25-μm pore size).<br />

4. Waring blender with a 2-L flask (SEB Moulinex, Ecully, France).<br />

5. Grinding buffer: 10 mM acetate buffer, pH 4.6, 0.4M sucrose, 0.2 % (v/v) protease<br />

inhibitor cocktail for plant (Sigma, St. Louis, MO, USA) (see Note 1).<br />

6. Cell wall purification buffers: 5 mM acetate buffer, pH 4.6, 0.6M or 1M sucrose,<br />

0.2% (v/v) protease inhibitor cocktail.<br />

7. Cell wall washing buffer: 5 mM acetate buffer, pH 4.6.<br />

2.2.2. Extraction <strong>and</strong> Separation of Proteins<br />

1. H1 protein extraction buffer: 5 mM acetate buffer, pH 4.6, 0.2M CaCl 2, 0.1%<br />

protease inhibitor cocktail for plant (Sigma, St. Louis, MO, USA).<br />

2. H2 protein extraction buffer: 5 mM acetate buffer, pH 4.6, 2M LiCl, 0.1% protease<br />

inhibitor cocktail.<br />

3. Econo-Pac ® 10DG desalting column (Bio-Rad, Hercules, CA,USA).<br />

4. Desalting column equilibration buffer: 50 mM ammonium formate.<br />

5. Bradford protein assay (Coomassie ® Protein assay Reagent Kit, Pierce, Rockford,<br />

IL, USA) (16).<br />

6. 1-DE sample buffer: 62 mM Tris pH 6.8 (HCl), 2% SDS, 10% glycerol, 5%<br />

mercapto-ethanol.<br />

3. Methods<br />

The choice of a protocol to extract CWP for proteomic analysis is dependent<br />

on the plant material <strong>and</strong> of the type of proteins to be released from cell walls.<br />

Working on living cells is probably the best solution to avoid intracellular


Isolation of Plant Cell Wall Proteins 193<br />

contamination. This is possible for cell suspension cultures or seedlings grown<br />

in liquid medium as well as for any plant organ that can be infiltrated under<br />

vacuum with extraction buffers. Both labile <strong>and</strong> weakly bound CWP can be<br />

released. When this is not possible, it is necessary to purify cell walls. The main<br />

problem is to avoid intracellular contaminants that will stick nonspecifically to<br />

cell walls. Only weakly bound CWP can be extracted from purified cell walls<br />

because labile CWP are lost during cell wall preparation.<br />

Another important point is the choice of the extraction solution. For example,<br />

a solution of 0.3M mannitol infiltrated in living tissues such as leaves can<br />

solubilize a few CWP expected to be located only in intercellular spaces. Indeed,<br />

identified proteins are acidic, suggesting no ionic interactions with negatively<br />

charged cell wall components (9). NaCl is usually used for extraction of proteins<br />

retained by ionic interactions in the cell wall. LiCl can extract hydroxyprolinerich<br />

glycoproteins from intact cells in Chlamydomonas reinhardii (18). Calcium<br />

chloride is probably the most efficient salt to extract CWP (9,19). The ability<br />

of acidic <strong>and</strong> neutral carbohydrates to strongly chelate calcium (20,21) might<br />

explain, through a competition mechanism, that proteins or glycoproteins<br />

weakly bound to cell wall polysaccharides can be selectively solubilized by<br />

CaCl 2. CDTA, a chelating agent, solubilizes Ca 2+ -pectate. It releases a small<br />

number of proteins having domains of interaction with polysaccharides, notably<br />

proteins showing homology to lectins. This suggests an interaction of these<br />

proteins with polysaccharides associated to pectins (9).<br />

3.1. Extraction of Labile or Weakly Bound CWP by Nondestructive<br />

techniques<br />

3.1.1. Liquid Culture Medium of Seedlings<br />

1. Soak A. thaliana ecotype Columbia seeds (see Note 2) in tap water for 2 h, then<br />

sterilize in diluted bleach (2.4% w/v) for 45 min, <strong>and</strong> rinse several times with<br />

deionized water.<br />

2. Sterilized seeds (100 mg) are germinated <strong>and</strong> grown in MS liquid culture medium<br />

in ten 1-L flasks on a rotary shaker (90 rpm) at 26°C in the dark (22). Each flask<br />

contains 130 mL of culture medium. After 14 d, etiolated seedlings are harvested<br />

<strong>and</strong> the culture medium is filtered through nylon net (60 μm) to remove cell<br />

debris.<br />

3. Collect 900 mL of culture medium (see Note 3). Mix with 9 g PVPP. Shake the<br />

mixture at 4°C for at least 30 min, filter <strong>and</strong> centrifuge to pellet the insoluble<br />

residue. Dialyze against 10 L distilled water during 10–12 h at 4°C using a dialysis<br />

bag (MWCO: 12 kDa) with three changes. Reduce the volume of the sample by<br />

repeated centrifugations (3,500g for 15 min at 4°C) through a Centriprep ® device<br />

(MWCO: 10 kDa) to about 1 mL.<br />

4. Quantify proteins using the Bradford protein assay.


194 Jamet et al.<br />

5. Dilute 250 μg proteins extracted from culture medium with 250 μL 2-DE sample<br />

buffer. Proteins are separated by 2-DE using 13 cm- IPG gel strips pH 4–7 or<br />

6–11 for the first dimension.<br />

3.1.2. Cell Suspension Cultures<br />

1. A cell suspension culture of A. thaliana ecotype Columbia is grown on Gamborg<br />

liquid medium. From this culture, 50 mL (25 g) is transferred every 2 wk to 250<br />

mL fresh medium in 1-L Erlenmeyer flask <strong>and</strong> shaken at 70 rpm in an orbital<br />

shaker, under continuous light (30 μE.m −2 .s −1 ) at 22°C.<br />

2. Wash cells of 7-day-old A. thaliana suspension culture with water <strong>and</strong> pellet<br />

them by centrifugation at 200g. Plasmolyze by successive immersion in 25 %<br />

glycerol, 50 % glycerol for 10 min each, <strong>and</strong> finally wash in 50 % cold glycerol.<br />

All subsequent extractions are performed at 0°C except otherwise stated.<br />

3. Before protein extraction, wash cells with cell washing buffer to remove contaminant<br />

proteins coming from broken cells that nonspecifically stick to cell walls.<br />

4. Extract proteins by washings of the plasmolyzed cells under gentle stirring (30<br />

min) in the proportion of 25 mL of pelleted cells per 50 mL of solution. First<br />

extraction is performed with C1 buffer.<br />

5. Wash cells with the same extraction buffer, then with 50 % glycerol before<br />

centrifugation at 200g for 5 min.<br />

6. Extract weakly bound CWP with C2, C3, C4, or C5 buffer in the same way (see<br />

Note 4).<br />

7. Exhaustively dialyze protein extracts at 4°C against 20 L H 2O using a dialysis bag<br />

(MWCO: 2 kDa). Measure the protein content of each extract using the Bradford<br />

protein assay.<br />

8. Dialyze against Hi-Trap SP equilibration buffer <strong>and</strong> apply to a Hi-Trap SP<br />

Sepharose column equilibrated with Hi-Trap SP equilibration buffer at 1 mL/min.<br />

Elute the retained basic proteins with Hi-Trap SP elution buffer at 1 mL/min.<br />

Desalt the basic proteins on an Econo-Pac ® 10DG desalting column equilibrated<br />

with desalting column equilibration buffer. Freeze-dry the eluate. Resuspend the<br />

dry residue in 40 μL 1-DE sample buffer <strong>and</strong> separate proteins by 1-DE on a<br />

10–17% gradient polyacrylamide gel (16.5 × 13.5 × 0.15 cm).<br />

9. Freeze-dry the acidic <strong>and</strong> neutral proteins from the Hi-Trap SP Sepharose column<br />

effluent. Solubilize the dry residue with a minimal volume of resuspending<br />

solution <strong>and</strong> desalt on an Econo-Pac ® 10DG desalting column. Freeze-dry the<br />

proteins, dissolve in 2-DE sample buffer <strong>and</strong> perform a 2-DE using a 7 cm-IPG<br />

gel strip pH 4–7 for the first dimension.<br />

3.1.3. Rosette Leaves<br />

3.1.3.1. Extraction of Proteins<br />

1. Sterilize A. thaliana ecotype Columbia seeds by soaking in diluted bleach (2.6%<br />

w/v) for 5 min <strong>and</strong> rinse several times with deionized water. Sow the seeds on


Isolation of Plant Cell Wall Proteins 195<br />

humid compost in 10 × 10 cm pots <strong>and</strong> cover the pots with a plastic film. Remove<br />

the plastic film after 48 h <strong>and</strong> transfer the cultures in a growth chamber at 70%<br />

humidity, with a photoperiod of 9 h light at 110 μE.m −2 s −1 at 22°C, <strong>and</strong> 15 h<br />

dark at 20°C. Plants should be moderately watered with a nutrient solution once<br />

a week.<br />

2. Remove carefully 4- to 5-week-old plants (Fig. 2A) from the pots <strong>and</strong> wash<br />

compost off with deionized water. Cotyledons <strong>and</strong> yellowish leaves should be<br />

systematically removed from plants. Process whole plants for vacuum-infiltration<br />

as follows. Make a small noose with a piece of string <strong>and</strong> pass the root through<br />

the noose. Tighten the noose around the collar then twist the root around the<br />

string <strong>and</strong> wrap in parafilm. In a large beaker, immerse completely the rosettes<br />

first in distilled water for a few seconds in recovering solution. Put the beaker<br />

with the immersed rosettes in a dessicator connected to a vacuum pump (Fig. 2B).<br />

Vacuum-infiltrate the rosettes for 2 min after starting the pump. Reintroduce<br />

carefully air in the dessicator after vacuum breakage (Fig. 2C). Transfer the<br />

infiltrated plants to a centrifuge tube, with the collar at about 1 cm at the edge<br />

of the tube (Fig. <strong>2D</strong>). Paste the lower part of the root outside of the tube with<br />

adhesive tape. Introduce at the bottom of the centrifuge tube 300 μL of recovering<br />

solution. Centrifuge infiltrated plants in swinging buckets at 200g for 17 min at<br />

Fig. 2. Vacuum-infiltration of rosette leaves. Four steps of the procedure are illustrated:<br />

(A) 4–5 wk-old plants; (B) vacuum-infiltration of immersed rosettes in a dessicator<br />

connected to a vacuum pump; (C) rosette leaves after vacuum-infiltration, note<br />

the darker part of a leaf after successful infiltration (black arrow); (D) infiltrated plant<br />

transferred to a centrifuge tube, note the drop of solution containing the protease<br />

inhibitor cocktail at the bottom of the tube (black arrow).


196 Jamet et al.<br />

20°C (see Note 5). Collect the apoplastic washing fluids with a micropipet <strong>and</strong><br />

estimate the volume. Vacuum-infiltration <strong>and</strong> centrifugation should be repeated<br />

twice.<br />

3. Assay the apoplastic fluids for malate dehydrogenase (MDH) activity to detect<br />

cytoplasmic contaminations. Measure MDH activity at room temperature in 3 mL<br />

MDH assay mixture <strong>and</strong> one-twentieth of the volume of the recovered apoplastic<br />

fluids. Reduction of NADP is followed at = 340 nm. Pool only those apoplastic<br />

washing fluids with no detectable MDH activity.<br />

4. Vacuum-infiltrate rosettes with R1, R2, R3 or R4 buffer. Check for MDH activity<br />

on the recovered apoplastic fluids as described above. Discard any apoplastic<br />

washing fluids with MDH activity. Pool the remaining apoplastic washing fluids<br />

free of MDH activity.<br />

3.1.3.2. Analysis of Labile CWP by 2-DE<br />

1. Exhaustively dialyze the apoplastic washing fluids from rosettes infiltrated with<br />

the recovering solution at 4°C against deonized water in low binding 2 kDa<br />

cutoff Spectra/Por ® CE dialysis bags. Freeze-dry the dialysates. Resuspend the<br />

dry residues in 3 mL of resuspending solution <strong>and</strong> desalt on an Econo-Pac ® 10DG<br />

desalting column equilibrated with desalting column equilibration buffer for the<br />

complete removal of mannitol. Freeze-dry the eluate.<br />

2. Solubilize the dry residue in 2-DE sample buffer <strong>and</strong> separate proteins by 2-DE<br />

using a 7 cm-IPG gel strip pH 4–7 for the first dimension.<br />

3.1.3.3. Analysis of Weakly Bound CWP<br />

1. Exhaustively dialyze the apoplastic washing fluids from rosettes infiltrated with<br />

R1, R2, R3, or R4 buffer against Hi-Trap SP equilibration buffer as described<br />

above. Apply to a Hi-Trap SP Sepharose column equilibrated with Hi-Trap SP<br />

equilibration buffer at 1 mL/min. Elute the retained basic proteins with Hi-Trap<br />

SP elution buffer at 1 mL/min. Desalt the basic proteins on an Econo-Pac ® 10DG<br />

desalting column equilibrated with desalting column equilibration buffer. Freezedry<br />

the eluate. Resuspend the dry residue in 40 μL 1-DE sample buffer <strong>and</strong><br />

separate proteins by 1-DE on a 10–17% gradient SDS-polyacrylamide gel (16.5<br />

× 13.5 × 0.15 cm).<br />

2. Freeze-dry the acidic <strong>and</strong> neutral proteins in the Hi-Trap SP Sepharose column<br />

effluent. Solubilize the dry residue with a minimal volume of resuspending<br />

solution <strong>and</strong> desalt on an Econo-Pac ® 10DG desalting column equilibrated with<br />

desalting column equilibration buffer. Freeze-dry the proteins <strong>and</strong> perform a 2-DE.<br />

3.2. Extraction of Weakly Bound CWP by Destructive Techniques<br />

3.2.1. Cell Wall <strong>Preparation</strong><br />

1. Soak A. thaliana ecotype Columbia seeds (see Note 2) in tap water for 2 h,<br />

then sterilize in diluted bleach (2.4 %) for 45 min, <strong>and</strong> rinse several times with<br />

deionized water. Sow the seeds (150 mg) in a Magenta box (6 × 6 cm) containing


Isolation of Plant Cell Wall Proteins 197<br />

50 mL of solid MS medium. Grow seedlings at 23°C in the dark for 11 d (see<br />

Note 6).<br />

2. Harvest hypocotyls (around 2 cm high) of an average of 20 Magenta boxes as<br />

follows. First, remove carefully the solid MS medium carrying the seedlings from<br />

each box. Then, cut hypocotyls below cotyledons <strong>and</strong> above root with a pair of<br />

scissors. Wash the 1-cm-long hypocotyls with distilled water onto a nylon net<br />

(1.5-mm pore size) to remove all the cut cotyledons <strong>and</strong> seed coats that stick<br />

to hypocotyls (see Note 7). Transfer the hypocotyls into 500 mL of grinding<br />

buffer <strong>and</strong> add PVPP (1g/10g fresh weight of hypocotyls) to complex phenolic<br />

compounds.<br />

3. Grind the mixture in cold room using a Waring blender at full speed for 15 min.<br />

4. Separate cell walls from soluble cytoplasmic fluid by centrifugation of the<br />

homogenate for 15 min at 1,000g <strong>and</strong> 4°C. Further purify the pellet by two<br />

successive centrifugations in 500 mL of cell wall purification buffers, 0.6M <strong>and</strong><br />

1M sucrose respectively.<br />

5. Wash the residue with 3 L of cell wall washing buffer on a nylon net (25-μm pore<br />

size) to eliminate all soluble compounds. Grind the resulting cell wall fraction in<br />

liquid nitrogen in a mortar with a pestle before lyophilization. Starting with 16 g<br />

fresh weight of hypocotyls, this procedure usually results in 1.3 g dry powder.<br />

3.2.2. Extraction of Proteins<br />

1. Typically, 0.65 g of lyophilized cell walls is used for one experiment. Extract<br />

proteins by successive salt solutions in this order: two extractions with 6 mL H1<br />

buffer, followed by two extractions with 6 mL H2 buffer. Resuspend cell walls<br />

by vortexing for 5-10 min at room temperature, <strong>and</strong> then centrifuge for 15 min<br />

at 4,000g <strong>and</strong> 4°C. Supernatants from the same extracting buffer are pooled.<br />

2. Desalt supernatants using Econo-Pac ® 10DG desalting columns equilibrated with<br />

desalting column equilibration buffer. Lyophilize the extracts <strong>and</strong> resuspend in<br />

H2O2. 3. Quantify proteins using the Bradford protein assay.<br />

4. Add 1-DE sample buffer. Separate proteins by 1-DE on a 12.5% SDSpolyacrylamide<br />

gel.<br />

3.3. Analysis of CWP<br />

3.3.1. Specific Constraints for Separation by Electrophoresis <strong>and</strong><br />

Protein Identification by Mass Spectrometry<br />

The separation of CWP by classical two-dimensional gel electrophoresis<br />

(2-DE) is difficult. Because most CWP are basic glycoproteins (Fig. 1C), they<br />

are poorly resolved by this technique (22). They are better separated by 1-DE.<br />

However, protocols including chromatographic steps to separate proteins before<br />

1-DE are available (24,25). In this chapter, a method able to separate acidic


198 Jamet et al.<br />

<strong>and</strong> basic CWP is proposed for a better resolution of these two types of CWP<br />

in 2-DE or 1-DE respectively.<br />

Frequently, in 1-DE, proteins are not well separated from one<br />

another, <strong>and</strong> a protein sample can contain a mixture of proteins.<br />

However, the peptide mass mapping technology using high resolution<br />

(< 20 ppm) MALDI-TOF mass spectrometry (MS) permits the identification<br />

of several proteins from a mixture. Search engines such as MS-<br />

FIT from Protein Prospector (http://prospector.ucsf.edu/ucsfhtml4.0/msfit.htm)<br />

or MASCOT (http://www.matrixscience.com/search_form_select.html) allow<br />

multiple searches. In case of difficulties, proteins can also be identified by<br />

peptide sequencing using LC (liquid chromatography)-MS/MS systems (9).<br />

3.3.2. Use of Bioinformatics for the Evaluation of the Efficiency<br />

of an Extraction Protocol<br />

The reliability of protein profiling for a compartment like the cell wall,<br />

strongly depends on the quality of the preparation. Unfortunately, the classical<br />

methods to check the purity of a particular fraction are not conclusive<br />

for proteomic studies, because the sensibility of the analysis by mass<br />

spectrometry is 10–1,000 times more sensitive than enzymatic or immunological<br />

tests using specific markers. Our experience in the field has shown<br />

that the most efficient way to evaluate the quality of a cell wall preparation<br />

is (1) to identify all the proteins extracted from the cell wall by<br />

mass spectrometry, <strong>and</strong> (2) to perform extensive bioinformatic analysis to<br />

determine if the identified proteins contain a signal peptide, <strong>and</strong> no retention<br />

signals for other cell compartments. Comparison of the results obtained<br />

with different programs is necessary to ensure a reliable prediction: PSORT<br />

allows predicting any subcellular localization (http://psort.ims.u-tokyo.ac.jp/<br />

form.html); TargetP looks for the presence of signal peptides for protein<br />

secretion or of transit peptides for mitochondrion or chloroplast targeting<br />

(http://www.cbs.dtu.dk/services/TargetP/); Aramemnon compares the results of<br />

several programs predicting the presence of signal peptides <strong>and</strong> transmembrane<br />

domains (http://aramemnon.botanik.uni-koeln.de/). It is then possible to<br />

conclude about the quality of the cell wall preparation by calculating the ratio<br />

of predicted secreted proteins to intracellular ones.<br />

4. Notes<br />

1. Protease inhibitor cocktail for plant is required to prevent proteolysis during<br />

the extraction procedure. Proteolysis induces the production of smaller broken<br />

proteins that can be spread over 1-D or 2-D polyacrylamide gels. Thus, proteolysis<br />

can prevent the identification of both broken proteins <strong>and</strong> other proteins of interest


Isolation of Plant Cell Wall Proteins 199<br />

by mass spectrometry. Moreover, the occurrence of these polypeptides is a great<br />

problem for quantitative <strong>and</strong> comparative proteomics.<br />

2. Seeds germinate in culture media that are favourable to development of bacteria<br />

or fungi. Because of the high amount of seeds (150 mg) introduced in a culture<br />

flask or in a Magenta box, the possible contamination events are multiplied. So,<br />

seeds should be carefully sterilized, <strong>and</strong> the healthy state of plants should be very<br />

good during their production in greenhouses.<br />

3. Culture media should be processed immediately after recovery. Otherwise, proteolysis<br />

may occur even if they are stored frozen.<br />

4. No more than two successive extractions with salt solutions should be performed.<br />

Otherwise, cells are damaged <strong>and</strong> intracellular contaminants are released in the<br />

culture medium <strong>and</strong> can stick non-specifically to cell walls (10).<br />

5. Be careful setting minimal acceleration to avoid seriously damaging the vacuuminfiltrated<br />

plants during the centrifugation step. Imperatively centrifuge in<br />

swinging buckets to get undamaged plants during spinning.<br />

6. All seedlings should grow at about the same rate to reach the same size after 11<br />

d. If germination is not homogeneous, place the boxes at 6°C during 2dtoallow<br />

all seeds to start germination without growth. Then, all boxes can be put at 23°C<br />

for 11 d.<br />

7. Cotyledons should be carefully removed. They contain few protein species but<br />

each of them in a huge amount. Because of their density, cotyledons co-sediment<br />

with cell walls. As a consequence, few cotyledons induce a significant contamination<br />

during extraction of cell wall proteins, especially by storage proteins.<br />

This contamination prevents the identification of proteins of interest by mass<br />

spectrometry.<br />

Acknowledgments<br />

The authors are grateful to the Université Paul Sabatier (Toulouse III, France)<br />

<strong>and</strong> the CNRS for support.<br />

References<br />

1. Hunter, T. C., Andon, N. L., Koller, A., Yates, J. R. <strong>and</strong> Haynes, P. A. (2002) The<br />

functional proteomics toolbox: methods <strong>and</strong> applications. J. Chromatogr. B 782,<br />

161–181.<br />

2. Carpita, N. <strong>and</strong> Gibeaut, D. (1993) Structural models of primary cell walls in<br />

flowering plants: consistency of molecular structure with the physical properties<br />

of the walls during growth. Plant J. 3, 1–30.<br />

3. Cosgrove, D. J. (2005) Growth of the plant cell wall. Nat. Rev. Mol. Cell. Biol. 6,<br />

850–861.<br />

4. Jamet, E., Canut, H., Boudart, G. <strong>and</strong> Pont-Lezica, R. F. (2006) Cell wall proteins:<br />

a new insight through proteomics. Trends Plant Sci. 11, 33–9.<br />

5. Varner, J. E. <strong>and</strong> Lin, L.-S. (1989) Plant cell wall architecture. Cell 56, 231–39.


200 Jamet et al.<br />

6. Brady, J. D., Sadler, I. H., <strong>and</strong> Fry, S.C. (1996) Di-isodityrosine, a novel tetrameric<br />

derivative of tyrosine in plant cell wall proteins: a new potential cross-link.<br />

J. Biochem. 315, 323–27.<br />

7. Schnabelrauch, L. S., Kieliszewski, M. J., Upham, B. L., Alizedeh, H. <strong>and</strong><br />

Lamport, D. T. A. (1996) Isolation of pI 4.6 extensin peroxidase from tomato cell<br />

suspension cultures <strong>and</strong> identification of Val-Tyr-Lys as putative intermolecular<br />

cross-link site. Plant J. 9, 477–89.<br />

8. Shah, K., Penel, C., Gagnon, J., <strong>and</strong> Dun<strong>and</strong>, C. (2004) Purification <strong>and</strong> identification<br />

of a Ca +2 -pectate binding peroxidase from Arabidopsis leaves. Phytochem.<br />

65, 307–12.<br />

9. Boudart, G., Jamet, E., Rossignol, M., et al. (2005) Cell wall proteins in apoplastic<br />

fluids of Arabidopsis thaliana rosettes: Identification by mass spectrometry <strong>and</strong><br />

bioinformatics. Proteomics 5, 212–21.<br />

10. Borderies, G., Jamet, E., Lafitte, C., et al. (2003) Proteomics of loosely bound cell<br />

wall proteins of Arabidopsis thaliana cell suspension cultures: a critical analysis.<br />

Electrophoresis 24, 3421–32.<br />

11. Charmont, S., Jamet, E., Pont-Lezica, R., <strong>and</strong> Canut, H. (2005) Proteomic analysis<br />

of secreted proteins from Arabidopsis thaliana seedlings: improved recovery<br />

following removal of phenolic compounds. Phytochem. 66, 453–61.<br />

12. Held, M. A., Tan, L., Kamyab, A., Hare, M., Shpak, E. <strong>and</strong> Kieliszewski, M. J.<br />

(2004) Di-isodityrosine is the intermolecular cross-link of isodityrosine-rich<br />

extensin analogs cross-linked in vitro. J. Biol. Chem. 279, 55474–82.<br />

13. Miller, J. G. <strong>and</strong> Fry, S. C. (1992) Production <strong>and</strong> harvesting of ionically wallbound<br />

extensin from living cell suspension cultures. Plant Cell Tissue Organ Cult.<br />

31, 61–66.<br />

14. Murashige, T. <strong>and</strong> Skoog, F. (1962) A revised medium for rapid growth <strong>and</strong><br />

bioassays with tobacco tissue culture. Physiol. Plant. 15, 473–97.<br />

15. Loomis, W. D. (1974) Overcoming problems of phenolics <strong>and</strong> quinones in the<br />

isolation of plant enzymes <strong>and</strong> organelles. Meth. Enzymol. 31, 528–45.<br />

16. Ramagli, L. S. <strong>and</strong> Rodriguez, L. V. (1985) Quantitation of microgram amounts<br />

of protein in two-dimensional polyacrylamide electrophoresis sample buffer.<br />

Electrophoresis 6, 559–63.<br />

17. Axelos, M., Curie, C., Mazzolini, L., Bardet, C. <strong>and</strong> Lescure, B. (1992) A protocol<br />

for transient gene expression in Arabidopsis thaliana protoplasts isolated from cell<br />

suspension cultures. Plant Physiol. Biochem. 30, 123–28.<br />

18. Voigt, J. (1985) Extraction by lithium chloride of hydroxyproline-rich glycoproteins<br />

from intact cells of Chlamydomonas reinhardii. Planta 164, 379–89.<br />

19. Smith, J., Muldoon, E., <strong>and</strong> Lamport, D. (1984) Isolation of extensin precursors by<br />

direct elution of intact tomato cell suspension cultures. Phytochem. 23, 1233–39.<br />

20. Angyal, S. (1989) Complexes of metal cations with carbohydrates in solution. Adv.<br />

Carbohydr. Chem. Biochem. 47, 1–44.<br />

21. van Buren, J. (1991) Function of pectin in plant tissue structure <strong>and</strong> firmness in,<br />

The Chemistry <strong>and</strong> Technology of Pectin (Walter, R. H. ed.), Academic Press,<br />

New York, pp. 1–22.


Isolation of Plant Cell Wall Proteins 201<br />

22. Bardy, N., Carrasco, A., Galaud, J. P., Pont-Lezica, R. <strong>and</strong> Canut, H. (1998)<br />

Free-flow electrophoresis for fractionation of Arabidopsis thaliana membranes.<br />

Electrophoresis 19, 1145–53.<br />

23. Rabilloud, T. (2002) Two-dimensional gel electrophoresis in proteomics: old, old<br />

fashioned, but still climbs up the mountains. Proteomics 2, 3–10.<br />

24. Stasyk, T. <strong>and</strong> Huber, L. A. (2004) Zooming in fractionation strategies in<br />

proteomics. Proteomics 4, 3704–16.<br />

25. Lescuyer, P., Hochstrasser, D. F., Sanchez, J. C. (2004) Comprehensive<br />

proteome analysis by chromatographic protein prefractionation. Electrophoresis 25,<br />

1125–1135.


18<br />

Isolation <strong>and</strong> <strong>Fractionation</strong> of the Endoplasmic<br />

Reticulum from Castor Bean (Ricinus communis)<br />

Endosperm for Proteomic Analyses<br />

William J. Simon, Daniel J. Maltman, <strong>and</strong> Antoni R. Slabas<br />

Summary<br />

This chapter describes the preparation <strong>and</strong> isolation of highly purified endoplasmic<br />

reticulum (ER) from the endosperm of developing <strong>and</strong> germinating castor bean (Ricinus<br />

communis) seeds to provide a purified organelle fraction for differential proteomic<br />

analyses. The method uses a two-step ultracentrifugation protocol first described by<br />

Coughlan (1) <strong>and</strong> uses sucrose density gradients <strong>and</strong> a sucrose flotation step to yield<br />

purified ER devoid of other contaminating endomembrane material. Using a combination<br />

of one dimensional (1D) <strong>and</strong> two dimensional (<strong>2D</strong>) gel electrophoresis the complexity<br />

<strong>and</strong> reproducibility of the protein profile of the purified organelle is evaluated prior to<br />

detailed proteomic analyses using mass spectrometry based techniques.<br />

Key Words: Castor bean; electrophoresis; endoplasmic reticulum; proteomics;<br />

Ricinus communis.<br />

1. Introduction<br />

The endoplasmic reticulum (ER) is a specialized endomembrane system<br />

within all cells responsible for a number of important biological processes<br />

including, protein folding, protein sorting, <strong>and</strong> secretion (2), <strong>and</strong> protein Nglycosylation<br />

(3). In the seeds of higher plants the ER is the processing site for<br />

the synthesis of storage proteins (4) <strong>and</strong> is also the site for fatty acid modification<br />

(5–7), triacyglycerol (TAG) biosynthesis (8), complex lipid biosynthesis<br />

<strong>and</strong> the primary site for membrane biogenesis.<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

203


204 Simon et al.<br />

Underst<strong>and</strong>ing the processes of TAG biosynthesis <strong>and</strong> complex lipid partitioning,<br />

particularly in the major commercially grown oil producing plant<br />

species, is an important step towards maximizing both oil quality <strong>and</strong> yield<br />

from these crops. This is particularly so if advances in molecular biology are<br />

to be used to rationally design new crops using transgenic technologies.<br />

Although the plant ER plays a central role in metabolism there have been<br />

a limited number of studies on the organelle due to the difficulties associated<br />

with its isolation <strong>and</strong> purification.<br />

In order to overcome this we have taken advantage of the liquid properties<br />

of castor bean endosperm to prepare preparative amounts of ER using modifications<br />

of the method first described by Beevers (9).<br />

The ER of developing seeds is the major site of synthesis of both lipid <strong>and</strong><br />

storage proteins – such biosynthetic capacity is lost during germination. In order<br />

to elucidate the changes in the ER during seed development <strong>and</strong> germination we<br />

have prepared material from both to allow for a detailed differential proteomic<br />

analysis of the two tissues (10).<br />

2. Materials<br />

2.1. Tissue Homogenization <strong>and</strong> ER Purification<br />

1. 5% Hypochlorite solution in water (see Note 1) for seed surface sterilization.<br />

2. Homogenization medium: 500 mM sucrose, 10 mM KCl, 1 mM EDTA, 1 mM<br />

MgCl2 2 mM dithiothreitol (DTT), 0.1 mM phenylmethyl-sulfonyl fluoride<br />

(PMSF), 150 mM Tricine-KOH pH 7.5. Keep at 4°C on ice.<br />

3. Sucrose solutions for gradient formation: prepare fresh in sterile water 50 mL of<br />

each of the following, 20%, 30%, 40%, <strong>and</strong> 60% sucrose each containing 1 mM<br />

EDTA <strong>and</strong> 0.1 mM PMSF. Keep at 4°C on ice.<br />

4. 10% glycerol solution for –80°C storage of purified ER fractions.<br />

5. The following Beckman centrifuges, rotors <strong>and</strong> centrifuge tubes:<br />

Avanti 30 centrifuge, L-70, <strong>and</strong> Optima TLX ultra-centrifuges, F0850, SW28,<br />

SW41, <strong>and</strong> TL100.4 rotors, 326823 <strong>and</strong> 344061 ultra-clear tubes <strong>and</strong> 343776<br />

micro tubes.<br />

2.2. Protein Estimations<br />

1. Protein st<strong>and</strong>ard: Bovine serum albumin (BSA) 1.0 mg/mL in water.<br />

2. Protein assay kit (modified Bradford) from BIO-RAD.<br />

2.3. SDS-Polyacrylamide Gel Electrophoresis (SDS-<strong>PAGE</strong>) Analysis<br />

of Purified ER<br />

The SDS-<strong>PAGE</strong> method described is a modification of that originally<br />

described by Laemmli (11). All reagents are from BIO-RAD.


Isolation <strong>and</strong> <strong>Fractionation</strong> of the Endoplasmic Reticulum 205<br />

1. SDS-<strong>PAGE</strong> resolving gel (10%): 3.33 mL Acrylamide-Bis solution (37.5:1), 2.5<br />

mL 1.5M Tris-HCl pH 8.8, 100 μL of 10% SDS, 100 μL of 10% ammonium<br />

persulphate, 10 μL of TEMED in a final volume of 10 mL water.<br />

2. SDS-<strong>PAGE</strong> stacking gel (4%): 1.3 mL Acrylamide-Bis solution (37.5:1), 2.5 mL<br />

0.5M Tris-HCl pH 6.8, 100 μL of 10% SDS, 100 μL of 10% ammonium persulphate,<br />

10 μL of TEMED (added last once all other constituents are mixed) in a<br />

final volume of 10 mL water.<br />

3. Water saturated isobutanol solution: Mix together equal volumes of water <strong>and</strong><br />

isobutanol in a glass bottle <strong>and</strong> shake vigorously to mix. Allow to settle <strong>and</strong> use<br />

the top layer.<br />

4. Gel running buffer: 25 mM Tris-HCl pH 8.3, 250 mM glycine, 0.1% SDS (see<br />

Note 2).<br />

5. <strong>Sample</strong> loading buffer: 50 mM Tris-HCl pH 6.8, 100mM DTT, 2% SDS, 0.1%<br />

bromophenol blue, 10% glycerol (see Note 3).<br />

6. Dalton Mark VII molecular weight markers (Sigma) made up in sample loading<br />

buffer as the manufacturers instructions.<br />

2.4. Mass Spectrometer Compatible Silver Staining<br />

Prepare all reagents fresh immediately before use.<br />

1. Fixing solution: 10% acetic acid, 40% methanol.<br />

2. Sensitizing solution: 75 mL methanol, 10 mL sodium-thiosulphate (5 %), 17 g<br />

sodium-acetate in 250 mL water.<br />

3. Washing solution: water.<br />

4. Silver solution: 0.25% silver nitrate in water.<br />

5. Developing solution: 6.25 g sodium carbonate, 100 μL formaldehyde in 250mL<br />

water.<br />

6. Stop solution: 3.65 g ethylenediaminetetracetic acid (EDTA) in 250 mL water.<br />

2.5. Mini 2 Dimensional Gels (<strong>2D</strong> gels)<br />

1. The method described in this text uses the Multiphor II electrophoresis unit<br />

combined with the Immobiline Dry Strip (IPG) kit <strong>and</strong> the Immobiline Dry<br />

Strip Reswelling Tray from GE Healthcare for the first dimension isoelectric<br />

focusing (IEF) step of <strong>2D</strong> electrophoresis. The method should be adaptable to<br />

IEF kit from any manufacturer that uses IPG strips, however focusing parameters<br />

may need to optimized. For a useful guide to <strong>2D</strong> electrophoresis see (12).<br />

2. Immobiline Dry Strip 7 cm ready made IPG strips pH 3–10 from GE Health care.<br />

3. Lysis-rehydration buffer: 9M urea, 2M thiourea, 4% CHAPS (3-[(3<br />

cholamidopropyl)dimethylammonio]-1-propanesulfonate), 1% DTT, 2% carrier<br />

ampholytes pH 3–10 (GE Healthcare). Buffer is made up without DTT <strong>and</strong><br />

carrier ampholytes, aliquoted <strong>and</strong> stored at –80°C. Once thawed DTT <strong>and</strong> carrier<br />

ampholytes are added immediately before use.


206 Simon et al.<br />

4. Equilibration buffer 1: 50 mM Tris-HCl pH 8.8, 6M urea, 30% glycerol, 10%<br />

SDS, 1% DTT. Prepare 10mL.<br />

5. Equilibration buffer 2: 50 mM Tris-HCl pH 8.8, 6M urea, 30% glycerol, 10%<br />

SDS, 4.8% iodoacetamide, 0.01% bromophenol blue. Prepare 10 mL.<br />

6. Low melting point (LMP) agarose solution: 5% made up in SDS-<strong>PAGE</strong> running<br />

buffer. Prepare 20 mL for each mini <strong>2D</strong> gels being run by heating the solution to<br />

100°C immediately before use.<br />

7. SDS-<strong>PAGE</strong> gel reagents as described in Section 2.3.<br />

8. Mass spectrometer compatible silver staining reagents as described in Section 2.4.<br />

3. Methods<br />

3.1. Isolation of ER Membranes from Germinating <strong>and</strong> Developing<br />

Castor Bean Endosperm<br />

1. In order to make a proteomic comparison between the ER from germinating <strong>and</strong><br />

developing castor bean, material must be prepared from the seeds at both growth<br />

stages. For the germinating sample mature seeds are first surface sterilized for<br />

1.0 min in 5% hypochlorite solution <strong>and</strong> then washed overnight in running tap<br />

water. The soaked seeds are then sown in moist vermiculite <strong>and</strong> germinated in the<br />

dark at 30°C for 3–4 d before dissecting out the endosperm. For the developing<br />

material seedpods are harvested from castor plants 25 d after flowering (see<br />

Note 4) <strong>and</strong> individual seeds are removed for processing.<br />

2. Take 40–50 seeds (see Note 5) from each of the germinating or developing<br />

samples <strong>and</strong> carefully dissect out the endosperm <strong>and</strong> remove the embryo <strong>and</strong><br />

cotyledons. Place the endosperm halves into a large glass Petri dish containing<br />

10 mL ice-cold homogenization buffer <strong>and</strong> manually chop them for 10 min<br />

on ice using two single-sided razor blades (see Note 6). Carry out all further<br />

procedures at 4°C unless otherwise stated.<br />

3. Filter the crude homogenate through a 100-μm nylon mesh to remove large debris<br />

<strong>and</strong> transfer the filtrate to 50-mL ultra-clear centrifuge tubes <strong>and</strong> centrifuge in a<br />

Avanti 30 bench-top centrifuge using a F0850 rotor at 1,000g at 4°C for 15 min.<br />

4. During this centrifugation step prepare in two Beckman Ultraclear 25 × 89-mm<br />

centrifuge tubes, discontinuous sucrose density gradients consisting of 7 mL of<br />

20% sucrose carefully layered on top of 13 mL of 30% sucrose. Both sucrose<br />

solutions contain 1 mM EDTA <strong>and</strong> 0.1 mM PMSF.<br />

5. With a glass rod carefully remove <strong>and</strong> discard the fat pad (see Note 7) which<br />

has formed on top of the supernatant during centrifugation of the homogenate<br />

<strong>and</strong> divide the supernatant into two halves each of which is carefully layered<br />

onto the top of the prepared sucrose gradients (step 4 above).<br />

6. Transfer the tubes to the buckets of the Beckman SW28 rotor <strong>and</strong> centrifuge at<br />

100,000g for2hat2°CinaBeckman L70 ultra-centrifuge.<br />

7. Following centrifugation mount the tubes vertically in a laboratory clamp<br />

positioned under a lamp <strong>and</strong> the membranes should be clearly visible as a<br />

distinct b<strong>and</strong> at the 20-30% sucrose interface (Fig. 1). Without disturbing the


Isolation <strong>and</strong> <strong>Fractionation</strong> of the Endoplasmic Reticulum 207<br />

Fig. 1. Following centrifugation at 100,000g for 2 h the centrifuge tube is carefully<br />

removed from the rotor without disturbing the sucrose gradient <strong>and</strong> mounted in a<br />

laboratory clamp positioned under a lamp. This enables the membrane layer to be<br />

clearly seen (marked by the arrow) at the 20–30% sucrose interface (∼1.12 g/cm 3<br />

density). As shown a hypodermic needle is used to pierce the centrifuge tube wall just<br />

below the membrane layer <strong>and</strong> the membranes are removed using a syringe.<br />

sucrose gradient carefully pierce the wall of the tube with a hypodermic needle<br />

immediately below the interface <strong>and</strong> using a syringe remove the membrane layer<br />

(Fig. 1).<br />

8. Pool the membrane fractions from both tubes <strong>and</strong> mix with an equal volume of<br />

ice cold 60% sucrose containing 1 mM EDTA <strong>and</strong> 0.1 mM PMSF.<br />

9. Pipet 4.0 mL of diluted membrane fraction into the bottom of Beckman Ultraclear<br />

14 × 89 mm centrifuge tubes <strong>and</strong> carefully overlay with 3 mL of 40% sucrose<br />

solution, 3 mL of 30% sucrose solution <strong>and</strong> 2 mL of 20% sucrose all containing<br />

1mM EDTA <strong>and</strong> 0.1 mM PMSF.


208 Simon et al.<br />

10. Transfer the tubes to the buckets of the Beckman SW41 rotor <strong>and</strong> centrifuge<br />

at 250,000g for 22 h at 2°C in a Beckman L70 ultra-centrifuge. During this<br />

centrifugation step the ER fraction will float through the dense sucrose layers<br />

to resolve as a highly purified fraction at the 20–30% sucrose interface (∼1.12<br />

g/cm 3 density.<br />

11. Carefully remove the purified ER from each tube as described in step 7 <strong>and</strong><br />

pool together. Dilute with an equal volume of ice-cold water <strong>and</strong> aliquot into<br />

Beckman thick walled 500-μL polycarbonate centrifuge tubes.<br />

12. Transfer the tubes to the Beckman TL100.4 rotor <strong>and</strong> pellet the membranes<br />

by centrifugation in a Beckman Optima TLX ultracentrifuge at 250,000g for<br />

45 min.<br />

Pour off the supernatant <strong>and</strong> re-suspend the membrane pellet in a minimum<br />

volume of 10% (v/v) glycerol aliquot into tubes suitable for storage, snap freeze<br />

in liquid nitrogen <strong>and</strong> store at –80°C (see Note 8).<br />

3.2. Protein Estimation – Modified Bradford<br />

1. Using the 1.0 mg/mL BSA stock solution (Section 2.2.1) prepare a calibration<br />

curve as outlined in Table 1. Vortex solutions <strong>and</strong> incubate at room temperature<br />

for 15 min.<br />

2. In duplicate mix 2 μL of sample with 798 μL of water <strong>and</strong> 200 μL of Bradford<br />

reagent. Vortex solutions <strong>and</strong> incubate at room temperature for 15 min.<br />

3. Using a spectrophotometer measure the absorbance of the st<strong>and</strong>ards <strong>and</strong> samples<br />

at 595 nm.<br />

Plot the calibration curve <strong>and</strong> estimate the protein concentration of the<br />

samples from this curve.<br />

3.3. (SDS-<strong>PAGE</strong>) Analysis of Purified ER from Developing<br />

<strong>and</strong> Germinating Castor Bean Endosperm<br />

1. For SDS-<strong>PAGE</strong> analyses we use the mini Protean gel kit from BIO-RAD<br />

although any gel format may be used including commercially available<br />

precast gels.<br />

Table 1<br />

Bradford assay calibration curve st<strong>and</strong>ard dilutions.<br />

BSA Series (μg) 0 1 2 5 10 15 20 40<br />

1mg/mLBSA (μL) - 1 2 5 10 15 20 40<br />

H 2O (μL) 800 799 798 795 790 785 780 760<br />

Bradford reagent (μL) 200 200 200 200 200 200 200 200


Isolation <strong>and</strong> <strong>Fractionation</strong> of the Endoplasmic Reticulum 209<br />

2. Assemble the gel kit with spacers for 0.75 mm gels <strong>and</strong> prepare 10 mL of<br />

resolving gel solution as outlined in Section 2.3.1. Using a Pasteur pipet carefully<br />

pipet the gel solution between the plates avoiding air bubbles. Leave sufficient<br />

space between the top of the gel solution <strong>and</strong> the top of the glass plates to allow<br />

for the gel comb <strong>and</strong> approximately 1 cm of stacker gel.<br />

3. Overlay the gel solution with water-saturated isobutanol <strong>and</strong> allow the gel to<br />

polymerize at room temperature which should take around 30 min.<br />

4. Pour off the isobutanol <strong>and</strong> rinse the top of the gel thoroughly with several<br />

washes of water.<br />

5. Prepare 10 mL of stacking gel solution as described in Section 2.3.2 <strong>and</strong> using a<br />

Pasteur pipet carefully overlay this gel solution onto the top of the polymerized<br />

resolving gel. Insert the gel comb avoiding the formation of air bubbles <strong>and</strong><br />

allow the stacking gel to polymerize, which should take around 30 min at room<br />

temperature.<br />

6. While the stacking gel is polymerizing prepare 800 mL of 1× gel running buffer<br />

as described in Section 2.3.4.<br />

7. Once the stacking gel has set carefully remove the gel comb <strong>and</strong> rinse out the<br />

wells with running buffer. Assemble the gel kit <strong>and</strong> fill the upper <strong>and</strong> lower<br />

buffer compartments with running buffer.<br />

8. For SDS-<strong>PAGE</strong> analyses of purified ER fractions using mass spectrometer<br />

compatible silver stain dissolve an aliquot of the purified preparation equivalent<br />

to between 10 <strong>and</strong> 20 μg of total protein in SDS-<strong>PAGE</strong> sample loading buffer<br />

(see Note 9), centrifuge for 5 min in a bench-top microfuge at maximum speed<br />

<strong>and</strong> load directly into the washed wells of the gel.<br />

9. Include at least one lane of SDS-VII molecular weight markers as described in<br />

Section 2.3.6. on every gel.<br />

10. Connect the electrophoresis tank to the power supply <strong>and</strong> run the electrophoresis<br />

at 100 volts until the bromophenol blue dye front passes through the stacker<br />

gel into the resolving gel. At this point increase the voltage to 200 volts <strong>and</strong><br />

continue until the dye front reaches the end of the gel.<br />

11. Switch off the power supply, disassemble the gel kit <strong>and</strong> carefully transfer the<br />

gel to a clean polythene container for staining.<br />

3.4. Mass Spectrometer Compatible Silver Staining<br />

1. Mass spectrometric methods for proteomic analyses are highly sensitive<br />

techniques capable of detecting <strong>and</strong> identifying subfemtomole amounts of protein<br />

in a sample. For this reason contamination of samples particularly with human<br />

keratin can be a major problem during analyses. To avoid this as much as possible<br />

use freshly prepared staining reagents, h<strong>and</strong>le gels as little as possible <strong>and</strong> use<br />

clean containers for all staining procedures.<br />

2. Following electrophoresis transfer the gel to a clean staining tray <strong>and</strong> fix the<br />

proteins in the gel by incubating the gel in 100 mL of fixing (Section 2.4.1)<br />

solution (see Note 10) for 15 min with gentle shaking (see Note 11) at room<br />

temperature. Repeat this step with a second 100 mL of fixing solution.


210 Simon et al.<br />

3. Discard the fixing solution <strong>and</strong> add 100 mL of sensitizing solution (Section 2.4.2).<br />

Incubate the gel for 30 min at room temperature with gentle shaking.<br />

4. Discard the sensitizing solution <strong>and</strong> wash the gel thoroughly with 3 × 5 min<br />

washes with fresh water.<br />

5. Pour off the last water wash, add 100 mL of silver solution (Section 2.4.4) <strong>and</strong><br />

incubate the gel for 20 min at room temperature with gentle shaking.<br />

6. Discard the silver solution <strong>and</strong> wash the gel thoroughly with 3 × 1 min washes<br />

with fresh water.<br />

7. Pour off the last water wash, add 100 mL of developing solution (Section 2.4.5)<br />

<strong>and</strong> incubate the gel for 4 minutes at room temperature with gentle shaking.<br />

Monitor the intensity of the staining during this incubation period <strong>and</strong> if necessary<br />

lengthen or shorten the developing time to achieve b<strong>and</strong>s of reasonable intensity<br />

without staining the gel background.<br />

8. Discard the developing solution, add 100 mL of stopping solution (Section 2.4.6)<br />

<strong>and</strong> incubate the gel for 10 min at room temperature with gentle shaking.<br />

9. Discard the stopping solution <strong>and</strong> wash the gel thoroughly with 3 × 5 min washes<br />

with fresh water. This staining procedure should result in discrete protein b<strong>and</strong>s<br />

stained from brown to black, depending on protein concentration, against a clear<br />

gel background. A representative 1D-SDS <strong>PAGE</strong> gel showing three independent<br />

kDa<br />

66<br />

45<br />

36<br />

29<br />

24<br />

20<br />

14<br />

S<br />

Developing ER Germinating ER<br />

1<br />

2<br />

3<br />

Fig. 2. Protein estimations were made on each preparation using the modified<br />

Bradford procedure as described in Section 3.2. Protein samples (10 μg) were<br />

mixed with SDS-<strong>PAGE</strong> sample buffer vortexed at room temperature for 10 min <strong>and</strong><br />

microfuged at maximum speed before loading into wells. Electrophoresis was carried<br />

out as described in Section 18.3.3. Lane S contains molecular weight markers. The<br />

protein profiles are highly reproducible between preparations <strong>and</strong> clear differences are<br />

seen between the developing <strong>and</strong> germinating ER samples on these gels.<br />

1<br />

2<br />

3


Isolation <strong>and</strong> <strong>Fractionation</strong> of the Endoplasmic Reticulum 211<br />

castor bean ER preparations from both developing <strong>and</strong> germinating endosperm<br />

stained using this protocol is shown in Fig. 2.<br />

3.5. Mini <strong>2D</strong> Gel Protein Profiling of Purified ER from Developing<br />

<strong>and</strong> Germinating Castor Bean Endosperm<br />

1. For differential proteomic analyses of the developing <strong>and</strong> germinating ER a<br />

number of independent biological samples need to be prepared. This is to allow<br />

for the true biological differences between the two tissue types to be compared<br />

<strong>and</strong> to enable the elimination of technical artifacts such as gel to gel variability,<br />

sample solubilization <strong>and</strong> staining artifacts. The use of mini <strong>2D</strong> gels allows<br />

the rapid <strong>and</strong> detailed evaluation of the reproducibility <strong>and</strong> complexity of the<br />

protein profiles of the purified ER fraction from independent preparations before<br />

beginning large-scale proteomic profiling experiments. Fig. 3 shows the mini<br />

<strong>2D</strong> gel profiles from three independent biological preparations of germinating<br />

<strong>and</strong> developing ER prepared using the protocols outlined in this text.<br />

2. To an aliquot of purified ER material equivalent to 50 μg of total protein from<br />

each preparation add 4 volumes of ice-cold absolute acetone <strong>and</strong> incubate on<br />

ice for 60 min. Pellet the proteins from this 80% acetone precipitation step by<br />

centrifugation for 10 min in a microfuge at maximum speed.<br />

3. Allow the pellet to air dry but do not over-dry.<br />

4. Add 125 μL (see Note 12) of <strong>2D</strong> lysis-rehydration buffer (Section 2.5.1) to<br />

the pellet <strong>and</strong> gently disperse it using a pipetman fitted with a 100 μL pipet<br />

tip. Sonicate the sample for 10 min in a sonicating water bath <strong>and</strong> incubate on<br />

a vortex shaker at 30°C for 60 min. Centrifuge for 10 min in a microfuge at<br />

maximum speed.<br />

5. Using the reswelling tray rehydrate an IPG strip with the entire solubilized<br />

sample for a minimum of 6 h but ideally overnight (see Note 13).<br />

6. Assemble the Multiphor kit for isolelectric focusing.<br />

7. Using forceps remove the re-hydrated IPG strip from the reswelling tray, rinse<br />

with water <strong>and</strong> gently blot dry.<br />

8. Place the rehydrated IPG strip in the plastic insert of the IEF tray, position the<br />

electrodes <strong>and</strong> connect them to the power supply.<br />

9. Fill the IEF tray with mineral oil so that the gel strip is completely covered.<br />

10. Fit the lid to the Multiphor unit <strong>and</strong> begin isoelectric focusing (IEF).<br />

11. IEF is typically carried out in three phases or voltage steps (see Note 14). For<br />

7-cm IPG strips with a sample loading of 50 μg of purified ER fraction these<br />

steps are outlined in Table 2.<br />

12. While the IEF gel is running prepare an SDS-<strong>PAGE</strong> mini gel for the second<br />

dimension. For this gel no stacking gel is required <strong>and</strong> the gel should be prepared<br />

with 1-mm spacers to allow for the IPG strip to be positioned on the gel surface.<br />

13. Prepare 10 mL of resolving gel solution as described in Section 2.3.1. Using a<br />

Pasteur pipet carefully pipette the gel solution between the gel plates avoiding<br />

air bubbles until the solution is about 0.8 cm from the top of the lower glass<br />

plate.


212 Simon et al.<br />

Germinating Developing<br />

pH 3.0 pH 10 S pH 3.0 pH 10 S<br />

Fig. 3. ER was independently purified from developing <strong>and</strong> germinating castor bean<br />

in three separate experiments using the protocols described in the text. Molecular<br />

weight st<strong>and</strong>ards were loaded adjacent to the basic end of the IEF strip <strong>and</strong> are marked<br />

S. An aliquot equivalent to 50 μgoftotal protein from each preparation was evaluated<br />

on mini <strong>2D</strong> gels (pH 3.0–10) <strong>and</strong> stained with mass spectrometer compatible silver<br />

stain. As can be clearly seen the independent preparations are reproducible <strong>and</strong> there<br />

are significant differences in the proteomic profiles between the two tissues.<br />

14. Overlay the gel solution with water-saturated isobutanol <strong>and</strong> allow the gel to<br />

polymerize at room temperature until the IEF is completed.<br />

15. Following IEF disconnect the gel kit from the power supply <strong>and</strong> disassemble<br />

the unit (see Note 15).<br />

16. Place the focused IPG strip into equilibration buffer 1 (Section 2.5.4) <strong>and</strong><br />

incubate at room temperature for 15 min with gentle shaking.<br />

17. Transfer strip to equilibration buffer 2 (Section 2.5.5) <strong>and</strong> incubate at room<br />

temperature for 15 min with gentle shaking.


Isolation <strong>and</strong> <strong>Fractionation</strong> of the Endoplasmic Reticulum 213<br />

Table 2<br />

Isoelectric focusing parameters a<br />

step volts mA Watts Time (h) kVh<br />

1 200 2 5 0.01<br />

2 3500 2 5 1.30 2.8<br />

3 3500 2 5 1.00 2.2<br />

a Step 1 is a low voltage step which minimizes sample aggregation. Voltage is then gradually<br />

increased during step 2 to the final focusing voltage <strong>and</strong> held at this voltage during step 3 until<br />

focusing is complete.<br />

18. During this second equilibration step pour of the isobutanol solution from the<br />

polymerized second dimension gel, wash the gel surface extensively with water<br />

<strong>and</strong> blot dry.<br />

19. Prepare low melting point agarose solution as described in Section 2.5.6.<br />

20. Remove the gel strip from the equilibration solution <strong>and</strong> place it resting on<br />

its edge on a piece of moistened filter paper to allow any excess equilibration<br />

solution to drain away.<br />

21. Using forceps position the strip between the glass plates of the second dimension<br />

gel with its backing strip against one of the glass plates <strong>and</strong> gently push the<br />

IPG strip down until the entire lower edge is in contact with the SDS-<strong>PAGE</strong><br />

gel surface. Ensure there are no air bubbles trapped between the IPG strip <strong>and</strong><br />

the gel.<br />

22. If required SDS-VII molecular weight markers can be spotted onto a small piece<br />

of filter paper <strong>and</strong> positioned in contact with the SDS-<strong>PAGE</strong> gel surface at one<br />

end of the IPG strip.<br />

23. Overlay the IPG strip <strong>and</strong> with low melting point agarose solution that has<br />

cooled to between 40–50°C.<br />

24. Assemble the gel kit, connect it to the power supply <strong>and</strong> begin running the 2nd<br />

dimension electrophoresis at 60 volts for 10 min. This will allow the proteins<br />

to migrate from the IPG strip into the SDS-<strong>PAGE</strong> gel. Increase the voltage to<br />

150 volts <strong>and</strong> continue the electrophoresis until the bromophenol dye front has<br />

reached the end of the gel.<br />

25. Following electrophoresis stain the gels using the mass spectrometer compatible<br />

silver protocol described in Section 3.4.<br />

4. Notes<br />

1. Unless otherwise stated “Water” in this text refers to high purity water with a<br />

resistivity of 18 M-cm.<br />

2. SDS-<strong>PAGE</strong> running buffer can be made up as a 10× stock solution, stored at<br />

room temperature <strong>and</strong> diluted as required for use.


214 Simon et al.<br />

3. SDS-<strong>PAGE</strong> sample loading buffer can be conveniently made up for use as a 5×<br />

stock solution, divided into aliquots <strong>and</strong> stored at –20°C.<br />

4. In order to ensure that developing seeds are collected at the correct developmental<br />

stage plants are monitored daily during growth <strong>and</strong> flowers are tagged<br />

as they emerge. The seedpods are then allowed to develop <strong>and</strong> are harvested<br />

after 25 d.<br />

5. The protocol described here has been optimised to yield the maximum amount of<br />

purified ER devoid of contamination with other plant endomembrane material.<br />

Our attempts to scale up this procedure using higher seed numbers as starting<br />

material have only resulted in contaminated preparations. We recommend that<br />

for scale up use the protocol as described but increase the number of preparations.<br />

6. Carefully chopping the endosperm with single sided razor blades allows for<br />

successful tissue homogenisation without significant membrane disruption.<br />

7. During the low speed centrifugation step of the crude homogenate to remove<br />

cell debris a fat pad will form on top of the supernatant in the centrifuge tube.<br />

It is important that this fat pad is carefully removed without disruption before<br />

the supernatant containing the membrane fraction is loaded onto the sucrose<br />

gradient.<br />

8. The typical yield of purified ER from a Ricinus germinating or developing<br />

preparation is approx 1.0 mg. We usually re-suspend the final pellet of purified<br />

ER in 500 μL of 10% (v/v) glycerol <strong>and</strong> divide into 5× 100-μL aliquots for<br />

storage at –80°C.<br />

9. <strong>Sample</strong>s for SDS-<strong>PAGE</strong> analyses are best prepared by adding a volume of 5 x<br />

sample buffer to an aliquot of sample equivalent to 10–20 μg of total protein<br />

<strong>and</strong> then diluting to 5 times the volume to give a sample ready for loading in<br />

1× sample buffer.<br />

10. For mass spectrometric compatible silver staining use 100mL of each solution<br />

for each mini gel being processed.<br />

11. Gels can be left in fixing solution overnight or longer if necessary until staining<br />

is required. During staining procedures gels are shaken gently on a gel rocker<br />

platform or on an orbital shaker set at low speed.<br />

12. The volume of <strong>2D</strong> lysis-rehydration buffer required to solubilize a sample is<br />

dependent on the dimensions of the IPG strip being re-hydrated. For mini gels<br />

using 7-cm IPG strips the volume is 125 μL, for 11-cm IPG strips 200 μL, for<br />

13-cm IPG strips 250 μL <strong>and</strong> for 18-cm large format gels 350 μL.<br />

13. Using the IEF reswelling tray up to 12 IPG can be rehydrated with samples at<br />

the same time.<br />

14. The voltage <strong>and</strong> duration of IEF is dependent on the pH <strong>and</strong> length of the IPG<br />

strip being used <strong>and</strong> on the total amount of protein loaded onto the gel. For<br />

guidelines on the parameters used with different IPG strips see the technical<br />

data sheets supplied with the IPG strips by the manufacturers.<br />

15. Following IEF gel strips can be equilibrated immediately ready for the SDS-<br />

<strong>PAGE</strong> second dimension step or they can be stored at –20°C <strong>and</strong> the equilibration<br />

<strong>and</strong> second dimension done at a later date.


Isolation <strong>and</strong> <strong>Fractionation</strong> of the Endoplasmic Reticulum 215<br />

References<br />

1. Coughlan, S., Hastings, C., <strong>and</strong> Winfrey Jnr, R. J. (1996) Molecular charcterisation<br />

of plant endoplasmic reticulum – identification of protein disulphide-isomerase as<br />

the major reticuloplasmin. Eur. J. Biochem 235, 215–24.<br />

2. Vitale, A. <strong>and</strong> Denecke, J. (1999) The endoplasmic reticulum – gateway of the<br />

secretory pathway. The Plant Cell 11, 614–28.<br />

3. Helenius, A. <strong>and</strong> Aebi, M. (2004) The role of N-linked glycans in the endoplasmic<br />

reticulum. Ann. Rev. Biochem, 73, 1019–49.<br />

4. Galil, G., Sengupta-Gopalan, C., <strong>and</strong> Ceriotti, A. (1998) The endoplasmic reticulum<br />

<strong>and</strong> its role in protein maturation <strong>and</strong> biogenesis of oil bodies. Plant. Mol. Biol 38,<br />

1–29.<br />

5. Cassagne, C., Lessire R., Bessoule, J., et al. (1994) Biosynthesis of very long chain<br />

fatty acids in plants. Prog. Lipid Res. 33, 55–69.<br />

6. Van de Loo, F.J., Broun, P., Turner, S., <strong>and</strong> Somerville, C. (1995) Expressed<br />

sequence tags from developing castor seeds. Proc. Natl. Acad. Sci. USA. 92,<br />

6743–47.<br />

7. Shanklin, J. <strong>and</strong> Cahoon, E. B. (1998) Desaturation <strong>and</strong> related modifications of<br />

fatty acids. Annu. Rev. Plant Physiol. Plant Mol. Biol. 49, 611–41.<br />

8. Kennedy, E. P. (1961) Biosynthesis of complex lipids. Fed. Proc. Fed. Am. Soc.<br />

Exp. Biol. 20, 934–40.<br />

9. Beevers, H. (1979) Microbodies in higher plants. Annu. Rev. Plant Physiol. 30,<br />

159–73.<br />

10. Maltman, D. J., Simon, W. J., Wheeler, C. H., Dunn, M. J., Wait, R., <strong>and</strong><br />

Slabas, A. R. (2002) Proteomic analysis of the endoplasmic reticulum from developing<br />

<strong>and</strong> germinating seed of castor (Ricinus communis). Electrophoresis 23,<br />

626–39.<br />

11. Laemmli, U. K. (1970) Cleavage of structural proteins during the assembly of the<br />

head of bacteriophage T4. Nature 227, 680–85.<br />

12. Berkelman, T. <strong>and</strong> Stenstedt, T. (ed.) (1998) 2-D Electrophoresis using immobilized<br />

pH gradients – principles <strong>and</strong> methods. Amersham Pharmacia Biotech h<strong>and</strong>book.


19<br />

Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics<br />

Aida Pitarch, César Nombela, <strong>and</strong> Concha Gil<br />

Summary<br />

The cell wall is an external envelope shared by yeasts <strong>and</strong> filamentous fungi that<br />

defines the interface between the microorganism <strong>and</strong> its environment. It is an extremely<br />

complex structure consisting of an elastic framework of microfibrillar polysaccharides<br />

(glucans <strong>and</strong> chitin) that surrounds the plasma membrane <strong>and</strong> to which a wide array of<br />

different proteins, often heavily glycosylated, are anchored in various ways. Intriguingly,<br />

these cell wall proteins (CWPs) play a key role in morphogenesis, adhesion, pathogenicity,<br />

antigenicity, <strong>and</strong> as a promising target for antifungal drug design. However, the CWPs are<br />

difficult to analyze because of their low abundance, low solubility, hydrophobic nature,<br />

extensive glycosylation, covalent attachment to the wall polysaccharide skeleton, <strong>and</strong><br />

high heterogeneity. We describe a typical procedure of cell wall fractionation to isolate<br />

<strong>and</strong> solubilize different CWP species from yeasts <strong>and</strong> filamentous fungi according to<br />

the type of linkages that they establish with other wall components <strong>and</strong> under suitable<br />

conditions for following reproducible proteomic analyses. CWPs retained noncovalently or<br />

by disulfide bonds are first extracted from isolated yeast or fungal cell walls by detergents<br />

<strong>and</strong> reducing agents. Subsequently, CWPs covalently linked to or closely entrapped within<br />

the internal glucan-chitin network are sequentially released either by mild alkali conditions<br />

or by enzymatic treatments first with glucanases <strong>and</strong> then with chitinases. This strategy<br />

is a powerful tool not only for obtaining an overview of the sophisticated cell wall<br />

proteome of yeasts <strong>and</strong> filamentous fungi, but also for characterizing mechanisms of<br />

incorporation, assembly <strong>and</strong> retention of CWPs into this intricate cellular compartment<br />

<strong>and</strong> their interactions with structural wall polysaccharides.<br />

Key Words: Cell wall; cell wall proteins; fractionation; fungus; GPI proteins; PIR<br />

proteins; proteomics; yeast.<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

217


218 Pitarch et al.<br />

Fig. 1. Schematic representations of the cell wall from S. cerevisiae <strong>and</strong> C. albicans,<br />

<strong>and</strong> molecular modules of CWPs solubilized by the present procedure of cell wall<br />

fractionation. The cell wall of S. cerevisiae <strong>and</strong> C. albicans basically consists of -1,3<br />

<strong>and</strong> -1,6-glucans, chitin, mannoproteins <strong>and</strong> proteins. -1,3-glucan <strong>and</strong> chitin form<br />

an elastic microfibrillar polysaccharide skeleton surrounding the plasma membrane to<br />

which mannoproteins are attached through -1,6-glucan, alkali-sensitive bonds, <strong>and</strong>/or<br />

other hitherto uncharacterized linkages. Cell wall proteins (CWPs; mannoproteins <strong>and</strong>


Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics 219<br />

1. Introduction<br />

The cell wall is an intricate structure common to yeasts <strong>and</strong> filamentous fungi<br />

that surrounds the plasma membrane <strong>and</strong> is strategically placed at the interface<br />

between the cell <strong>and</strong> its environment, including the host (1–3). This external<br />

envelope, accountable for 20–30% of the cell dry weight, is essential for the<br />

survival of the microorganism. In fact, it is involved in many vital functions,<br />

such as physical protection, osmotic stability, selective permeability barrier,<br />

immobilized enzyme support, cell-cell interactions (e.g., cell recognition <strong>and</strong><br />

adhesion) <strong>and</strong> morphogenesis, to name but a few. In pathogen fungi, this<br />

cellular compartment further takes an active part in virulence, pathogenicity,<br />

antigenicity, immunomodulation of the immune response, <strong>and</strong> adhesion to host<br />

substrates (2). Most importantly, its essential nature <strong>and</strong> its fungal specificity<br />

(given its absence in mammalian cells) interestingly make the cell wall an<br />

attractive target site (see Note 1) to design antifungal drugs with selective<br />

toxicity for human pathogen fungi, such as C<strong>and</strong>ida albicans or Aspergillus<br />

fumigatus, among others (4–6).<br />

Although the cell wall structure <strong>and</strong> organization have been investigated<br />

most extensively in the prototype yeast Saccharomyces cerevisiae, a similar<br />

molecular model is also applicable for other ascomycetes, <strong>and</strong> in particular<br />

for C. albicans, a dimorphic fungus capable of growing either in yeast form<br />

or as hyphae (see Note 2) (1,7,8). The cell wall of S. cerevisiae is mainly<br />

composedofglucans(with-1,3<strong>and</strong>-1,6linkages),chitin(N-acetylglucosamine<br />

polymers), <strong>and</strong> proteins (often highly O- <strong>and</strong>/or N-mannosylated) interconnected<br />

by covalent <strong>and</strong>/or non-covalent bonds, leading to an elevated complexity (see<br />

Fig. 1). -1,3-glucan, the major component of the cell-wall (electron-transparent)<br />

inner layer, forms an elastic three-dimensional microfibrillar framework, encircling<br />

the cell, to which other wall constituents are covalently anchored. Chitin<br />

is often cross-linked to the -1,3-glucan microfibrillar backbone on its inner<br />

side (close to the plasma membrane) <strong>and</strong>, to a lesser extent, to short sidechains<br />

of -1,6-glucan. This N-acetylglucosamine polymer presents low levels<br />

(see Note 2), except in the budding neck ring, in the primary septum, in <strong>and</strong><br />

around the bud scars, or under stress conditions. Both -1,3-glucan <strong>and</strong> chitin<br />

(structural wall polysaccharides) provide mechanical strength <strong>and</strong> elasticity<br />

◭<br />

Fig. 1. proteins) can also be loosely associated, either by non-covalent bonds or<br />

through disulfide bridges, with other covalently linked CWPs. See Introduction for<br />

further information. The callouts depict details for potential mechanisms of CWP<br />

retention into the cell wall on the basis of the procedure of cell wall fractionation<br />

described in this chapter to isolate <strong>and</strong> solubilize different CWP species from yeasts<br />

<strong>and</strong> filamentous fungi.


220 Pitarch et al.<br />

to the cell wall. -1,6-glucan, a flexible minor wall component, interconnects<br />

certain cell wall proteins (CWPs), the so-called glycosyl phosphatidylinositol<br />

(GPI)-CWPs, with -1,3-glucan (∼90% of GPI-CWPs) or chitin (∼10%<br />

of GPI-CWPs) through a phosphodiester bridge in their GPI remnant (see<br />

Note 3). The CWPs are mostly located on the outside of this -1,3-glucanchitin<br />

network (i.e., at the cell-wall electron-dense outer layer) <strong>and</strong>, in minor<br />

amounts, throughout the cell wall, determining its porosity. These CWPs can be:<br />

1. Loosely associated, either noncovalently or through disulfide bonds, with other<br />

cell wall components. This group of CWPs comprises (1) soluble precursor<br />

forms of covalently linked CWPs, (2) proteins related to the biosynthesis <strong>and</strong><br />

modulation of wall constituents, such as -1,3-glucosyltransferase (Bgl2p), exoglucanase<br />

(Exg1p) <strong>and</strong> chitinase (Cts1p), <strong>and</strong> (3) noncanonical proteins<br />

“classically” considered to be confined to the intracellular compartment because<br />

they lack the conventional secretory signal sequence (2,9–11). Nevertheless, the<br />

mechanism by which these nonconventional proteins are targeted to the cell<br />

surface remains enigmatic (11–13). This array of loosely associated proteins is<br />

commonly found at the cell surface but also, in a smaller ratio, in the cell-wall<br />

inner layers. These CWPs can be extracted using detergents <strong>and</strong> reducing agents<br />

(see Fig. 1).<br />

2. Covalently linked to -1,3-glucan:<br />

a. Directly via an alkali-labile linkage (speculatively through a O-linked sidechain),<br />

such as PIR-CWPs (CWPs with internal repeats). The PIR-CWPs are<br />

highly O-mannosylated proteins with one or more internal repeat regions, a<br />

N-terminal signal peptide, a Kex2 proteolytic processing site, <strong>and</strong> a C-terminal<br />

sequence with four cysteine residues at highly conserved positions (1,14–17).<br />

They are normally located in the cell-wall inner layer (18). Other CWPs belong<br />

to this category are also present in the yeast cell walls (see the following <strong>and</strong><br />

Fig. 2). This type of CWPs can be solubilized under mild alkali conditions or<br />

by enzymatic treatment with -1,3-glucanases but not with -1,6-glucanases<br />

(see Figs. 1–3).<br />

b. Indirectly by a -1,6-glucan moiety through their GPI remnant, such as GPI-<br />

CWPs. The GPI-CWPs are highly O-glycosylated proteins with an N-terminal<br />

signal peptide, a C-terminal GPI anchor addition signal, <strong>and</strong> serine- <strong>and</strong><br />

threonine-rich regions (see Note 3) (1,7,17,19). These CWPs are predominantly<br />

placed in the cell-wall outer layer. This group of CWPs can be released<br />

either by enzymatic treatment with -1,3 or -1,6-glucanases or by using<br />

hydrofluoric acid (HF)-pyridine, which cleavages the phosphodiester bond in<br />

the GPI remnant (20) (GPI-CWP 1 in Figs. 1–3).<br />

3. Covalently anchored to chitin by a -1,6-glucan moiety via their GPI remnant,<br />

such as some GPI-CWPs (21,22). This type of CWP-polysaccharide complex is<br />

largely found in the lateral walls or under stress conditions. These CWPs can be


Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics 221<br />

Fig. 2. Schematic representations of the different known types of covalently linked<br />

CWPs in S. cerevisiae <strong>and</strong> C. albicans, <strong>and</strong> the most commonly used methods to<br />

solubilize them from isolated cell walls. These diagrams are based on information from<br />

references (1,14,18-21,23). See Introduction for further details. PIR-CWPs, a small<br />

number of hybrid GPI-CWPs (e.g., Cwp1p (23); GPI-CWPs 2 <strong>and</strong> 3) at acidic pHs, <strong>and</strong><br />

other CWPs (25,32) are directly attached to -1,3-glucan through an alkali-sensitive<br />

linkage (ASL). GPIr denotes GPI remnant, <strong>and</strong> ASL alkali-sensitive linkage.<br />

extracted either by enzymatic treatment with chitinases or -1,6-glucanases or by<br />

using HF-pyridine (GPI-CWP 4 in Figs. 1–3).<br />

However, it is unsurprising that other types of linkages, hitherto uncharacterized,<br />

among CWPs <strong>and</strong> structural wall components are also present in the<br />

yeast cell wall (see Fig. 1). Be that as it may, there is no doubt that the molecular<br />

model of the yeast cell wall is even more sophisticated than that outlined above.<br />

This is because certain CWPs can simultaneously be retained into the -1,3glucan/chitin<br />

skeleton in various ways. For instance, the Cwp1p, a S. cerevisiae<br />

GPI-CWP, is double-anchored to the -1,3-glucan framework both through an<br />

alkali-sensitive linkage <strong>and</strong> by its -1,6-glucan moiety, playing an important<br />

role in stress response (23). Hence, two additional GPI-CWP-polysaccharide<br />

complexes are defined (GPI-CWPs 2 <strong>and</strong> 3 in Figs. 2 <strong>and</strong> 3).<br />

Taking into account the special architecture <strong>and</strong> nature of the cell wall, the<br />

isolation <strong>and</strong> solubilization of CWPs from this complex cellular compartment<br />

is not therefore an evident <strong>and</strong> easy affair. Indeed, CWPs from yeasts


222 Pitarch et al.<br />

Fig. 3. Venn diagram summarizing the most commonly used methods to extract the<br />

different known types of CWPs in S. cerevisiae <strong>and</strong> C. albicans. This chart is based<br />

on data from references (1,10,14,18–21,23). The GPI-CWP numbers refer to those<br />

indicated in Fig. 2.<br />

<strong>and</strong> filamentous fungi are tricky enough to resolve by two-dimensional<br />

electrophoresis (2-DE) gels, because of their low abundance, low solubility,<br />

hydrophobicity, high heterogeneity, extensive glycosylation (especially, O<strong>and</strong>/or<br />

N-mannosylation), <strong>and</strong> covalent attachment to the wall polysaccharide<br />

(-1,3-glucan/chitin) skeleton (11,24). These problems can, at least to some<br />

degree, be solved by sequential solubilization of CWPs on the basis of the type<br />

of attachments that they establish to other cell wall components (11,17,25).<br />

This procedure implies breakage of the covalent linkages between CWPs <strong>and</strong><br />

wall polysaccharides (see Chapter 20). Intriguingly, cell wall fractionation is<br />

an appropriate paradigm system to:<br />

1. Reduce the intricacy of the cell wall.<br />

2. Enrich samples for CWPs <strong>and</strong> thus increase the detection of low-abundance<br />

species by removing the most abundant soluble gene products.<br />

3. Enhance the solubility of large, low abundance, <strong>and</strong>/or hydrophobic CWPs.<br />

4. Define (map <strong>and</strong> identify) the proteins that make up the cell wall (the cell wall<br />

proteome), <strong>and</strong> elaborate a comprehensive <strong>and</strong> integrated view of the complex<br />

CWP composition. The CWP resolution can be increased using the cell wall<br />

fractionation procedure described here, because the heterogeneous population of<br />

protein species present in the yeast <strong>and</strong> fungal cell envelope can be distributed<br />

over several 2-DE gels (11).<br />

5. Characterize mechanisms of incorporation, assembly <strong>and</strong> retention of CWPs into<br />

the cell wall.<br />

6. Elucidate the CWP interactions with cell wall polysaccharides.


Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics 223<br />

7. Study protein-protein interactions or regulatory networks exclusive to the cell<br />

wall.<br />

8. Monitor abnormal protein expression localized to this external envelope.<br />

9. Discover novel diagnostic/prognostic markers, antifungal targets <strong>and</strong>/or therapeutic<br />

c<strong>and</strong>idates for human mycoses.<br />

This chapter will integrate a typical procedure of cell wall fractionation to<br />

extract different CWP species from isolated cell walls of yeasts or filamentous<br />

fungi according to their interactions with other wall components. The resulting<br />

selectively enriched CWP fractions can then directly be (1) analyzed by 2-<br />

DE (see Note 4) <strong>and</strong> mass spectrometry (MS) (11,17) or (2) digested with<br />

trypsin, followed by liquid chromatography (LC) in t<strong>and</strong>em with MS analyses<br />

(25) to circumvent some of the difficulties associated with in-gel digestion of<br />

the heavily glycosylated CWPs (12). The purity <strong>and</strong> quality of these enriched<br />

fractions of CWPs should be screened by using bona fide markers both of the<br />

cell wall <strong>and</strong> of intracellular compartments before carrying out any interpretation<br />

of the results. However, the unambiguous evidence for their cell wall<br />

location will only be established after (1) their in situ immunolocalization (i.e.,<br />

immunoelectron microscopy or immunofluorescence studies) <strong>and</strong>/or (2) the use<br />

of tagged fusion proteins (e.g., c-myc-tag or green fluorescence protein (GFP)<br />

fusion proteins) based on the fusion of the protein in question to modified<br />

versions of extracellular enzymes that rely on a detectable phenotype.<br />

2. Materials<br />

Growth media, solutions <strong>and</strong> buffers should be sterilized by autoclaving<br />

before use when working under sterile conditions. Their labile components<br />

should be sterilized separately using a 0.22-μm filter (Millipore, Bedford, MA)<br />

<strong>and</strong> then added to the other autoclaved ingredients. All solutions <strong>and</strong> buffers<br />

should be prepared with ultrapure water, as provided by Nanopure or Milli-Q<br />

18 M/cm resistivity systems (Millipore), <strong>and</strong> precooled when the procedure<br />

is carried out at 4°C.<br />

2.1. Cell Wall Isolation from Yeasts <strong>and</strong> Filamentous Fungi<br />

1. Yeast-Peptone-D-glucose (YPD) plates: 1% (w/v) yeast extract (Difco Laboratories,<br />

Detroit, MI), 2% (w/v) peptone (Difco), 2% (w/v) d-glucose, 2% (w/v)<br />

agar (Difco).<br />

2. YPD medium: 1% (w/v) yeast extract, 2% (w/v) peptone, 2% (w/v) d-glucose.<br />

3. Bead mill homogenizer (model MSK; Braun Biotech International GmbH,<br />

Melsungen, Germany).<br />

4. Liquid carbon dioxide (CO2).


224 Pitarch et al.<br />

5. 0.40- to 0.60-mm chilled, acid-washed glass beads (Sartorius, Goettingen,<br />

Germany) (see Note 5).<br />

6. PMSF stock solution: 0.1 M in isopropanol. Dissolve 174 mg of phenylmethylsulfonyl<br />

fluoride (PMSF; Fluka, Chelmsford, MA) in a final volume of 10 mL<br />

isopropanol, <strong>and</strong> store at –20°C (see Note 6). PMSF should be h<strong>and</strong>led with<br />

caution because it is highly toxic. Weigh this hazardous compound in a fume<br />

hood, <strong>and</strong> wear gloves, goggles <strong>and</strong> a mask.<br />

7. Lysis buffer: 10 mM Tris-HCl, pH 7.4, 1 mM PMSF.<br />

8. YPD-chloramphenicol plates: 1% (w/v) yeast extract, 2% (w/v) peptone, 2%<br />

(w/v) d-glucose, 2% (w/v) agar (Difco), 10-μg/mL chloramphenicol. When the<br />

1-L autoclaved YPD <strong>and</strong> agar solution cools to ∼65°C, add 1 mL of 10×<br />

chloramphenicol solution.<br />

9. Chloramphenicol solution (10X): Dissolve 10 mg of chloramphenicol in a final<br />

volume of 1 mL ethanol, <strong>and</strong> sterilize using a 0.2-μm filter.<br />

10. Wash solution A: 1 mM PMSF.<br />

11. Wash solution B: 5% (w/v) NaCl, 1 mM PMSF.<br />

12. Wash solution C: 2% (w/v) NaCl, 1 mM PMSF.<br />

13. Wash solution D: 1% (w/v) NaCl, 1 mM PMSF.<br />

2.2. Protein Solubilization from Isolated Yeast <strong>and</strong> Fungal Cell Walls<br />

2.2.1. By Detergents <strong>and</strong> Reducing Agents<br />

1. Wash buffer: 50 mM Tris-HCl, pH 8.0, 1 mM PMSF.<br />

2. Extraction buffer: 50 mM Tris-HCl, pH 8.0, 0.1 M EDTA, 2% (w/v) SDS, 10 mM<br />

DTT (dithiothreitol; see Note 7).<br />

2.2.2. Under Mild Alkali Conditions<br />

1. Wash solution: 1 mM PMSF.<br />

2. Wash buffer: 0.1 M sodium acetate, pH 5.5, 1 mM PMSF.<br />

3. Extraction solution: 30 mM NaOH, 1 mM PMSF.<br />

4. Stop solution: acetic acid.<br />

2.2.3. By -1,3-Glucanase Treatment<br />

1. Wash solution: 1 mM PMSF.<br />

2. Wash buffer: 50 mM Tris-HCl, pH 7.5, 1 mM PMSF.<br />

3. Extraction buffer: 1,500 U Quantazyme ylgTM (Quantum Biotechnologies Inc,<br />

Montreal, Canada; see Note 8) per gram of wet weight of cell walls, in 2 mL of<br />

a solution containing 50 mM Tris-HCl, pH 7.5, 10 mM DTT, 1 mM PMSF (see<br />

Note 9).<br />

4. Stop solution: 10% SDS.


Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics 225<br />

2.2.4. By Exochitinase Treatment<br />

1. Wash solution: 1 mM PMSF.<br />

2. Wash buffer: 50 mM sodium phosphate buffer, pH 6.3.<br />

3. Extraction buffer: 0.3 U exochitinase (Sigma, St. Louis, MO) per gram of wet<br />

weight of cell walls, in 2 mL of a solution containing 50 mM sodium phosphate<br />

buffer, pH 6.3 (see Note 10).<br />

4. Stop solution: 10% SDS.<br />

2.3. Protein Precipitation.<br />

1. 100% Trichloroacetic acid (TCA) solution: Dissolve 100 g of TCA in sufficient<br />

water to yield a final volume of 100 mL (see Note 11). TCA should be h<strong>and</strong>led<br />

with caution, because it is extremely caustic. Protect eyes <strong>and</strong> avoid contact with<br />

skin when working with TCA solutions.<br />

2. Acetone precooled to –20°C.<br />

3. Neutralizing solution: 0.1 N NaOH.<br />

3. Methods<br />

The protocols described below outline a typical procedure of cell wall<br />

fractionation to isolate <strong>and</strong> solubilize different CWP species from yeasts <strong>and</strong><br />

filamentous fungi on the basis of the type of linkages that they establish<br />

with other wall components. This method involves (1) cell homogenization<br />

by physical disruption techniques, (2) isolation of cell walls by differential<br />

centrifugation, (3) sequential solubilization of CWPs from isolated cell walls<br />

using different chemical agents (detergents, reducing agents, <strong>and</strong> alkalis) <strong>and</strong><br />

enzymes (glucanases <strong>and</strong> chitinases), <strong>and</strong> (4) precipitation of the resulting<br />

selectively enriched CWP fractions under suitable conditions for subsequent<br />

proteomic analyses. A flowchart of the strategy presented here is shown in<br />

Fig. 4. This is based on earlier methods described by Kapteyn et al. (20,21)<br />

<strong>and</strong> Mrsa et al. (26), <strong>and</strong> on recent modifications reported by Pitarch et al. (11)<br />

that have proved effective at properly extracting CWPs from S. cerevisiae <strong>and</strong><br />

C. albicans. Nevertheless, adaptation of growth <strong>and</strong> extraction conditions may<br />

be required for other species of yeasts <strong>and</strong> filamentous fungi.<br />

3.1. Cell Wall Isolation from Yeasts <strong>and</strong> Filamentous Fungi (see<br />

Note 12)<br />

Although cell disintegration can be accomplished using a wide variety of<br />

techniques, mechanical breakage of cells using glass beads is certainly one of<br />

the most quick <strong>and</strong> reliable procedures to disrupt the cell walls <strong>and</strong> plasma<br />

membranes of yeasts <strong>and</strong> filamentous fungi (see Note 13). After cell disruption,


226 Pitarch et al.<br />

Fig. 4. Flowchart of a typical procedure of cell wall fractionation to isolate <strong>and</strong><br />

solubilize different CWP species from yeasts <strong>and</strong> filamentous fungi according to the<br />

type of attachments that they establish to other cell wall components.<br />

cell walls can be separated from other cytosolic <strong>and</strong> membranous components<br />

by differential centrifugation of the cell homogenate at relatively low speed<br />

(see Note 14).


Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics 227<br />

1. Grow yeast or fungal cells on an YPD plate (stock maintenance medium) at<br />

30°C for 2 days. Use a single colony to inoculate 50 mL of YPD (or selective)<br />

medium in a 250-mL flask (see Note 15), <strong>and</strong> grow overnight at 30ºC with<br />

vigorous rotary shaking (200 rpm).<br />

2. Use this 50-mL preculture to inoculate four 2-L flasks containing 500 mL fresh<br />

YPD medium each, <strong>and</strong> grow at 30°C in a shaking incubator (200 rpm) until<br />

the culture reaches early-log phase growth (OD 600nm = 0.5–1; see Note 16).<br />

3. Harvest the cells by centrifugation at 4,500g for 5 min (see Note 17) <strong>and</strong> discard<br />

the supernatant.<br />

4. Resuspend the cell pellet in 200 mL ice-cold water, <strong>and</strong> centrifuge 5 min at<br />

4,500g. Decant the supernatant.<br />

5. Resuspend the cell pellet in 200 mL ice-cold lysis buffer, <strong>and</strong> centrifuge 5 min<br />

at 4,500g. Discard the supernatant.<br />

6. Resuspend the cells in 3 volumes of ice-cold lysis buffer, <strong>and</strong> add 3–4 volumes<br />

of 0.5-mm acid-washed glass beads (see Note 18). Transfer the cell suspension<br />

to an appropriate sized shaking flask (see Note 19).<br />

7. Grind the suspension at maximum speed for 30–60 s using a bead mill homogenizer<br />

<strong>and</strong> cooling with liquid CO 2 (for example, in a CO 2-refrigerated MSK<br />

homogenizer), <strong>and</strong> then place the shaking flask on ice for 1–2 min (see Note 20).<br />

Repeat this step until complete cell breakage. Monitor the degree of cell breakage<br />

with a phase-contrast microscope <strong>and</strong> by plating on YPD-chloramphenicol plates<br />

(see Note 21).<br />

8. Enable the glass beads to settle out <strong>and</strong> collect the supernatant carefully<br />

(see Note 22). Wash the glass beads with ice-cold lysis buffer <strong>and</strong> collect the<br />

washings until they are clear (see Note 23). Pool the supernatant <strong>and</strong> all the<br />

washings.<br />

9. Centrifuge the pooled supernatant <strong>and</strong> washings (cell homogenate) at 1,000g for<br />

10 min at 4°C. Discard the supernatant carefully (see Note 24).<br />

10. Resuspend the isolated cell walls in 200 mL ice-cold wash solution A. Centrifuge<br />

10 min at 1,000g <strong>and</strong> at 4°C. Carefully decant the supernatant. Repeat this step<br />

two more times.<br />

11. Resuspend the cell walls in 200 mL ice-cold wash solution B. Centrifuge 10 min<br />

at 1,000g <strong>and</strong> at 4°C. Carefully decant the supernatant. Repeat this step four<br />

more times (see Note 25).<br />

12. Resuspend the walls in 200 mL ice-cold wash solution C. Centrifuge 10 min<br />

at 1,000g <strong>and</strong> at 4°C. Carefully remove the supernatant. Repeat this step four<br />

more times.<br />

13. Resuspend the pellet in 200 mL ice-cold wash solution D. Centrifuge 10 min at<br />

1,000g <strong>and</strong> at 4°C. Carefully decant the supernatant. Repeat this step four more<br />

times (see Note 26).<br />

14. Resuspend the walls in 200 mL ice-cold wash solution A. Centrifuge 10 min at<br />

1,000g <strong>and</strong> at 4°C. Carefully discard the supernatant. Repeat this step two more<br />

times using preweighed centrifuge bottles.<br />

15. Weigh the wet wall pellet (see Note 27).


228 Pitarch et al.<br />

3.2. Protein Solubilization from Isolated Yeast <strong>and</strong> Fungal Cell Walls<br />

CWPs can then be solubilized sequentially from isolated cell walls by a wide<br />

variety of reagents (e.g., detergents, reducing agents, alkalis, <strong>and</strong> hydrolytic<br />

enzymes, among others) in connection with their attachments to other wall<br />

components (see Fig. 4). All procedures are performed at 4°C with prechilled<br />

solutions, reagents <strong>and</strong> apparatus (see Note 28) unless otherwise indicated.<br />

3.2.1. By Detergents <strong>and</strong> Reducing Agents<br />

Detergents, such as sodium dodecyl sulfate (SDS) or n-octylglucoside, can<br />

be used to extract CWPs that are associated noncovalently with other wall<br />

components. The use of reducing agents, such as dithiothreitol (DTT) or mercaptoethanol<br />

(ME), (1) enables the solubilization of CWPs that are loosely<br />

associated, either by disulfide bridges or through non-covalent bonds, with other<br />

covalently linked CWPs, (2) results in an increase in the cell wall porosity, <strong>and</strong> (3)<br />

facilitates the subsequent action of wall degrading enzymes (see Figs. 1 <strong>and</strong> 4).<br />

1. Resuspend the purified cell walls (prepared as described in Subheading 3.1) in<br />

200 mL ice-cold wash buffer. Centrifuge 10 min at 1,000g <strong>and</strong> carefully decant<br />

the supernatant (see Note 24).<br />

2. Add 5 mL of extraction buffer for each wet gram of cell walls <strong>and</strong> resuspend. Boil<br />

the cell wall suspension at 100°C for 10 min, <strong>and</strong> centrifuge 10 min at 1,000g.<br />

3. Collect the supernatant <strong>and</strong> store the pellet.<br />

4. Precipitate the supernatant (containing SDS/DTT-extractable CWPs) <strong>and</strong> store<br />

at –80°C (see Note 29 <strong>and</strong> Fig. 4).<br />

5. Repeat step 2 with the stored pellet <strong>and</strong> carefully discard the supernatant (see<br />

Note 30).<br />

6. Weigh the wet wall pellet (containing SDS/DTT-resistant cell walls) <strong>and</strong> divide<br />

it equally between two tubes (see Fig. 4 <strong>and</strong> Notes 27 <strong>and</strong> 31). Store them for<br />

further extractions (see Subheadings 3.2.2, 3.2.3 <strong>and</strong> 3.2.4).<br />

3.2.2. Under Mild Alkali Conditions<br />

Treatment of SDS/DTT-resistant cell walls under mild alkali conditions<br />

(using 30 mM NaOH) results in the extraction of CWPs linked to -1,3-glucan<br />

through an alkali-sensitive linkage (of unknown nature) by the -elimination<br />

process (1, 14–17). PIR-CWPs, some GPI-CWPs, <strong>and</strong> other CWPs belong this<br />

group (see Figs. 1–4).<br />

1. Resuspend one tube of the purified SDS/DTT-resistant cell walls (prepared as<br />

described in Subheading 3.2.1) in 200 mL ice-cold wash solution. Centrifuge<br />

10 min at 1,000g <strong>and</strong> carefully decant the supernatant (see Note 24). Repeat this<br />

step two more times.<br />

2. Resuspend the walls in 200 mL ice-cold wash buffer. Centrifuge 10 min at 1,000g<br />

<strong>and</strong> carefully discard the supernatant. Repeat this step five to seven more times.


Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics 229<br />

3. Add 4 mL of ice-cold extraction solution for each wet gram of cell walls <strong>and</strong><br />

resuspend. Incubate the cell wall suspension at 4°C for 17 h with gentle shaking<br />

(see Note 32).<br />

4. Stop the chemical reaction by adding neutralizing amounts of acetic acid (see<br />

Note 33).<br />

5. Centrifuge 10 min at 1,000g <strong>and</strong> collect the supernatant (containing alkalisensitive<br />

CWPs). Precipitate or dialyze the clear supernatant (see Note 33) <strong>and</strong><br />

store at –80°C.<br />

3.2.3. By -1,3-Glucanase Treatment<br />

-1,3-glucanases (commercially available) can be used to extract CWPs<br />

covalently anchored to -1,3-glucan. This group of CWPs released from<br />

SDS/DTT-resistant cell walls by these wall hydrolytic enzymes contains (1)<br />

GPI-CWPs, which are indirectly attached to -1,3-glucan, via -1,6-glucan,<br />

through a phosphodiester bridge <strong>and</strong> can alternatively be solubilized either by -<br />

1,6-glucanases (see Note 34) or by using HF-pyridine, (2) alkali-sensitive CWPs<br />

(including PIR-CWPs <strong>and</strong> some GPI-CWPs, among others), which are anchored<br />

to -1,3-glucan through uncharacterized linkages (perhaps by a O-linked sidechain)<br />

<strong>and</strong> can also be released under mild alkali conditions (using 30 mM<br />

NaOH; see Subheading 3.2.2), <strong>and</strong> (3) other CWPs linked to -1,3-glucan<br />

through other types of hitherto unidentified bridges (see Figs. 1–4).<br />

1. Resuspend the other tube of the purified SDS/DTT-resistant cell walls (prepared<br />

as described in Subheading 3.2.1) in 200 mL ice-cold wash solution. Centrifuge<br />

10 min at 1,000g <strong>and</strong> carefully decant the supernatant (see Note 24). Repeat this<br />

step two more times.<br />

2. Resuspend the walls in 200 mL ice-cold wash buffer. Centrifuge 10 min at 1,000g<br />

<strong>and</strong> carefully remove the supernatant. Repeat this step five to seven more times.<br />

3. Add 2 mL of extraction buffer for each wet gram of cell walls <strong>and</strong> resuspend (see<br />

Note 34). Incubate the cell wall suspension at 37°C for 17 h with gentle shaking.<br />

4. Stop the enzymatic reaction by adding the stop solution at a final concentration<br />

of 0.4% (w/v) <strong>and</strong> heating at 100°C for 3–5 min.<br />

5. Centrifuge 10 min at 1,000g.<br />

6. Collect the supernatant (containing -1,3-glucanase-extractable CWPs) <strong>and</strong> store<br />

the pellet (containing SDS/DTT <strong>and</strong> -1,3-glucanase-resistant cell walls) for<br />

further extractions (see Subheading 3.2.4).<br />

7. Precipitate the clear supernatant <strong>and</strong> store at –80°C.<br />

3.2.4. By Exochitinase Treatment<br />

Enzymatic treatment of the SDS/DTT- <strong>and</strong> -1,3-glucanase-resistant cell<br />

walls with exochitinases leads to the solubilization of CWPs covalently<br />

anchored to chitin. These comprise (1) a small subgroup of GPI-CWPs, which


230 Pitarch et al.<br />

are indirectly linked to chitin, via -1,6-glucan, through their GPI remnant <strong>and</strong><br />

can also be extracted either by -1,6-glucanases or by using HF-pyridine, <strong>and</strong><br />

(2) other CWPs attached to chitin through other types of hitherto uncharacterized<br />

linkages (see Figs. 1–4).<br />

1. Resuspend the SDS/DTT <strong>and</strong> -1,3-glucanase-resistant cell walls (prepared as<br />

described in Subheading 3.2.3) in 200 mL ice-cold wash solution. Centrifuge<br />

10 min at 1,000g <strong>and</strong> carefully decant the supernatant (see Note 24). Repeat this<br />

step two more times.<br />

2. Resuspend the walls in 200 mL ice-cold wash buffer. Centrifuge 10 min at 1,000g<br />

<strong>and</strong> carefully discard the supernatant. Repeat this step five to seven more times.<br />

3. Add 2 mL of extraction buffer for each wet gram of cell walls <strong>and</strong> resuspend.<br />

Incubate the cell wall suspension at 37°C for 17 h with gentle shaking.<br />

4. Stop the enzymatic reaction by adding the stop solution at a final concentration<br />

of 0.4% (w/v) <strong>and</strong> heating for 3–5 min at 100°C.<br />

5. Centrifuge 10 min at 1,000g <strong>and</strong> collect the supernatant (containing<br />

-1,3-glucanase-resistant <strong>and</strong> exochitinase extractable CWPs).<br />

6. Precipitate the clear supernatant <strong>and</strong> store at –80°C.<br />

3.3. Protein Precipitation.<br />

The different selectively enriched CWP fractions obtained in the<br />

Subheading 3.2 should be concentrated <strong>and</strong> desalted before carrying out further<br />

proteomic analyses. These can be (1) dialyzed against a volatile buffer <strong>and</strong><br />

dried, or (2) precipitated with TCA as described below (see Note 35), among<br />

other methods.<br />

1. Add 1/9th the total volume of the protein sample of an ice-cold 100% TCA<br />

solution to a final concentration of 10%.<br />

2. Mix thoroughly <strong>and</strong> incubate on ice for 30 min.<br />

3. Centrifuge the suspension at 10,000g for 15 min, <strong>and</strong> discard the supernatant (see<br />

Note 36).<br />

4. Wash protein pellet twice with cold acetone to remove residual TCA.<br />

5. Air dry for 30 min.<br />

6. Add neutralizing amounts of 0.1N NaOH (see Note 37) <strong>and</strong> store at –80°C.<br />

4. Notes<br />

1. Supporting this enterprise, -1,3-glucan is targeted by a new antifungal drug<br />

class in recent clinical use, i.e., echinoc<strong>and</strong>ins (including caspofungin <strong>and</strong><br />

micafungin), which blocks the biosynthesis of this cell wall polysaccharide (4–6).<br />

2. It must be borne in mind that unlike S. cerevisiae, many filamentous fungi<br />

contain further -glucans <strong>and</strong> a high chitin content in their cell walls (1).<br />

3. The GPI-proteins are translocated into the endoplasmic reticulum (ER), where<br />

(1) the N-terminal signal peptide (secretion signal necessary to enter the classical


Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics 231<br />

ER-Golgi secretory pathway) is cleaved, (2) the C-terminal GPI anchor addition<br />

signal is replaced with a GPI anchor, <strong>and</strong> (3) O- <strong>and</strong>/or N-linked core glycosylation<br />

takes place. Remarkably, further glycosylation also then occurs in the<br />

Golgi apparatus. These GPI-anchored proteins are directed through the secretory<br />

pathway to the outer side of the plasma membrane, where they are attached<br />

through their C-terminal GPI anchors (see Fig. 5). Intriguingly, some of them are<br />

released from the plasma membrane by cleavage of their GPI anchors, resulting<br />

in GPI anchor remnants (a truncated, lipidless GPI anchors) (1,19,27,28). These<br />

proteins are then covalently incorporated into the cell wall, by the attachment<br />

of their GPI remnants to -1,6-glucan (19).<br />

4. It is convenient to use gradient gels that enable protein separation up to at<br />

least 500 kDa, because the extensive glycosylation (especially, O- <strong>and</strong>/or Nmannosylation)<br />

of a large proportion of CWPs results in extremely high apparent<br />

molecular masses on SDS-<strong>PAGE</strong> or 2-DE gels (7).<br />

5. The size of the glass beads is crucial to achieve an efficient cell disruption.<br />

Optimal bead size for spores is 0.1 mm <strong>and</strong> for yeast <strong>and</strong> mycelia 0.5 mm.<br />

6. The PMSF can also be solubilized in ethanol, methanol <strong>and</strong> 1,2-propanediol. It<br />

is unstable in aqueous solution. PMSF is added to reduce possible proteolytic<br />

processes. It inhibits serine proteases (e.g., trypsin, chymotrypsin, <strong>and</strong> thrombin)<br />

<strong>and</strong> thiolproteases (e.g., papain).<br />

7. 40 mM -mercaptoethanol may be substituted for 10 mM DTT. mercaptoethanol<br />

or DTT should be added just before use.<br />

8. Quantazyme ylg TM is a recombinant yeast -1,3-glucanase purified from E. coli<br />

that is completely free of protease, endo- <strong>and</strong> exonuclease activity. It is highly<br />

stable in solution for months at 4°C, maintaining its whole activity.<br />

9. The use of reducing agents facilitates the ability of Quantazyme ylg TM to degrade<br />

the cell wall -1,3-glucan. 40 mM -mercaptoethanol or 10 mM cysteine may<br />

be substituted for 10 mM DTT.<br />

10. Exochitinase is a cell wall lytic enzyme isolated from Serratia marcescens that<br />

catalyzes the progressive degradation of chitin, starting at its nonreducing end.<br />

This preparation contains phosphate buffer salts <strong>and</strong> shows an optimum pH<br />

of 6.0.<br />

11. Because TCA is very hygroscopic, the whole content of a newly opened TCA<br />

bottle should be used to prepare the TCA stock solution.<br />

12. Perform all procedures from this subheading until isolated cell walls are obtained<br />

under sterile conditions. Use sterile centrifuge bottles.<br />

13. Mechanical cell breakage using a bead mill homogenizer is considered the<br />

technique of choice for disrupting cells with cell walls, especially spores, yeasts<br />

<strong>and</strong> fungi, but it also works successfully with algae, bacteria, plant, <strong>and</strong> animal<br />

tissue culture in suspension. Cell disruption takes place by the crushing action<br />

of the glass beads, which are vigorously agitated by shaking or stirring, after<br />

crashing with the cells. The Braun MSK cell homogenizer combines two types<br />

of motions in the shaking flask: rotation <strong>and</strong> tumbling. For small-scale preparations,<br />

a Fast-Prep cell breaker (Q-Biogene, Carlsbad, CA) can be used in lieu


232 Pitarch et al.<br />

Fig. 5. Putative model of the incorporation of GPI-anchored proteins into the cell<br />

wall. This is based on information from references (19,27,28). Their GPI anchor is<br />

processed at the plasma membrane leading to a GPI remnant (a truncated, lipidless GPI<br />

anchor). This is cross-linked to -1,6-glucan when these GPI proteins are covalently<br />

incorporated into the cell wall. See Note 3 for further details.


Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics 233<br />

of the Braun MSK cell homogenizer. Cell breakage of filamentous fungi can<br />

alternatively be achieved by (1) grinding freeze-dried mycelium in a mortar <strong>and</strong><br />

pestle, (2) grinding mycelium in liquid nitrogen, or (3) homogenizing thick cell<br />

pastes. When h<strong>and</strong>ling liquid nitrogen, use insulated gloves, avoid any potential<br />

contact because of the risk of frostbite, <strong>and</strong> never utilize glass containers because<br />

they may break.<br />

14. Differential centrifugation in sucrose density gradients may alternatively be used<br />

to isolate the cell wall fraction.<br />

15. Liquid cultures should be grown in a flask that is at least 4–5 times larger than<br />

the culture volume.<br />

16. It is important that the yeast culture is in early-log phase growth<br />

(∼ OD 600nm= 0.5–1) because it is easier to disrupt their cell walls with the bead<br />

mill homogenizer than those close to or in stationary phase growth. It must be<br />

borne in mind that the composition of the cell wall changes with the growth<br />

stage <strong>and</strong> culture conditions (growth temperature, external pH, oxygen levels,<br />

<strong>and</strong> composition of the growth medium (1,29)), among others. Hence, it may be<br />

necessary to adjust the cell density according to the specific objectives of the<br />

experiment.<br />

17. Overall, yeasts are successfully harvested by centrifugation, whereas vacuumassisted<br />

filtration, rather than centrifugation, is often preferred for harvesting<br />

filamentous fungi.<br />

18. The wet weight (in grams) of the cell pellet is nearly equal to the packed cell<br />

volume (in milliliters). Add about 3 mL of ice-cold lysis buffer for each wet<br />

gram of cell pellet <strong>and</strong> resuspend.<br />

19. The shaking flasks can be made of glass or stainless steel. The latter are better at<br />

transferring heat. It is essential to exclude all air from the shaking flask before<br />

the cell breakage to avoid foaming <strong>and</strong> denaturation of proteins.<br />

20. It is extremely important that the temperature of the cell suspension remains at<br />

4°C during the cell disruption to prevent heat inactivation <strong>and</strong> denaturation of<br />

proteins. Use liquid CO 2 to cool the protein sample during cell breakage.<br />

21. This procedure is carried out until complete cell breakage, which will vary with<br />

yeast/fungal strain <strong>and</strong> growth stage. The degree of cell breakage should be<br />

examined:<br />

a. Before proceeding: by observing cell lysis under a phase-contrast microscope.<br />

b. After proceeding: by plating an aliquot of the cell suspension after <strong>and</strong><br />

before cell disruption on YPD-chloramphenicol plates <strong>and</strong> growing at 30°C.<br />

The ratio of CFUs after to before cell breakage is then calculated to<br />

estimate the efficiency of cell disruption <strong>and</strong>, therefore, potential intracellular<br />

contamination in the subsequent steps. Failure of cells to grow on<br />

YPD-chloramphenicol plates should be evidenced.<br />

22. The supernatant (cell homogenate) <strong>and</strong> following washings (residual cell<br />

homogenate) can be collected (1) by decanting after allowing the glass beads to<br />

settle out by gravity, or (2) by straining through a perforated tube or “strainer”


234 Pitarch et al.<br />

(see Fig. 6). To perform the last method, transfer the cell homogenate containing<br />

the glass beads to a plastic tube <strong>and</strong> make holes of a diameter less than 0.4–0.6<br />

mm in its bottom with a flamed needle to obtain a “strainer.” After straining<br />

the cell homogenate off, collect it <strong>and</strong> wash the glass beads with ice-cold lysis<br />

buffer. Collect the washing, <strong>and</strong> wash again until the collected washings are<br />

clear. Pool the cell homogenate <strong>and</strong> all the washings. The washing step is critical<br />

Fig. 6. Proposed procedure for removing the glass beads from the cell homogenate.<br />

In the method described here, the cell homogenate is strained through a perforated tube<br />

with holes of a diameter less than 0.4–0.6 mm in its bottom, whereas the glass beads<br />

(with a diameter of 0.4–0.6 mm) are retained in the perforated tube or “strainer” (see<br />

Note 22). Successive washings with ice-cold lysis buffer should then be performed to<br />

increase the recovery of cell homogenate, <strong>and</strong> thus of cell wall material.


Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics 235<br />

to avoid any loss of material while removing the glass beads from the cell<br />

homogenate.<br />

23. To reuse the glass beads, rinse them by soaking in concentrated nitric acid for<br />

1 h, <strong>and</strong> then flush through with water. Dry them in a baking oven, cool, <strong>and</strong><br />

store at 4°C (see Fig. 6).<br />

24. It is important to decant the supernatant carefully, because the cell-wall pellet<br />

is less compact than the preceding cell pellets. In general, care should be taken<br />

during the following steps of this procedure (especially during decanting of the<br />

washings) to prevent any loss of cell walls, <strong>and</strong> therefore of CWPs. Be that as<br />

it may, this potential loss of protein material does not interestingly result in a<br />

preferential deficiency of certain CWP species or families.<br />

25. The purpose of extensively washing the isolated cell walls with solutions<br />

of decreasing concentrations of NaCl is to remove potential extracellular,<br />

membranous <strong>and</strong>/or cytosolic protein contaminants that can be adhered to them<br />

through nonspecific ionic interactions (see Fig. 7). The number of washing steps<br />

will therefore rely on the objectives of the experiment.<br />

26. A further washing step with 0.1 M Na 2CO 3 overnight at 4°C under gentle shaking<br />

may reduce potential cytoplasmic contamination that is found into microsomes,<br />

because this treatment allows them to be opened <strong>and</strong> washed (30,31).<br />

27. The wet weight (in grams) of cell walls in the pellet can be calculated by taking<br />

the weight of the centrifuge bottle (which has been previously weighed) away<br />

from the total weight (i.e., the weight of wall pellet plus centrifuge bottle).<br />

28. Centrifugation must be performed in refrigerated centrifuges at 4°C, with<br />

prechilled rotors, to avoid undesirable proteolytic activity.<br />

29. This fraction is not a pure preparation of cell wall, but rather is enriched in CWPs<br />

(loosely associated with other wall components) <strong>and</strong> may potentially contain a<br />

small amount of membrane proteins.<br />

30. This extra step is important to remove any remaining SDS/DTT-extractable<br />

CWPs <strong>and</strong> potential membranous components from the isolated cell walls, which<br />

will be used in subsequent CWP extraction steps (see Fig. 4).<br />

31. This pellet, containing SDS/DTT-resistant cell walls, is divided equally between<br />

two tubes to independently extract alkali-sensitive CWPs <strong>and</strong> -1,3-glucanaseextractable<br />

CWPs in the following steps (see Fig. 4).<br />

32. It is convenient to place the cell wall suspension into a container with ice in a<br />

cool room at 4°C (see Fig. 4).<br />

33. The chemical reaction must be stopped by acid neutralization. This can be<br />

performed:<br />

a. by adding neutralizing amounts of acetic acid to the wall suspension. The<br />

clear supernatant containing alkali-sensitive CWPs should then be (i) precipitated<br />

by adding nine volumes of ice-cold methanol (18) or (ii) dialyzed<br />

against water or 20 mM bis-Tris, pH 6.0 (32).<br />

b. by subsequent protein precipitation of the clear supernatant containing alkalisensitive<br />

CWPs with TCA (see Subheading 3.3).


236 Pitarch et al.<br />

Fig. 7. Basic principle of the procedure used for washing the isolated cell walls<br />

of yeasts <strong>and</strong> filamentous fungi. Potential extracellular, membranous <strong>and</strong>/or cytosolic<br />

protein contaminants that may nonspecifically associate with the isolated cell walls<br />

through ionic interactions can be removed under relatively more stringent conditions.<br />

Extensive washings of the isolated cell walls with high ionic strength (e.g., 5% NaCl)<br />

can disrupt the nonspecific ionic interactions between the isolated cell walls <strong>and</strong> these<br />

putative contaminants.


Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics 237<br />

34. Endo--1,6-glucanase isolated from Trichoderma harzianum (33) can be used<br />

to release GPI-CWPs (0.8 U/g wet weight of cell walls in 100 mM sodium<br />

acetate pH 5.5) (18,20). Subsequently, -1,6-glucanase-digested cell walls can<br />

be treated with Quantazyme or ice-cold 30 mM NaOH to extract (1) the -<br />

1,6-glucanase-resistant PIR-CWPs, (2) the -1,6-glucanase-resistant GPI-CWPs<br />

(GPI-CWPs linked to the -1,3-glucan through a alkali-sensitive linkage), <strong>and</strong><br />

(3) hybrid GPI-CWPs, such as Cwp1p (see Fig. 2) (23).<br />

35. The protein concentration should be higher than 100 μg/mL before precipitating<br />

with TCA. If the amount of protein precipitated is less than 1 nmole, the protein<br />

sample should be concentrated <strong>and</strong> desalted by other ways (e.g., by ultrafiltration<br />

or forced dialysis), because the pellet is imperceptible.<br />

36. The supernatant should be stored in case the protein did not precipitate.<br />

37. Protein preparation can then be resuspended in a small volume of buffer suitable<br />

for the subsequent analytical procedures (e.g. sample buffer for SDS-<strong>PAGE</strong> or<br />

2-DE).<br />

Acknowledgments<br />

We thank the Merck, Sharp & Dohme (MSD) Special Chair in Genomics<br />

<strong>and</strong> Proteomics, European Community (STREP LSHB-CT-2004-511952),<br />

Comunidad de Madrid (S-SAL-0246/2006) <strong>and</strong> Comisión Interministerial de<br />

Ciencia y Tecnología (BIO-2003-00030 <strong>and</strong> BIO-2006-01989) for financial<br />

support of our laboratory.<br />

References<br />

1. Klis, F. M., Boorsma, A. <strong>and</strong> de Groot, P. W. (2006) Cell wall construction in<br />

Saccharomyces cerevisiae. Yeast, 23, 185–202.<br />

2. Chaffin, W. L., Lopez-Ribot, J. L., Casanova, M., Gozalbo, D. <strong>and</strong> Martinez, J. P.<br />

(1998) Cell wall <strong>and</strong> secreted proteins of C<strong>and</strong>ida albicans: identification, function,<br />

<strong>and</strong> expression. Microbiol Mol.Biol Rev. 62, 130–80.<br />

3. Bernard, M. <strong>and</strong> Latge, J. P. (2001) Aspergillus fumigatus cell wall: composition<br />

<strong>and</strong> biosynthesis. Med.Mycol. 39, 9–17.<br />

4. Odds, F. C., Brown, A. J., <strong>and</strong> Gow, N. A. (2003) Antifungal agents: mechanisms<br />

of action. Trends Microbiol. 11, 272–79.<br />

5. Morrison, V. A. (2006) Echinoc<strong>and</strong>in antifungals: review <strong>and</strong> update.<br />

Expert.Rev.Anti.Infect.Ther. 4, 325–42.<br />

6. Deresinski, S. C. <strong>and</strong> Stevens, D. A. (2003) Caspofungin. Clin.Infect.Dis. 36,<br />

1445–57.<br />

7. de Groot, P., Ram, A. F., <strong>and</strong> Klis, F. M. (2005) Features <strong>and</strong> functions of<br />

covalently linked prtoeins in fungal cell walls. Fungal.Genet.Biol. 42, 657–75.<br />

8. Ruiz-Herrera, J., Elorza, M. V., Valentin, E <strong>and</strong> Sent<strong>and</strong>reu, R. (2006) Molecular<br />

organization of the cell wall of C<strong>and</strong>ida albicans <strong>and</strong> its relation to pathogenicity.<br />

FEMS Yeast Res. 6, 14–29.


238 Pitarch et al.<br />

9. Klis, F. M., de Groot, P. <strong>and</strong> Hellingwerf, K. (2001) Molecular organization of the<br />

cell wall of C<strong>and</strong>ida albicans. Med.Mycol. 39, 1–8.<br />

10. Cappellaro, C., Mrsa, V. <strong>and</strong> Tanner, W. (1998) New potential cell wall glucanases<br />

of Saccharomyces cerevisiae <strong>and</strong> their involvement in mating. J.Bacteriol. 180,<br />

5030–37.<br />

11. Pitarch, A., Sanchez, M., Nombela, C. <strong>and</strong> Gil, C. (2002) Sequential fractionation<br />

<strong>and</strong> two-dimensional gel analysis unravels the complexity of the dimorphic fungus<br />

C<strong>and</strong>ida albicans cell wall proteome. Mol.Cell Proteomics 1, 967–82.<br />

12. Pitarch, A., Nombela, C., <strong>and</strong> Gil, C. (2006) C<strong>and</strong>ida albicans biology <strong>and</strong><br />

pathogenicity: Insights from proteomics, in Microbial Proteomics: Functional<br />

Biology of Whole Organisms (Humphery-Smith,I. <strong>and</strong> Hecker,M., eds), Wiley-Vch,<br />

Hoboken, NJ, pp. 285–330.<br />

13. Nombela, C., Gil, C. <strong>and</strong> Chaffin, W. L. (2005) Non-conventional protein secretion<br />

in yeast. Trends Microbiol. 14, 15–21.<br />

14. Mrsa, V. <strong>and</strong> Tanner, W. (1999) Role of NaOH-extractable cell wall proteins<br />

Ccw5p, Ccw6p, Ccw7p <strong>and</strong> Ccw8p (members of the Pir protein family) in stability<br />

of the Saccharomyces cerevisiae cell wall. Yeast 15, 813–20.<br />

15. Kapteyn, J. C., Van Egmond, P., Sievi, E., Van Den, E. H., Makarow, M. <strong>and</strong><br />

Klis, F. M. (1999) The contribution of the O-glycosylated protein Pir2p/Hsp150<br />

to the construction of the yeast cell wall in wild-type cells <strong>and</strong> beta-1,6-gluc<strong>and</strong>eficient<br />

mutants. Mol.Microbiol. 31, 1835–44.<br />

16. K<strong>and</strong>asamy, R., Vediyappan, G. <strong>and</strong> Chaffin, W. L. (2000) Evidence for the<br />

presence of Pir-like proteins in C<strong>and</strong>ida albicans. FEMS Microbiol Lett. 186,<br />

239–43.<br />

17. Weig, M., Jansch, L., Gross, U., De Koster, C. G., Klis, F. M., <strong>and</strong> de Groot, P. W.<br />

(2004) Systematic identification in silico of covalently bound cell wall proteins<br />

<strong>and</strong> analysis of protein-polysaccharide linkages of the human pathogen C<strong>and</strong>ida<br />

glabrata. Microbiology 150, 3129–44.<br />

18. Kapteyn, J. C., Hoyer, L. L., Hecht, J. E., et al. (2000) The cell wall architecture of<br />

C<strong>and</strong>ida albicans wild-type cells <strong>and</strong> cell wall-defective mutants. Mol.Microbiol.<br />

35, 601–11.<br />

19. Kollar, R., Reinhold, B. B., Petrakova, E., et al. (1997) Architecture of the yeast<br />

cell wall. -1,6-glucan interconnects mannoprotein, -1,3-glucan, <strong>and</strong> chitin. J.Biol<br />

Chem. 272, 17762–75.<br />

20. Kapteyn, J. C., Montijn, R. C., Vink, E., et al. (1996) Retention of Saccharomyces<br />

cerevisiae cell wall proteins through a phosphodiester-linked -1,3-/-1,6-glucan<br />

heteropolymer. Glycobiology 6, 337–45.<br />

21. Kapteyn, J. C., Ram, A. F., Groos, E. M., et al. (1997) Altered extent of<br />

cross-linking of -1,6-glucosylated mannoproteins to chitin in Saccharomyces<br />

cerevisiae mutants with reduced cell wall -1,3-glucan content. J.Bacteriol. 179,<br />

6279–84.<br />

22. Sestak, S., Hagen, I., Tanner, W. <strong>and</strong> Strahl, S. (2004) Scw10p, a cell-wall<br />

glucanase/transglucosidase important for cell-wall stability in Sacccharomyces<br />

cerevisiae. Microbiology, 150, 3197–3208.


Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics 239<br />

23. Kapteyn, J. C., ter Riet, B., Vink, E., et al. (2001) Low external pH induces HOG1dependent<br />

changes in the organization of the Saccharomyces cerevisiae cell wall.<br />

Mol.Microbiol. 39, 469–479.<br />

24. Pitarch, A., Sanchez, M., Nombela, C. <strong>and</strong> Gil, C. (2003): Analysis of the<br />

C<strong>and</strong>ida albicans proteome. I. Strategies <strong>and</strong> applications. J.Chromatogr.B<br />

Analyt.Technol.Biomed.Life Sci. 787, 101–128.<br />

25. de Groot, P. W., de Boer, A. D., Cunningham, J., et al. (2004) Proteomic analysis of<br />

C<strong>and</strong>ida albicans cell walls reveals covalently bound carbohydrate-active enzymes<br />

<strong>and</strong> adhesins. Eukaryot.Cell 3, 955–65.<br />

26. Mrsa, V., Seidl, T., Gentzsch, M. <strong>and</strong> Tanner, W. (1997) Specific labelling<br />

of cell wall proteins by biotinylation. Identification of four covalently linked<br />

O-mannosylated proteins of Saccharomyces cerevisiae. Yeast 13, 1145–54.<br />

27. Lipke, P. N. <strong>and</strong> Kurjan, J. (1992) Sexual agglutination in budding yeasts. Structure,<br />

function, <strong>and</strong> regulation of adhesion glycoproteins. Microbiol. Rev. 56, 180–94.<br />

28. Roh, D. H., Bowers, B., Riezman, H. <strong>and</strong> Cabib, E. (2002) Rho1p mutations<br />

specific for regulation of -1,3-glucan synthesis <strong>and</strong> the order of assembly of the<br />

yeast cell wall. Mol.Microbiol. 44, 1167–83.<br />

29. Aguilar-Uscanga, B. <strong>and</strong> Francois, J. M. (2003) A study of the yeast cell wall<br />

composition <strong>and</strong> structure in response to growth conditions <strong>and</strong> mode of cultivation.<br />

Lett.Appl.Microbiol. 37, 268–74.<br />

30. Fujiki, Y., Hubbard, A. L., Fowler, S. <strong>and</strong> Lazarow, P. B. (1982) Isolation of<br />

intracellular membranes by means of sodium carbonate treatment: application to<br />

endoplasmic reticulum. J.Cell Biol. 93, 97–102.<br />

31. Lopez-Villar, E., Monteoliva, L., Larsen, M. R., et al. (2006) Genetic <strong>and</strong> proteomic<br />

evidences support the localization of yeast enolase in the cell surface. Proteomics<br />

6, S107–S118.<br />

32. Yin, Q. Y., de Groot, P., Dekker, H. L., de Jong, L., Klis, F. M. <strong>and</strong> de Koster, C. G.<br />

(2005) Comprehensive proteomic analysis of Saccharomyces cerevisiae cell walls:<br />

Identification of proteins covalently attached via glycosylphosphatidylinositol<br />

remnants or mild alkali-sensitive linkages. J.Biol Chem. 280, 20894–901.<br />

33. De La Cruz, J., Pintor-Toro, J. A., Benitez, T. <strong>and</strong> Llobell, A. (1995) Purification<br />

<strong>and</strong> characterization of an endo--1,6-glucanase from Trichoderma harzianum that<br />

is related to its mycoparasitism. J.Bacteriol. 177, 1864–71.


20<br />

Collection of Proteins Secreted from Yeast Protoplasts<br />

in Active Cell Wall Regeneration<br />

Aida Pitarch, César Nombela, <strong>and</strong> Concha Gil<br />

Summary<br />

The yeast cell wall is a dynamic <strong>and</strong> complex matrix of polysaccharides (glucans,<br />

mannans, <strong>and</strong> chitin), proteins <strong>and</strong> minor amounts of lipids that isolate the yeast from<br />

the extracellular medium, protecting it against osmotic <strong>and</strong> physical injuries. Removal of<br />

this essential structure for cell integrity <strong>and</strong> viability by controlled enzymatic digestion<br />

in an iso-osmotic medium brings about protoplast formation. When yeast protoplasts<br />

are incubated in an osmotically stabilized liquid nutrient medium, cell wall precursors<br />

are secreted into the culture medium to de novo synthesize the cell wall. During the<br />

early stages of the regeneration process of protoplast walls, many wall protein precursors<br />

(presumably structural proteins along with remodeling <strong>and</strong> cross-linking enzymes) are<br />

shed into the extracellular medium but not covalently incorporated into the nascent cell<br />

wall, intriguingly enabling their easy isolation <strong>and</strong> solubilization. We have developed a<br />

method to collect proteins secreted from yeast protoplasts in active cell wall regeneration<br />

under conditions that are suitable for subsequent proteomic analyses. This procedure<br />

circumvents some of the troubles intrinsically related to other extraction protocols of cell<br />

wall proteins, such as chemical or enzymatic modifications, <strong>and</strong> poor quality in protein<br />

resolution <strong>and</strong> identification because of linkages to glucan/chitin residues. It further offers<br />

a valuable model system to underst<strong>and</strong> how the de novo cell wall biosynthesis occurs in<br />

the yeast cell or how the yeast cell wall participates in morphogenesis.<br />

Key Words: C<strong>and</strong>ida albicans; cell wall; protoplasts; regeneration; Saccharomyces<br />

cerevisiae; yeast.<br />

1. Introduction<br />

Yeasts are unicellular eukaryotic organisms that, unlike mammalian cells,<br />

are surrounded by an elastic <strong>and</strong> highly dynamic structure, namely the cell wall.<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

241


242 Pitarch et al.<br />

This external envelope, basically composed of -1,3 <strong>and</strong> -1,6-glucans, chitin,<br />

mannoproteins, <strong>and</strong> proteins (1,2), is crucial for preserving the morphology<br />

<strong>and</strong> osmotic integrity of the yeast cell <strong>and</strong>, therefore, essential for cell viability<br />

(see chapter on Cell Wall <strong>Fractionation</strong> for Yeast <strong>and</strong> Fungal Proteomics for<br />

further details). The yeast cell wall can be completely eliminated by controlled<br />

enzymatic digestion in an iso-osmotic medium, resulting in the formation of<br />

protoplasts (see Fig. 1). Yeast protoplasts are uniquely enveloped by the plasma<br />

membrane, which displays typical invaginations <strong>and</strong> dictates their distinctive<br />

spherical shape on the basis of physical laws. When yeast protoplasts are cultivated<br />

in osmotically stabilized nutrient media, these are intriguingly able to<br />

synthesize a new cell wall <strong>and</strong> revert to normal cells, which are capable of proliferating<br />

<strong>and</strong> inducing proper morphogenesis (see Note 1) (3–6). Both processes,<br />

cell wall regeneration <strong>and</strong> protoplast reversion, are intrinsically associated with<br />

yeast cell survival (see Note 2) (5).<br />

Chitin is the first cell wall component to be deposited on the surface of<br />

regenerating protoplasts (7–9). The microfibrils of nascent chitin, distributed<br />

irregularly over the plasma membrane at the early stages of reversion, undergo<br />

a gradual rise in density during the subsequent stages of cell wall regeneration,<br />

leading to the formation of a regular fibrillar mesh. This de novo chitin skeleton,<br />

located around the plasma membrane of reverting protoplasts, is soon overlaid<br />

with -1,3-glucan. Remarkably, although cell wall proteins (i.e., mannoproteins<br />

<strong>and</strong> proteins) begin to be synthesized early in the course of protoplast reversion<br />

to normal cells, their covalent incorporation into this de novo glucan-chitin<br />

framework is delayed until an adequate amount of -1,3-glucan molecules is<br />

assembled on the nascent polysaccharide lattice. In fact, cell wall proteins are<br />

the last components to be effectively bound to the regenerating wall. For this<br />

reason, during the initial stages of the reversion process, cell wall proteins<br />

(biologically intended for being anchored to the wall polysaccharide network)<br />

are not covalently retained into the nascent wall of regenerating protoplasts <strong>and</strong>,<br />

consequently, are shed into the extracellular medium. A schematic representation<br />

of the dynamics of protoplast formation <strong>and</strong> de novo wall construction<br />

in reverting protoplasts is shown in Fig. 1.<br />

The use of regenerating protoplasts <strong>and</strong>, in particular the collection of<br />

proteins secreted from yeast protoplasts in active cell wall regeneration (i.e.,<br />

during the first stages of the regeneration process of protoplast walls) into the<br />

culture medium, are a good <strong>and</strong> simple model system to:<br />

1. Easily isolate <strong>and</strong> solubilize proteins of the complex yeast cell wall, because these<br />

are freely released from actively reverting protoplasts into the culture medium.<br />

2. Reduce the complexity of this subcellular compartment (see Chapter 19 for<br />

details on intricate structure <strong>and</strong> molecular organization of the yeast cell wall) <strong>and</strong>


Collection of Proteins Secreted from Regenerating Yeast Protoplasts 243<br />

Fig. 1. Dynamics of protoplast formation, cell wall regeneration <strong>and</strong> protoplast<br />

reversion to normal yeast cells. Elimination of the yeast cell wall by controlled<br />

enzymatic digestion in an iso-osmotic medium leads to the protoplast formation (see<br />

Note 14). Yeast protoplasts have typical intrinsic features, such as (1) spherical shape,<br />

(2) invaginations of the plasma membrane in many areas, <strong>and</strong> (3) ability to synthesize<br />

new cell walls <strong>and</strong> revert to normal growing cells in iso-osmotic regenerating conditions<br />

(see Note 1). Regenerating protoplasts in an osmotically stabilized nutrient medium


244 Pitarch et al.<br />

facilitate further related analytical procedures, exploiting the fact that the yeast<br />

cell is deprived of its cell wall <strong>and</strong> stimulated to resynthesize it step by step.<br />

3. Bypass some of the problems inherently associated with chemical <strong>and</strong>/or<br />

enzymatic treatments to solubilize cell wall proteins (see Chapter 19), such as:<br />

a. Potential protein modifications. These are avoided with the method described<br />

here because cell wall proteins are collected from the growth medium <strong>and</strong><br />

directly analyzed in ensuing proteomic studies without the use of chemical<br />

agents or enzymes that may in some measure modify them.<br />

b. The extraction of proteins bearing glucan <strong>and</strong>/or chitin side-chain<br />

residues, which hinder their subsequent resolution by two-dimensional gel<br />

electrophoresis (2-DE) <strong>and</strong>/or identification by mass spectrometry (MS) (10,<br />

11). This is circumvented using the present approach, because this does not<br />

break the covalent linkages between proteins <strong>and</strong> structural wall polysaccharides<br />

(glucan/chitin).<br />

4. Characterize cell wall protein precursors before their incorporation into the cell<br />

wall (i.e., those gene expression products involved in the de novo wall biosynthesis).<br />

These mainly include structural proteins, as well as remodelling <strong>and</strong><br />

cross-linking enzymes (10,12–14)).<br />

5. Study the de novo generation <strong>and</strong>, therefore, all steps of the biosynthesis of the<br />

cell wall of yeasts, because this stratagem yields information not only about<br />

the composition of cell wall precursors but also about their interactions (at the<br />

different stages of protoplast reversion to normal cells) (7–9,12).<br />

6. Elucidate the role of the yeast wall in cell morphogenesis, given that cell wall<br />

regeneration by reverting protoplasts is a de novo process of morphogenesis<br />

(3–5,15).<br />

◭<br />

Fig. 1. (Continued) first form a fibrillar chitin network, whereupon -1,3-glucan<br />

molecules are promptly deposited. Mannoproteins <strong>and</strong> proteins are the last components<br />

to be assembled on the nascent fibrillar mesh, because their covalent incorporation<br />

into the regenerating cell wall only occurs after the establishment of a structural<br />

glucan-chitin matrix around the plasma membrane of reverting protoplasts. Consequently,<br />

during the early stages of the regeneration process of protoplast walls, cell<br />

wall protein precursors are not retained into the nascent wall but are secreted into the<br />

culture medium. It follows that deposition of the fibrillar component (polysaccharide<br />

framework in the inner wall layer) <strong>and</strong> amorphous component (protein precursors in<br />

the outer wall layer) into the de novo cell wall is therefore desynchronized (5). The<br />

filasomes, located in the reverting site of the protoplasts, appear to be involved in<br />

the transport of cell wall components from the cytoplasm to the plasma membrane<br />

in association with secretory vesicles. Interestingly, actin plays a key role in (1) the<br />

initiation of cell wall regeneration, (2) the correct deposition of cell wall precursors,<br />

<strong>and</strong> (3) preservation of the proper shape of the regenerating protoplasts (see Note 2)<br />

(15,23).


Collection of Proteins Secreted from Regenerating Yeast Protoplasts 245<br />

7. Discover new diagnostic/prognostic markers <strong>and</strong>/or therapeutic targets for human<br />

mycoses (14).<br />

8. Increase the productivity <strong>and</strong> facilitate downstream processing of useful (recombinant<br />

or nonrecombinant) proteins extracellularly expressed in yeasts (16),<br />

because the use of regenerating protoplasts eliminates the physical barrier of the<br />

cell wall, which may limit their excretion. However, it is worth mentioning that<br />

this stratagem cannot be exploited for long-term production processes because of<br />

(1) the extreme fragility of yeast protoplasts, <strong>and</strong> (2) complete cell wall regeneration<br />

around the active protoplasts during long-term culture periods (see Note 3).<br />

In this chapter, we describe a method for obtaining proteins secreted into<br />

the culture medium during the early stages of the regeneration process of<br />

protoplast walls under suitable conditions for subsequent proteomic analyses<br />

(10,13,14,17,18). We also provide a rapid <strong>and</strong> straightforward protocol for<br />

checking the degree of purity of the cell wall proteins excreted from yeast protoplasts<br />

in active cell wall regeneration. Proteins involved in cell wall construction<br />

(such as -1,3-glucosyltransferases, exoglucanases, glycosyl phosphatidylinositol<br />

(GPI)-proteins, <strong>and</strong> proteins with internal repeats [PIR], among others),<br />

heat shock proteins, glycolytic enzymes <strong>and</strong> other proteins have successfully<br />

been identified using the procedure presented here in combination with 2-<br />

DE <strong>and</strong> MS (10,13,14,17). Given the nature of the protein sample, (multidimensional)<br />

liquid chromatography techniques in t<strong>and</strong>em with MS could also<br />

alternatively be applied. Although other methods to isolate <strong>and</strong> solubilize cell<br />

wall proteins from yeast species have been reported in the Chapter 19, the<br />

choice between these or the one outlined here, which each have advantages<br />

<strong>and</strong> disadvantages, will depend on the specific application.<br />

2. Materials<br />

All solutions <strong>and</strong> buffers should be prepared with ultrapure water (doubledistilled,<br />

deionized water with a resistivity >18M/cm), <strong>and</strong> prechilled when<br />

working at 4°C. Growth media, solutions <strong>and</strong> buffers should be sterilized<br />

by autoclaving before use. Their labile components should be filter-sterilized<br />

separately <strong>and</strong> added to the other ingredients after autoclaving.<br />

2.1. Collection of Proteins Secreted from Yeast Protoplasts in Active<br />

Cell Wall Regeneration<br />

2.1.1. <strong>Preparation</strong> of Yeast Protoplasts<br />

1. Yeast-Peptone-D-glucose (YPD) plates: 1% (w/v) yeast extract (Difco Laboratories,<br />

Detroit, MI), 2% (w/v) peptone (Difco), 2% (w/v) d-glucose, 2% (w/v)<br />

agar (Difco).


246 Pitarch et al.<br />

2. YPD medium: 1% (w/v) yeast extract, 2% (w/v) peptone, 2% (w/v) d-glucose.<br />

3. Pretreatment buffer: 10 mM Tris-HCl, pH 9.0, 5 mM EDTA, 1% (v/v)<br />

-mercaptoethanol.<br />

4. 1 M sorbitol solution: Dissolve 182.17 g of sorbitol in sufficient water to yield a<br />

final volume of 1 L.<br />

5. Glusulase ® (Du Pont; NEN Life Science Products, Boston, MA).<br />

2.1.2. Active Cell Wall Regeneration of Yeast Protoplasts<br />

1. Complete minimal (CM) medium: 0.17% (w/v) yeast nitrogen base (YNB) without<br />

amino acids or ammonium sulfate (Difco), 0.5% (w/v) (NH4) 2SO4, 2% (w/v)<br />

d-glucose, 0.13% (w/v) dropout powder (see Note 4).<br />

2. Dropout powder: 2.5 g adenine, 1.2 g l-arginine, 6.0 g l-aspartic acid, 6.0 g lglutamic<br />

acid, 1.2 g l-histidine, 3.6 g l-leucine, 1.8 g l-lysine, 1.2 g l-methionine,<br />

3.0 g l-phenylalanine, 22.5 g l-serine, 12.0 g l-threonine, 2.4 g l-tryptophan,<br />

1.8 g l-tyrosine, 9.0 g l-valine, <strong>and</strong> 1.2 g uracil.<br />

3. Lee medium (19): 0.02% (w/v) MgSO4, 0.25% (w/v) K2HPO4, 0.5% (w/v) NaCl,<br />

0.5% (w/v) (NH4) 2SO4, 0.05% (w/v) l-alanine, 0.13% (w/v) l-leucine, 0.1%<br />

(w/v) l-lysine, 0.01% (w/v) l-methionine, 0.007% (w/v) l-ornitine, 0.05% (w/v)<br />

l-phenylalanine, 0.05% (w/v) l-proline, 0.05% (w/v) l-threonine (see Note 5). After<br />

autoclaving, add 25 mL of 50% glucose solution <strong>and</strong> 2 mL of 0.1% biotin solution.<br />

4. 50% glucose solution: Dissolve 12.5 g of d-glucose in a final volume of 25 mL<br />

water <strong>and</strong> sterilize using a 0.22-μm filter (Millipore, Bedford, MA).<br />

5. 0.1% biotin solution: Dissolve 2 mg of biotin in a final volume of 2 mL water<br />

<strong>and</strong> sterilize using a 0.22-μm filter.<br />

2.1.3. Recovery of Proteins Secreted from Regenerating Protoplasts<br />

1. Phenylmethylsulfonyl fluoride (PMSF) stock solution: 0.1 M PMSF in<br />

isopropanol. Dissolve 174 mg of PMSF (Fluka, Chelmsford, MA) in a final<br />

volume of 10 mL isopropanol, <strong>and</strong> store at –20 ºC (see Note 6). PMSF should be<br />

h<strong>and</strong>led with caution, because it is highly toxic. Weigh this hazardous chemical<br />

in a fume hood, <strong>and</strong> wear gloves, goggles <strong>and</strong> a mask.<br />

2. Antipain stock solution: Dissolve 5 mg of antipain (Sigma, St. Louis, MO) in a<br />

final volume of 1 mL water, <strong>and</strong> store at –20 ºC (see Note 7).<br />

3. Leupetin stock solution: Dissolve 5 mg of leupeptin (Sigma) in a final volume of<br />

1 mL water, <strong>and</strong> store at –20 ºC (see Note 8).<br />

4. Pepstatin stock solution: Dissolve 2.5 mg of pepstatin (Sigma) in a final volume<br />

of 1 mL methanol, <strong>and</strong> store at –20 ºC (see Note 9).<br />

5. Protease inhibitor mix: 0.1 mM PMSF, 2 μg/mL antipain, 2 μg/mL leupeptin, <strong>and</strong><br />

1 μg/mL pepstatin (i.e., for 450 mL of protein solution, add 450 μL of PMSF stock<br />

solution <strong>and</strong> 180 μL each of antipain, leupeptin <strong>and</strong> pepstatin stock solutions).<br />

Mix just before use.<br />

6. Filtration unit (Millipore).<br />

7. 0.22-μm pore-size nitrocellulose filter (Millipore).


Collection of Proteins Secreted from Regenerating Yeast Protoplasts 247<br />

2.1.4. Concentration of Proteins Secreted from Regenerating<br />

Protoplasts<br />

1. Ultrafiltration apparatus (300-mL stirred ultrafiltration cell system; Amicon;<br />

Beverly, MA).<br />

2. YM-10 Diaflo ® ultrafiltration membrane (pre-equilibrated 10,000-Da pore-size<br />

ultrafilter; Amicon).<br />

3. Magnetic stirrer.<br />

4. Oxygen-free nitrogen with pressure regulator (see Note 10).<br />

5. Membrane wash solution: 1–2 M NaCl.<br />

6. Membrane store solution: 10% ethanol.<br />

7. Lyophilizer.<br />

2.2. Assay of Cell Wall Protein Purity: Alkaline Phosphatase Assay<br />

1. Lysis buffer: 50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1 mM dithiothreitol (DTT),<br />

0.5 mM PMSF <strong>and</strong> 5 μg/mL each of antipain, leupeptin <strong>and</strong> pepstatin (see Notes 6–9).<br />

2. NaCl-glycine buffer (alkaline buffer): 0.1 M NaCl, 0.1 M glycine, pH 9.7 (adjusted<br />

with 1 N NaOH).<br />

3. Stock p-nitrophenol (PNP) solution: 8 mM PNP (Sigma) in NaCl-glycine buffer.<br />

4. Substrate solution: 20 mM disodium p-nitrophenyl phosphate (PNPP; Sigma) in<br />

NaCl-glycine buffer.<br />

5. Stop solution: 0.05 M NaOH.<br />

6. Spectrophotometer.<br />

3. Methods<br />

3.1. Collection of Proteins Secreted from Yeast Protoplasts in Active<br />

Cell Wall Regeneration<br />

The flowchart in Fig. 2 summarizes the different steps in the collection of<br />

proteins excreted into the osmotically stabilized liquid nutrient medium during<br />

the first stages of the regeneration process of protoplast walls. These involve<br />

(1) the complete elimination of the yeast cell wall by controlled enzymatic<br />

digestion (i.e., protoplast formation), (2) active regeneration of the cell wall<br />

<strong>and</strong> ensuing secretion of wall protein precursors into the growth medium,<br />

(3) recovery of the culture medium with secreted proteins by centrifugation<br />

<strong>and</strong> filtration, <strong>and</strong> (4) concentration of secreted proteins by ultrafiltration <strong>and</strong><br />

lyophylization. This method to isolate <strong>and</strong> solubilize protein precursors of<br />

the yeast cell walls is taken from the published protocols in Saccharomyces<br />

cerevisiae <strong>and</strong> C<strong>and</strong>ida albicans by Pardo et al. (10) <strong>and</strong> Pitarch et al. (17),<br />

respectively, <strong>and</strong> is based on earlier methodology developed by Elorza et al.<br />

(20). Although this procedure has been used successfully <strong>and</strong> reproducibly on


248 Pitarch et al.<br />

Fig. 2. Flowchart of basic steps in the collection of proteins secreted from yeast<br />

protoplasts in active cell wall regeneration into the osmotically stabilized liquid nutrient<br />

medium.


Collection of Proteins Secreted from Regenerating Yeast Protoplasts 249<br />

these yeast species for subsequent proteomic analyses (10,13,14,17), it may be<br />

necessary to adjust it for other yeast species or even for filamentous fungi.<br />

3.1.1. <strong>Preparation</strong> of Yeast Protoplasts (see Note 11)<br />

1. Grow yeast cells on a YPD plate (stock maintenance medium) at 28–30 ºC for<br />

2 d. Inoculate 50 mL of YPD medium in a 250-mL flask (see Note 12) with a<br />

single colony from the YPD plate, <strong>and</strong> grow overnight at 28–30 ºC in a shaking<br />

incubator (200 rpm).<br />

2. Use this 50-mL preculture to inoculate two 2-L flasks containing 500 mL<br />

fresh YPD medium each, <strong>and</strong> grow at 28–30 ºC with vigorous rotary shaking<br />

(200 rpm) until the culture reaches mid-log phase growth (OD 600nm= 4;see<br />

Note 13).<br />

3. Harvest the yeast cells by centrifugation at 4,500g for 5 min <strong>and</strong> discard the<br />

supernatant.<br />

4. Resuspend the cell pellet in 150 mL water, <strong>and</strong> centrifuge 5 min at 4,500g.<br />

Decant the supernatant.<br />

5. Gently resuspend the yeast cells in 100 mL of pretreatment buffer to a density<br />

of 1–2 × 10 9 cells/mL, <strong>and</strong> incubate at 28 ºC with gentle rotary shaking (80<br />

rpm) for 30 min (see Note 14). Centrifuge 10 min at 600g, <strong>and</strong> discard the<br />

supernatant.<br />

6. Gently resuspend the cell pellet in 150 mL of a 1 M sorbitol solution (see<br />

Note 15). Centrifuge 10 min at 600g to harvest cells, <strong>and</strong> discard the supernatant.<br />

7. Gently resuspend the cell pellet in a 1 M sorbitol solution to a density of 5 ×<br />

10 8 cells/mL, <strong>and</strong> add 30 μL/mL ice-cold Glusulase ® (see Note 16).<br />

8. Incubate cells with very gentle shaking (80 rpm; see Note 17) at 28 ºC until<br />

more than 90–95% of them are protoplasts (∼45 min to 1 h). Monitor the degree<br />

of protoplast formation with a phase-contrast microscope (see Note 18).<br />

9. Harvest protoplasts by very gentle centrifugation at 600g for 15 min. Decant the<br />

supernatant carefully (see Note 19).<br />

10. Gently wash the protoplast pellet with 150 mL of a 1 M sorbitol solution (see<br />

Note 20). Centrifuge 15 min at 600g <strong>and</strong> decant the supernatant carefully.<br />

11. Repeat this step two more times to eliminate any trace of Glusulase ® (see Note 21).<br />

12. Remove a small aliquot of the protoplast preparation for use in Subheading 3.2<br />

to evaluate enzymatic activity of alkaline phosphatase (see Note 22).<br />

3.1.2. Active Cell Wall Regeneration of Yeast Protoplasts (see Notes 11<br />

<strong>and</strong> 17)<br />

1. Gently resuspend the protoplasts in the regenerating buffer supplemented with<br />

1 M sorbitol to a density of 3×108cells/mL (see Notes 20 <strong>and</strong> 23).<br />

2. Incubate protoplasts with gentle rotary shaking (80 rpm) at 28 ºC for 2htoinduce<br />

their active cell wall regeneration.


250 Pitarch et al.<br />

3.1.3. Recovery of Proteins Secreted from Regenerating Protoplasts<br />

(see Notes 17 <strong>and</strong> 24)<br />

1. Centrifuge the regenerating protoplasts at 600g for 15–20 min at 4 ºC, <strong>and</strong> collect<br />

the supernatant carefully (see Note 19 <strong>and</strong> 25).<br />

2. Add the protease inhibitor mix.<br />

3. Filter the supernatant through a 0.22-μm pore-size nitrocellulose filter without<br />

any external device in ice at 4 ºC (see Note 26).<br />

4. Remove a small aliquot of cell-free culture filtrate to further monitor the cell lysis<br />

by quantitative determination of alkaline phosphatase (see Subheading 3.2).<br />

3.1.4. Concentration of Proteins Secreted from Regenerating<br />

Protoplasts (see Note 24)<br />

1. Wash an YM-10 Diaflo ® ultrafiltration membrane by floating it skin (glossy) side<br />

down in a beaker of water for 1h(see Note 27). Change water at least three<br />

times.<br />

2. Concentrate the cell-free culture filtrate by ultrafiltration using the washed YM-10<br />

ultrafilter (see Fig. 3A <strong>and</strong> Notes 10 <strong>and</strong> 28).<br />

3. Dilute with 300 mL water, <strong>and</strong> concentrate again. Repeat this step three more<br />

times to eliminate any trace of sorbitol (see Note 29).<br />

4. Remove the ultraconcentrated protein solution (molecular weight fraction above<br />

10,000 Da) from the ultrafiltration cell (see Fig. 3B <strong>and</strong> Note 30).<br />

5. Quick-freeze the ultraconcentrated protein solution at –80 ºC (see Note 31), <strong>and</strong><br />

concentrate it by lyophilization (see Note 32).<br />

6. Resuspend the freeze-dried protein sample in a small volume of water (see<br />

Note 33), <strong>and</strong> quantify the protein content by st<strong>and</strong>ard protein determination<br />

methods.<br />

3.2. Assay of Cell Wall Protein Purity: Alkaline Phosphatase Assay<br />

(see Note 34)<br />

The method given below outlines an easy <strong>and</strong> prompt screening procedure<br />

to check the efficiency, purity, <strong>and</strong> quality of the collection of proteins secreted<br />

from yeast protoplasts in active cell wall regeneration before performing further<br />

proteomic analyses <strong>and</strong> establishing any interpretation of the results. This is<br />

based on measurement of enzymatic activity of alkaline phosphatase, which is<br />

exclusive to the cytosol in yeasts <strong>and</strong> thus exploited as a marker of cytosolic<br />

contamination. This intracellular enzyme catalyzes the cleavage of phosphate<br />

ester bonds from many compounds (including the chromogenic phosphatase<br />

substrate used in this protocol, p-nitrophenylphosphate; PNPP) under alkaline<br />

conditions <strong>and</strong> an optimum temperature of 37 ºC. If there is protoplast lysis<br />

during sample preparation <strong>and</strong>, therefore, cytosolic contamination (containing<br />

alkaline phosphatase, among other intracellular proteins) into the sample of


Collection of Proteins Secreted from Regenerating Yeast Protoplasts 251<br />

Fig. 3. Schematic representation of a stirred ultrafiltration cell on a magnetic stirrer.<br />

Pressurized stirred ultrafiltration cells can efficiently be used to simultaneously concentrate<br />

<strong>and</strong> desalt samples of dilute proteins (from initial volumes of 3 mL–2 L to final<br />

concentrate volumes of 50 μL–60 mL). In the procedure described here, the cell-free<br />

culture filtrate is forced through an YM-10 Diaflo ® ultrafilter, located on a polymer<br />

grid at the bottom of the cell (see Note 28). Protein sample is separated into two groups<br />

according to molecular weight <strong>and</strong> size (see Note 27). The ultraconcentrate (molecular<br />

weight fraction above 10,000 Da) is subjected to lyophilization or centrifugal microconcentration<br />

(see Note 32) before carrying out further proteomic analyses. The protein<br />

content of the ultrafiltrate (molecular weight fraction below 10,000 Da) can also be<br />

quantified by st<strong>and</strong>ard protein determination methods to evaluate the final protein<br />

recovery.


252 Pitarch et al.<br />

wall protein precursors, then alkaline phosphatase will hydrolyze PNPP (which<br />

is colorless), releasing p-nitrophenol (PNP, which is yellow-colored at alkaline<br />

pH but colorless at acidic pH values) <strong>and</strong> inorganic phosphate (see Fig. 4).<br />

Accordingly, only protein solutions with intracellular contamination will turn<br />

yellow following the addition of PNPP. The enzyme activity of alkaline<br />

phosphatase can consequently be detected <strong>and</strong> quantified by spectrophotometry.<br />

Fig. 4. Biochemical principle of the alkaline phosphatase assay. The pnitrophenylphosphate<br />

(PNPP) is a chromogenic substrate for several phosphatases, such<br />

as acid phosphatases <strong>and</strong> alkaline phosphatases. Alkaline phosphatase hydrolyzes this<br />

artificial substrate, which is colorless, at alkaline pH values (pH = 9.7) <strong>and</strong> 37°C to form<br />

p-nitrophenol (PNP), which is yellow-colored in basic solutions <strong>and</strong> can be measured<br />

at 410 nm on a spectrophotometer. The intensity of the yellow color correlates with the<br />

amount of PNP issuing from the hydrolysis of PNPP catalyzed by phosphatase alkaline,<br />

according to Beer’s law. In contrast, acid phosphatase hydrolyzes PNPP under acidic<br />

conditions (pH = 4.8) <strong>and</strong> 37°C leading to PNP, which is colorless at acidic pH values.


Collection of Proteins Secreted from Regenerating Yeast Protoplasts 253<br />

The present method is extremely sensitive, because very small amounts of<br />

alkaline phosphatase generate sufficient PNP after an adequate incubation<br />

period, bringing a measurable color about. This protocol is modified from that<br />

of Cabib et al. (21), which was developed for rapid checking of colonies with<br />

lysed yeast cells (embedded into a gelled medium supplemented with PNPP at<br />

pH 9.7). These acquired a yellow color on <strong>and</strong> around colonies as a result of<br />

the release of alkaline phosphatase.<br />

Interestingly, the present assay is not affected by the acid phosphatases,<br />

which are located in the outermost <strong>and</strong> innermost cell wall layers of the yeast<br />

cells (1) <strong>and</strong>, therefore, detected in intact cells (see Fig. 4) . This is because<br />

of the fact that these extracellular enzymes are only able to hydrolyze PNPP<br />

under acidic conditions (optimum pH = 4.8) <strong>and</strong> the pH of the reaction buffer<br />

is highly alkaline (pH = 9.7).<br />

1. Resuspend the aliquot of protoplasts (see Subheadings 3.1.3 <strong>and</strong> Note 22)in1mL<br />

of ice-cold lysis buffer. Vortex for 1 min, <strong>and</strong> cool on ice for 1–2 min. Repeat this<br />

step until complete cell breakage (monitored with a phase-contrast microscope).<br />

Remove cell debris from the homogenate by centrifugation at 12,500g for 10 min<br />

at 4 ºC. Collect the supernatant <strong>and</strong> use it as a source of alkaline phosphatase for<br />

the positive control (see Note 35).<br />

2. Use known amounts of the reaction product PNP in NaCl-glycine buffer (10–110<br />

μg/mL) to obtain a st<strong>and</strong>ard curve. Mix thoroughly by careful vortexing. Measure<br />

<strong>and</strong> record the absorbance at 410 nm in the spectrophotometer. Use the same<br />

plastic cuvet for all samples, starting with the most dilute sample.<br />

3. Include controls of (1) NaCl-glycine buffer alone (buffer control), (2) cell-free<br />

culture filtrate alone (enzyme control), (3) substrate without cell-free culture<br />

filtrate (substrate control), <strong>and</strong> (4) substrate with protoplast lysate (positive<br />

control) as indicated in Fig. 5 to determine superfluous absorbance either<br />

from other compounds in buffer <strong>and</strong> cell-free culture filtrate, or from substrate<br />

breakdown (see Note 36).<br />

4. Place 500 μL of cell-free culture filtrate of products secreted from regenerating<br />

protoplasts in a 1.5-mL Eppendorf tube (see Subheadings 3.1.3 <strong>and</strong> Note 36),<br />

<strong>and</strong> add 500 μL of substrate solution (see Note 36).<br />

5. Vortex carefully <strong>and</strong> incubate in the dark at 37 ºC for 30 min. Protect from bright<br />

light.<br />

6. Stop the enzyme activity with 400 μL of stop solution. Mix carefully <strong>and</strong> cool<br />

the tubes to room temperature (25–35ºC).<br />

7. Measure <strong>and</strong> record the absorbance at 410 nm in the spectrophotometer. Use the<br />

same plastic cuvet for all tubes.<br />

8. Determine enzyme activities in all tubes using the st<strong>and</strong>ard curve (10–110 μg<br />

p-nitrophenol/mL). The units of enzyme (see Note 37) are calculated as:<br />

U.E. = 14.38 × c/t, where c is the PNP concentration in μg/mL <strong>and</strong> t is the reaction<br />

time in min.


254 Pitarch et al.<br />

Fig. 5. Schematic diagram of the basic steps in the alkaline phosphatase assay.<br />

4. Notes<br />

1. In all yeast species studied so far, protoplasts embedded into gelled nutrient<br />

media are always able to regenerate a complete cell wall <strong>and</strong> revert to normal<br />

reproducing cells (5). In contrast, the cell wall regeneration of reverting yeast<br />

protoplasts grown in liquid nutrient media is (1) incomplete in Saccharomyces<br />

cerevisiae <strong>and</strong> other budding yeasts, bringing about cells with aberrant walls<br />

(albeit with all cell wall components) incapable of dividing <strong>and</strong> inducing<br />

normal morphogenesis (5,12), but (2) complete in C<strong>and</strong>ida albicans (4,22),<br />

Schizosaccharomyces pombe (23,24), Nadsonia elongata (5) <strong>and</strong> Endomycopsis<br />

fibuliger (5), resulting in normal growing cells. Accordingly, certain physical


Collection of Proteins Secreted from Regenerating Yeast Protoplasts 255<br />

factors can actively be involved in the cell wall construction <strong>and</strong> protoplast<br />

reversion (5).<br />

2. The morphology of the de novo cell wall formed on the protoplast surface follows<br />

that of the protoplast (i.e., spherical shape) (5). On the contrary, the shape of<br />

new cells originated from the walled protoplasts is at least partly determined by<br />

the cell wall (i.e., oval shape; see Fig. 1).<br />

3. The complete reversion of S. pombe, N. elongata <strong>and</strong> C. albicans protoplasts to<br />

normal cell walls <strong>and</strong> growing cells is attained after 12, 20, <strong>and</strong> 24 h, respectively,<br />

of incubation in osmotically stabilized liquid nutrient media (4,5,23).<br />

4. To promote optimal regeneration of protoplast cell walls, it is convenient to<br />

use a 10× dropout powder solution that has been autoclaved separately or filter<br />

sterilized, <strong>and</strong> added to the remaining ingredients after autoclaving.<br />

5. To avoid precipitation of MgSO 4 <strong>and</strong> K 2HPO 4, these should be autoclaved<br />

separately or filter sterilized, <strong>and</strong> added to the other ingredients after autoclaving.<br />

6. The PMSF can also be solubilized in ethanol, methanol <strong>and</strong> 1,2-propanediol; <strong>and</strong><br />

is unstable in aqueous solution. PMSF inhibits serine proteases (e.g., trypsin,<br />

chymotrypsin, <strong>and</strong> thrombin) <strong>and</strong> thiolproteases (e.g., papain). It is added to<br />

reduce possible proteolytic processes (see Table 1).<br />

7. Antipain is also soluble in methanol <strong>and</strong> dimethylsulfoxide (DMSO). It inhibits<br />

papain <strong>and</strong> trypsin <strong>and</strong>, to a lesser extent, plasmin. It is added to reduce possible<br />

proteolytic processes (see Table 1).<br />

8. The inhibition specificity of leupeptin is of broad spectrum. It inhibits serine<br />

<strong>and</strong> thiol-proteases. It is added to reduce possible proteolytic processes (see<br />

Table 1).<br />

9. Pepstatin is insoluble in water. It inhibits acid proteases (e.g., pepsin, chymosin,<br />

cathepsin D <strong>and</strong> renin, among others). It is added to reduce possible proteolytic<br />

processes (see Table 1).<br />

10. It is important not to use compressed air because it can bring about (1) large<br />

pH changes as a result of carbon dioxide dissolution, <strong>and</strong>/or (2) oxidations in<br />

sensitive protein solutions.<br />

11. Perform all procedures from this subheading under sterile conditions. Use sterile<br />

centrifuge bottles.<br />

12. Liquid cultures should be grown in a flask that is at least 4–5 times larger than<br />

the culture volume.<br />

13. The cell density of inoculum <strong>and</strong> incubation time should be adjusted according<br />

to the yeast strain. It is important that the yeast culture is in mid-log phase<br />

growth (∼ OD 600nm= 4) because (1) yeast cells are more susceptible to wall<br />

lytic enzymes (Glusulase ® treatment) for complete protoplasting than those in<br />

stationary phase growth <strong>and</strong> (2) there is sufficient biomass accumulation.<br />

14. This step is critical for a successful protoplast preparation because mercaptoethanol<br />

pretreatment facilitates the subsequent action of wall lytic<br />

enzymes (Glusulase ® treatment) by (1) breaking disulfide bonds of cell<br />

wall proteins <strong>and</strong> (2) slightly disorganizing the cell wall. In fact, recent<br />

studies have demonstrated that a better protoplasting effect is attained in


Table 1<br />

Protease inhibitors<br />

Effective<br />

concentrations Stock solutions Soluble in Notes<br />

Inhibitor Specificity<br />

Isopropanol Ethanol<br />

Methanol<br />

17 mg/mL in<br />

isopropanol<br />

17–174 μg/mL (0.1–1<br />

mM)<br />

PMSF Serine proteases<br />

Thiolproteasesa Stable ∼1 month<br />

at –20 ºC<br />

1, 2-propanediol<br />

Water<br />

Methanol<br />

2–50 μg/mL 5 mg/mL in<br />

water<br />

Antipain Trypsinb Papainc DMSO d<br />

Water Stable ∼6 months<br />

at –20 ºC<br />

Stable ∼1 week at<br />

+4 ºC<br />

Methanol<br />

Ethanole Stable ∼1 week at<br />

+4 ºC<br />

0.5–2 μg/mL 5 mg/mL in<br />

water<br />

Leupeptin Serine proteases<br />

Thiolproteases<br />

Pepstatin Acid proteases 0.7–1 μg/mL 2.5 mg/mL in<br />

methanol<br />

a Reversible by dithiothreitol (DTT) treatment.<br />

b Serine protease.<br />

c Thiolprotease.<br />

d Dimethylsulfoxide.<br />

e Sit overnight.


Collection of Proteins Secreted from Regenerating Yeast Protoplasts 257<br />

-mercaptoethanol-pretreated yeast cells than in nonpretreated cells (25).<br />

Centrifugation for isolation of -mercaptoethanol-treated yeast cells should be<br />

carried out with the brake in 1.5.<br />

15. 1 M sorbitol is used as an osmotic stabilizer. In general, sugars <strong>and</strong> sugar alcohols<br />

(e.g., sorbitol, mannitol) are consistently used for yeast protoplast formation <strong>and</strong><br />

reversion, whereas MgSO 4 <strong>and</strong> KCl are commonly chosen for preparation <strong>and</strong><br />

regeneration of protoplasts from filamentous fungi.<br />

16. Viable protoplasts can be achieved by controlled autolysis of the yeast cell wall<br />

using snail or microbial lytic enzymes (see Table 2) (25). Glusulase ® is a<br />

preparation of the intestinal juice of the Roman garden snail Helix pomatia, <strong>and</strong><br />

consists of a mixture of lytic enzymes (-glucuronidase, sulfatase, <strong>and</strong> cellulase).<br />

It has proven particularly useful for obtaining protoplasts (i.e., for completely<br />

digesting yeast cell walls) in nearly all yeast species (5). Sterilized tweezers<br />

should be used to take the lid off the Glusulase ® bottle. It is important not to do<br />

it with the h<strong>and</strong>s. Gently mix Glusulase ® with cell suspension (without shaking<br />

it).<br />

17. Perform all procedures from this point on with very gentle shaking. It is very<br />

important to h<strong>and</strong>le the protoplasts gently in this protocol, because yeast protoplasts<br />

are extremely fragile. This can prevent further intracellular contamination<br />

into the sample of proteins secreted from actively reverting protoplasts.<br />

18. In general, 90–95% of protoplasts should be routinely achieved. The incubation<br />

time <strong>and</strong> amount of Glusulase ® needed to attain protoplasts (i.e., complete yeast<br />

cell wall lysis) can vary with yeast strain <strong>and</strong> growth stage <strong>and</strong> may therefore<br />

be necessary to adjust the incubation time <strong>and</strong> Glusulase ® amount given in the<br />

protocol. The degree of protoplast formation is assessed under a phase-contrast<br />

microscope by:<br />

a. Counting spherical cells.<br />

b. Observing cell lysis in hypotonic solution (e.g., after the addition of water;<br />

see Fig. 6). The number of osmotically-resistant cells (non-protoplasted cells)<br />

is determined by diluting an aliquot of the cell suspension after <strong>and</strong> before<br />

Glusulase ® treatment in water <strong>and</strong> plating on YPD plates (<strong>and</strong> growing at<br />

28–30 ºC). The ratio of CFUs after to before treatment is then calculated to<br />

estimate the efficiency of protoplast formation.<br />

19. Perform all centrifugations for protoplast isolation with the brakes off. It is<br />

important to decant the supernatant carefully, because the protoplast pellet is<br />

less compact than the preceding cell pellets.<br />

20. Protoplast resuspension is difficult <strong>and</strong> sticky. For this reason, a small volume<br />

should first be used to resuspend the protoplasts by gently swirling liquid across<br />

the surface of the pellet. Then add more solution until reaching the correct final<br />

volume.<br />

21. It is important to wash the protoplasts several times (before regenerating protoplast<br />

walls) to remove sulfatases, <strong>and</strong> on the whole any enzymatic activity,


Table 2<br />

Main preparations of lytic enzymes used for yeast cell wall digestion <strong>and</strong> subsequent production of viable protoplasts<br />

from yeast cells<br />

Types Source Location Composition Commercial name References<br />

(10, 14, 26)<br />

Glusulase ® - Du<br />

Pont; NEN Life<br />

Science Products,<br />

– -glucuronidase<br />

– sulfatase<br />

– cellulase<br />

Gut juice<br />

(digestive<br />

enzymes)<br />

Snail enzymes Helix pomatia<br />

(the Roman<br />

garden snail)<br />

Boston, MA, US<br />

Not available (25, 27)<br />

Achatina achatina<br />

(the giant African<br />

(20, 28)<br />

Zymolyase 20T ®<br />

– MP<br />

Biomedicals,<br />

Aurora, Ohio, US<br />

– Seikagaku<br />

Corporation,<br />

Tokyo, Japan<br />

– Miles<br />

Laboratories,<br />

Elkhart, Ill., US<br />

Gut juice<br />

– -glucuronidase<br />

(digestive<br />

– endo--glucanase<br />

enzymes)<br />

– arylsulfatase<br />

Culture fluid – -1,3-glucan<br />

laminaripentaohydrolase<br />

– -1,3-glucanase<br />

– protease<br />

– mannase<br />

– amylaseb – xylanaseb – phosphataseb snail)<br />

Arthrobacter<br />

luteus (bacterium)<br />

Microbial<br />

enzymes a<br />

a Very expensive for use in industrial processes.<br />

b Trace.


Collection of Proteins Secreted from Regenerating Yeast Protoplasts 259<br />

Fig. 6. Evaluation of the efficiency of protoplast formation. When an osmotically<br />

stabilized protoplast/cell suspension is diluted with water, the extracellular medium<br />

becomes hypotonic to cytosol (i.e., the free water concentration is greater outside the<br />

cell). This results in a passive movement of free water from extracellular medium to<br />

cytoplasm (an influx of free water). Consequently, protoplasts gain water, swell <strong>and</strong><br />

burst, whereas nonprotoplasted cells do not undergo cytolysis but an increase in turgor<br />

pressure because of the presence of the cell wall (see Note 18). In isotonic solutions,<br />

the movement of water into <strong>and</strong> out of the cell maintains equilibrium.<br />

present in the Glusulase ® preparation that may modify the protein precursors<br />

secreted into the medium.<br />

22. The protoplast preparation can be lysed to release intracellular proteins <strong>and</strong> used<br />

as a source of alkaline phosphatase (positive control; see Subheading 3.2).<br />

23. Lee medium is reliably used as a regenerating buffer for C<strong>and</strong>ida spp. (9,17),<br />

whereas complete minimal (CM) medium is preferred for Saccharomyces spp.<br />

(10). These regeneration media are chosen because they are rich <strong>and</strong> chemically


260 Pitarch et al.<br />

defined media. Hence, there are no substances in their composition that interfere<br />

in the subsequent proteomic analyses of the secreted proteins. YPD medium is<br />

a complex medium, <strong>and</strong> cannot be used because it contains proteins (from the<br />

yeast extract), which could result in misidentifications.<br />

24. Perform all procedures from this point on at 4 ºC (in a cool room at 4 ºC)<br />

with precooled solutions, reagents <strong>and</strong> apparatus to avoid undesirable proteolytic<br />

activity.<br />

25. It is probable that the protoplast pellet is resuspendend when there is a small<br />

volume of the supernatant left before being carefully collected. If this happens,<br />

then centrifuge once more at 600 g for 20 min, <strong>and</strong> gently collect the supernatant<br />

again.<br />

26. If there is not enough time to finish the entire protocol in one laboratory period,<br />

the procedure can be stopped after filling the filtration unit with the centrifuged<br />

medium (supernatant) <strong>and</strong> placing it in a container with ice in a cool room at<br />

4 ºC. Do not use any external device, because this may facilitate protoplast lysis<br />

<strong>and</strong> subsequent intracellular contamination in the sample of secreted proteins.<br />

27. The YM-10 Diaflo ® ultrafiltration membrane had a pore size of 10,000 Da<br />

to yield molecular weight fractions above <strong>and</strong> below 10,000 Da. It should be<br />

h<strong>and</strong>led carefully <strong>and</strong> only at the edge. The washing step is important to remove<br />

preservative substances (e.g., sodium azide).<br />

28. Place the YM-10 membrane in an ultrafiltration cell, skin (glossy) side toward<br />

cell-free culture filtrate, <strong>and</strong> then fill the cell with the culture filtrate. Place it on a<br />

magnetic stirrer, <strong>and</strong> connect the inlet line to a regulated nitrogen pressure source<br />

(see Note 10 <strong>and</strong> Fig. 3A). Pressurize the cell <strong>and</strong> pressure-check following the<br />

supplier’s instructions. Turn on the stirrer, <strong>and</strong> adjust the stirring rate. When<br />

the ultrafiltration is accomplished, depressurize <strong>and</strong> continue stirring for a few<br />

minutes to increase protein recovery (see Fig. 3B).<br />

29. It is essential to remove any trace of sorbitol because this interferes in the protein<br />

resolution of subsequent proteomic analyses.<br />

30. The ultrafiltration membrane should be washed with 1–2 mL of water to elute<br />

proteins potentially sticking to it. Add this volume to the ultraconcentrate. To<br />

reuse the ultrafilter, rinse it with 1–2 M NaCl, <strong>and</strong> then flush it through with<br />

water. Store the ultrafilter in 10% ethanol at 4 ºC.<br />

31. The ultraconcentrate can also be frozen quickly into liquid nitrogen.<br />

32. Centrifugal microconcentration techniques can alternatively be used to reconcentrate<br />

the ultraconcentrated protein sample.<br />

33. The protein sample can be used directly in two-dimensional gel electrophoresis<br />

(see Fig. 7) (10,13,14,17). Alternatively, freeze-dried protein preparation can be<br />

resuspended in a small volume of buffer suitable for the subsequent proteomic<br />

analyses.<br />

34. This assay should be h<strong>and</strong>led throughout with caution, because p-nitrophenol<br />

is a skin irritant. Wear gloves <strong>and</strong> a laboratory coat. Duplicates of each tube<br />

should be performed.


Collection of Proteins Secreted from Regenerating Yeast Protoplasts 261<br />

Fig. 7. Proteomics of proteins secreted from C. albicans protoplasts in active cell<br />

wall regeneration.<br />

35. The clarified supernatant (protoplast lysate) can be stored at –80 ºC <strong>and</strong> used in<br />

future assays.<br />

36. This assay can also be carried out on a microtiter plate, using 100 μL, rather<br />

500 μL, of each component.<br />

37. Although substantial enzyme activity is found in the absence of Mg 2+ ions, it<br />

is convenient to use 1.5 mM MgCl 2 or1mM MgSO 4 in the substrate solution,<br />

because these divalent cations stimulate alkaline phosphatase activity.<br />

38. One unit of enzyme (U.E.) is defined as the amount of enzyme that will produce<br />

1 nmol of p-nitrophenol per minute.


262 Pitarch et al.<br />

Acknowledgments<br />

We thank the Merck, Sharp & Dohme (MSD) Special Chair in Genomics<br />

<strong>and</strong> Proteomics, European Community (STREP LSHB-CT-2004-511952),<br />

Comunidad de Madrid (S-SAL-0246/2006) <strong>and</strong> Comisión Interministerial de<br />

Ciencia y Tecnología (BIO-2003-00030 <strong>and</strong> BIO-2006-01989) for financial<br />

support of our laboratory.<br />

References<br />

1. Chaffin, W. L., Lopez-Ribot, J. L., Casanova, M., Gozalbo, D. <strong>and</strong> Martinez,<br />

J. P. (1998) Cell wall <strong>and</strong> secreted proteins of C<strong>and</strong>ida albicans: identification,<br />

function, <strong>and</strong> expression. Microbiol Mol.Biol Rev. 62, 130–80.<br />

2. Klis, F. M., Boorsma, A., <strong>and</strong> de Groot, P. W. (2006) Cell wall construction in<br />

Saccharomyces cerevisiae. Yeast 23, 185–02.<br />

3. Osumi, M. (1998) The ultrastructure of yeast: cell wall structure <strong>and</strong> formation.<br />

Micron. 29, 207–33.<br />

4. Nishiyama, Y., Aoki, Y., <strong>and</strong> Yamaguchi, H. (1995) Morphological aspects of<br />

cell wall formation during protoplast regeneration in C<strong>and</strong>ida albicans. J.Electron<br />

Microsc. 44, 72–8.<br />

5. Necas, O. (1971) Cell wall synthesis in yeast protoplasts. Bacteriol.Rev. 35, 149–70.<br />

6. Takagi, T., Ishijima, S. A., Ochi, H., <strong>and</strong> Osumi, M., (2003) Ultrastructure <strong>and</strong><br />

behavior of actin cytoskeleton during cell wall formation in the fission yeast<br />

Schizosaccharomyces pombe. J.Electron Microsc. 52, 161–74.<br />

7. Rico, H., Carrillo, C., Aguado, C., Mormeneo, S., <strong>and</strong> Sent<strong>and</strong>reu, R. (1997) Initial<br />

steps of wall protoplast regeneration in C<strong>and</strong>ida albicans. Res.Microbiol. 148,<br />

593–603.<br />

8. Kapteyn, J. C., Dijkgraaf, G. J., Montijn, R. C., <strong>and</strong> Klis, F. M. (1995) Glucosylation<br />

of cell wall proteins in regenerating spheroplasts of C<strong>and</strong>ida albicans.<br />

FEMS Microbiol Lett. 128, 271–77.<br />

9. Elorza, M. V., Marcilla, A., Sanjuan, R., Mormeneo, S., <strong>and</strong> Sent<strong>and</strong>reu, R. (1994)<br />

Incorporation of specific wall proteins during yeast <strong>and</strong> mycelial protoplast regeneration<br />

in C<strong>and</strong>ida albicans. Arch.Microbiol. 161, 145–51.<br />

10. Pardo, M., Monteoliva, L., Pla, J., Sanchez, M., Gil, C., <strong>and</strong> Nombela, C. (1999)<br />

Two-dimensional analysis of proteins secreted by Saccharomyces cerevisiae regenerating<br />

protoplasts: a novel approach to study the cell wall. Yeast 15, 459–72.<br />

11. Pitarch, A., Sanchez, M., Nombela, C., <strong>and</strong> Gil, C. (2002) Sequential fractionation<br />

<strong>and</strong> two-dimensional gel analysis unravels the complexity of the dimorphic fungus<br />

C<strong>and</strong>ida albicans cell wall proteome. Mol.Cell Proteomics 1, 967–82.<br />

12. Klis, F. M. (1994) Review: cell wall assembly in yeast. Yeast, 10, 851–69.<br />

13. Pardo, M., Ward, M., Bains, S., et al. (2000) A proteomic approach for the study<br />

of Saccharomyces cerevisiae cell wall biogenesis. Electrophoresis 21, 3396–3410.<br />

14. Pitarch, A., Jimenez, A., Nombela, C., <strong>and</strong> Gil, C. (2006) Decoding serological<br />

response to C<strong>and</strong>ida cell wall immunome into novel diagnostic, prognostic, <strong>and</strong>


Collection of Proteins Secreted from Regenerating Yeast Protoplasts 263<br />

therapeutic c<strong>and</strong>idates for systemic c<strong>and</strong>idiasis by proteomic <strong>and</strong> bioinformatic<br />

analyses. Mol.Cell Proteomics 5, 79–96.<br />

15. Kobori, H., Yamada, N., Taki, A., <strong>and</strong> Osumi, M. (1989) Actin is associated<br />

with the formation of the cell wall in reverting protoplasts of the fission yeast<br />

Schizosaccharomyces pombe. J.Cell Sci. 94, 635–46.<br />

16. Tanaka, H., Kamogawa, T, Aoyagi, H., Kato, I. <strong>and</strong> Nakajima, R. (2000) Invertase<br />

production by Saccharomyces cerevisiae protoplasts immobilized in strontium<br />

alginate gel beads. J. Biosci. Bioeng. 89, 498–500.<br />

17. Pitarch, A., Pardo, M., Jimenez, A., et al. (1999) Two-dimensional gel<br />

electrophoresis as analytical tool for identifying C<strong>and</strong>ida albicans immunogenic<br />

proteins. Electrophoresis 20, 1001–10.<br />

18. Pitarch, A., Nombela, C., <strong>and</strong> Gil, C. (2006) C<strong>and</strong>ida albicans biology <strong>and</strong><br />

pathogenicity: Insights from proteomics, in Microbial Proteomics: Functional<br />

Biology of Whole Organisms (Humphery-Smith,I. <strong>and</strong> Hecker,M., eds), Wiley-Vch,<br />

Hoboken, NJ, pp. 285–330.<br />

19. Lee, K. L., Buckley, H. R., <strong>and</strong> Campbell, C. C. (1975) An amino acid liquid<br />

synthetic medium for the development of mycelial <strong>and</strong> yeast forms of C<strong>and</strong>ida<br />

albicans. Sabouraudia. 13, 148–53.<br />

20. Elorza, M. V., Rico, H., Gozalbo, D., <strong>and</strong> Sent<strong>and</strong>reu, R. (1983) Cell wall composition<br />

<strong>and</strong> protoplast regeneration in C<strong>and</strong>ida albicans. Antonie Van Leeuwenhoek<br />

49, 457–69.<br />

21. Cabib, E. <strong>and</strong> Duran, A. (1975) Simple <strong>and</strong> sensitive procedure for screening yeast<br />

mutants that lyse at nonpermissive temperatures. J.Bacteriol. 124, 1604–06.<br />

22. Murgui, A., Elorza, M. V., <strong>and</strong> Sent<strong>and</strong>reu, R. (1986) Tunicamycin <strong>and</strong> papulac<strong>and</strong>in<br />

B inhibit incorporation of specific mannoproteins into the wall of C<strong>and</strong>ida<br />

albicans regenerating protoplasts. Biochim.Biophys.Acta 884, 550–8.<br />

23. Osumi, M., Yamada, N., Kobori, H., et al. (1989) Cell wall formation in regenerating<br />

protoplasts of Schizosaccharomyces pombe: study by high resolution, low<br />

voltage scanning electron microscopy. J.Electron Microsc. 38, 457–68.<br />

24. Osumi, M., Sato, M., Ishijima, S. A., Konomi, M., Takagi, T., <strong>and</strong> Yaguchi, H.<br />

(1998) Dynamics of cell wall formation in fission yeast, Schizosaccharomyces<br />

pombe. Fungal.Genet.Biol. 24,178–206.<br />

25. Ezeronye, O. U. <strong>and</strong> Okerentugba, P. O. (2001) Optimum conditions for yeast<br />

protoplast release <strong>and</strong> regeneration in Saccharomyces cerevisiae <strong>and</strong> C<strong>and</strong>ida<br />

tropicalis using gut enzymes of the giant African snail Achatina achatina.<br />

Lett.Appl.Microbiol. 32, 190–93.<br />

26. Eddy, A. A. <strong>and</strong> Williamson, D. H. (1957) A method of isolating protoplasts from<br />

yeasts. Nature 179, 1252–53.<br />

27. Agogbua, S. O., Anosike, E. O <strong>and</strong> Ugochukwu, E. N. (1978) Partial purification<br />

<strong>and</strong> some properties of arylsulphatases from the gut of the giant African snail<br />

Achatina achatina. Comp. Biochem. Phys. 59B, 169–73.<br />

28. Kaneko, T., Kitamura, K. <strong>and</strong> Yamamoto, Y. (1973) Susceptibilities of yeast to<br />

yeast cell lytic enzyme of Arthrobacter luteus. Agric. Biol. Chem. 37, 2295–2302.


21<br />

<strong>Sample</strong> <strong>Preparation</strong> Procedure for Cellular Fungi<br />

Alois Harder<br />

Summary<br />

A crucial step in quantitative proteomics is an artefact free <strong>and</strong> reproducible sample<br />

preparation protocol, which has to be adapted <strong>and</strong> optimized to nearly all types of cells.<br />

Here we provide a sample preparation method for quantitative proteomics of cellular fungi.<br />

Two different protein extraction methods were compared with focus on reproducibility,<br />

minimized proteolytic degradation <strong>and</strong> protein losses during the sample preparation.<br />

In the first preparation the cells were lysed by sonication followed by protein solubilization<br />

in “st<strong>and</strong>ard” lysis buffer. The second preparation was performed with a SDSpresolubilization<br />

step followed by sonication <strong>and</strong> further boiling, before diluting the<br />

sample with lysis buffer. We have shown that the sample preparation for cellular fungi<br />

is performed with maximum protein solubilization, higher reproducibility <strong>and</strong> a reduced<br />

proteolytic activity by including a SDS-presolubilization step in the sample preparation<br />

protocol.<br />

Key Words: <strong>2D</strong>-electrophoresis; fungi; proteolytic activity; sample preparation;<br />

yeast.<br />

1. Introduction<br />

Fungal cells have nowadays found their way in nearly all areas of biochemistry<br />

<strong>and</strong> pharmacy. Famous examples are active medicaments like PerenterolTM with Saccharomyces boulardii as the active ingredient (1,2), yeast host cell<br />

systems like the vaccine production cycle or diverse secondary metabolite<br />

discovering systems like the Novobiocin synthesis to name only few (3,4). To<br />

push those applications or detect new fungal targets or metabolites, a solid<br />

basis of information in cell growth, cell cycle, gene- <strong>and</strong> protein data are<br />

essential. Whereas cell growth <strong>and</strong> cycle can be researched in conceivable<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

265


266 Harder<br />

time, gene <strong>and</strong> protein data are missing for the majority of the fungi. The<br />

finished <strong>and</strong> currently running fungal genome sequencing projects are mainly<br />

concentrating on model- or pathogen strains: Saccharomyces cerevisiae, Ashbya<br />

gossypii, Aspergillus fumigatus, C<strong>and</strong>ida parapsilosis, C<strong>and</strong>ida dubliniensis,<br />

Pneumocystis carinii, Phytophthora infestans, <strong>and</strong> Schizosaccharomyces pombe<br />

have been sequenced or will be finished soon <strong>and</strong> are available for the<br />

community (5,6).<br />

For identification of fungal proteins, genome data of the organism to be<br />

researched has to be available. Although the fungal protein databases are<br />

growing, the listed open reading frames are far from their complete <strong>and</strong> correct<br />

functional characterization.<br />

However,withthesequencedfungalgenomes,theavailableproteindata<strong>and</strong>the<br />

homology – search programs with a solid foundation for further proteome analysis<br />

where eumycotic cells are present. Fungal cells are easy to cultivate, to manipulate,<br />

<strong>and</strong> in most cases are non-toxic, but the preparation of a robust <strong>and</strong> artefactfree<br />

protein extract for a proteomics approach is a challenge. Major problems in<br />

fungal proteomics result from the cells strong proteolytic activity. Most of the<br />

fungalproteases,intensivelyresearchedintheeumycoticmodelorganismSaccharomyces<br />

cerevisiae, are located in the cytosol <strong>and</strong> the vacuole. Few proteases<br />

are located in the membrane, the endoplasmic reticulum, the mitochondria,<br />

<strong>and</strong> the Golgi complex (7,8). These include endoproteinases, carboxypeptidases,<br />

aminopeptidases, <strong>and</strong> dipeptidyl-aminopeptinase. By activation of these<br />

proteasesproteinmodificationswilloccur<strong>and</strong>canruleoutquantitativeproteomics<br />

research (9–10,11).<br />

Of the many cellular proteases, the lumenal vacuolar proteases comprise the<br />

major source of problems for protein analysis <strong>and</strong> proteome research. Endoproteinase<br />

A <strong>and</strong> B (PrA <strong>and</strong> PrB), carboxypeptidases Y <strong>and</strong> S, aminopeptidase<br />

I (LAP IV), <strong>and</strong> the aminopeptidase yscCo (ApCo) are found soluble in the<br />

vacuole (12,13). Polypeptide inhibitors of these vacuolar proteases are found<br />

in the cytosol (e.g., inhibitor for PrB is I B; inhibitor for yscCo is yscCo F).<br />

By preparing the crude extract all cell compartments are broken up, meaning<br />

that corresponding protease inhibitors will bind to their substrate by forming<br />

an inactive complex—in this state proteolytic activity is inhibited. As reported<br />

from Jones et al. the fungal proteases PrA <strong>and</strong> PrB, can be activated out of the<br />

crude extract by lowering the pH to a level of 4–5. The reason for this activation<br />

might be the hydrolysis of the polypeptide inhibitors (7,14). Further research<br />

on protease deficiency mutants showed that the addition of denaturating agents<br />

activates proteolysis by removing the corresponding inhibitor from the protease<br />

molecule. Due to the exigency in proteomics research, for working in strict


<strong>Sample</strong> <strong>Preparation</strong> Procedure for Cellular Fungi 267<br />

denaturation conditions, the necessity of a “non st<strong>and</strong>ard” sample preparation<br />

procedure for fungal cells is obvious.<br />

The fungal sample preparation for proteomics research has to<br />

• (BL)be carried out in strong denaturating conditions for its stability during<br />

the <strong>2D</strong> - electrophoretic separation<br />

• display robustness to guarantee reproducibility by repeating the experiments<br />

• solubilize low abundant protein as well as high <strong>and</strong> low molecular weight<br />

proteins in all pH ranges<br />

• effectively inactivate the strong proteolytic activity of fungal cells<br />

2. Materials<br />

2.1. Cell Culture<br />

1. St<strong>and</strong>ard aerobic incubator with an inbuilt horizontal rotary shaker. 300 mL<br />

Erlenmeyer flasks with silicone plugs are used for aerobic cell cultivation.<br />

2. Horizontal rotary shaker (e.g., HS 260, IKA).<br />

3. Incubation mask (e.g., Certomat H/HK).<br />

4. S. cerevisiae haploid wild type strain N318C (“Deutsche Sammlung für Mikroben<br />

und Zellinien”, DSMZ Braunschweig, Germany).<br />

5. Defined synthetic YNB medium (Difco Labs, San Francisco CA, USA) is<br />

dissolved to a concentration of 6.7 g/L in distilled water (


268 Harder<br />

2.4. Scintillation Counting <strong>and</strong> Protein Assay<br />

1. For S35-labelled cells, a scintillation counting is performed: 4.5 mL POPOP<br />

solution is required for one vial; 2 × 4 cm cut filter paper is used for the counting<br />

(see Note 1). As a calibration st<strong>and</strong>ard a C12 isotope (GE Healthcare) is applied.<br />

2. Lowry protein assay kit for SDS containing samples.<br />

3. The Bradford protein assay kit (Bio-Rad) for samples dissolved in lysis buffers.<br />

4. BSA (Bio-Rad) is used as a protein st<strong>and</strong>ard for both protein assays.<br />

UV-spectral photometer e.g., Beckman DU-64 (see Note 2).<br />

3. Methods<br />

3.1. S. cerevisiae Cell Culture<br />

1. The S. cerevisiae haploid wild type strain N318C is cultivated at 30 °C in defined<br />

synthetic YNB medium. First a preculture is grown, in which about 105 cells are<br />

inoculated in 30 mL culture medium (see previous section) <strong>and</strong> grown to a cell<br />

density of OD 0.6 measured at =610nm (see Note 3).<br />

2. Then the main culture (30 mL, 300-mL Erlenmeyer flasks) is started with an<br />

inoculum from 10 μL out of the preculture. The cells are harvested by centrifugation<br />

(5,000g, 15 min) in the late midlogarithmic phase, when the culture reached<br />

an OD of 1.1 followed by washing in ice-cold sterilized water (see Note 4).<br />

3. A control smear is performed on a Sabouraud agar plate to check for cellular<br />

contaminations. The incubation of the agar plate was done for 48 h at 37 °C in<br />

sterile conditions.<br />

4. Harvested yeast cells are then transferred into individual 1.5 mL Eppendorf tubes<br />

<strong>and</strong> stored at –78 °C, if not processed immediately.<br />

3.2. S-35 In Vitro Labeling (optional)<br />

1. The S-35 in vitro labelling can be done additionally to the st<strong>and</strong>ard cell growth<br />

procedure. For the in vitro S 35 - labelling experiment the main culture is grown<br />

to an OD of 1.0 (=610 nm)<br />

2. Then 10 mL of the cell suspension is inoculated with 100 μCi <strong>and</strong> kept shaking for a<br />

quarter of the fungal generation time (for S. cerevisiae in YNB – medium→ 30min).<br />

3. After the S-35 incubation the cells are harvested by centrifugation (5 min, 5,000g)<br />

<strong>and</strong> the supernatant is discarded to the radioactive waste. The amount of protein,<br />

which will be extracted out of the obtained S-35 labeled pellet will be approx<br />

5 mg protein solubilized in lysis buffer. Only methionine deficient mediums are<br />

suited for S-35 in vitro labeling.<br />

3.3. Cell Disruption <strong>and</strong> Protein Solubilization<br />

1. The yeast cell pellet from 10 mL culture OD 1.1 (see Section 21.3.1) is resuspended<br />

in 200 μL hot (95 °C) SDS buffer for presolubilization (see Note 5 <strong>and</strong> 6).


<strong>Sample</strong> <strong>Preparation</strong> Procedure for Cellular Fungi 269<br />

2. After brief vortexing the hot suspension is sonicated 10 times for 1s (60 W, 20<br />

kHz). The sonication has to be performed in such way that the probe is dipping<br />

as deep as possible in the suspension <strong>and</strong> not touching the sample cup during<br />

the agitation time. A 1.5-mL cup (conic bottom) is used for maximum abrasive<br />

agitation. Avoid foaming of the sample during this process (see Note 7).<br />

3. After cell lyses the sample is boiled additionally for 5 min <strong>and</strong> then cooled down<br />

in an ice cold water bath to a sample temperature of maximum 20 °C (see Note<br />

8 <strong>and</strong> 9).<br />

4. Then the suspension is diluted with 500 μL lysis buffer <strong>and</strong> kept shaking for 20<br />

min at room temperature.<br />

5. After spinning down for 5 min at 10,000g, the protein concentration is measured<br />

from the clear supernatant (see Section 21.3.4).<br />

6. The clear supernatant is stored at –78 °C or subjected to <strong>2D</strong>-electrophoresis.<br />

An example of four replicate gels (IPG 4-7, 250 μg protein load, S. cerevisiae,<br />

SDS presolubilisation) is shown in Fig. 2 compared to a “st<strong>and</strong>ard preparation<br />

protocol” without the SDS presolubilization step (Fig. 1).<br />

1 97kD 2<br />

67kD<br />

45kD<br />

29kD<br />

21kD<br />

12kD<br />

6kD<br />

3 4<br />

Fig. 1. Displaying four replicate <strong>2D</strong> gels (IPG 4-7L, 250 μg protein load, fluorescence<br />

stain) from the yeast proteome (S. cerevisiae) performed without the SDS -<br />

presolubilization step (see Note 12 <strong>and</strong> 13).


270 Harder<br />

5 6<br />

97kD<br />

67kD<br />

45kD<br />

29kD<br />

21kD<br />

12kD<br />

6kD<br />

7 8<br />

Fig. 2. Displaying four replicate <strong>2D</strong> gels (IPG 4-7L, 250 μg protein load, fluorescence<br />

stain) from the yeast proteome (S. cerevisiae) performed with the SDS - presolubilization<br />

step (see Note 14).<br />

7. An approximate 7 μg/μL protein concentration can be expected in that obtained<br />

700-μL sample volume (see Note 10)<br />

3.4. Scintillation Counting <strong>and</strong> Protein Assay<br />

1. 1. For S35 - labeled cells, the protein concentration is measured by removing<br />

1 μL of the lysis buffer extract to a filter. After drying the sample, the filter is<br />

inserted into a counting vial, which is filled with 4.5 mL POPOP - solution. The<br />

scintillation counter is calibrated to a C12 – st<strong>and</strong>ard, which was first solubilized<br />

in lysis buffer. The scintillation counting should be repeated three times for each<br />

sample. Approximately 3 × 106 cpm should be loaded onto an IPG strip 4–7,<br />

2.5×106counts onto the gradient 3–10. This method is working with <strong>and</strong> without<br />

the SDS presolubilization step (see Note 11).<br />

2. 20 μL of the SDS extract are used for the Lowry protein assay. 5 μL of the lysis<br />

buffer extract are taken for the Bradford protein assay.


<strong>Sample</strong> <strong>Preparation</strong> Procedure for Cellular Fungi 271<br />

4. Notes<br />

1. For S35 counting: after an appropriate amount of sample is pipeted onto the<br />

filter, the sample has to be dried for at least 30 min, due to water residues<br />

suppress the radio signal significantly.<br />

2. The determination of the protein content is optional, but an optimum <strong>and</strong> reproducible<br />

protein load is essential for successful protein quantification.<br />

3. Significantly increased proteolytic activity in fungi is observed in cells grown in<br />

minimal medium compared to cells grown in full medium, in cells harvested in<br />

their stationary phase (increases proteases activity to a level at least 100 times<br />

that of log phase cells), in vital cells stored or grown in nitrogen starvation <strong>and</strong><br />

in the presence of peptone in the culture medium.<br />

4. When you are researching fungal stress responses, washing the pellet with ice<br />

cold water will induce a deficient cell response. In that case the cell washing<br />

can be performed with 15 °C water as well.<br />

5. The whole procedure is carried out in one single Eppendorf tube. By establishing<br />

the yeast sample preparation procedure main focus was given to its<br />

reproducibility, robustness, <strong>and</strong> maximum protein solubility with respect to the<br />

yeast’s peculiarities like proteolytic activity <strong>and</strong> cell wall constitution.<br />

6. Dried pellets (for measuring the amount of cells) are not suited for further<br />

proteome analysis, due to heat shock responses of the cells. Homologue pellets<br />

should be used for protein extraction <strong>and</strong> <strong>2D</strong> separation.<br />

7. The correct sonication will provide a quantitative cell disruption <strong>and</strong> protein<br />

disaggregating by simultaneously cracking the fungal DNA <strong>and</strong> RNA str<strong>and</strong>s,<br />

which otherwise can produce horizontal streaks in the <strong>2D</strong> <strong>PAGE</strong>. The DNA <strong>and</strong><br />

RNA fragments should become visible as a thin white foam after the sonication<br />

step.<br />

8. By presolubilization in hot SDS buffer fungal proteases are denatured <strong>and</strong> inactivated<br />

during the cell breakage. Boiling the sample in SDS will increase protein<br />

solubility, especially for the high molecular weight <strong>and</strong> hydrophobic proteins,<br />

due to the high affinity of SDS to proteins in solution.<br />

9. It is crucial to cool down the SDS-boiled sample below 20 °C, to avoid carbamylation<br />

reactions with the urea containing lysis buffer. The final SDS concentration<br />

should not exceed 0.25% in the extract to be applied onto the IPG strip, due to<br />

the interference of the SDS with the isoelectric focussing, so be sure that during<br />

the SDS boiling step the total volume is kept.<br />

10. By starting with about 1×108cells (S. cerevisiae) the obtained protein concentration<br />

will be approximately 7μg/μL in a 700 μL volume.<br />

11. The protein concentration within this preparation method can be measured with<br />

the Lowry Kit after the SDS-presolubilization or with a scintillation counter (by<br />

S-35 labeled cells) out of the final extract. Determination of the dry weight or<br />

cell counting after cell harvesting should be done at least one time to check<br />

purity <strong>and</strong> approximate amount of cultivated cells.


272 Harder<br />

12. A major problem in fungal protein extraction is the strong fungal proteolytic<br />

activity. By applying a st<strong>and</strong>ard preparation protocol significant degradation can<br />

occur (see Fig. 1) <strong>and</strong> consequently a protein quantification is useless.<br />

13. In many cases proteolytic activity is not as obvious as in the gels 1–4 (Fig. 1),<br />

especially when degradation only begins. It can show up in disappearance of<br />

single spots or in the decrease of the quantities of some spots, which can be<br />

spuriously interpreted as a cell response. Therefore the complete <strong>and</strong> irreversible<br />

inactivation of the mycotic proteome is essential.<br />

14. Note, that the obtained “crude extract” will include proteins from the cytoplasm,<br />

the organelles, membrane bound proteins <strong>and</strong> surface proteins. Integral plasma<br />

membrane proteins as well as integral cell wall proteins are not extracted with<br />

that recipe (see Fig. 2).<br />

References<br />

1. Sougioultzis, S., Simeonidis, S., Bhaskar, K. R., et al. (2006) Saccharomyces<br />

boulardii produces a soluble anti-inflammatory factor that inhibits NF-kappaBmediated<br />

IL-8 gene expression. Biochem. Biophys Res Commun. 343, 69–76<br />

2. Dalmasso, G., Loubat, A., Dahan, S., Calle, G., Rampal, P., <strong>and</strong> Czerucka, D.<br />

(2006) Saccharomyces boulardii prevents TNF-alpha-induced apoptosis in EHECinfected<br />

T84 cells. Res. Microbiol. 164, 876–84.<br />

3. Hardwidge, P. R., Donohoe, S., Aebersold, R., <strong>and</strong> Finlay, B. B. (2006) Proteomic<br />

analysis of the binding partners to enteropathogenic Escherichia coli virulence<br />

proteins expressed in Saccharomyces cerevisiae. Proteomics. 6(7), 2174–79.<br />

4. Jenkins, J. R., Pocklington, M. J., <strong>and</strong> Orr, E. (1990) The F1 ATP synthetase<br />

beta-subunit: a major yeast novobiocin binding protein. Cell Sci. Aug 96 ( Pt 4),<br />

675–82.<br />

5. Hermida, L., Brachat, S., Voegeli, S., Philippsen, P., <strong>and</strong> Primig, M. (2005) The<br />

Ashbya Genome Database (AGD) - a tool for the yeast community <strong>and</strong> genome<br />

biologists. Nucleic Acids Res. 33, 348–52.<br />

6. [No authors listed]. (1997) The yeast genome directory.Nature 387 (6632 Suppl) 5.<br />

7. Jones, E. W. (1991) Tackling the protease problem in Saccharomyces cerevisiae.<br />

Methods Enzymol. 19, 428–53.<br />

8. Groll, M. <strong>and</strong> Huber, R. (2005) Purification, crystallization, <strong>and</strong> X-ray analysis of<br />

the yeast 20S proteasome. Methods Enzymol. 398, 329–36.<br />

9. McIntyre, J., Podlaska, A., Skoneczna, A., Halas, A., <strong>and</strong> Sledziewska-Gojska,<br />

E. (2006) Analysis of the spontaneous mutator phenotype associated with 20S<br />

proteasome deficiency in S. cerevisiae. Mutat Res. 593(1–2), 153–63.<br />

10. Garduno, E., Perez-Giraldo, C., Blanco, M.T., Hurtado, C., <strong>and</strong> Gomez-Garcia, A. C.<br />

(2005)Exposuretotherapeuticconcentrationsofritonavir,butnotsaquinavir,reduces<br />

secreted aspartyl proteinase of C<strong>and</strong>ida parapsilosis. Chemotherapy 51(5), 252–5.<br />

11. Schmidt, M., Haas, W., Crosas, B., et al. (2005) The HEAT repeat protein Blm10<br />

regulates the yeast proteasome by capping the core particle. Nat. Struct. Mol. Biol.<br />

4, 294–303.


<strong>Sample</strong> <strong>Preparation</strong> Procedure for Cellular Fungi 273<br />

12. Mima, J., Hayashidam, M., Fujii, T., et al. (2005) Structure of the carboxypeptidase<br />

Y inhibitor IC in complex with the cognate proteinase reveals a novel mode of the<br />

proteinase-protein inhibitor interaction. J. Mol. Biol. 346(5), 1323–34.<br />

13. Takai, T., Kato, T., Sakata, Y., et al. (2003) Recombinant Der p1<strong>and</strong>Derf1<br />

exhibit cysteine protease activity but no serine protease activity. Mol. Biol. 328(4),<br />

944–52.<br />

14. Fotedar, R. <strong>and</strong> Al-Hedaithy, S. S. (2005) Comparison of phospholipase <strong>and</strong><br />

proteinase activity in C<strong>and</strong>ida albicans <strong>and</strong> C. dubliniensis. Mycoses 48(1), 62–7.<br />

15. Görg, A., Weiss, W., <strong>and</strong> Dunn, M. J. (2004) Current two-dimensional<br />

electrophoresis technology for proteomics. Proteomics 04 (12), 3665–85.<br />

16. Williams, B. <strong>and</strong> Wilson, K. (1994) Methoden der Biochemie. Thieme-Verlag<br />

Stuttgart. New York, 3. Auflage 1994<br />

17. Johnson, T. M., Holady, s. K., Sun, Y., Subramaniam, P. S., Johnson H. M., <strong>and</strong><br />

Krishna, N. R. (1999) Purification, <strong>and</strong> characterization of interferon-tau produced<br />

in Pichia pastoris grown in a minimal medium. Interferon Cytokine Res. 19(6),<br />

631–6.<br />

18. Harder, A., Wildgruber, R., Nawrocki, A., Fey, S. J., Larsen, P. M., <strong>and</strong> Gorg,<br />

A. (1999) Comparison of yeast cell protein solubilization procedures for twodimensional<br />

electrophoresis. Electrophoresis 20 (4–5), 826–9.<br />

19. Wildgruber, R., Reil, G., Drews, O., Parlar, H., <strong>and</strong> Gorg, A. (2002) Web-based<br />

two-dimensional database of Saccharomyces cerevisiae proteins using immobilized<br />

pH gradients from pH 6 to pH 12 <strong>and</strong> matrix-assisted laser desorption/ionizationtime<br />

of flight mass spectrometry. Proteomics 2(6), 727–32.<br />

20. Luche, S., Santoni, V., <strong>and</strong> Rabilloud, T. (2003) Evaluation of nonionic <strong>and</strong><br />

zwitterionic detergents as membrane protein solubilizers in two-dimensional<br />

electrophoresis. Proteomics 3(3), 249–53.<br />

21. Görg, A. Two-Dimensional Electrophoresis of Proteins using Immobilized<br />

pH Gradients: online manual at: http://www.weihenstephan.de/<br />

blm/deg/manual/manfrm.htm<br />

22. Rabilloud, T., Strub, J. M., Luche, S., Dorsselaer, A., <strong>and</strong> Lunardi, J. (2001)<br />

Comparison between Sypro Ruby <strong>and</strong> ruthenium II tris (bathophenanthroline disulfonate)<br />

as fluorescent stains for protein detection in gels. Proteomics 1(5), 699–704.


22<br />

Isolation <strong>and</strong> Enrichment of Secreted Proteins<br />

from Filamentous Fungi<br />

Martha L. Medina <strong>and</strong> Wilson A. Francisco<br />

Summary<br />

Filamentous fungi have been recognized as extraordinary producers of secreted proteins<br />

<strong>and</strong> are known to produce novel proteins <strong>and</strong> enzymes through dispensable metabolic<br />

pathways. Here, methods are described for the isolation <strong>and</strong> enrichment of samples of<br />

secreted proteins from cultures of filamentous fungi for analysis by gel electrophoresis<br />

<strong>and</strong> mass spectrometry techniques. These methods can be readily applied to the study of<br />

differential protein expression <strong>and</strong> secretion <strong>and</strong> metabolic pathways in filamentous fungi<br />

by proteomic approaches.<br />

Key Words: Exoproteome; extracellular proteins; protein deglycosylation; protein<br />

precipitation; secreted proteins; secretome; gel electrophoresis.<br />

1. Introduction<br />

Protein secretion plays an important role in filamentous fungi, particularly in<br />

nutrition, as secreted enzymes degrade complex biological molecules to serve<br />

as carbon <strong>and</strong> nitrogen sources. Filamentous fungi are known for their ability<br />

to secrete a broad spectrum of enzymes, the majority of which are hydrolytic,<br />

into the extracellular matrix (1). This ability has been widely exploited by<br />

the biotechnology industry for the production of enzymes for commercial<br />

<strong>and</strong> industrial use. Most commonly, filamentous fungi secrete proteins via a<br />

classical secretory pathway (2) <strong>and</strong> most, if not all, of the secreted proteins<br />

are glycosylated, containing modifications such as oligomannose N- <strong>and</strong> Oglycans<br />

(3). These attached sugars increase the stability of the secreted proteins<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

275


276 Medina <strong>and</strong> Francisco<br />

<strong>and</strong> provide resistance to environmental influences, as well as increase their<br />

solubility in the culture media. Although many studies of protein secretion<br />

have been carried out in yeast <strong>and</strong> animal systems, studies on protein secretion<br />

by filamentous fungi are limited (4). Typical studies have focused on the<br />

identification, purification <strong>and</strong> characterization of secreted proteins, but only<br />

a few studies have been conducted on the global analysis of fungal extracellular<br />

proteomes. Proteomic studies on the analysis of secreted proteins from a<br />

number of fungi, including Aspergillus flavus (5,6), A. oryzae (7), Fusarium<br />

graminearum (8), <strong>and</strong> Pleurotus sapidus (9), have recently appeared in the<br />

literature. Much of the delay in the use of proteomic techniques for the study<br />

of fungal protein expression in general can be attributed to the fact that there is<br />

a lack of complete <strong>and</strong> publicly available genome sequences from filamentous<br />

fungi. It is expected that as the number of published fungal genomes increases,<br />

proteomic studies on both intracellular <strong>and</strong> extracellular proteins will follow.<br />

Within their high capacity to produce secreted proteins, filamentous fungi are<br />

able to express <strong>and</strong> secrete proteins for “dispensable” metabolic functions (10).<br />

These enzymes participate in pathways that are either not required for growth<br />

or are only required for growth under a limited range of conditions. These<br />

changes in protein secretion provide an excellent platform for the systematic<br />

study of the overall protein secretion expression (secretome or exoproteome) as<br />

a function of culture conditions. Proteomic analysis using two-dimensional gel<br />

electrophoresis (2-DE) <strong>and</strong> mass spectrometry (MS) has proven to be the most<br />

powerful <strong>and</strong> sensitive method for the identification of proteins in complex<br />

mixtures. 2-DE provides an excellent platform to assess differential secreted<br />

protein expression. Although the number of proteins that can be analyzed by 2-<br />

DE is still limited to 1,000–2,000 on one gel, the maximum number of secreted<br />

proteins by a filamentous fungi under a given set of conditions is well within<br />

this range, as a recent analysis of the genome of Aspergillus niger identified<br />

only about 400 putative secreted proteins from a total of about 5,100 genes (11).<br />

Obtaining MALDI-TOF MS data from tryptic digests of gel b<strong>and</strong>s <strong>and</strong> spots<br />

<strong>and</strong> searching against online protein databases can identify 2-DE separated<br />

proteins. The success of peptide mass fingerprinting depends on the detection of<br />

a representative set of peptide masses derived from a protein <strong>and</strong> that the protein<br />

in question is known (it exists in a protein database). Alternatively, proteins can<br />

be identified by MS/MS with custom databases that contain sequences of all<br />

publicly known fungal proteins or by de-novo sequencing using this technique.<br />

Several technical issues need to be considered when studying the secretome<br />

of filamentous fungi by proteomic analysis. First, although fungi have the<br />

ability to secrete large amounts of protein, these proteins are highly diluted in<br />

the culture medium. Second, glycosylation poses a problem for visualization<br />

of proteins separated by gel electrophoresis, as glycosylated samples tend to


Isolation of Secreted Proteins from Filamentous Fungi 277<br />

smear in gels. In addition, glycosylation can protect proteins against proteolysis,<br />

making the identification of these proteins more difficult as peptide mass<br />

fingerprinting relies on efficient protease digestion before mass spectrometry<br />

(12). Also, the attached sugars greatly increase the size of generated peptide<br />

fragments, adding another layer of complexity in their identification by mass<br />

spectrometry. These problems can be overcome by concentrating the media,<br />

most commonly by lyophilization or ultrafiltration, <strong>and</strong> by removing the<br />

sugars with commercially available glycosidases or by chemical deglycosylation<br />

methods.<br />

Here we describe a general procedure for the isolation <strong>and</strong> enrichment of<br />

secreted proteins from cultures of filamentous fungi for proteomic analysis<br />

by gel electrophoresis <strong>and</strong> mass spectrometry, including protocols for protein<br />

deglycosylation. The methods described here can be easily adopted for the<br />

isolation <strong>and</strong> characterization of the secretome of filamentous fungi grown<br />

under varied conditions.<br />

2. Materials<br />

2.1. Filtration <strong>and</strong> Lyophilization<br />

1. Miracloth (Calbiochem, EMD Biosciences, Inc., San Diego, CA) or No. 2<br />

Whatman filter paper.<br />

2. Lyophilizer<br />

2.2. Centrifugation <strong>and</strong> Ultrafiltration<br />

1. Centricon Plus-70 Centrifugal Filter Device (Millipore, Billerica, MA).<br />

2. Centrifuge<br />

2.3. Precipitation with Trichloroacetic Acid<br />

1. 20% (w/v) Trichloroacetic acid (TCA) (Sigma-Aldrich, St. Louis, MO), stored<br />

at 4°C.<br />

2. 70% Ethanol, stored at –20°C.<br />

3. Acetone<br />

2.4. Precipitation with Methanol <strong>and</strong> Chloroform<br />

1. Methanol<br />

2. Chloroform<br />

2.5. Enzymatic Deglycosylation<br />

1. Ultrafree-0.5 centrifugal filter devices with Biomax-5 membranes 5,000 NMWL<br />

(Millipore, Billerica, MA).


278 Medina <strong>and</strong> Francisco<br />

2. Peptide: N-glycosidase F (PNGase F) (New Engl<strong>and</strong> BioLabs, Inc., Ipswich, MA).<br />

PNGase F is provided with 10× glycoprotein denaturing buffer (0.5% SDS, 1%<br />

-mercaptoethanol), 10× G7 reaction buffer (50 mM sodium phosphate, pH 7.5),<br />

10% Nonidet P-40 (NP-40), <strong>and</strong> a solution of PNGase F (see Note 1).<br />

2.6. Chemical Deglycosylation (see Note 2)<br />

1. Reacti-Vial Reaction Vials (5 mL) (Pierce Chemical Co., Rockford, IL).<br />

2. Trifluoromethanesulfonic acid (TFMS) (Sigma/Aldrich, St. Louis, MO, USA)<br />

(see Note 3).<br />

3. Anisole, anhydrous (Sigma/Aldrich, St. Louis, MO), stored at 4°C.<br />

4. 60% (v/v) Pyridine solution, stored at –20°C.<br />

5. Diethyl ether, stored at –20°C.<br />

6. 95% Ethanol, stored at 4°C.<br />

2.7. SDS-<strong>PAGE</strong><br />

1. SDS <strong>Sample</strong> Reducing Buffer: Mix 4.05 mL of deionized water, 1.25 mL 0.5M<br />

Tris-HCl, pH 6.8, 2.50 mL glycerol, 2.00 mL 10% SDS <strong>and</strong> 0.20 mL 0.5% (w/v)<br />

bromophenol blue.<br />

2. -Mercaptoethanol<br />

2.8. Two-Dimensional Gel Electrophoresis<br />

1. 2-DE Buffer: 8M urea, 2% (w/v) CHAPS, 50 mM dithiothreitol, 0.2% (w/v) 100×<br />

Bio-Lyte 3–10 ampholytes (Bio-Rad Laboratories, Inc., Hercules, CA), 0.001%<br />

(w/v) bromophenol blue.<br />

3. Methods<br />

This chapter describes general-purpose sample preparation methods for the<br />

isolation <strong>and</strong> enrichment of secreted proteins from cultures of filamentous<br />

fungi. The methods described in the following section can be divided into (1)<br />

isolation <strong>and</strong> concentration by filtration <strong>and</strong> lyophilization or ultrafiltration,<br />

(2) precipitation, <strong>and</strong> (3) deglycosylation of concentrated protein samples by<br />

enzymatic (PNGase F) or chemical deglycosylation using TFMS acid (13).<br />

The samples can then be further analyzed by gel electrophoresis <strong>and</strong> mass<br />

spectrometry by established techniques described in other chapters. A schematic<br />

representation of these protocols is shown in Fig. 1.<br />

3.1. Isolation <strong>and</strong> Concentration of Supernatant Broth<br />

1. After the studied filamentous fungus has been grown in liquid culture media for<br />

the desired time, the broth containing the secreted proteins is collected by filtration


Isolation of Secreted Proteins from Filamentous Fungi 279<br />

Fig. 1. Simplified schematic for the isolation <strong>and</strong> enrichment of secreted proteins<br />

from cultures of filamentous fungi for proteomic analysis (see Methods section for<br />

details).<br />

through a Miracloth or No. 2 Whatman filter paper. Alternatively, the fungal<br />

mycelia can be separated from the supernatants by centrifugation at 10,000g for<br />

10 min. (see Note 4).<br />

2. The supernatants are concentrated by lyophilization (steps 3–5) or ultrafiltration<br />

(steps 6–8).<br />

3. For lyophilization, place the filtered supernatants in individual round bottom<br />

flasks <strong>and</strong> freeze in liquid nitrogen. (see Note 5).


280 Medina <strong>and</strong> Francisco<br />

4. Place the round bottom flasks on the lyophilizer until they are completely dry.<br />

5. Redissolve the contents of the round bottom flasks in a minimal amount of<br />

deionized water, <strong>and</strong> store at –20°C until further analysis.<br />

6. For ultrafiltration, place the filtered supernatant in a Centricon Plus-70 centrifugal<br />

filter device to a maximum of 70 mL.<br />

7. Centrifuge at 3,500g until the desired final volume is achieved.<br />

8. The concentrated supernatant can be washed with an appropriate buffer (e.g.,<br />

50 mM phosphate buffer, pH 6.0) <strong>and</strong> reconcentrated to remove excess salts,<br />

pigments, <strong>and</strong> metabolites. (see Note 6).<br />

3.2. Protein Precipitation<br />

Protein precipitation is an efficient method for the removal of most contaminants,<br />

including detergents, salts, peptides, lipids, <strong>and</strong> phenolic compounds<br />

from protein samples. Either one of the methods described below can be used<br />

to precipitate the secreted proteins from the concentrated broth supernatant in<br />

preparation for gel electrophoresis analysis. It should be noted that no method<br />

will precipitate all proteins <strong>and</strong> some proteins will be difficult to resuspend<br />

following precipitation.<br />

3.2.1. Precipitation with Trichloroacetic Acid (see Note 7)<br />

1. Pipet 0.3 mL sample of concentrated broth supernatant into a 1.5-mL siliconized<br />

microcentrifuge tube <strong>and</strong> add an equal amount of cold TCA solution.<br />

2. The mixture is incubated for 2hat–20°C to allow the proteins to precipitate.<br />

After 2 h, allow samples to thaw if lightly frozen.<br />

3. Centrifuge for 10 min at 14,000g.<br />

4. The samples are decanted, <strong>and</strong> 1 mL of cold 70% ethanol is added, vortexed, <strong>and</strong><br />

recentrifuged for 3 min.<br />

5. Step 4 is repeated 3 times.<br />

6. To completely dry the secreted protein pellet, add 1 mL of acetone, vortex <strong>and</strong><br />

centrifuge for 1 min. The acetone is decanted, <strong>and</strong> the pellet is allowed to air dry<br />

for 30 min.<br />

7. The dried pellet can be stored at –20°C until further analysis.<br />

8. For electrophoresis analysis, the pellet is dissolved in SDS-<strong>PAGE</strong> or 2-DE sample<br />

buffer, as described below (see Subheading 3.5 <strong>and</strong> Note 8).<br />

3.2.2. Precipitation with Chloroform/Methanol<br />

1. To 100 μL of the concentrated supernatant in a siliconized microcentrifuge tube,<br />

add 400 μL of methanol, 100 μL of chloroform, <strong>and</strong> 300 μL of H2O, <strong>and</strong> mix<br />

well.<br />

2. Incubate at 4°C for 5 min <strong>and</strong> centrifuge at 9,000g at 4°C for 2 min.


Isolation of Secreted Proteins from Filamentous Fungi 281<br />

3. The upper phase is carefully removed <strong>and</strong> discarded. Add another 300 μL of<br />

methanol to the rest of the lower chloroform phase <strong>and</strong> the interphase with the<br />

precipitated protein <strong>and</strong> mix well.<br />

4. Incubate at 4°C for 5 min <strong>and</strong> pellet the proteins by centrifugation at 13,000g for<br />

5 min at 4°C. The supernatant is removed <strong>and</strong> the protein pellet is dried under a<br />

stream of air.<br />

5. The dried pellet can be stored at –20°C until further analysis.<br />

6. For electrophoresis analysis, the pellet is dissolved in SDS-<strong>PAGE</strong> or 2-DE sample<br />

buffer (see Subheading 3.5).<br />

3.3. Protein Deglycosylation<br />

3.3.1. Enzymatic Protein Deglycosylation with PNGase F<br />

1. Concentrate a 500 μL sample of the concentrated supernatant broth to 50 μL<br />

using a Ultrafree-0.5 with a Biomax-5 membrane (5,000 NMWL) centrifugal<br />

filter device by centrifugation at 12,000g at 4°C. To desalt the sample, add 400 μL<br />

of deionized water to the concentrated sample <strong>and</strong> centrifuge again until a final<br />

volume of 50 μL is reached.<br />

2. The concentrated, desalted solution is removed from the centrifugal filter device<br />

<strong>and</strong> placed in a 600 μL siliconized microcentrifuge tube.<br />

3. Enzymatic digestion is carried out as follows <strong>and</strong> according to the manufacturer’s<br />

instructions (New Engl<strong>and</strong> BioLabs, Inc.): 15 μL of denaturing buffer is added to<br />

the sample <strong>and</strong> boiled at 100°C for 10 min. To this sample, 3.5 μL of G7 buffer<br />

<strong>and</strong> 3.5 μL of NP-40 buffer are added, <strong>and</strong> the samples are digested with 1,000 U<br />

of PNGase F (see Note 9) for 18 hours at 37°C.<br />

4. Following deglycosylation, the samples are precipitated as described above (see<br />

Subheading 3.3).<br />

3.3.2. Chemical Protein Deglycosylation<br />

with Trifluoromethanesulfonic Acid<br />

1. Glycoprotein samples should be relatively free of salts, minerals <strong>and</strong> detergents.<br />

<strong>Sample</strong>s must also be completely dried. Lyophilize protein sample in a 5-mL<br />

Reacti-vial.<br />

2. Incubate lyophilized sample with 0.3 mL of cold anisole <strong>and</strong> 0.6 mL of cold<br />

TFMS at 0°C in an ice-bath for 4 h under nitrogen with occasional shaking.<br />

3. The reaction mixture is cooled to below –20°C by placing in a dry ice-ethanol<br />

bath <strong>and</strong> neutralized by slowly adding 1.2 mL of cold 60% aqueous pyridine<br />

solution.<br />

4. The deglycosylated peptides are freed of reagents <strong>and</strong> low-molecular weight<br />

sugars by adding 2 mL of cold diethyl ether. The suspension is vortexed <strong>and</strong><br />

extracted twice with cold ether.


282 Medina <strong>and</strong> Francisco<br />

5. The aqueous layer is lyophilized, redissolved in deionized water, <strong>and</strong> precipitated<br />

as described in Subheading 3.3.<br />

3.4. <strong>Preparation</strong> of <strong>Sample</strong>s for Gel Electrophoresis Analysis (see<br />

Note 10)<br />

3.4.1. SDS-<strong>PAGE</strong><br />

1. Add 50 μL of -mercaptoethanol to 950 μL SDS Reducing Buffer before use.<br />

2. Dissolve the precipitated protein pellet in the appropriate volume of SDS reducing<br />

sample buffer determined by the size of the gel <strong>and</strong> the system used.<br />

3. Boil sample for 4 min.<br />

3.4.2. Two-dimensional Gel Electrophoresis<br />

1. Dissolve the precipitated protein pellet in the appropriate volume of 2-DE Buffer<br />

determined by the size of the IPG strip <strong>and</strong> the system used for isoelectric focusing<br />

(see Note 11).<br />

2. Incubate the sample for2hatroom temperature <strong>and</strong> remove any insoluble material<br />

by centrifugation at 10,000g for 10 min.<br />

4. Notes<br />

1. PNGase F is an amidase that cleaves between the innermost N-acetylglucosamine<br />

<strong>and</strong> asparagine residues of high mannose, hybrid, <strong>and</strong> complex oligosaccharides<br />

from N-linked glycoproteins. PNGase F is the enzyme of choice for<br />

removing most N-linked oligosaccharides. Although secreted proteins could<br />

contain O-linked oligosaccharides, a recent study done on commercial cellulose<br />

enzyme preparation from the filamentous fungus Trichoderma reesei demonstrated<br />

that PNGase F treatment was superior to other enzymatic or chemical<br />

deglycosylation treatements in terms of yielding peptides through MALDI-MS<br />

that resulted in actual protein identification when searched against a database<br />

(12).<br />

2. For chemical deglycosylation, the use of a commercially available kit is recommended.<br />

Two of such kits are the GlycoProfile IV, Chemical Deglycosylation<br />

Kit from Sigma/Aldrich (Sigma/Aldrich, St. Louis, MO, USA) <strong>and</strong> Glycofree<br />

Chemical Deglycosylation Kit from ProZyme, Inc. (San Le<strong>and</strong>ro, CA, USA).<br />

3. Trifluoromethanesulfonic acid is a strong acid, highly corrosive <strong>and</strong> hygroscopic.<br />

Protective goggles, laboratory coat <strong>and</strong> gloves should be worn when working<br />

with TFMS.<br />

4. Typically, centrifuging is used to separate cells, in this case mycelia, from the<br />

supernatants. However, centrifuging does not work as well for filamentous fungi,<br />

as mycelia do not pack well <strong>and</strong> can float to the surface of the supernatant;<br />

therefore, filtering is preferred over centrifugation.


Isolation of Secreted Proteins from Filamentous Fungi 283<br />

5. Culture supernatants in the round bottom flasks should be frozen by spinning<br />

the flask while immersing in liquid nitrogen to ensure even distribution of the<br />

supernatant in the flask. This will allow better lyophilization of the samples.<br />

6. An alternate method for desalting is dialysis. Concentrated supernatant broth<br />

can be dialyzed against water or any appropriate buffer at 4°C.<br />

7. An alternate protocol for precipitation of fungal secreted proteins by<br />

trichloroacetic acid has been described by Suárez et al. (2005) (14). In this<br />

protocol, the concentrated, dialyzed <strong>and</strong> lyophilized culture filtrate is resuspended<br />

in 20% TCA in acetone containing 0.2% dithiothreitol (DTT), stored at<br />

–20°C. The suspension is kept at –20°C overnight. The sample is centrifuged at<br />

16,000g at 4 C for 10 min <strong>and</strong> the resulting pellet is washed three times with<br />

acetone containing 0.2% DTT. The supernatants are removed by centrifugation<br />

<strong>and</strong> the pellet is dried overnight at room temperature. The pellet is dissolved<br />

in SDS-<strong>PAGE</strong> or 2-DE sample buffer before gel electrophoresis analysis (see<br />

Subheading 3.5). In this protocol, acetone is used to increase the solubility of<br />

interfering organic compounds <strong>and</strong> increase protein precipitation <strong>and</strong> DTT is<br />

included to prevent protein modification.<br />

8. TCA precipitation allows for further concentration of the proteins in the sample,<br />

as well as for removal of non-protein substances, salts <strong>and</strong> other agents that may<br />

interfere with electrophoresis separation. Care should be taken to make sure the<br />

protein pellet is completely dry before adding sample buffer, as any leftover<br />

TCA will turn the Bromophenol Blue in the sample buffer yellow.<br />

9. The majority of the proteins secreted by filamentous fungi can have carbohydrate<br />

contents reaching up to 50% of the total molecular weight of the protein (1).<br />

This heavy glycosylation is thought to be responsible for the tendency of these<br />

proteins to show “smearing” on SDS-<strong>PAGE</strong> gels (12). Enzymatic protein deglycosylation<br />

using PNGase F or chemical deglycosylation using TFMS acid will<br />

help to obtain better resolution of the protein b<strong>and</strong>s on the SDS-<strong>PAGE</strong> <strong>and</strong> 2-DE<br />

gels. Deglycosylation can also aid in identification of proteins by MALDI-MS<br />

by obtaining better-resolved peaks in the mass spectra.<br />

10. The preparation of samples for gel electrophoresis requires prior knowledge<br />

of protein concentration to determine the amount of protein to be loaded. The<br />

protein concentration in the concentrated supernatant broths can be determined<br />

according to the method of Bradford (15) using the Bio-Rad Protein Assay<br />

Reagent (Bio-Rad Laboratories, Inc., Hercules, CA), or any other commercially<br />

available kit. As fungal culture broths may contain phenolic compounds, it is<br />

important to note that many of these compounds interfere with several of the<br />

most popular methods for protein determination. It is suggested that protein<br />

concentration is determined following protein precipitation <strong>and</strong> resolubilization<br />

in SDS Reducing Buffer or 2-DE sample buffer. Unfortunately, several components<br />

of these buffers may also cause problems in the assessment of protein<br />

concentration. Two commercially available protein determination kits that can<br />

be used with samples prepared for electrophoresis techniques are 2-D Quant


284 Medina <strong>and</strong> Francisco<br />

Kit (GE Healthcare Bio-Sciences Corp., Piscataway, NJ) <strong>and</strong> Advanced Protein<br />

Assay (Cytoskeleton, Inc., Denver, CO).<br />

11. Resolubilization of the precipitated protein pellet in 2-DE buffer may require<br />

vortexing <strong>and</strong>/or sonication. It has been recently shown that adding 20–30 μL<br />

of 0.2M NaOH to the TCA precipitated pellet for 2 min, before adding the<br />

solubilization buffer, increases the amount of soluble protein in the sample<br />

buffer (16).<br />

Acknowledgments<br />

The techniques described here were adapted from the appropriate original<br />

papers. This work was supported by NSF grant MCB-0317126 to W.A.F.<br />

M.L.M. was supported in part by a fellowship through the Research Training<br />

Group in Optical Biomolecular Devices provided under NSF grant DBI-<br />

9602258-003 <strong>and</strong> the NSF-funded MGE@MSA Program, an Alliance for<br />

Graduate Education <strong>and</strong> the Professoriate, headquartered at Arizona State<br />

University.<br />

References<br />

1. Peberdy, J. F. (1994) Protein secretion in filamentous fungi - trying to underst<strong>and</strong><br />

a highly productive black-box, Trends Biotechnol. 12, 50–7.<br />

2. Conesa, A., Punt, P. J., van Luijk, N., <strong>and</strong> van den Hondel, C. A. A. J. J. (2001)<br />

The secretion pathway in filamentous fungi: A biotechnological view, Fungal.<br />

Genet. Biol. 33, 155–71.<br />

3. Archer, D. B. <strong>and</strong> Peberdy, J. F. (1997) The molecular biology of secreted enzyme<br />

production by fungi, Crit. Rev. Biotechnol. 17, 273–306.<br />

4. Wallis, G. L. F., Swift, R. J., Hemming, F. W., Trinci, A. P. J., <strong>and</strong> Peberdy, J. F.<br />

(1999) Glucoamylase overexpression <strong>and</strong> secretion in Aspergillus niger: analysis<br />

of glycosylation, BBA-Gen. Subjects 1472, 576–86.<br />

5. Medina, M. L., Haynes, P. A., Breci, L., <strong>and</strong> Francisco, W. A. (2005) Analysis of<br />

secreted proteins from Aspergillus flavus, Proteomics 5, 3153–61.<br />

6. Medina, M. L., Kiernan, U. A., <strong>and</strong> Francisco, W. A. (2004) Proteomic analysis of<br />

rutin-induced secreted proteins from Aspergillus flavus, Fungal. Genet. Biol. 41,<br />

327–335.<br />

7. Zhu, L. Y., Nguyen, C. H., Sato, T., <strong>and</strong> Takeuchi, M. (2004) Analysis of secreted<br />

proteins during conidial germination of Aspergillus oryzae RIB40, Biosci. Biotech.<br />

Bioch. 68, 2607–12.<br />

8. Phalip, V., Delal<strong>and</strong>e, F., Carapito, C., et al. (2005) Diversity of the exoproteome<br />

of Fusarium graminearum grown on plant cell wall, Curr. Genet. 48, 366–79.<br />

9. Zorn, H., Peters, T., Nimtz, M., <strong>and</strong> Berger, R. G. (2005) The secretome of<br />

Pleurotus sapidus, Proteomics 5, 4832–38.<br />

10. Keller, N. P. <strong>and</strong> Hohn, T. M. (1997) Metabolic pathway gene clusters in<br />

filamentous fungi, Fungal. Genet. Biol. 21, 17–29.


Isolation of Secreted Proteins from Filamentous Fungi 285<br />

11. Semova, N., Storms, R., John, T., et al. (2006) Generation, annotation, <strong>and</strong> analysis<br />

of an extensive Aspergillus niger EST collection, BMC Microbiol. 6:7.<br />

12. Fryksdale, B. G., Jedrzejewski, P. T., Wong, D. L., Gaertner, A. L., <strong>and</strong> Miller, B. S.<br />

(2002) Impact of deglycosylation methods on two-dimensional gel electrophoresis<br />

<strong>and</strong> matrix assisted laser desorption/ionization-time of flight-mass spectrometry<br />

for proteomic analysis, Electrophoresis 23, 2184–93.<br />

13. Edge, A. S. B., Faltynek, C. R., Hof, L., Reichert, L. E., <strong>and</strong> Weber, P. (1981)<br />

Deglycosylation of glycoproteins by trifluoromethanesulfonic acid, Anal. Biochem.<br />

118, 131–7.<br />

14. Suarez, M. B., Sanz, L., Chamorro, M. I., et al. (2005) Proteomic analysis of<br />

secreted proteins from Trichoderma harzianum - Identification of a fungal cell<br />

wall-induced aspartic protease, Fungal Genet. Biol. 42, 924–34.<br />

15. Bradford, M. M. (1976) Rapid <strong>and</strong> sensitive method for quantitation of microgram<br />

quantities of protein utilizing principle of protein-dye binding, Anal. Biochem. 72,<br />

248–54.<br />

16. N<strong>and</strong>akumar, M. P., Shen, J., Raman, B., <strong>and</strong> Marten, M. R. (2003) Solubilization<br />

of trichloroacetic acid (TCA) precipitated microbial proteins via NaOH for<br />

two-dimensional electrophoresis, J. Proteome Res. 2, 89–93.


23<br />

Isolation <strong>and</strong> Solubilization of Cellular Membrane<br />

Proteins from Bacteria<br />

Kheir Zuobi-Hasona <strong>and</strong> L. Jeannine Brady<br />

Summary<br />

Membrane proteins are rarely identified in two-dimensional electrophoretic (2-DE)<br />

proteomics maps. This is because of low abundance, poor solubility, <strong>and</strong> inherent<br />

hydrophobicity. In this study, membrane preparations from the Gram-positive bacterium<br />

Streptococcus mutans were isolated from protoplasts <strong>and</strong> by mechanical grinding.<br />

Membrane proteins were extracted using a mixture of trifluroethanol <strong>and</strong> chloroform,<br />

solubilized using highly chaotropic buffer containing ASB-14 <strong>and</strong> Triton X-100 <strong>and</strong><br />

subjected to two-dimensional gel electrophoresis.<br />

Key Words: Gram-positive; membrane proteins; solubilization; streptococcus<br />

mutans; two-dimensional gel electrophoresis.<br />

1. Introduction<br />

Membrane proteins are notably limited in proteomic analysis of bacteria<br />

(1–4). The primary reason is their intrinsically hydrophobic nature leading to<br />

poor solubility (1,5). This hydrophobicity likely causes self-aggregation during<br />

isoelectric focusing leading to poor resolution <strong>and</strong> streaking in 2-D gels. Many<br />

approaches have been reported to improve recovery of hydrophobic proteins<br />

including fractionation for enrichment of proteins of interest <strong>and</strong> reduction of<br />

sample complexity (6,7), experimenting with different detergents to extract <strong>and</strong><br />

solubilize these proteins (8–10), <strong>and</strong> extraction using organic solvents (11–13).<br />

While extraction of proteins from eukaryotic membranes <strong>and</strong> analysis by 2-<br />

D electrophoresis has been technically feasible, such an approach to analyze<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

287


288 Zuobi-Hasona <strong>and</strong> Brady<br />

bacterial membrane proteins, particularly those from Gram-positive organisms<br />

has proved much more challenging. In this study membrane-associated proteins<br />

of the Gram-positive bacterium Streptococcus mutans were prepared using two<br />

techniques, one by mechanical grinding (14) <strong>and</strong> the other from protoplasts (15,<br />

16). Both methods involve differential centrifugation to enrich for membranes<br />

within the preparation. The membrane associated proteins were extracted using<br />

a mixture of trifluroethanol <strong>and</strong> chloroform (13). This treatment produced three<br />

separate phases: An upper aqueous phase containing most of the proteins, an<br />

insoluble interphase containing few proteins <strong>and</strong> a lower chloroformic phase<br />

containing low concentration of proteins with low molecular weight (17). The<br />

extracted proteins were solubilized using highly chaotropic buffer containing<br />

ASB-14 <strong>and</strong> Triton X-100 <strong>and</strong> subjected to two-dimensional gel electrophoresis<br />

(Fig. 1). These techniques were efficient <strong>and</strong> highly reproducible for analysis<br />

of membrane proteins in S. mutans <strong>and</strong> suggest the potential use for other<br />

streptococci <strong>and</strong> Gram-positive bacteria in general.<br />

2. Materials<br />

2.1. Cell Culture <strong>and</strong> Lysis<br />

1. UA-159, Streptococcus mutans, wild-type strain.<br />

2. Todd-Hewitt broth supplemented with 0.3% yeast extract (THYE).<br />

3. TM buffer: 50 mM maleate buffer, pH 6.0, containing 20 mM MgCl2. 4. Protease inhibitor cocktail (Sigma), reconstitute with DMSO <strong>and</strong> water, aliquot<br />

<strong>and</strong> store at –20°C.<br />

5. Alumina Powder (A-5, Sigma).<br />

6. Prechilled mortar <strong>and</strong> pestle (–20°C).<br />

7. Wash buffer: 20 mM Tris-HCl, pH 6.8, containing 1 mM MgCl2 <strong>and</strong> protease<br />

inhibitor cocktail at a 1:200 v/v ratio.<br />

2.2. <strong>Preparation</strong> of Membranes from Protoplasts<br />

1. Phosphate-buffered saline (PBS).<br />

2. Buffer A: 20% sucrose, 20 mM Tris-HCl, pH 7.0, <strong>and</strong> 10 mM MgCl2. Store<br />

at 4°C.<br />

2. Mutanolysin (INC Corp), store at -20°C.<br />

3. Lysozyme (Sigma), store at –20°C.<br />

4. Buffer B: 10 mM Tris-HCl, pH 8.1, 50 mM MgCl2 <strong>and</strong> 10 mM Glucose. Store<br />

at 4°C.<br />

5. Buffer C: 10 mM Tris-HCl, pH 8.1, 50 mM NaCl <strong>and</strong> 20 mM MgCl2. Store<br />

at room temperature.<br />

6. Buffer D: 20 mM Tris-HCl, pH 7.2, <strong>and</strong> 10 mM MgCl2


Isolation <strong>and</strong> Solubilization of Cellular Membrane Proteins 289<br />

2.3. Trifluroethanol/Chloroform Extraction <strong>and</strong> Analysis<br />

1. 50 mM ammonium bicarbonate pH 11.0, store at 4°C.<br />

2. Protease inhibitor cocktail (Sigma).<br />

3. Trifluroethanol/chloroform mixture (2:1 v/v).<br />

4. Ready Prep 2-D Cleanup Kit (Bio-Rad).<br />

5. Solubilization buffer: 7M urea, 2M thiourea, 2% Triton X-100, 0.5% ASB-14,<br />

50 mM dithiothreitol (DTT) <strong>and</strong> 0.2% Bio-Lytes pH 3–10. Aliquot <strong>and</strong> store<br />

at –70°C.<br />

6. RC DC protein assay kit (Bio-Rad).<br />

7. St<strong>and</strong>ard electrophoresis equipment from Bio-Rad: PROTEAN IEF Cell, Criterion<br />

Cell.<br />

3. Methods<br />

3.1. Cell Culture <strong>and</strong> Lysis<br />

1. Transfer 100 mL of overnight culture of Streptococcus mutans, strain UA159<br />

into 2 L of prewarmed Todd-Hewitt broth supplemented with 0.3% yeast extract.<br />

(THYE).<br />

2. Incubate at 37°C with gentle agitation, until an absorbance reading of 0.7 at 600<br />

nm is reached.<br />

3. Place the culture immediately on ice for 20 min.<br />

4. Harvest the cells by centrifugation at 12,000g for 10 min at 4°C.<br />

5. Wash the cell pellet twice in TM buffer containing protease inhibitor at 1:30 v/v<br />

ratio.<br />

6. Resuspend the cell pellet with 6 mL of the same buffer divide into three aliquots<br />

<strong>and</strong> store as slurries (see Note 1).<br />

7. Transfer each frozen slurry to a prechilled mortar.<br />

8. Add 3-g Alumina powder (A-5, Sigma). (see Note 2)<br />

9. Use a prechilled pestle to grind the cells for 20 min.<br />

10. Transfer the mixture into a 15 mL conical tube <strong>and</strong> centrifuge at 3,000g for 5<br />

min at 4°C to remove the Alumina.<br />

11. Centrifuge the supernatant at 16,000g for 10 min at 4°C to remove intact cells<br />

<strong>and</strong> cell debris. Discard the pellet.<br />

12. Ultracentrifuge the supernatant at 100,000g for 18 h at 4°C to pellet the<br />

membranes.<br />

13. Rinse the pellet three times with wash buffer (total volume 15 mL).<br />

14. Resuspend the pellet in 5 ml wash buffer <strong>and</strong> centrifuge at 45,000g for5h<br />

at 4°C.<br />

15. Remove the supernatant by pipette; it represents the cell cytoplasm <strong>and</strong> store<br />

at –70°C.<br />

16. Keep the pellet in the centrifuge tube covered with parafilm, place in a box <strong>and</strong><br />

store at –70°C until used. This represents the membrane-containing fraction.


290 Zuobi-Hasona <strong>and</strong> Brady<br />

(A)<br />

(B)<br />

Fig. 1. Silver stained 2-D gel of Streptococcus mutans. (A) Membrane proteins<br />

obtained using the grinding method. (B) Cytoplasmic proteins. The 2-D gel profile<br />

of cytoplasmic proteins showed an entirely different distribution of protein spots as<br />

compared with the gel of extracted membrane proteins. Nanoelectrospray quadrupole<br />

time of flight (QTOF)-t<strong>and</strong>em mass spectrometry (MS/MS) analysis of several protein<br />

spots isolated from this gel (B) identified proteins predicted to be cytoplasmically<br />

localized, including translation elongation factor (spot #8), the 10 kDa chaperone GroES<br />

(spot #9), <strong>and</strong> ribosome recycling factor (spot #10). None of the protein spots identified


Isolation <strong>and</strong> Solubilization of Cellular Membrane Proteins 291<br />

3.2. <strong>Preparation</strong> of Membranes from Protoplasts<br />

1. Transfer 100 ml of overnight culture of Streptococcus mutans, strain UA159<br />

into 2 L of prewarmed Todd-Hewitt broth supplemented with 0.3% yeast extract<br />

(THYE).<br />

2. Incubate at 37°C with gentle agitation, until an absorbance reading of 0.7 at<br />

600 nm is reached.<br />

3. Harvest the cells by centrifugation at 12,000g for 10 min at 4°C.<br />

4. Wash the pellet with phosphate-buffered saline (PBS).<br />

5. Re-suspend the cells with 50 mL Buffer A containing 150 μL protease inhibitor<br />

cocktail, 1 mg mutanolysin <strong>and</strong> 18 mg lysozyme. Swirl gently.<br />

6. Incubate the mixture for additional 3hat37°C, swirling gently every 10 min.<br />

7. Examine by light microscope under high power (see Note 3).<br />

8. Collect the protoplast at 12,000g for 10 min at 4°C. Store supernatant at –20°C.<br />

8. Wash the protoplasts with 30 mL Buffer B.<br />

10. Add 100 μL protease inhibitor cocktail <strong>and</strong> passage twice using French Press<br />

under 10,000 psi.<br />

11. Remove debris <strong>and</strong> unbroken protoplasts by centrifugation at 6,000g for 10 min.<br />

at 4°C.<br />

12. Ultra-centrifuge the supernatant for 30 min. at 45,000g at 4°C.<br />

13. Label the supernatant as cytoplasmic proteins <strong>and</strong> store at –70°C.<br />

14. Resuspend the pellet in 3 mL Buffer C at room temperature.<br />

15. Ultracentrifuge at 100,000g for 45 min at 4°C.<br />

16. Decant the supernatant <strong>and</strong> wash the pellet twice with 2 mL Buffer D <strong>and</strong> decant<br />

the wash.<br />

17. Store the pellet at –70°C until used.<br />

3.3. Trifluoroethanol/Chloroform Extraction <strong>and</strong> Analysis<br />

1. Re-suspend each membrane pellet (from Sections 3.1 <strong>and</strong> 3.2) in 150 μL of<br />

50 mM ammonium bicarbonate (pH 11), containing 15 μL protease inhibitor<br />

cocktail <strong>and</strong> vortex well for one minute.<br />

2. Add 1 mL of trifluroethanol/chloroform mixture (2:1 v/v) <strong>and</strong> vortex well for<br />

one minute.<br />

3. Maintain the mixture on ice for 1 h <strong>and</strong> vortex every 5 min for 10 s each time.<br />

◭<br />

Fig. 1. (Continued) from membrane extracts were observed in the 2-D gel of<br />

cytoplasmic proteins. On the other h<strong>and</strong> QTOF/MS/MS analysis of several spots<br />

isolated from gel (A) revealed known membrane <strong>and</strong> surface-associated proteins,<br />

including Enolase (spot #1), Biotin carboxyl carrier protein (spot #2), LemA-like protein<br />

(spot #3), Glucose 6-phosphate isomerase (spot #4), Phosphoglycerate kinase (spot<br />

#5), Glyeraldehyde 3-phosphate dehydrogenase (spot #6) <strong>and</strong> 50S ribosomal protein<br />

L7/L12 (spot #7).


292 Zuobi-Hasona <strong>and</strong> Brady<br />

4. Centrifuge at 10,000g for 5 min. at 4°C to separate into three phases.<br />

5. Carefully transfer the aqueous upper phase <strong>and</strong> the chloroformic lower phase<br />

into separate microcentifuge tubes. Keep the middle phase in the original microcentrifuge.<br />

6. Dry using vacuum centrifuge for 2 h.<br />

7. Solubilize each residue with 200 μL solubilization buffer, vortex for 1 min; let<br />

it set at RT then repeat vortexing twice.<br />

8. Precipitate membrane proteins with the Bio-Rad clean-up kit (see Note 4).<br />

9. Re-suspend the resulting pellets in solubilization buffer (from step 7), vortex<br />

well as in step 7.<br />

10. Centrifuge at 13,000g for 5 min at room temperature (see Note 5).<br />

11. Measure the protein concentration using RC DC protein assay <strong>and</strong> adjust to<br />

∼250 μg/mL solubilization buffer from step 7 (see Note 6).<br />

12. Analyze membrane fraction by st<strong>and</strong>ard two-dimensional electrophoresis<br />

protocols <strong>and</strong> silver stain the resulting 2-D gels.<br />

4. Notes<br />

1. Passage the cells through a syringe few times. The slurries can be placed in<br />

weighing boats wrapped, <strong>and</strong> stored frozen at –70°C until needed to be used.<br />

2. Use mask to avoid inhaling the Alumina, it’s recommended to do the grinding in<br />

an ice pocket to avoid protein degradation.<br />

3. More than 90% of cell wall digestion occurs after 3hofincubation. Continuous<br />

gentle agitation can speed the process.<br />

4. This step should be done using centrifuge at 4°C. Avoid over-drying the pellets,<br />

as it will be difficult to resuspend, resulting in loss of certain proteins.<br />

5. It is necessary to clarify the protein sample from insoluble particles that might<br />

cause streaking in <strong>2D</strong> gel. The supernatant can be used directly for isoelectricfocusing<br />

(IEF) in IPG strips. Store any remaining protein sample at –70°C for<br />

later analysis.<br />

6. RC DC Protein Assay (Bio-Rad cat # 500-0121) or Plus One 2-D Kit (Amersham)<br />

can be used for protein quantitation. Dilute as necessary with solubilization buffer<br />

to yield the desired quantity of protein (50–100 μg, when using silver stain).<br />

References<br />

1. Santoni, V., Molloy, M. P. <strong>and</strong> Rabilloud, T. (2000) Membrane proteins <strong>and</strong><br />

proteomics: un amour impossible? Electrophoresis 21, 1054–70.<br />

2. Molloy, M. P., Herbert, B. R., Slade, M. B., Rabilloud, T., et al. (2000) Proteomics<br />

analysis of the Escherichia coli outer membrane. Eur. J. Biochem. 267, 2871–88.<br />

3. Nouwens, A. S., Cordwell, S. J. Larsen, M. R., Molloy, M. P. et al. (2000) Complementing<br />

genomics with proteomics: the membrane subproteome of Pseudomonas<br />

aeruginosa PAO1. Electrophoresis, 21, 3797–3809.


Isolation <strong>and</strong> Solubilization of Cellular Membrane Proteins 293<br />

4. Wilkins, M. R., Gasteiger, E., Sanchez, J. –C., Bairoch, A. <strong>and</strong> Hochstrasser, D. F.<br />

(1998) Two-dimensional gel electrophoresis for proteome projects: the effect of<br />

protein hydrophobicity <strong>and</strong> copy number. Electrophoresis , 19, 1501–05.<br />

5. Pasquali, C. Fialka, I., <strong>and</strong> Huber, L. A. (1997) Preparative two-dimensional gel<br />

electrophoresis of membrane proteins. Electrophoresis 18, 2573–81.<br />

6. Molloy, M. P., Herbert, B. R., Walsh, B. J., Tyler, M. I., et al. (1998) Extraction<br />

of membrane proteins by differential solubilization for separation using twodimensional<br />

gel electrophoresis. Electrophoresis 19, 2573–81.<br />

7. Lehner, I., Niehot, M., <strong>and</strong> Borlak, J., (2003) An optimized method for the isolation<br />

<strong>and</strong> identification of membrane proteins. Electrophoresis 24, 1795–1808.<br />

8. Rabilloud, T., Blisnick, T., Heller, M., Luche, S., et al. (1999) Analysis of<br />

membrane proteins by two-dimensional electrophoresis: comparison of the proteins<br />

extracted from normal or Plasmodium faciparum-infected erythrocyte ghosts.<br />

Electrophoresis 20, 3603–10.<br />

9. Wissing, J., Heim, S., Flohe, L., Bilitewski, U., <strong>and</strong> Frank, R. (2000) Enrichment<br />

of hydrophobic proteins via Triton X-114 phase partitioning <strong>and</strong> hydroxyapatite<br />

column chromatography for mass spectrometry. Electrophoresis 21, 2589–93.<br />

10. Wasinger, V. C., Pollack, J. B., <strong>and</strong> Humphery-Smith, I., (2000) The proteome<br />

of Mycoplasma genitalium. Chaps-soluble component. Eur. J. Biochem. 267,<br />

1571–82.<br />

11. Molloy, M. P., Herbert, B., Williams, K. L., <strong>and</strong> Gooley, A. A., (1999) Extraction<br />

of Escherichia coli proteins with organic solvents before two-dimensional<br />

electrophoresis. Electrophoresis 20, 701–4.<br />

12. Seigneurin-Berny, D., Roll<strong>and</strong>, N., Garin, J., <strong>and</strong> Joyard, J., (1999) Technical<br />

Advance: Differential extraction of hydrophobic proteins from chloroplast envelope<br />

membranes: a subcellular specific proteomic approach to identify rare intrinsic<br />

membrane proteins. Plant. J. 19, 217–228.<br />

13. Deshusses, J. M. P., Burgess, J. A., Scherl, A., Wenger, Y., Walter, N., et al. (2003)<br />

Exploitation of specific properties of trifluroethanol for extraction <strong>and</strong> separation<br />

of membrane proteins. Proteomics, 3, 1418–24.<br />

14. Vadeboncoeur, C., St Martin, S., Brochu, D., <strong>and</strong> Hamilton, I. R., (1991) Effect of<br />

growth rate <strong>and</strong> pH on intracellular levels <strong>and</strong> activities of the components of the<br />

phosphoenolpyruvate: sugar phosphotransferase system in streptococcus mutans<br />

Ingbritt. Infect. Immun. 59, 900–6.<br />

15. Wessel, D. <strong>and</strong> Flugge, U. I., (1984) A method for the quantitative recovery of<br />

protein in dilute solution in the presence of detergents <strong>and</strong> lipids. Anal. Biochem.<br />

138, 141–3.<br />

16. Bordier, C. (1981) Phase separation of integral membrane proteins in Triton X-114<br />

solution. J. Biol. Chem. 256, 1604–7.<br />

17. Zuobi-Hasona, K., Crowley, P. J., Hasona, A., Bleiweis, A. S., <strong>and</strong> Brady, L. J.<br />

(2005) Solubilization of cell membrane proteins from Streptococcus mutans for<br />

two-dimensional gel electrophoresis. Electrophoresis 26, 1200–6.


24<br />

Isolation <strong>and</strong> Solubilization of Gram-Positive Bacterial<br />

Cell Wall-Associated Proteins<br />

Jason N. Cole, Steven P. Djordjevic, <strong>and</strong> Mark J. Walker<br />

Summary<br />

This chapter describes a simple, rapid <strong>and</strong> reproducible method to prepare bacterial cell<br />

wall extracts for two-dimensional gel electrophoresis (<strong>2D</strong>E). The extraction process uses<br />

mutanolysin, an N-acetylmuramidase, to gently solubilize cell wall-associated proteins<br />

from Gram-positive prokaryotes. The cells are first washed with buffer <strong>and</strong> resuspended in<br />

a solution containing mutanolysin. Following incubation at 37 °C, the sample is centrifuged<br />

<strong>and</strong> the supernatant containing the soluble cell wall-associated proteins is harvested.<br />

Following a brief precipitation step, the pellet is solubilized in sample buffer ready for<br />

isoelectric focusing <strong>and</strong> <strong>2D</strong>E analysis.<br />

Key Words: Bacterial proteome; cell wall-associated; Gram-positive; group A streptococcus;<br />

mutanolysin; Streptococcus pyogenes; two-dimensional gel electrophoresis.<br />

1. Introduction<br />

Gram-positive bacteria are bounded by a thick cell wall composed of<br />

primarily of peptidoglycan, a large macromolecule of acetamido sugars <strong>and</strong><br />

amino acids (1). The glycan chains of peptidoglycan consist of alternating<br />

units of N-acetylglucosamine (GlcNAc) <strong>and</strong> N-acetylmuramic acid (MurNAc)<br />

in -1,4 linkage (2). Short cell wall peptides are cross-linked to the glycan<br />

chains to form a three-dimensional molecular network. The major function of<br />

the cell wall envelope is to provide a rigid exoskeleton for protection against<br />

mechanical <strong>and</strong> osmotic lysis. The cell wall also maintains a defined cell shape<br />

<strong>and</strong> plays an integral role in the anchoring of proteins to the cell surface (3,4).<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> <strong>Fractionation</strong>, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

295


296 Cole et al.<br />

Most Gram-positive cell walls are resistant to dissolution with lysozyme (5).<br />

Mutanolysin, a muralytic enzyme derived from Streptomyces globisporus 1829,<br />

cleaves the -1,4 linkage of the N-acetylmuramyl-N-acetylglucosamine in the<br />

glycan backbone of the peptidoglycan-polysaccharide polymer, which is highly<br />

conserved among bacterial species (6). Although mutanolysin has the same<br />

specificity as lysozyme, it has a wider range of activity among peptidoglycanpolysaccharide<br />

from bacterial strains resistant to lysozyme, including Streptococcus<br />

pyogenes (group A streptococcus), a Gram-positive pathogen responsible<br />

for numerous human diseases.<br />

In this chapter, the authors describe a convenient <strong>and</strong> efficient mutanolysin<br />

extraction method for the solubilization of cell wall-associated proteins from<br />

S. pyogenes. A detailed discussion is presented on bacterial growth conditions,<br />

cell wall purification, preparative SDS-<strong>PAGE</strong> analysis, sample preparation,<br />

isoelectric focusing <strong>and</strong> <strong>2D</strong>E.<br />

2. Materials<br />

Unless stated otherwise, all solutions were prepared with glass distilled<br />

water <strong>and</strong> high purity electrophoresis grade reagents. Review the manufacturers’<br />

safety data sheets (MSDS) <strong>and</strong> follow the safety precautions <strong>and</strong> general<br />

h<strong>and</strong>ling procedures for hazardous materials.<br />

2.1. Bacterial Culture<br />

1. Horse blood agar plates (BioMérieux). Store at 4 °C.<br />

2. THBY medium: 30 g/L Todd-Hewitt broth, 10 g/L yeast extract (Difco).<br />

Autoclave <strong>and</strong> store at room temperature.<br />

2.2. Extraction of Cell Wall-Associated Proteins<br />

1. TE buffer: 50 mM Tris-HCl, 1 mM EDTA, pH 8.0. Autoclave <strong>and</strong> store at 4 °C.<br />

2. TE-Sucrose (TES) buffer: 20% (w/v) sucrose in TE buffer (pH 8.0). Autoclave<br />

<strong>and</strong> store at 4 °C.<br />

3. Phenylmethylsulfonyl fluoride (PMSF, Sigma) dissolved at 1 mM in chilled TES<br />

buffer (see Note 1). PMSF is very toxic <strong>and</strong> rapidly degrades in aqueous solutions.<br />

Prepare immediately before use <strong>and</strong> place on ice.<br />

4. Mutanolysin: Resuspend 10,000 units of chromatographically purified mutanolysin<br />

from Streptomyces globisporus ATCC 21553 (Sigma) in 2 mL of chilled filtersterilized<br />

0.1 M K2HPO4 (pH 6.2) for a working solution of 5,000 units/mL. Store<br />

200 μL aliquots at –20 °C.<br />

5. Lysozyme dissolved in chilled TES buffer at 100 mg/mL. Avoid foaming by<br />

gently pipeting up <strong>and</strong> down. Prepare fresh each time <strong>and</strong> store on ice before use.


Isolation of Gram-Positive Bacterial Cell Wall Proteins 297<br />

6. Mutanolysin mix: 1 mL TES buffer, 100 μL lysozyme (100 mg/mL in TES),<br />

50 μL mutanolysin (5,000 U/mL in 0.1 M K 2HPO 4, pH 6.2). Prepare immediately<br />

before use <strong>and</strong> place on ice.<br />

2.3. Bicinchoninic Acid (BCA) Protein Assay<br />

1. Bicinchoninic Acid Protein Assay Kit (Sigma): Store Reagents A <strong>and</strong> B at room<br />

temperature.<br />

2. Bovine serum albumin (BSA, Sigma).<br />

3. TES buffer: 20% (w/v) sucrose in TE buffer (50 mM Tris-HCl, 1 mM EDTA, pH<br />

8.0). Autoclave <strong>and</strong> store at 4 °C.<br />

4. Microtiter plate reader capable of measuring absorbance in the 560 nm region.<br />

5. Microtiter plate <strong>and</strong> sealing film.<br />

2.4. SDS-Polyacrylamide Gel Electrophoresis (SDS-<strong>PAGE</strong>)<br />

1. 0.5 M Tris-HCl (pH 6.8) <strong>and</strong> 1.5 M Tris-HCl (pH 8.8) prepared in Milli-Q ®<br />

water. Adjust pH with concentrated HCl. Store at 4 °C.<br />

2. 10% (w/v) SDS dissolved in Milli-Q ® water. Store at room temperature.<br />

3. 40% acrylamide/bis solution (37.5:1, Amresco). Acrylamide monomer is a<br />

neurotoxin <strong>and</strong> suspected carcinogen; care should be taken to avoid exposure.<br />

Store in dark at 4 °C.<br />

4. N,N,N ′ ,N ′ -Tetramethylethylenediamine (TEMED, Amresco). Store at room<br />

temperature.<br />

5. Ammonium persulfate (APS, Amresco) prepared at 10% (w/v) in Milli-Q ® water<br />

(see Note 2).<br />

6. Gel overlay solution: 70% (v/v) ethanol. Store at room temperature.<br />

7. 10× running buffer: 250 mM Tris-HCl, 1.92 M glycine, 1% (w/v) SDS, pH 8.3.<br />

Do not adjust pH with acid or base. Dilute to 1× working solution <strong>and</strong> mix<br />

thoroughly before use. Store at room temperature.<br />

8. 1 M Dithiothreitol (DTT) dissolved in 0.01 M sodium acetate (pH 5.2). Filtersterilize<br />

<strong>and</strong> store 1 mL aliquots at –20 °C.<br />

9. 5× loading buffer: 225 mM Tris-HCl (pH 6.8), 50% (v/v) glycerol, 5% (w/v)<br />

SDS, 0.05% (w/v) Bromophenol Blue, 250 mM DTT (see Note 3). Store at<br />

room temperature. Dilute sample 1:5 with loading buffer <strong>and</strong> heat at 95 °C for<br />

10 min.<br />

10. Molecular weight markers: PageRuler Protein Ladder (Fermentas). Store<br />

at –20 °C.<br />

11. Coomassie Blue staining solution: 0.2% (w/v) Coomassie Blue R250, 40%<br />

(v/v) methanol, 10% (v/v) acetic acid. Store at room temperature.<br />

12. Rapid destain solution: 40% (v/v) methanol, 10% (v/v) acetic acid. Store at room<br />

temperature.<br />

13. Final destain solution: 4% (v/v) glycerol, 10% (v/v) acetic acid. Store at room<br />

temperature.


298 Cole et al.<br />

2.5. Trichloroacetic Acid (TCA) Precipitation<br />

1. 10% (v/v) TCA (Sigma): Protect from light <strong>and</strong> store at room temperature. TCA<br />

is a highly corrosive acid. Use safety equipment when h<strong>and</strong>ling to avoid exposure.<br />

2. Ethanol 100% (v/v) analytical reagent stored at –20 °C.<br />

3. <strong>Sample</strong> solubilization solution: 8 M urea, 100 mM DTT, 4% (w/v) CHAPS, 0.8%<br />

(v/v) Bio-Lyte ® 3/10 ampholyte (Bio-Rad), 40 mM Tris-HCl. Make up to volume<br />

with Milli-Q ® water (see Note 4). Store 1 mL aliquots at –20 °C. Thaw required<br />

number of aliquots <strong>and</strong> discard leftover solution.<br />

2.6. Two-Dimensional (<strong>2D</strong>) Gel Electrophoresis<br />

2.6.1. <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> Rehydration<br />

1. <strong>Sample</strong> solubilization solution: 8 M urea, 100 mM DTT, 4% (w/v) CHAPS, 0.8%<br />

(v/v) Bio-Lyte ® 3/10 ampholyte (Bio-Rad), 40 mM Tris-HCl. Make up to volume<br />

with Milli-Q ® water. Store 1 mL aliquots at –20 °C. Thaw required number of<br />

aliquots <strong>and</strong> discard leftover solution.<br />

2. Water-bath sonicator or equivalent.<br />

3. Bromophenol Blue solution 1% (w/v) in Milli-Q ® water. Pass through a 0.22-μm<br />

filter unit (Millipore) to remove particulate matter <strong>and</strong> store at room temperature.<br />

4. 11-cm ReadyStrip linear pH 4-7 IPG strips (Bio-Rad).<br />

5. Disposable plastic rehydration tray (Bio-Rad).<br />

6. Mineral oil (Bio-Rad). Store at room temperature.<br />

2.6.2. First Dimension Isoelectric Focusing (IEF)<br />

1. Electrode wicks (Bio-Rad).<br />

2. PROTEAN ® IEF Cell <strong>and</strong> 11-cm focusing tray (Bio-Rad).<br />

3. Rehydrated 11-cm ReadyStrip linear pH 4–7 IPG strip (Bio-Rad).<br />

4. Mineral oil (Bio-Rad). Store at room temperature.<br />

2.6.3. Casting <strong>2D</strong> Gels<br />

1. 1.5 M Tris-HCl (pH 8.8) prepared in Milli-Q ® water. Adjust pH with concentrated<br />

HCl. Store at 4 °C.<br />

2. 10% (w/v) SDS dissolved in Milli-Q ® water. Store at room temperature.<br />

3. 40% acrylamide/bis solution (37.5:1, Amresco). Store in dark at 4 °C.<br />

4. TEMED (Amresco). Store at room temperature.<br />

5. 10% (w/v) APS in Milli-Q ® . Prepare fresh each time.<br />

6. Water-saturated n-butanol gel overlay solution: Combine equal volumes of<br />

n-butanol <strong>and</strong> Milli-Q ® water <strong>and</strong> shake to mix. Allow to settle <strong>and</strong> use top<br />

(aqueous layer) to overlay gels. Store at room temperature.<br />

7. Gel storage solution: 0.375 M Tris-HCl (pH 8.8), 0.1% (w/v) SDS. Store at 4 °C.


Isolation of Gram-Positive Bacterial Cell Wall Proteins 299<br />

2.6.4. Second Dimension SDS-<strong>PAGE</strong><br />

1. Disposable plastic equilibration tray (Bio-Rad).<br />

2. Equilibration buffer: 6 M urea, 2% (w/v) SDS, 375 mM Tris-HCl (pH 8.8), 20%<br />

(v/v) glycerol, 2.5% (v/v) acrylamide, 130 mM DTT. Store 4 mL aliquots at<br />

–20 °C. Thaw required number of aliquots <strong>and</strong> discard leftover solution.<br />

3. Molecular weight markers: PageRuler Prestained Protein Ladder (Fermentas).<br />

Store at –20 °C.<br />

4. 10× running buffer: 250 mM Tris-HCl, 1.92 M glycine, 1% (w/v) SDS, pH 8.3.<br />

Do not adjust pH. If the pH is not accurate remake buffer. Dilute to 2× <strong>and</strong> 1×<br />

working concentrations <strong>and</strong> mix thoroughly before use. Store at room temperature.<br />

5. Bromophenol Blue solution 1% (w/v) in Milli-Q ® water. Pass through a 0.22 μm<br />

filter unit (Millipore) to remove particulate matter <strong>and</strong> store at room temperature.<br />

6. Agarose gel overlay solution: 1% (w/v) agarose in 1× running buffer (pH 8.3).<br />

Do not adjust pH. Add 0.1 mL of 1% (w/v) Bromophenol Blue per 100 mL<br />

of overlay solution <strong>and</strong> heat in a microwave oven until completely melted (see<br />

Note 5). Allow to cool slightly (60 °C) before using. Store at room temperature<br />

after use.<br />

2.6.5. Protein Detection <strong>and</strong> Gel Documentation<br />

1. Colloidal Coomassie stain: 17% (w/v) ammonium sulfate, 3% (v/v) phosphoric<br />

acid, 34% (v/v) methanol, 0.1% (w/v) Coomassie G250 (see Note 6). Store at<br />

room temperature.<br />

2. Destain solution: 1% (v/v) glacial acetic acid. Store at room temperature.<br />

3. Methods<br />

For full information pertaining to chemical <strong>and</strong> electrical hazards refer to the<br />

manufacturers’ material safety data sheets (MSDS) <strong>and</strong> instruction manuals.<br />

S. pyogenes (UN2814) is a potential human pathogen <strong>and</strong> safety precautions<br />

must be taken to avoid exposure.<br />

3.1. Bacterial Culture<br />

1. Sixteen-streak the S. pyogenes strain onto a fresh horse blood agar plate (see<br />

Note 7). Seal with Parafilm ® <strong>and</strong> incubate at 37 °C overnight (approx 16 h).<br />

2. Inoculate a single, well-isolated hemolytic colony (see Note 8) intoa5mLsterile<br />

tube containing 2 mL of THBY medium. Incubate at 37 °C overnight for 16 h<br />

without shaking.<br />

3. To a 250 mL conical flask containing 98 mL of THBY, add the entire volume<br />

of overnight culture <strong>and</strong> incubate at 37 °C without agitation until late stationary<br />

phase is reached (approx 16 h) (see Note 9).


300 Cole et al.<br />

3.2. Extraction of Cell Wall-Associated Proteins<br />

1. Transfer the overnight culture to a sterile centrifuge tube <strong>and</strong> harvest the bacterial<br />

cells by centrifugation at 7,560g for 20 min at 4 °C.<br />

2. Carefully decant the culture supernatant <strong>and</strong> discard. Place the bacterial pellet on<br />

ice for 5 min.<br />

3. Resuspend the pellet in 5 mL of chilled TE buffer containing 1 mM PMSF (see<br />

Note 10) by pipetting up <strong>and</strong> down. Take care to avoid foaming <strong>and</strong> ensure no<br />

bacterial clumps are visible. Centrifuge at 7,560g for 20 min at 4 °C <strong>and</strong> discard<br />

the supernatant.<br />

4. Repeat step 3. Resuspend the pellet in 1.15 mL of ice-cold mutanolysin mix by<br />

pipeting up <strong>and</strong> down. Take care to avoid foaming <strong>and</strong> ensure no bacterial clumps<br />

are visible.<br />

5. Transfer to a sterile microcentrifuge tube <strong>and</strong> incubate for 2hat37°Cwith<br />

shaking (200 rpm) (see Note 11).<br />

6. Centrifuge at 14,000g for 5 min at room temperature in a bench-top microcentrifuge.<br />

7. Collect the supernatant (solubilized cell wall-associated proteins) by aspiration.<br />

Store 500 μL aliquots at –20 °C (see Note 12).<br />

8. Determine the protein concentration of the cell wall extract using the BCA protein<br />

assay described below.<br />

9. Prepare for SDS-<strong>PAGE</strong> analysis by diluting 20 μL of 5× loading buffer with 80<br />

μL of cell wall extract in a microcentrifuge tube. Pierce the lid <strong>and</strong> boil for 10<br />

min. Allow to cool to room temperature before loading (see Note 13).<br />

3.3. BCA Protein Assay<br />

1. Prepare the required amount of BCA Working Reagent by mixing 50 parts<br />

of Reagent A (Sigma proprietary solution containing BCA, sodium carbonate,<br />

sodium tartrate <strong>and</strong> sodium bicarbonate in 0.1 M NaOH, pH 11.25) with 1 part<br />

of Reagent B, a 4% (w/v) solution of copper(II) sulfate pentahydrate. Mix by<br />

vortexing until the solution is a uniform light green color (see Note 14).<br />

2. Prepare BSA protein st<strong>and</strong>ards ranging from 0.2 to 1 mg/mL in TES buffer (see<br />

Note 15). Include a blank containing buffer with no protein. Protein st<strong>and</strong>ards<br />

may be stored at –20 °C.<br />

3. Dilute the cell wall extract 1:5 in TES buffer to ensure the concentration is within<br />

the linear range of 0.2 to 1 mg/mL.<br />

4. Add 25 μL of each st<strong>and</strong>ard, cell wall extract <strong>and</strong> blank to a microtiter plate<br />

before adding 200 μL of Working Reagent (see Note 16).<br />

5. Seal the plate, incubate at 37 °C for 30 min <strong>and</strong> cool to room temperature (see<br />

Note 17).<br />

6. Measure the absorbance at 562 nm with a microtiter plate reader <strong>and</strong> estimate<br />

the protein concentration of unknown samples from the BSA st<strong>and</strong>ard curve (see<br />

Note 18).


Isolation of Gram-Positive Bacterial Cell Wall Proteins 301<br />

3.4. SDS-<strong>PAGE</strong><br />

This procedure is for use of the discontinuous Laemmli system (7) in a<br />

Mini-PROTEAN ® 3 Cell (Bio-Rad) <strong>and</strong> can be readily adapted for other gel<br />

formats.<br />

1. Thoroughly clean the glass plates with a laboratory detergent, rinse completely<br />

with distilled water <strong>and</strong> air dry.<br />

2. Prepare a 12% resolving gel by mixing 3 mL of 40% acrylamide/bis solution<br />

with 2.5 mL of 1.5 M Tris-HCl (pH 8.8), 4.35 mL Milli-Q ® water, 100 μL 10%<br />

(w/v) SDS, 15 μL TEMED <strong>and</strong> 50 μL 10% (w/v) APS (see Note 19). Pour a 0.75<br />

mm thick resolving gel to the required level <strong>and</strong> immediately overlay with 70%<br />

(v/v) ethanol to give a flat gel surface (see Note 20). Allow the gel to polymerize<br />

for45minto1hatroom temperature (see Note 21). The resolving gel can be<br />

submerged in 1.5 M Tris-HCl (pH 8.8) diluted 1:4 in distilled water <strong>and</strong> stored<br />

at 4 °C for up to 1 wk.<br />

3. Decant the ethanol <strong>and</strong> thoroughly rinse the gel surface with distilled water.<br />

Remove excess water between the glass plates above the resolving gel with a<br />

piece of filter paper, taking care to avoid the gel surface.<br />

4. Prepare the 4% stacking gel by mixing 0.5 mL of 40% acrylamide/bis solution<br />

with 2.5 mL of 0.5 M Tris-HCl (pH 6.8), 3.18 mL Milli-Q ® water, 50 μL 10<br />

% (w/v) SDS, 5 μL TEMED <strong>and</strong> 25 μL 10% (w/v) APS. Pour the stacking gel,<br />

insert the comb <strong>and</strong> allow the stacking gel to polymerize for 30–45 min at room<br />

temperature.<br />

5. Prepare the running buffer by diluting 100 mL of 10× running buffer with 900<br />

mL of distilled water. Mix thoroughly before use.<br />

6. After the stacking gel has set, gently remove the comb <strong>and</strong> thoroughly rinse the<br />

wells with running buffer. Place the gel into the electrophoresis module <strong>and</strong> add<br />

running buffer to the upper <strong>and</strong> lower chambers. Load 15 μL of sample (approx<br />

20 μg protein) <strong>and</strong> molecular weight markers for size comparison (see Note 22).<br />

7. Connect the lid of the electrophoresis unit to a suitable power supply <strong>and</strong> run at<br />

a constant 200 V for approximately 45 min, or until the dye front reaches the<br />

bottom of the gel (see Note 23).<br />

8. Once electrophoresis is complete, discard the running buffer <strong>and</strong> remove the<br />

stacking gel from the resolving gel. Place the resolving gel in a plastic tray <strong>and</strong><br />

submerge in Coomassie Blue staining solution (see Note 24). Cover the tray to<br />

minimize exposure to vapors, microwave (600 W for 15 s) <strong>and</strong> shake at room<br />

temperature for1horleave overnight. Decant stain (see Note 25) <strong>and</strong> add enough<br />

destain solution to completely cover the gel. Microwave for 15 s <strong>and</strong> shake for 20<br />

min at room temperature. Repeat with fresh destain solution until the background<br />

is clear (see Note 26).<br />

9. Discard destain <strong>and</strong> submerge gel in final destain solution ready for documentation<br />

with the GS-800 calibrated densitometer (Bio-Rad) or equivalent (see Note 27).<br />

A representative SDS-<strong>PAGE</strong> gel of cell wall extracts from S. pyogenes is shown<br />

in Fig. 1.


302 Cole et al.<br />

Fig. 1. Coomassie Blue stained 12% SDS-<strong>PAGE</strong> reducing gel of a mutanolysin<br />

cell wall extract harvested from S. pyogenes strain 5448 (serotype M1) after growth<br />

at 37 °C to late stationary phase in THBY medium without agitation. Molecular mass<br />

markers are given in kilo-Daltons (kDa).<br />

3.5. TCA Precipitation<br />

1. Add 1 mL of 10% (v/v) TCA to 1 mL of cell wall extract <strong>and</strong> mix immediately<br />

by vortexing at maximum output (see Note 28). Incubate on ice for 20 min.<br />

2. Centrifuge at 14,000g for 15 min at room temperature in a bench-top microcentrifuge.<br />

3. Discard supernatant <strong>and</strong> wash pellet with 1 mL of ice-cold 100% (v/v) ethanol to<br />

remove residual TCA (see Note 29).<br />

4. Centrifuge at 14,000g for 5 min at room temperature in a bench-top microcentrifuge.<br />

5. Discard the supernatant <strong>and</strong> dry the pellet for 30–60 min at room temperature.<br />

Store TCA precipitated pellets at –20 °C before <strong>2D</strong>E (see Note 30).<br />

3.6. <strong>2D</strong> Gel Electrophoresis<br />

3.6.1. <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> Rehydration<br />

1. Completely resuspend the TCA precipitated pellet in 500 μL of sample solubilization<br />

solution by pipeting up <strong>and</strong> down, taking care to avoid foaming (see<br />

Note 31).


Isolation of Gram-Positive Bacterial Cell Wall Proteins 303<br />

2. Sonicate at 14 W for 30 sec <strong>and</strong> vortex for 15 sec at room temperature to aid<br />

solubilization. Repeat this step a further 3 times (see Note 32).<br />

3. Centrifuge at 14,000g for 15 min at room temperature in a bench-top microcentrifuge<br />

(see Note 33).<br />

4. Transfer the supernatant to a new microcentrifuge tube <strong>and</strong> discard the pellet<br />

(see Note 34).<br />

5. Calculate the volume of sample required for 200 μg protein <strong>and</strong> dispense into<br />

a new microcentrifuge tube. Adjust the volume to 250 μL with sample solubilization<br />

solution (see Note 35).<br />

6. Add 2 μL of Bromophenol Blue solution <strong>and</strong> mix by briefly vortexing (see<br />

Note 36).<br />

7. Pipette 250 μL of sample along the length of a channel in an 11 cm rehydration<br />

tray. Eliminate air bubbles using forceps to ensure even distribution of the<br />

sample in the IPG strip.<br />

8. Carefully place the IPG strip onto the sample gel side down. Eliminate air<br />

bubbles under the strip by gently lifting the strip from one end with forceps.<br />

9. Carefully overlay the strip with 2 mL of mineral oil to prevent evaporation (see<br />

Note 37). Attach the rehydration tray lid <strong>and</strong> rehydrate overnight (11–16 h) at<br />

room temperature (see Note 38). The sample will be passively absorbed by the<br />

strip during rehydration.<br />

10. Remove the IPG strip from the rehydration tray with forceps (see Note 39).<br />

Remove the excess oil by holding the strip vertically <strong>and</strong> gently touching the<br />

tip of the backing strip to blotting paper (see Note 40).<br />

3.6.2. First Dimension IEF<br />

The following IEF method assumes the use of the PROTEAN ® IEF Cell<br />

(Bio-Rad) (see Note 41).<br />

1. Equilibrate four electrode wicks per IPG strip in Milli-Q ® water (see Note 42).<br />

2. Remove excess water by briefly blotting the wicks on filter paper. Place two<br />

hydrated wicks directly on top of the cathode <strong>and</strong> anode electrode wires of the<br />

focusing tray.<br />

3. Place the IPG strip gel side down onto the wicks, with the acidic (positive)<br />

end of the strip at the anode (positive) electrode of the focusing tray (see<br />

Note 43).<br />

4. Overlay the IPG strip with 2 mL of mineral oil <strong>and</strong> eliminate air bubbles under<br />

the strip by gently lifting it from one end with forceps (see Note 44). Place the lid<br />

on the focusing tray <strong>and</strong> place into the IEF Cell, ensuring good contact between<br />

the electrodes of the focusing tray <strong>and</strong> the electrodes of the IEF Cell.<br />

5. Program the IEF Cell with the following focusing conditions: 100 V for 1 h;<br />

300 V for 1 h; 600 V for 1 h; 1,000 V for 1 h; 2,000 V for 1 h; 4,000 V for<br />

40,000 Volt-hours (Vh) (approx 10 h); <strong>and</strong> 100 V hold (12 h) (see Note 45). Use<br />

an IEF Cell temperature of 20 °C with a maximum current of 50 μA per IPG<br />

strip.


304 Cole et al.<br />

6. Remove the IPG strip from the IEF tray <strong>and</strong> drain the excess mineral oil with<br />

blotting paper as described in step 10 above. Proceed directly to the equilibration<br />

step (see Note 46).<br />

3.6.3. Casting <strong>2D</strong> Gels<br />

This protocol is for the casting of homogenous polyacrylamide gels with the<br />

Ettan DALTsix Gel Caster (GE Biosciences).<br />

1. Lay the Ettan DALTsix Gel Caster on its back. Remove the faceplate <strong>and</strong> place<br />

the triangular rubber wedge in the V-shaped base.<br />

2. Place a separator sheet against the back wall of the caster (square corners against<br />

the rubber wedge) followed by a 1.0 mm gel casting cassette. Fill the caster by<br />

alternately layering separator sheets <strong>and</strong> gel cassettes, ending with a separator<br />

sheet. Add thicker filler sheets until the stack of cassettes is flush with the edge<br />

of the caster (see Note 47).<br />

3. Carefully place the faceplate onto the caster, clamp with six spring clips <strong>and</strong><br />

tighten the faceplate screws (see Note 48).<br />

4. Attach the cap to the filler port on the faceplate to prevent leakage. Place the Gel<br />

Caster in an upright position on a level surface ready for casting.<br />

5. Prepare the 12.5% gel solution by mixing 156 mL of 40% acrylamide/bis with<br />

125 mL of 1.5 M Tris-HCl (pH 8.8), 6 mL 10% (w/v) SDS <strong>and</strong> 208 mL Milli-Q ®<br />

water. Mix well with a magnetic stirrer before adding 6 mL 10% (w/v) APS <strong>and</strong><br />

100 μL TEMED (see Note 49). To cast a homogenous 1.0-mm-thick gel, add<br />

the APS followed by TEMED <strong>and</strong> slowly pour the gel solution into the filling<br />

channel located at the back of the caster. Continuing pouring until the level is<br />

approx 4 cm below the top edge of the short plate (see Note 50).<br />

6. Immediately overlay each gel with 4 mL of water-saturated n-butanol to exclude<br />

air <strong>and</strong> ensure a level surface. Cover the top of the Gel Caster with plastic wrap<br />

<strong>and</strong> allow gels to polymerize for 3–4 h at room temperature.<br />

7. Pour off the gel overlay solution <strong>and</strong> rinse the surface of each gel with Milli-Q ®<br />

water to remove residual n-butanol <strong>and</strong> unpolymerized acrylamide. Add 5 mL of<br />

gel storage solution to the top of each gel. Seal the top of the Caster with plastic<br />

wrap <strong>and</strong> store at 4 °C overnight (see Note 51).<br />

3.6.4. Second Dimension SDS-<strong>PAGE</strong><br />

The following procedure is for the running of large format (26 × 20 cm)<br />

acrylamide gels with the vertical Ettan DALTsix Electrophoresis Unit (GE<br />

Biosciences) (see Note 52).<br />

1. Turn on the MultiTemp III heat exchanger <strong>and</strong> equilibrate to 10 °C.<br />

2. Place the IPG strip gel side up into an 11 cm equilibration tray <strong>and</strong> add 2 mL of<br />

Equilibration buffer. Rock gently for 20 min at room temperature (see Note 53).


Isolation of Gram-Positive Bacterial Cell Wall Proteins 305<br />

3. During the equilibration step, rinse the glass cassettes with water to remove<br />

adhering acrylamide. Rinse the surface of each gel with Milli-Q ® water <strong>and</strong><br />

invert to drain (see Note 54).<br />

4. Remove the IPG strips from the equilibration tray <strong>and</strong> wash each side with 3<br />

mL of 1× running buffer (see Note 55).<br />

5. Hold the gel cassette in a horizontal position with the short glass plate side up.<br />

Using forceps, place the IPG strip onto the long glass plate with the plastic<br />

backing against the plate. By carefully pushing against the plastic backing of<br />

Fig. 2. Examination of the cell wall proteome of S. pyogenes strain 5448 (serotype<br />

M1). The mutanolysin cell wall extract was harvested after growth at 37 °C to late<br />

stationary phase in THBY medium without agitation. The extracts were concentrated<br />

by TCA precipitation, isoelectric focused over a pH range of 4–7 <strong>and</strong> resolved with<br />

a 12.5% SDS-<strong>PAGE</strong> gel. The gel was stained with colloidal Coomassie <strong>and</strong> several<br />

l<strong>and</strong>mark proteins identified by MALDI-TOF peptide mass fingerprinting analysis.<br />

Identified protein spots are denoted by numbered arrows, which correspond to the<br />

proteins in Table 1. Molecular mass markers are given in kilo-Daltons (kDa).


306 Cole et al.<br />

the strip with a spatula, slide the strip between the glass plates down to the gel<br />

surface. Take care not to damage the gel matrix with the spatula during this step.<br />

The strip should rest gently against the gel surface with the positive (acidic) end<br />

against the left edge of the glass plate (see Note 56).<br />

6. Apply 10 μL of prestained molecular weight markers to a piece of filter paper<br />

or electrode wick paper. Allow the markers to soak in <strong>and</strong> position on the gel<br />

surface next to one end of the IPG strip with forceps.<br />

7. Overlay the IPG strip <strong>and</strong> prestained markers with 3 mL of molten agarose gel<br />

overlay solution (see Note 57) <strong>and</strong> allow to set for 1 min.<br />

8. Insert the gels into the cassette carrier <strong>and</strong> add blank casset inserts to empty gel<br />

slots. Secure the gel cassets with the upper buffer chamber seal <strong>and</strong> place the<br />

assembly into the lower buffer chamber.<br />

9. Add approximately 4.0 L of 1× running buffer to the lower chamber <strong>and</strong> approximately<br />

0.8 L of 2× running buffer to the upper chamber. Adjust the buffer levels<br />

of both chambers until they are equal <strong>and</strong> between the minimum <strong>and</strong> maximum<br />

fill lines marked on the tank (see Note 58).<br />

10. Attach the lid of the Electrophoresis Unit <strong>and</strong> connect to a suitable power supply.<br />

Run at 2.5 W per acrylamide gel for 30 min; 40 W for 1 h; <strong>and</strong> 80 W for 4–5 h<br />

(see Note 59).<br />

3.6.5. Protein Detection <strong>and</strong> Gel Documentation<br />

1. Carefully remove the IPG strips from the top of the gel <strong>and</strong> trim the gel bottom<br />

(see Note 60).<br />

2. Submerge the gel in colloidal Coomassie stain <strong>and</strong> incubate overnight with<br />

gentle rocking (see Note 61).<br />

3. Discard the stain <strong>and</strong> rock the gel in destain solution overnight. Repeat at least<br />

once with fresh destain solution until the background is clear.<br />

4. Document gel with the GS-800 calibrated imaging densitometer (Bio-Rad) or<br />

equivalent. The <strong>2D</strong>E pattern of a representative S. pyogenes cell wall extract<br />

is presented in Fig. 2. The mutanolysin extracts are highly enriched with cell<br />

wall-associated proteins, as evidenced by the MALDI-TOF mass spectrometry<br />

identification of several well-characterized cell wall proteins of S. pyogenes<br />

(Table 1).<br />

5. Store gel at 4 °C in a sealed plastic bag containing water (short term) or water<br />

with 0.005% (w/v) sodium azide for long term storage.<br />

4. Notes<br />

1. Dissolve the required amount of PMSF in a small volume of ice-cold isopropanol<br />

<strong>and</strong> adjust to final volume with chilled TES buffer.<br />

2. APS breaks down in water resulting in rapid loss of reactivity. APS solutions<br />

should be prepared fresh each time.<br />

3. Add required volume of 1 M DTT to buffer just before use.


Table 1<br />

L<strong>and</strong>mark cell wall-associated proteins identified by MALDI-TOF peptide mass fingerprinting analysis for S. pyogenes<br />

strain 5448 (Serotype M1)<br />

Spot Protein Function or pathway Accession no. a Molecular mass (kDa) b pI b Peptidematch c Coverage (%) d<br />

1 Enolase Virulence factor P69950 47.2 4.74 24 68.9<br />

(SEN)<br />

2 GAPDH Virulence factor Q5XDW3 35.8 5.34 19 57.0<br />

3 Manganese- Virulence factor Q8P0D4 22.5 4.87 9 57.0<br />

dependent<br />

Superoxide<br />

dismutase<br />

a Swiss-Prot/TrEMBL accession number.<br />

b Theoretical values obtained from Swiss-Prot/TrEMBL database.<br />

c Number of tryptic peptides detected by MALDI-TOF MS that could be matched to the protein.<br />

d Percentage of protein sequence covered by the matched peptides.


308 Cole et al.<br />

4. Bio-Lyte ® 3/10 ampholyte (Bio-Rad) is recommended for all pH ranges.<br />

5. Bromophenol Blue tracking dye allows monitoring of electrophoresis.<br />

6. Combine ammonium sulfate, methanol <strong>and</strong> phosphoric acid. Bring to volume<br />

with Milli-Q ® water <strong>and</strong> dissolve by vigorous stirring. Add the Coomassie <br />

G250 <strong>and</strong> stir vigorously with heating. Do not filter solution in order to retain<br />

colloidal dye particles. Discard stain after each use.<br />

7. To minimize contamination <strong>and</strong> ensure cell viability, streak from glycerolized<br />

stocks of S. pyogenes stored at –80 °C rather than plates stored at 4 °C. Use<br />

fresh blood agar plates so that hemolysis is clearly visible.<br />

8. Hold plate up to light or use a light box to ensure the colony is hemolytic.<br />

9. The optical density at 600 nm (OD 600) for late stationary phase cultures of<br />

S. pyogenes is approx 1.2.<br />

10. PMSF irreversibly inhibits serine proteases <strong>and</strong> some cysteine proteases which<br />

may be liberated during the extraction process.<br />

11. Seal the microcentrifuge tube lid with Parafilm ® <strong>and</strong> secure to incubator in a<br />

horizontal position to ensure thorough mixing.<br />

12. Mutanolysin cell wall extracts can be stored for at least 2 yr at –20 °C.<br />

13. This step is used to verify the extraction of cell wall-associated proteins before<br />

sample preparation for <strong>2D</strong>E.<br />

14. The BCA assay is more sensitive than the Lowry assay <strong>and</strong> has less variability<br />

than the Bradford assay. Protein quantitation should be performed before <strong>2D</strong>E<br />

as the solubilization solution reagents interfere with the BCA assay. Working<br />

Reagent is stable for one day at room temperature.<br />

15. St<strong>and</strong>ards should be prepared using the same diluent as the cell wall extracts to<br />

compensate for buffer interference.<br />

16. To facilitate mixing, add each sample to the microtiter plate before adding the<br />

Working Reagent.<br />

17. A purple colored BCA-copper complex is generated in the BCA protein assay.<br />

The samples may also be incubated at 60 °C for 15 min or at room temperature<br />

(25°C)from2htoovernight. Incubation at higher temperatures will increase<br />

the sensitivity of the assay.<br />

18. An absorbance range of 540–590 nm may also be used with minimal loss of<br />

signal. A st<strong>and</strong>ard curve should be determined for each assay.<br />

19. Atmospheric oxygen inhibits the polymerization of acrylamide. For consistent<br />

results, degas the solution without the APS <strong>and</strong> TEMED under vacuum for at<br />

least 15 min. Add APS <strong>and</strong> TEMED <strong>and</strong> swirl the solution gently but thoroughly<br />

to minimize the introduction of oxygen.<br />

20. The 12% resolving gel has an approximate separation range of 14.4 kDa to<br />

120 kDa.<br />

21. Remove the ethanol overlay after 1htoprevent dehydration of the gel surface.<br />

22. Slowly load the sample to avoid a diffuse loading zone <strong>and</strong> loss of b<strong>and</strong><br />

sharpness. Load 15 μL of 1× loading buffer in unused wells to prevent the lateral<br />

spread of adjoining samples.


Isolation of Gram-Positive Bacterial Cell Wall Proteins 309<br />

23. The voltage <strong>and</strong> current used for SDS-<strong>PAGE</strong> is potentially lethal. Use all<br />

equipment in accordance with the manufacturer’s instructions.<br />

24. The detection limit for Coomassie Blue staining is 0.1–0.5 μg protein.<br />

25. The Coomassie Blue stain can be reused several times.<br />

26. The addition of a piece of sponge or paper towel to a corner of the tray will<br />

facilitate the removal of excess Coomassie Blue stain. Rapid destain solution<br />

can be recycled by filtering through activated carbon (Sigma). If loss of b<strong>and</strong><br />

intensity occurs because of excessive destaining, restain with Coomassie <strong>and</strong><br />

destain as described.<br />

27. The addition of glycerol to the final destain solution prevents the cracking of<br />

gels during drying.<br />

28. TCA is a hazardous substance <strong>and</strong> care should be taken to avoid exposure. The<br />

TCA precipitation step is used to increase protein concentration before <strong>2D</strong>E.<br />

29. The pellet is usually invisible, but can sometimes be seen as a white smear. To<br />

avoid disturbing the pellet, carefully add the ethanol <strong>and</strong> gently tap the tube to<br />

mix.<br />

30. Avoid over-drying as this will make the pellet difficult to resolubilize. TCA<br />

precipitated cell wall proteins can be stored for at least 2 yr at –20 °C.<br />

31. Poor protein solubilization may cause horizontal or vertical streaking in <strong>2D</strong> gels.<br />

Do not heat to resolubilize. <strong>Sample</strong>s containing urea must not be heated above<br />

35 °C to avoid protein carbamylation.<br />

32. A water-bath sonicator is suitable for this step. Alternatively, a sonicator with a<br />

microtip may be used.<br />

33. Centrifugation removes particulate material which can block the gel pores of<br />

the IPG strip, resulting in poor focusing <strong>and</strong> horizontal streaking.<br />

34. The protocol may be stopped at this point <strong>and</strong> the sample stored at –20 °C.<br />

35. Load up to 1 mg protein for Coomassie stained <strong>2D</strong> gels. <strong>Sample</strong> overload may<br />

cause horizontal or vertical streaking. Do not exceed a total loading volume of<br />

250 μL per strip.<br />

36. A trace amount of Bromophenol Blue is included for monitoring of IPG strip<br />

rehydration <strong>and</strong> subsequent electrophoresis.<br />

37. Evaporation will cause the urea to precipitate.<br />

38. The minimum rehydration time is 11 h.<br />

39. Thoroughly cleaned rehydration trays may be re-used.<br />

40. Drainage of the oil removes unabsorbed protein <strong>and</strong> minimizes horizontal<br />

streaking in <strong>2D</strong> gels. The IPG strip may be wrapped in plastic wrap <strong>and</strong> stored<br />

at –80 °C.<br />

41. Always wear laboratory gloves when h<strong>and</strong>ling IPG strips <strong>and</strong> all apparatus/<br />

solutions used in their preparation to prevent contamination from skin keratin.<br />

42. Electrode wicks are highly recommended because they remove salts <strong>and</strong> other<br />

contaminants in the sample.<br />

43. The majority of S. pyogenes cell wall-associated proteins have a pI between 4<br />

<strong>and</strong> 7 (8). The appropriate pH range for other Gram-positive prokaryotes should<br />

be determined empirically.


310 Cole et al.<br />

44. Covering the strip with mineral oil prevents evaporation <strong>and</strong> carbon dioxide<br />

absorption during focusing.<br />

45. The maximum permissible voltage is 8,000 V. The focusing range for 11 cm<br />

IPG strips is 15,000–60,000 Vh. Incomplete or excessive isoelectric focusing<br />

may cause horizontal or vertical streaking in <strong>2D</strong> gels. Focusing time should be<br />

kept to the minimum necessary <strong>and</strong> must be determined empirically.<br />

46. If required, the IPG strip may be wrapped in plastic wrap <strong>and</strong> stored indefinitely<br />

at –80 °C without having a detrimental effect on the final <strong>2D</strong> pattern. Thaw at<br />

room temperature for 10–15 min before equilibration.<br />

47. Firmly push the separator sheets <strong>and</strong> cassettes against the bottom of the caster<br />

to ensure a good seal. If a full set of gels is not required, use blank cassette<br />

inserts with separator sheets to occupy the extra space.<br />

48. Push down firmly during this step to ensure the faceplate forms a tight seal.<br />

49. A homogenous gel containing 12.5% total acrylamide has a separation size range<br />

of 14 to 100 kDa.<br />

50. For consistent results, degas the monomer solution (excluding the APS <strong>and</strong><br />

TEMED) under vacuum for 15 min before adding the APS <strong>and</strong> TEMED.<br />

Degassing is not essential, but will accelerate polymerization.<br />

51. For longer term storage, unload the gel cassettes from the caster, wrap in paper<br />

towel <strong>and</strong> completely submerge in gel storage buffer. Store in a sealed container<br />

at 4 °C for up to 3 months.<br />

52. Wear gloves when preparing buffers <strong>and</strong> h<strong>and</strong>ling <strong>2D</strong>E gels to prevent protein<br />

contamination, primarily with skin keratin.<br />

53. IPG strip equilibration should be undertaken before second dimension separation.<br />

This step enhances the solubility of focused proteins <strong>and</strong> allows the binding<br />

of SDS for second dimension separation. Ineffective equilibration may cause<br />

vertical streaking in <strong>2D</strong> gels. Thoroughly cleaned equilibration trays may be<br />

re-used.<br />

54. Ensure the glass casset <strong>and</strong> gel surface are dry before loading the IPG strips.<br />

Remove excess water from the wells with blotting paper, taking care to avoid<br />

the gel surface.<br />

55. This step lubricates the strip, preventing it from adhering to the glass plates<br />

during loading.<br />

56. Two 11 cm strips or three 7 cm strips fit onto a 26 × 20 cm gel. Avoid air bubbles<br />

between the strip <strong>and</strong> the gel surface or between the strip backing <strong>and</strong> glass<br />

plate. A gap between strip <strong>and</strong> gel, or damage to the strip during application,<br />

may cause vertical streaking in <strong>2D</strong> gels.<br />

57. Melt the agarose gel overlay solution in a microwave oven. Allow to cool slightly<br />

(60 °C) to prevent the decomposition of urea in the equilibration buffer. Pipet<br />

slowly to avoid introducing air bubbles under or behind the strip, which may<br />

interfere with protein resolution. Air bubbles should be removed immediately<br />

with a spatula.<br />

58. Depletion of ions in the running buffer can result in poor resolution <strong>and</strong> vertical<br />

streaking. To avoid this, discard the running buffer after approx 3 runs.


Isolation of Gram-Positive Bacterial Cell Wall Proteins 311<br />

59. Gels can be run overnight at 1.5 W per gel. The Bromophenol Blue in the<br />

agarose gel overlay solution is used to monitor migration.<br />

60. Trim around the sides of the gel to ensure it does not adhere to the spacers <strong>and</strong><br />

tear.<br />

61. Colloidal Coomassie can detect down to 100 ng/protein spot.<br />

Acknowledgments<br />

This work was supported by the National Health <strong>and</strong> Medical Research<br />

Council of Australia. We wish to thank Stuart Cordwell (School of Molecular<br />

<strong>and</strong> Microbial Biosciences, The University of Sydney, NSW, Australia) for<br />

assistance with the MALDI-TOF MS analysis.<br />

References<br />

1. Ton-That, H., Marraffini, L. A., <strong>and</strong> Schneewind, O. (2004) Protein sorting to<br />

the cell wall envelope of Gram-positive bacteria. Biochim. Biophys. Acta 1694,<br />

269–78.<br />

2. van Heijenoort, J. (2001) Formation of the glycan chains in the synthesis of<br />

bacterial peptidoglycan. Glycobiology 11, 25R–36R.<br />

3. Sjoquist, J., Movitz, J., Johansson, I. B., <strong>and</strong> Hjelm, H. (1972) Localization of<br />

protein A in the bacteria. Eur. J. Biochem. 30, 190–4.<br />

4. Navarre, W. W., <strong>and</strong> Schneewind, O. (1999) Surface proteins of Gram-positive<br />

bacteria <strong>and</strong> mechanisms of their targeting to the cell wall envelope. Microbiol.<br />

Mol. Biol. Rev. 63, 174–229.<br />

5. Chassy, B. M., <strong>and</strong> Giuffrida, A. (1980) Method for the lysis of Gram-positive,<br />

asporogenous bacteria with lysozyme. Appl. Environ. Microbiol. 39, 153–8.<br />

6. Yokogawa, K., Kawata, S., Takemura, T., <strong>and</strong> Yoshimura, Y. (1975) Purification<br />

<strong>and</strong> properties of lytic enzymes from Streptomyces globisporus 1829. Agric. Biol.<br />

Chem. 39, 1533–43.<br />

7. Laemmli, U. K. (1970) Cleavage of structural proteins during the assembly of the<br />

head of bacteriophage T4. Nature 227, 680–5.<br />

8. Cole, J. N., Ramirez, R. D., Currie, B. J., Cordwell, S. J., Djordjevic, S. P.,<br />

<strong>and</strong> Walker, M. J. (2005) Surface analyses <strong>and</strong> immune reactivities of major cell<br />

wall-associated proteins of group A Streptococcus. Infect. Immun. 73, 3137–46.


25<br />

Cell <strong>Fractionation</strong> of Parasitic Protozoa<br />

W<strong>and</strong>erley de Souza, José Andrés Morgado-Diaz,<br />

<strong>and</strong> Narcisa L. Cunha-e-Silva<br />

Summary<br />

Cell fractionation, a methodological strategy for obtaining purified organelle preparations,<br />

has been applied successfully to parasitic protozoa by a number of researchers.<br />

These studies have provided new information of the cell biology of these parasites <strong>and</strong><br />

have supported investigators to assume that some of the protozoa form the roots of the<br />

evolutionary tree of eukaryotic cells. The cell fractionation usually starts with disruption of<br />

the plasma membrane, using conditions that minimize damage to the membranes bounding<br />

intracellular organelles. An important requirement for successful cell fractionation is the<br />

evaluation of the isolation procedure that can be made by morphological <strong>and</strong> biochemical<br />

methods. The morphological approaches use light <strong>and</strong> electron microscopy of thin section<br />

of different fractions obtained, <strong>and</strong> the biochemical methods are based on the quantification<br />

of marker enzymes or other molecules (for instance, a special type of lipid, an<br />

antigen, etc.). Here we will present our experience in the isolation <strong>and</strong> characterization of<br />

some structures found in trypanosomatids <strong>and</strong> trichomonads.<br />

Key Words: Cell fractionation; electron microscopy; marker enzymes; parasitic<br />

protozoa; trichomonads; trypanosomatids.<br />

1. Introduction<br />

Parasitic protozoa are responsible for a large number of diseases affecting<br />

millions of people throughout the world. Some are caused by protozoa, which<br />

establish an initial binding to cells from the host <strong>and</strong> then invade them<br />

using several alternative mechanisms. Among these protozoa we can mention<br />

members of the Trypanosomatidae family, causative agents of Chagas’ disease<br />

<strong>and</strong> leishmaniasis, <strong>and</strong> members of the Apicomplexa group, which include<br />

From: Methods in Molecular Biology, vol. 425: <strong>2D</strong> <strong>PAGE</strong>: <strong>Sample</strong> <strong>Preparation</strong> <strong>and</strong> Fractioation, Volume 2<br />

Edited by: A. Posch © Humana Press, Totowa, NJ<br />

313


314 de Souza et al.<br />

agents of malaria <strong>and</strong> toxoplasmosis. Other protozoa exert their parasitic<br />

activity via interaction with epithelial cells from the gastrointestinal <strong>and</strong><br />

urogenital cavities. Examples include the agents of giardiasis, trichomoniasis,<br />

<strong>and</strong> amebiasis. To exert a parasitic activity these protozoa present<br />

several biosynthetic <strong>and</strong> metabolic pathways, which make them different from<br />

mammalian cells.<br />

Transmission electron microscopy of thin sections has provided most of the<br />

available information on the structural organization of parasitic protozoa. Structures<br />

typically found in mammalian cells, such as the nucleus, endoplasmic<br />

reticulum, Golgi complex, mitochondria <strong>and</strong> lysosomes are found in most,<br />

but not in all protozoa. Some lack structures such as the Golgi complex<br />

<strong>and</strong> mitochondria. Others present special secretory organelles, such as the<br />

micronemes, rhoptries, <strong>and</strong> dense granules found in Apicomplexa, or metabolic<br />

organelles, such as the hydrogenosome, found in trichomonads. One approach<br />

that has been used to obtain new information on structure <strong>and</strong> composition of<br />

structures <strong>and</strong> organelles found in parasitic protozoa is cell fractionation. Here<br />

we will present our experience in the isolation <strong>and</strong> characterization of some<br />

structures found in trypanosomatids <strong>and</strong> trichomonads. Figs. 1 <strong>and</strong> 2 show<br />

general views of the structural organization of Trypanosoma cruzi <strong>and</strong> Tritrichomonas<br />

foetus, respectively, based on information obtained by transmission<br />

electron microscopy. As can be seen in the figures these protozoa present well<br />

Fig. 1. Longitudinal section of a Trypanosoma cruzi epimastigote depicting the<br />

the typical positions of the nucleus (N), kinetoplast (K), anterior flagellum (F) <strong>and</strong><br />

reservosomes (R) at the posterior region of the parasite. Top inset shows a detail of<br />

reservosome structure <strong>and</strong> bottom inset shows Golgi complex (GC), always positioned<br />

next to the flagellar pocket, but that was not at the plane of the longitudinal section.<br />

Bars: 0.5 μm.


Cell <strong>Fractionation</strong> of Parasitic Protozoa 315<br />

Fig. 2. Longitudinal section of Tritrichomonas foetus showing the nucleus (N), Golgi<br />

complex (G), hydrogenosomes (H) <strong>and</strong> glycogen particles (asterisk) (Micrograph by<br />

M. Benchimol). Bar = 0.5 μm.<br />

elaborated cytoskeletal structures, known as the subpellicular microtubules <strong>and</strong><br />

the pelta-axostyle system, which make them resistant to cell breakage.<br />

2. Materials<br />

2.1. Cell Culture <strong>and</strong> St<strong>and</strong>ard Equipment<br />

1. Trypanosomatids: Liver Infusion Trypticase Medium (LIT) (1) supplemented with<br />

10% heat-inactivated bovine serum (Gibco/BRL).<br />

2. Trichomonads: Trypticase Yeast Maltose Medium (TYM) (2) supplemented with<br />

10% heat-inactivated bovine serum (Gibco/BRL).<br />

3. Ultrasonic apparatus (Sigma, GEX 600 Model) with st<strong>and</strong>ard probe (13 mm<br />

radiating diameter.<br />

4. Sorvall RC 5B centrifuge with GSA <strong>and</strong> SS-34 Rotors.


316 de Souza et al.<br />

5. Ultracentrifuge with swinging-bucket (Beckman SW 50.1, SW 28 <strong>and</strong> fixed angle<br />

Type 65) rotors.<br />

6. Potter-type homogenizer with Teflon pestle.<br />

2.2. Electron Microscopy<br />

1. Glutaraldehyde grade II (Ted Pella Inc.) is stored at 25% at –20 °C. Working<br />

solutions of 2.5% is prepared by diluting in Cacodylate buffer 0.1M, pH 7.2<br />

(Sigma).<br />

2. Osmium tetroxide (Ted Pella Inc.) is dissolved at 2% in distillated water <strong>and</strong><br />

stored in a single use aliquot at –20 °C. Working solutions is prepared by dilution<br />

in cacodylate buffer plus 5 mM CaCl2 <strong>and</strong> 0.8% potassium ferrocyanide.<br />

3. Acetone p.a: solutions of 30, 50, 70, 90, <strong>and</strong> 100%.<br />

4. Epoxy resin: Polybed 812 (Electron Microscopy Sciences, EMS).<br />

5. Aqueous solutions of uranyl acetate (Electron Microscopy Sciences, EMS) <strong>and</strong><br />

lead citrate.<br />

2.3. Isolation <strong>and</strong> Subfractionation of the Flagellum<br />

of Trypanosomatids<br />

1. Buffer A: 25 mM Tris–HCl, pH 7.4, 0.2 mM EDTA, 5 mM MgCl2,12mM μ-mercaptoethanol, 320 mM sucrose.<br />

2. Sonication buffer: buffer A supplemented with 1% bovine serum albumin (BSA)<br />

<strong>and</strong>1mMCaCl2 <strong>and</strong> protease inhibitors cocktail (Sigma).<br />

3. Sucrose gradient solutions for the isolation of flagellum: 0.8, 1.65, <strong>and</strong> 1.85M<br />

prepared in buffer A without sucrose.<br />

4. 2% Triton X-100 prepared in buffer A.<br />

5. Sucrose gradient to isolation of the flagellar membrane: 1.3, 1.6, 1.9, <strong>and</strong> 2.2M<br />

prepared in buffer A without sucrose.<br />

6. 0.0001% trypsin (type XIII-treated, Sigma).<br />

7. Soybean trypsin inhibitor (Sigma).<br />

8. Continuous sucrose gradient ranging from 1.8 to 2.2M<br />

2.4. Isolation of the Reservosome<br />

1. TMS buffer: 20 mM Tris-HCl, pH 7.2, 2mM MgCl 2, 250 mM sucrose.<br />

2. Sucrose solutions: 2.3, 1.2, 1.0, <strong>and</strong> 0.8 M prepared in TMS buffer without sucrose<br />

(TM buffer).<br />

3. TMS buffer supplemented with a cocktail of protease inhibitors (Sigma).<br />

2.5. Isolation of the Golgi Complex<br />

1. Buffer G: 10 mM Tris-HCl, pH 7.2, 0.25M sucrose, 2 mM MgCl2. 2. Hypotonic solution: G buffer without sucrose containing a cocktail of protease<br />

inhibitors.


Cell <strong>Fractionation</strong> of Parasitic Protozoa 317<br />

3. Gradient sucrose solutions: 2.3, 1.2, 1.0, <strong>and</strong> 0.8M prepared in G buffer without<br />

sucrose.<br />

4. 150 mM sodium carbonate, pH 11.5.<br />

2.6. Isolation <strong>and</strong> Subfractionation of the Hydrogenosomes<br />

1. H buffer: 10 mM Tris-HCl, pH 7.2, 0.25M sucrose <strong>and</strong> 2 mM MgCl 2.<br />

2. Percoll gradient solutions (Pharmacia): 53, 45, 24, <strong>and</strong> 18% prepared in G buffer.<br />

3. V buffer: 2% Triton X-100, 50 mM 3-(N-morpholino)propanesulfonic acid<br />

(MOPS))-NaOH, pH 7.0, 0.1 mM MgCl 2,1nM dithiothreitol (Sigma) <strong>and</strong> 10%<br />

sucrose.<br />

4. Proteinase K (Sigma) at a final concentration 0.5 mg/mL.<br />

5. Quench solution: phenylmethylsulphonyl fluoride (PMSF) (final concentration 40<br />

μg/mL).<br />

3. Methods<br />

3.1. Isolation of Trypanosomatid Structures<br />

3.1.1. Isolation <strong>and</strong> Subfractionation of the Flagellum<br />

of Trypanosomatids<br />

Trypanosomatid flagellum exhibits a huge lattice-like filamentous structure<br />

called paraflagellar rod (PFR), running alongside a canonical 9+2 axoneme<br />

(3,4). The flagellar membrane, which has been considered a special domain,<br />

although continuous with cell body membrane, surrounds both structures. PFR<br />

plays a role in protozoa movement, as was indicated by a Trypanosoma brucei<br />

mutant that did not assemble the structure <strong>and</strong> was immotile (5). Flagellar<br />

membrane has a crucial participation in the interaction of T. cruzi with invertebrate<br />

host <strong>and</strong> parasite differentiation (6). The knowledge concerning the role<br />

played by trypanosomatid flagellum in the life cycle of these parasites <strong>and</strong><br />

in the pathogenesis they cause in their vertebrate hosts has advanced using<br />

flagellar fractions as immunogen (7) or for biochemical <strong>and</strong> ultrastructural<br />

studies (8). There are two successful protocols for obtaining purified flagellar<br />

fractions: a classical one (9), deflagellating the protozoa <strong>and</strong> isolating the<br />

detached flagella by sucrose gradients, <strong>and</strong> a more simple method (10) that<br />

extracts the membranes of the whole protozoa <strong>and</strong> depolymerizes the cell body<br />

cytoskeleton by high salt treatment. The advantage of the latter method is that<br />

it does not take long, but it has a low yield, being limited to 105 parasites at<br />

a time. Here we describe the classical method that, although laborious, can be<br />

applied to 1012 parasites in a single experiment, yielding an abundant highly<br />

purified flagellar fraction.<br />

PFR is a highly conserved structure among trypanosomatids (11), allowing<br />

the adoption of the PFR from Herpetomonas megaseliae, a nonpathogenic


318 de Souza et al.<br />

trypanosomatid, as a model for ultrastructural <strong>and</strong> biochemical studies. The<br />

protocols presented below have already been published for whole flagella purification<br />

(9), PFR (12), <strong>and</strong> flagellar membrane isolation from both H. megaseliae<br />

<strong>and</strong> T. cruzi (8).<br />

1. Start with 10 11 –10 12 protozoa. Harvest protozoa by centrifugation <strong>and</strong> wash<br />

twice in ice cold buffer A. All subsequent steps should be performed in an ice<br />

bath (see Note 1).<br />

2. Washed protozoa (the pellet of the last washing step) are resuspended in<br />

sonication buffer: buffer A supplemented with 1% bovine serum albumin (BSA)<br />

<strong>and</strong>1mM CaCl 2 <strong>and</strong> protease inhibitors cocktail. Cell density is very important<br />

in this step. If you started with 8×10 11 protozoa, resuspend in 200 mL <strong>and</strong><br />

sonicate in 50-mL aliquots (see Note 2).<br />

3. We use an ultrasonic apparatus adjusted to 8% of maximum amplitude output.<br />

Typically, 6–8 sonication cycles of 10 s on <strong>and</strong> 5 s off are sufficient to deflagellate<br />

most cells (see Note 3).<br />

4. Whole cells, deflagellated cell bodies <strong>and</strong> liberated nuclei are pelleted with a<br />

low speed centrifugation at 120g, 10 min (see Note 4).<br />

5. Supernatants are centrifuged at 6,800g, 15 min, the pellet resuspended to 15 mL<br />

with buffer A <strong>and</strong> divided in six aliquots.<br />

6. Each aliquot is equilibrated on top of 0.8M sucrose cushions <strong>and</strong> spun at 1,080g<br />

for 20 min (see Note 5).<br />

7. Upper layer of each cushion is carefully removed <strong>and</strong> pelleted all together at<br />

17,300g. This is the crude flagellar fraction (see Note 6).<br />

8. The purification is achieved by laying 1-mL aliquots of crude flagellar fraction<br />

on top of a 1.65M/1.85M sucrose gradient, each step with 2 mL, <strong>and</strong> centrifuging<br />

at 130,000g for3h.<br />

9. Flagella equilibrate at the interface 1.65M/1.85M that must be carefully removed<br />

with a long <strong>and</strong> thin Pasteur pipet. All the interfaces together are diluted with<br />

buffer A without sucrose <strong>and</strong> pelleted at 17,300g for 20 min. The pellet is<br />

resuspended in a small volume of buffer A (0.5 mL) <strong>and</strong> contains purified<br />

flagella.<br />

10. Phase contrast microscopy can be used for initial examination of the flagellar<br />

fraction, but the only way of evaluating purity is electron microscopy. A small<br />

aliquot can be fixed by adding glutaraldehyde to the final concentration of 2.5%<br />

in buffer A <strong>and</strong> processed for electron microscopy (Fig. 3). (see Note 7).<br />

11. H. megaseliae or T. cruzi flagella purified as above are submitted to detergent<br />

extraction by doubling sample volume with ice cold 2% Triton X-100 in buffer<br />

A <strong>and</strong> incubated for 15 min. on ice with intermittent agitation.<br />

12. <strong>Sample</strong> is laid on top of a 1.3/1.6/1.9/2.2M sucrose <strong>and</strong> centrifuged at 130,000g<br />

for3h.<br />

13. Purified flagellar membrane that equilibrates at the interface 1.3/1.6M is carefully<br />

removed, diluted with buffer A without sucrose, pelleted at 17,300g for 20 min,<br />

<strong>and</strong> resuspended in a small volume (0.05 mL) (Fig. 4).


Cell <strong>Fractionation</strong> of Parasitic Protozoa 319<br />

Fig. 3. Flagellar fractions from Herpetomonas megaseliae. Shows purified whole<br />

flagella, with preserved axonemes <strong>and</strong> PFRs enclosed by loose-fitting membrane<br />

profiles. Bar = 0.2 μm; Adapted from (8).<br />

14. H. megaseliae or T. cruzi purified flagella are submitted to extensive membrane<br />

solubilization with three rounds of incubation in 2% Triton X-100 or Nonidet<br />

P-40 for 15 min at 4°C. After each round flagella are pelleted at 17,300g, 20 min.<br />

15. Demembranated flagella are resuspended in buffer A <strong>and</strong> incubated very briefly<br />

(30 s) at 28°C with 0.001% trypsin (type XIII, TPCK-treated, Sigma). The<br />

reaction is stopped by addition of a 20-fold excess of specific trypsin inhibitor<br />

(soybean trypsin inhibitor, Sigma). Shake in a Vortex at maximum speed for 1<br />

Fig. 4. Flagellar fractions from Herpetomonas megaseliae. Shows flagellar<br />

membranes purified from Fig. 3 preparation. Bar = 0.2 μm; Adapted from (8).


320 de Souza et al.<br />

min, divide in six aliquots <strong>and</strong> apply on a continuous sucrose gradient ranging<br />

from 1.8 to 2.2M.<br />

16. Centrifuge at 130,000g for 3 h. Collect 0.8-mL aliquots from top to bottom,<br />

dilute with buffer A without sucrose <strong>and</strong> pellet at 17,300g for 30 min. The<br />

third <strong>and</strong> fourth fractions from top to bottom are highly purified PFRs, as can<br />

be evaluated by electron microscopy (Fig. 5) <strong>and</strong> by using antibodies against<br />

major PFR proteins (Fig. 6B) <strong>and</strong> alpha-tubulin (Fig. 6C) in Western blots.<br />

SDS-<strong>PAGE</strong> analysis of purified PFRs (Fig. 6A) allowed for the first time the<br />

identification of some minor proteins that constitute this complex structure.<br />

3.1.2. Isolation of the Reservosome<br />

Reservosomes are storage organelles from T. cruzi epimastigotes that concentrate<br />

proteases <strong>and</strong> are essential for differentiation into trypomastigote forms.<br />

They were considered prelysosomes but the lack of a molecular marker has<br />

precluded a more detailed studied of their role in T. cruzi life cycle. Their<br />

purification (13) opened up the possibility of defining the necessary marker.<br />

1. Start with 3×10 10 epimastigotes from mid log phase cultures (see Note 8).<br />

2. Harvest protozoa by centrifugation at 4,800g, 10 min, 4°C <strong>and</strong> wash in cold<br />

TMS buffer (see Note 9).<br />

3. Resuspend in 45 mL of TMS (about 6.5 × 10 8 parasites/mL) <strong>and</strong> divide in three<br />

aliquots.<br />

4. Sonicate each aliquot at 10% of maximum amplitude of ultrasonic apparatus, in<br />

about 15 cycles of 2son<strong>and</strong>1soff. (see Note 3).<br />

5. Centrifuge at 2,450g, 10 min, pour the supernatant carefully into another tube<br />

<strong>and</strong> discard the pellet, containing non-ruptured cells, nuclei, kinetoplasts etc.<br />

Fig. 5. Flagellar fractions from Herpetomonas megaseliae. Shows that PFRs remain<br />

organized after purification from demembranated H. megaseliae purified flagella.<br />

Bar = 0.1 μm; Adapted from (12).


Cell <strong>Fractionation</strong> of Parasitic Protozoa 321<br />

Fig. 6. (A) SDS-<strong>PAGE</strong> gel of fractions from the PFR purification gradient. Lanes<br />

1–4 represent fractions 3–6, in this order. Besides the major PFR proteins (arrowhead),<br />

fractions 3 <strong>and</strong> 4 are clearly enriched in a number of minor b<strong>and</strong>s whose positions<br />

<strong>and</strong> estimated molecular masses are indicated on the left. Molecular mass markers are<br />

indicated on the right. (B <strong>and</strong> C) Western blots of different steps <strong>and</strong> gradient fractions<br />

from the paraflagellar rod (PFR) purification procedure probed with anti PFR major<br />

proteins antibody (panel B), <strong>and</strong> antialpha-tubulin monoclonal antibody (panel C). In<br />

both panels lanes 1–4 represent fractions 3–6, in this order, from the PFR purification<br />

gradient. Molecular mass markers are indicated on the left. Adapted from (12).


322 de Souza et al.<br />

6. Mix the supernatant with an equal volume of 2.3M sucrose in TMS <strong>and</strong> divide<br />

in two Beckman SW28 centrifuge tubes (usually 12 mL in each tube) (see<br />

Note 10).<br />

7. Repeating the procedure with the other two aliquots, all six tubes of the rotor<br />

will be used.<br />

8. Overlay carefully with 10 mL of 1.2M sucrose in TMS, 9 mL of 1.0M sucrose<br />

<strong>and</strong>8mLof0.8M sucrose.<br />

9. Centrifuge at 97,000g for 150 min, without brake.<br />

10. Collect the interfaces 0.8M/1.0M (B1) <strong>and</strong> 1.0M/1.2M (B2), 1.2M/1.27M (B3)<br />

<strong>and</strong> the pellet (P), dilute with TM (TMS without sucrose) <strong>and</strong> pellet at 90,000g<br />

for 20 min. Reservosomes are highly purified in B1 (Fig. 7) <strong>and</strong> very enriched<br />

in B2. Microsomes <strong>and</strong> glycosomes are the major components of B3. Acidocalcisomes<br />

are enriched in the pellet. Fig 7 here<br />

11. A small aliquot of each fraction is fixed by adding glutaraldehyde directly<br />

in TMS for electron microscopy (see Note 7), another aliquot is reserved for<br />

marker enzyme assays (see next step), <strong>and</strong> protease inhibitors are added to a<br />

third aliquot for SDS-<strong>PAGE</strong> <strong>and</strong> Western blot assays. Purified fractions must<br />

be stored below –70°C.<br />

12. To evaluate the purity of the fractions marker enzymes for the organelles that<br />

could be contaminants of reservosome fraction should be assayed: succinate<br />

cytochrome c reductase for mitochondria (14), pyrophosphatase for acidocalcisomes<br />

(Table 1) <strong>and</strong> hexokinase for glycosomes (15).<br />

Fig. 7. Trypanosoma cruzi reservosomes purified fraction. Adapted from (13).<br />

Bar = 1 μm.


Cell <strong>Fractionation</strong> of Parasitic Protozoa 323<br />

3.1.3. Isolation of the Golgi Complex<br />

In parasitic protozoan the Golgi complex is much less well organized, less<br />

prominent, <strong>and</strong> correspondingly more difficult to isolate than in mammalian<br />

cell. The success of the organelle isolation is dependent on choosing a good cell<br />

disruption method, which is dependent on the morphological characteristics of<br />

the parasite. Morphological studies have shown that the Golgi of trypanosomatids,<br />

<strong>and</strong> in particular of Trypanosoma cruzi, is made up of 4–10 stacked<br />

cisternae localized at the anterior region of the cell, close to the flagellar pocket<br />

(16). A method that preserves the Golgi with minimum damage during cell<br />

disruption was developed resulting in a fraction highly enriched in stacked Golgi<br />

cisternae <strong>and</strong> vesicles (19), as determined by electron microscopy (Fig. 8), <strong>and</strong><br />

also in galactosyltransferase, O-- GlcNA transferase <strong>and</strong> acid phosphatase<br />

activities. Contamination with other organelles (endoplasmic reticulum, plasma<br />

membrane, mitochondria, <strong>and</strong> glycosomes) is minimal, as analyzed by their<br />

respective marker enzymes (Table 2).<br />

All centrifugation steps <strong>and</strong> other operations are performed at 4 °C or in an<br />

ice bath.<br />

1. Harvest cells in a GSA Rotor at 1,000g for 10 min <strong>and</strong> wash three times in G<br />

buffer.<br />

2. Resuspend the cells in a hypotonic solution (G buffer without sucrose) containing<br />

a cocktail of protease inhibitors <strong>and</strong> incubated in this solution for 10 min.<br />

3. Homogenize the cells by sonication on ice with 15 cycles of 2 s with 1 s rest<br />

between cycles in an ultrasonic apparatus. Monitor the cell disruption by phasecontrast<br />

microscopy (see Note 3).<br />

4. Immediately add to the homogenate a concentrated sucrose solution, prepared in<br />

G buffer to a final concentration of 0.25M sucrose to minimize osmotic damage.<br />

Table 1<br />

Enzyme assays in the epimastigote subcellular fractions of the purification of<br />

reservosome of Trypanosoma cruzi<br />

Succinate cytochrome c reductase a Pyrophosphatase b<br />

Total homogenate 0.030 0.11<br />

Supernatant from 2,450g 0.043 0.14<br />

B1 0 0<br />

B2 0.006 0.02<br />

B3 0.044 0<br />

B4 0.830 0.15<br />

a Specific activity in mM min −1 mg protein −1 .<br />

b Specific activity in μM pyrophosphate consumed min −1 mg protein −1 .


324 de Souza et al.<br />

Fig. 8. General view of the Golgi fraction of Trypanosoma cruzi showing Golgi<br />

complex cisternae <strong>and</strong> several Golgi cisternae profiles. After (19). Bar = 0.5 μm.<br />

5. Centrifuge at 2,500g for 15 min <strong>and</strong> discard the pellet containing unbroken cells,<br />

nuclei, <strong>and</strong> kinetoplasts.<br />

Table 2<br />

Enzyme activities in Golgi fraction <strong>and</strong> other subcellular fractions of<br />

Trypanosoma cruzi. a<br />

Assay<br />

Enzyme Activity bc<br />

Homogenate fraction PNS GF<br />

NADPH cyt c reductase b 025 034 0043<br />

Acid phosphatase b 30 45 152<br />

5’-nucleotidase b 0101 0157 006<br />

Succinate cyt c reductase b 007 004 003<br />

Hexokinase b 0032 00436 00034<br />

O-a-GlcNAc transferase c 0076 0248 2044<br />

Gal transferase c 128 656 11480<br />

PNS, post-nuclear supernatant; GF, Golgi fraction<br />

a The results are of a typical experiment in which each number represents the means of<br />

duplicate determinations. Minor modifications were observed in at least three independent experiments.<br />

b Specific activity expressed as μM of product released min −1 mg protein −1 .<br />

c Specific activity expressed as pM of [ 3 H]GlcNAc or [ 3 H]Gal transferred h −1 mg protein −1 .


Cell <strong>Fractionation</strong> of Parasitic Protozoa 325<br />

6. Harvest the postnuclear supernatant (PNS), add an equal volume of 2.3M sucrose<br />

<strong>and</strong> homogenize for 10 min.<br />

7. Load 15 mL of PNS onto the bottom of Beckman SW 28 Rotor ultracentrifuge<br />

tubes <strong>and</strong> overlay sequentially with 7 mL each of 1.2M, 1.0M, <strong>and</strong> 0.8M sucrose.<br />

8. Centrifuge the gradient at 95,000 x g for 1h 30 min. The b<strong>and</strong> at the interface of<br />

1.0/1.2M sucrose corresponds to highly enriched Golgi complex fraction.<br />

9. Collect the fraction by diluting with G buffer without sucrose <strong>and</strong> centrifuging at<br />

80,000g for 45 min (Beckman Type 65 Rotor).<br />

3.2. Isolation of Trichomonad Structures<br />

3.2.1. Isolation of the Golgi Complex<br />

Tritrichomonas foetus, a member of the Trichomonadidae family, possesses a<br />

well developed Golgi complex, consisting of tightly packed elongated cisternae<br />

often with dilated terminal regions, localized at the anterior portion of the<br />

cell <strong>and</strong> in close association with the parabasal filament (18). The following<br />

protocol yields two subfractions containing light (GF1) <strong>and</strong> heavy (GF2) Golgi<br />

membranes (17). At the ultrastructural level GF1 fraction is constituted only<br />

of smooth-membrane structures, particularly cisternae <strong>and</strong> secretory vesicles<br />

(Fig. 9A) <strong>and</strong> contains a 20-fold enrichment of galactosyltransferase activity.<br />

GF2 fraction is composed of profiles of stacked Golgi membranes with three or<br />

more tightly apposed cisternae with dilated borders <strong>and</strong> associated to vesicles<br />

(Fig. 9B). This latter fraction contains a 7-fold enrichment in galactosyltransferase<br />

activity. Minimal contamination with other organelles, such as<br />

endoplasmic reticulum, hydrogenosomes, <strong>and</strong> plasma membrane is observed in<br />

these fractions, as quantified by their respective enzymatic markers (Table 3).<br />

All centrifugation steps <strong>and</strong> other operations are performed at 4 °C or in an<br />

ice bath.<br />

1. Harvest the cells in a GSA Rotor at 1,000g for 10 min <strong>and</strong> wash three times in<br />

G buffer.<br />

2. Resuspend the cells in G buffer containing a cocktail of protease inhibitors <strong>and</strong><br />

homogenize using 180 strokes of a Potter-type homogenizer. Stop the homogenization<br />

procedure when about 90% cell breakage is achieved as ascertained by<br />

phase-contrast microscopy.<br />

3. Centrifuge at 2,500g for 10 min to discard the pellet containing unbroken cells,<br />

nuclei, <strong>and</strong> hydrogenosomes.<br />

4. Add to the supernatant (PNS) an equal volume of 2.3M sucrose to make a 1.4M<br />

sucrose final concentration <strong>and</strong> homogenize for 10 min.<br />

5. Load 15 mL of PNS onto the bottom of SW28 ultracentrifuge tubes <strong>and</strong> overlay<br />

in succession with 7 mL each of 1.2M, 1.0M, <strong>and</strong> 0.8M sucrose.<br />

6. Centrifuge at 95,000g for 2.5 h <strong>and</strong> carefully remove two b<strong>and</strong>s from the top at<br />

0.8/1.0 (GF1) <strong>and</strong> 1.0/1.2M (GF2).


326 de Souza et al.<br />

Fig. 9. General aspect of the Golgi fractions of Tritrichomonas foetus. (A) GF1<br />

fraction showing several vesicles (arrowheads) <strong>and</strong> cisternae profiles (arrows). (B) GF2<br />

fraction showing several intact stacks of Golgi complex (arrowheads). After (17). Bars<br />

= 0.2 μm (A) <strong>and</strong> 0.5 μm (B).<br />

7. Dilute these fractions in G buffer without sucrose <strong>and</strong> collect them by centrifugation<br />

at 80,000g for 30 min (Beckman Type 65 Rotor).<br />

8. Further fractionation of these two compartments can be attempted by alkaline<br />

treatment to obtain the Golgi content <strong>and</strong> Golgi membrane subfraction. Blend the<br />

Table 3<br />

Distribution of Galactosyltransferase <strong>and</strong> other enzyme markers in Golgi<br />

fractions of Tritrichomonas foetus. a<br />

Fractions Total<br />

protein<br />

(mg)<br />

Galactosyl<br />

transferase<br />

(pmole/30<br />

min/mg<br />

protein)<br />

NADPH cit c<br />

reductase<br />

(μmole/min/mg<br />

protein)<br />

NADP-malic<br />

enzyme<br />

(μmole/min/mg<br />

protein)<br />

5’Nucleotidase<br />

Homogenate 3920 4434 0035 0012 0.93<br />

PNS 1040 6804 00079 0026 0.89<br />

GF2 20 31494 00029 0006 1.50<br />

GF1 10 87400 n.d. n.d. 0.66<br />

a Results are expressed as the means of duplicate determinations. n.d., not detectable; Number<br />

of experiments is given in parenthesis; PNS, postnuclear supernatant; GF2, heavy Golgi fraction;<br />

GF1, light Golgi fraction.


Cell <strong>Fractionation</strong> of Parasitic Protozoa 327<br />

Golgi fractions (GF1 <strong>and</strong> GF2), collect by centrifugation for 30 min at 80,000g<br />

(Beckman Type 65 Rotor), <strong>and</strong> treat with 150 mM sodium carbonate, pH 11.5,<br />

for 30 min.<br />

9. Collect the subfractions, again as in step 7.<br />

3.2.2. Isolation <strong>and</strong> Subfractionation of the Hydrogenosome<br />

T. foetus is a facultative anaerobic protist that lacks mitochondria, but<br />

contains another organelle, designated as the hydrogenosome. This organelle<br />

appears as a spherical or slightly elongated structure with a mean diameter<br />

of 0.5 μm, reaching 2 μm in dividing organelles. A common characteristic of<br />

most hydrogenosomes is the presence of a peripheral vesicle, which has been<br />

suggested to be a specialized sub-compartment (20). Additional information on<br />

this important organelle, which seems to be the main target of drugs used in<br />

the chemotherapy of trichomoniasis, can be obtained using hydrogenosomes<br />

isolated in a way that preserves their structure. Using Percoll gradients, we<br />

developed a procedure (21), which yielded the purest fraction obtained for<br />

hydrogenosomes of T. foetus (Fig. 10A) <strong>and</strong> allowed further sub-fractionation<br />

to isolate the peripheral vesicle (Fig. 10B). It is enriched about 12-fold in malic<br />

enzyme activity <strong>and</strong> free of other contaminant organelles (Table 4). Major<br />

hydrogenosome proteins were identified by SDS-<strong>PAGE</strong> (Fig. 10C).<br />

All centrifugation steps <strong>and</strong> other operations are performed at 4 °C or in an<br />

ice bath.<br />

1. Harvest the cells in a GSA Rotor at 1,000g for 10 min <strong>and</strong> wash three times in<br />

H buffer.<br />

2. Resuspend the cells in 30 mL of H buffer <strong>and</strong> homogenize using about 180 strokes<br />

of a Potter-type homogenizer. Stop the homogenization while some unbroken<br />

cells are still present.<br />

3. Dilute the homogenate in the proportion 1:10 with H buffer <strong>and</strong> centrifuge at<br />

1,500g for 10 min. Discard the pellet containing unbroken cells, nuclei <strong>and</strong> costa.<br />

4. Centrifuge the supernatant at 4,000g for 10 min. The pellet of this centrifugation<br />

corresponds to an enriched hydrogenosomal fraction.<br />

5. Resuspend the pellet in ice-cold H buffer <strong>and</strong> layer aliquots of 1 mL on the top<br />

of a Percoll gradient of 53, 45, 24, <strong>and</strong> 18% in H buffer (1 mL each solution).<br />

6. Centrifuge the gradient using a swinging bucket rotor (Beckman SW 50.1) at<br />

36,000g for 30 min, without using the brake. The pure fraction containing wellpreserved<br />

hydrogenosomes is recovered from the pellet of the Percoll gradient.<br />

7. Incubate a sample of the purified hydrogenosomal fraction in V buffer on ice for 1<br />

h. <strong>and</strong> centrifuge the suspension at 25,000g for 20 min (Beckman Type 65 Rotor).<br />

The supernatant (TX-supernatant) is made up of solubilized hydrogenosome<br />

membranes <strong>and</strong> the pellet (TX-pellet) of hydrogenosomal matrix attached to the<br />

peripheral vesicle.


328 de Souza et al.<br />

Fig. 10. (A) Electron micrograph showing the purified hydrogenosomes of Tritrichomonas<br />

foetus. The hydrogenosome matrix is homogeneous <strong>and</strong> the peripheral<br />

vesicles present an electron-dense content. Bar = 1 μm (B) Purified fraction of<br />

hydrogenosomal flat vesicles Bar = 0.5 μm (C) SDS-<strong>PAGE</strong> of the fractions obtained<br />

during the fractionation of T. foetus. Lane 1: whole homogenate, Lane 2: fraction before<br />

the Percoll gradient, Lane 3: purified hydrogenosomes. After (21).<br />

Table 4<br />

Specific activity <strong>and</strong> recovery of malic dehydrogenase in fractions of the<br />

purification of hydrogenosomes of Tritrichomonas foetus. a<br />

Fractions Total protein<br />

(mg)<br />

Specific activity (μmol<br />

min −1 mg protein)<br />

Enrichment<br />

(-fold)<br />

Yield (%)<br />

Whole homogenate 4500 0035 10 100<br />

Po fraction 580 0147 42 54<br />

Pellet gradient 80 0421 120 22<br />

a Po is the fraction before the Percoll gradient. Results are expressed as the means of duplicate<br />

determinations in three representative experiments.


Cell <strong>Fractionation</strong> of Parasitic Protozoa 329<br />

8. Wash a sample of the TX-pellet twice in H buffer <strong>and</strong> incubate for 45 min at<br />

room temperature with proteinase K (final concentration 0.5 mg/mL).<br />

9. Quench the mixture with PMSF (final concentration 40 μg/mL) on ice, for 15<br />

min <strong>and</strong> centrifuge at 350,000g for 30 min (Beckman Type 65 Rotor). The pellet<br />

of this centrifugation corresponds to the purified peripheral vesicle.<br />

4. Notes<br />

1. Three liters of axenic culture of H. megaseliae promastigotes in Warren’s<br />

medium supplemented with 5% fetal calf serum are enough for obtaining 8<br />

×1011 protozoa, after 30–36 h at 28°C. The same cell density of T. cruzi<br />

epimastigotes can be obtained with 5Lofparasites cultivated for 5dat28°C<br />

in LIT medium supplemented with 10% fetal calf medium (1). A good rotor to<br />

harvest a large volume culture is GSA of Sorvall ultracentrifuges.<br />

2. It is not necessary to use purified BSA to prepare sonication buffer; fraction V<br />

grade is good enough. The composition of protease inhibitors cocktail may vary,<br />

it is very important that it contains an irreversible cysteine protease inhibitor,<br />

such as E-64, because this class of protease is the most active in trypanosomatids.<br />

We use protease inhibitors cocktail for general use, from Sigma.<br />

3. It is essential to control deflagellation by phase contrast microscopy <strong>and</strong> stop<br />

the procedure when the majority, but not all the protozoa are deflagellated,<br />

otherwise some nuclei may rupture <strong>and</strong> free chromatin aggregates the organelles,<br />

including free flagella.<br />

4. Turn off centrifuge brake because this loose pellet resuspends easily. This step<br />

<strong>and</strong> the next are performed with Sorvall SS-34 fixed angle rotor. The supernatant<br />

of the low speed centrifugation should be controlled by phase contrast<br />

microscopy <strong>and</strong> this step should be repeated twice to clean the supernatant from<br />

whole cells <strong>and</strong> deflagellated cell bodies.<br />

5. The sucrose cushion must be spun in a swinging bucket rotor, as Beckman<br />

SW50.1, which must have each tube filled up to total volume (5 mL). In this<br />

step as well as in the gradient, the rotor should stop without using the brake.<br />

6. To improve the isolation yield, the cushion can be repeated using the bottom<br />

layer as the starting sample of step 6.<br />

7. A brief protocol for sample processing for electron microscopy: as fractions are<br />

easy to fix, 30 min of incubation in fixative solution is enough. On the other<br />

h<strong>and</strong>, fractions are not easy to wash, as the usual centrifugation of few minutes<br />

in a microfuge may not be sufficient to pellet the sample adequately <strong>and</strong> thus<br />

you would be losing your sample in the washing steps. To avoid this, wash in the<br />

ultracentrifuge at the same speed used to obtain the fraction until before osmium<br />

incubation when the fraction becomes denser. If you cannot process the samples<br />

in a short time (next day), change the fixative solution to 2.5% glutaraldehyde<br />

in 0.1 M cacodylate buffer pH 7.2, as Tris buffers are not good for electron<br />

microscopy. After fixation, samples are washed twice in 0.1M cacodylate buffer<br />

<strong>and</strong> post fixed with 1% osmium tetroxide in 0.1M cacodylate buffer containing


330 de Souza et al.<br />

0.8% potassium ferrocyanide <strong>and</strong> 5 mM calcium chloride for 20 min at room<br />

temperature in the dark (wear gloves <strong>and</strong> work in a fume hood!). Wash three<br />

times (or until the supernatant is clear) in 0.1M cacodylate buffer. Dehydrate in<br />

acetone series from 30 to 100%, 10 min incubation in each bath, repeating the<br />

last one twice. Infiltrate slowly in Epon (acetone + Epon 1:1 overnight, Epon<br />

for 6 h). Embed in Epon at 60 o Cfor2d.<br />

8. Epimastigotes from 5-day-old cultures are ideal for reservosome purification.<br />

Three liters of culture in LIT medium supplemented with 10% fetal calf serum<br />

(1) maintained at 28°C under gentle orbital agitation are enough to obtain 3 ×<br />

10 10 parasites.<br />

9. It is very important to work fast <strong>and</strong> in an ice bath, because reservosomes start to<br />

degrade their contents as soon as parasites are washed out from culture medium.<br />

It is adequate to add protease inhibitors to TMS buffer (mainly E-64, inhibitor<br />

of cysteine proteases, highly concentrated in the organelle), unless you just want<br />

to study the properties of reservosome proteases <strong>and</strong> their inhibitors.<br />

10. The bottom step of this gradient contains the sample <strong>and</strong> corresponds to 1.27M<br />

sucrose, very close in density to the next step. So, take care when preparing<br />

<strong>and</strong> measuring volumes of sucrose solutions before mixing sample with 2.3M<br />

sucrose.<br />

Acknowledgments<br />

The authors dedicate this chapter to the memory of Luiz Henrique Monteiro<br />

Leal, who greatly contributed to improve the methods of cell fractionation of<br />

parasitic protozoa, <strong>and</strong> passed away during the elaboration of this manuscript.<br />

References<br />

1. Camargo, E. P. (1964) Growth <strong>and</strong> differentiation in Trypanosoma cruzi. I. Origin<br />

of metacyclic trypanosomes in liquid media. Rev. Inst. Med. Trop. Sao Paulo 6,<br />

93–100.<br />

2. Diamond, L. S. (1957) The establishment of various trichomonads of animals <strong>and</strong><br />

man in axenix cultures. J. Parasitol. 43, 458–90.<br />

3. Fuge, H. (1969) Electron microscopic studies on the intra-flagellar structures of<br />

trypanosomes. J. Protozool. 16, 460–6.<br />

4. Farina, M., Attias, M., Souto-Padrón, T., <strong>and</strong> De Souza, W. (1986) Further studies<br />

on the organization of the paraxial rod of trypanosomatids. J. Protozool. 33, 552–7.<br />

5. Bastin, P., Sherwin, T., <strong>and</strong> Gull, K. (1998) Paraflagellar rod is vital for<br />

trypanosome motility. Nature 391, 548–548.<br />

6. De Souza, W. (1984) Cell biology of Trypanosoma cruzi. Int. Rev. Cytol. 86,<br />

197–285.<br />

7. Ruiz, A. M., Esteva, M., Riarte, A., Subias, E., <strong>and</strong> Segura, E.L. (1986) Immunoprotection<br />

of mice against Trypanosoma cruzi with a lyophilized flagellar fraction<br />

of the parasite plus adjuvant. Immunol Lett. 12, 1–4.


Cell <strong>Fractionation</strong> of Parasitic Protozoa 331<br />

8. Cunha-e-Silva, N. L., Hasson-Voloch, A., <strong>and</strong> De Souza, W. (1989) Isolation <strong>and</strong><br />

characterization of a highly purified flagellar membrane fraction from trypanosomatids.<br />

Mol. Biochem. Parasitol. 37, 129–36.<br />

9. Cunha, N. L., De Souza, W., <strong>and</strong> Hasson-Voloch, A. (1984) Isolation of<br />

the flagellum <strong>and</strong> characterization of the paraxial structure of Herpetomonas<br />

megaseliae. J. Submicrosc. Cytol. 16, 705–13.<br />

10. Robinson, D., Beattie, P., Sherwin, T., <strong>and</strong> Gull, K. (1991) Microtubules, tubulin,<br />

<strong>and</strong> microtubule-associated proteins of trypanosomes. Methods Enzymol. 196,<br />

285–99.<br />

11. Gadelha, C., Lebowitz, J. H., Manning, J., Seebeck, T., <strong>and</strong> Gull, K. (2004)<br />

Relationships between the major kinetoplastid paraflagellar rod proteins: a consolidating<br />

nomenclature. Mol. Biochem. Parasitol. 136, 113–115.<br />

12. Moreira-Leite, F.F., De Souza, W. <strong>and</strong> Cunha-e-Silva, N.L. (1999) Purification of<br />

the paraflagellar rod of the trypanosomatid Herpetomonas megaseliae <strong>and</strong> identification<br />

of some of its minor components. Mol. Biochem. Parasitol. 104, 131–40.<br />

13. Cunha-e-Silva, N. L., Atella, G. C., Porto-Carreiro, I. A., Morgado-Diaz, J. A.,<br />

Pereira, M. G., <strong>and</strong> De Souza, W. (2002) Isolation <strong>and</strong> characterization of a<br />

reservosome fraction from Trypanosoma cruzi. FEMS Microbiol. Lett. 214, 7– 12.<br />

14. King, T. E. (1967) The Keilin-Hartree heart-muscle preparation. Methods Enzymol.<br />

10, 202–8.<br />

15. Rodrigues, C. O., Scott, D. A., <strong>and</strong> Docampo, R. (1999) Characterization of a<br />

vacuolar pyrophosphatase in Trypanosoma brucei <strong>and</strong> its localization to acidocalcisomes.<br />

Mol. Cell. Biol. 19, 7712–23.<br />

16. Figueiredo, R. C. B. Q. <strong>and</strong> Soares, M. J. (1995) The Golgi complex of<br />

Trypanosoma cruzi epimastigote forms. J. Submic. Cytol. Pathol. 27, 209–15.<br />

17. Morgado-Díaz, J. A., Monteiro-Leal, L. H., <strong>and</strong> De Souza, W. (1996) Tritrichomonas<br />

foetus: Isolation <strong>and</strong> Characterization of the Golgi Complex. Exp.<br />

Parasitol. 83, 174–183.<br />

18. Benchimol, M., Ribeiro, K. C., Mariante, R. M., <strong>and</strong> Alderete, J. F. (2001) Structure<br />

<strong>and</strong> division of the Golgi complex in Trichomonas vaginalis <strong>and</strong> Tritrichomonas<br />

foetus. Eur. J. Cell Biol. 80, 593–607.<br />

19. Morgado-Díaz, J. A., Nakamura, C. V., Agrellos, O. A. A., et al. (2001) Isolation<br />

<strong>and</strong> characterization of the Golgi complex of the protozoan Trypanosoma cruzi.<br />

Parasitology 123, 33–43.<br />

20. Benchimol, M., Almeida, J. C. A., <strong>and</strong> De Souza, W. (1996) Further studies on<br />

the organization of the hydrogenosomes in Tritrichomonas foetus. Tiss. Cell. 28,<br />

287– 299<br />

21. Morgado-Díaz, J. A. <strong>and</strong> De Souza, W. (1997) Purification <strong>and</strong> biochemical characterization<br />

of the hydrogenosomes of the flagellate protozoan Tritrichomonas foetus.<br />

Eur. J. Cell Biol. 74, 85–91.


Index<br />

A<br />

Acrylamide-Bis solution, 205<br />

Affinity Depletion Cartridge for removal<br />

of HSA, 91<br />

Albumin <strong>and</strong> immunoglobulin-depleted<br />

plasma proteins<br />

1D gel electrophoresis assessment of<br />

fractions, 20–21<br />

UV absorbance assessment of<br />

fractions, 21–22<br />

Albumin (BSA) st<strong>and</strong>ards color response<br />

curve using BCA assay, 123<br />

Albumin-depleted plasma proteins<br />

1D gel electrophoresis assessment of<br />

fractions, 18–19<br />

UV absorbance assessment of<br />

fractions, 19–20<br />

Alkaline phosphatase assay, biochemical<br />

principle, 252, 254<br />

Anti-HSA POROS cartridge, 23<br />

Anti-human albumin monoclonal<br />

antibody, 15<br />

Arabidopsis thaliana chloroplasts,<br />

isolation <strong>and</strong> preparation,<br />

171–172<br />

chloroplast isolation, 174–176,<br />

178–181<br />

establishing yield <strong>and</strong> intactness of<br />

chloroplasts, 176<br />

growth of Arabidopsis seedlings,<br />

173–174, 177<br />

preparation for proteomics, 176–177,<br />

182–183<br />

B<br />

BCA Protein Assay Kit, 117<br />

Beconase AQ pump aspirator spray<br />

device, 79, 80<br />

333<br />

Bicinchoninic acid protein assay, 30,<br />

121, 297<br />

Biological Variation Analysis mode<br />

(BVA), 11<br />

Bovine serum albumin (BSA), 204<br />

Bradford protein assay, 121<br />

Bronchoalveolar lavage fluid (BALF)<br />

cellular pattern of, 72–73<br />

2-DE analysis, 71–72<br />

during fiber-optic bronchoscopy, 67<br />

proteome analysis for, 68<br />

sample preparing<br />

dialysis-lyophilization, 71<br />

ultra filtration, 71<br />

C<br />

C. albicans protoplasts in active cell wall<br />

regeneration, proteomics of<br />

proteins secreted from, 261<br />

Castor bean endosperm isolation <strong>and</strong><br />

fractionation of ER for<br />

proteomic analysis, 203–214<br />

isolation of, 206–208<br />

mass spectrometer compatible silver<br />

staining, 205, 209–211<br />

<strong>2D</strong> gel protein profiling of, 205–206,<br />

211–213<br />

SDS-<strong>PAGE</strong> analysis of developing<br />

<strong>and</strong> germinating of, 208–209<br />

SDS-<strong>PAGE</strong> analysis of purified ER,<br />

204–205<br />

tissue homogenization <strong>and</strong> ER<br />

purification, 204<br />

Cell physiology <strong>and</strong> disease molecular<br />

study, 139<br />

cell culture <strong>and</strong> lysis, 142<br />

isolation of nickel-binding proteins,<br />

142, 143–144


334 Index<br />

preparing samples for isolation of<br />

nickel-binding<br />

proteins, 143<br />

silver staining of metal-affinity<br />

enriched proteins compatible for<br />

mass spectrometry (MS),<br />

142–144<br />

Cell wall incorporation of GPI-anchored<br />

proteins model, 232<br />

Cell wall proteins (CWP), 187<br />

analysis<br />

<strong>and</strong> extraction protocol by<br />

bioinformatics, 198<br />

separation <strong>and</strong> identification of<br />

protein, 197–198<br />

extraction <strong>and</strong> analysis from liquid<br />

culture medium of<br />

seedlings, 189<br />

Cellular fungi sample preparation<br />

procedure, 261–266<br />

cell culture, 267<br />

cell disruption <strong>and</strong> protein<br />

solubilization, 267, 268–270<br />

S-35 in vitro cell labeling, 267–268<br />

scintillation counting <strong>and</strong> protein<br />

assay, 268, 270<br />

Cerebrospinal fluid (CSF)<br />

LC/MS/MS analysis processing, 63<br />

processing for 2-Dimensional gel<br />

electrophoresis, 59–60<br />

proteomics, 53–56<br />

gel electrophoresis, 57<br />

LC-MS/MS mass spectrometry, 58<br />

MALDI-TOF mass<br />

spectrometry, 58<br />

processing for 2-dimensional gel<br />

electrophoresis, 59–60<br />

processing for LC/MS/MS analysis,<br />

57–58<br />

silver staining <strong>and</strong> in-gel<br />

digestion, 57<br />

simplification by removal of<br />

abundant proteins, 58–59<br />

Chicken IgY antibodies, immunoaffinity<br />

fractionation of plasma proteins<br />

IgY-12 <strong>and</strong> LC2/ LC10 high capacity<br />

spin column kit, 43<br />

IgY-12 microbeads for 96-well filter<br />

plates, 43<br />

SDS gel electrophoresis, 43–44<br />

Chloramphenicol, 224<br />

Chloroplast<br />

isolation buffer (CIB), 174<br />

isolation procedure, 175<br />

light <strong>and</strong> electron micrographs, 180<br />

lysis <strong>and</strong> solubilization, 182<br />

protein samples by DIGE<br />

<strong>2D</strong>-<strong>PAGE</strong>, 182<br />

proteomic studies, 173<br />

CLUSTAW program in PIR, 84<br />

Collision-induced dissociation (CID), 136<br />

Cysteine-specific protein labeling using<br />

CyDye DIGE fluor saturation<br />

dyes, 2<br />

Cytoplasmatic proteins<br />

buffers for western blot analysis, 103<br />

cell culture <strong>and</strong> wash buffer, 102<br />

cell harvest, 104<br />

comparison of <strong>2D</strong>E pattern with<br />

pattern of whole cell<br />

protein, 109<br />

coomassie brilliant blue staining<br />

solutions, 104<br />

isolation from cultured cells 2-D gel<br />

electrophoresis, 101, 107–108<br />

buffers <strong>and</strong> reagents for, 102–103<br />

precipitation of cytoplasmatic<br />

proteins, 104–105<br />

silver staining solutions, 103<br />

western blot analysis for detection of<br />

purity, 105–107<br />

D<br />

DeCyder software, 10–11<br />

Depleted protein samples TCA<br />

precipitation for <strong>2D</strong> gel<br />

electrophoresis, 22–23


Index 335<br />

Differential in-gel analysis (DIA), 11<br />

DIGE saturation labeling conditions, 7<br />

DNA electrophoresis, 156<br />

DryStrip Reswilling tray, 70<br />

Dulbecco’s Phosphate Buffered Saline<br />

(D-PBS), 116<br />

E<br />

Econo-Pac ® 10DG desalting column, 190<br />

Electrospray quadrupole time of flight<br />

mass spectrometer, 80, 83<br />

Eosin stain, 133<br />

Epithelium lining fluid (ELF), 67<br />

S-Ethanol-cysteine residue, 63<br />

Etioplasts, dark-grown plants, 173<br />

F<br />

FFPE tissue, 137<br />

Filamentous fungi secreted proteins<br />

isolation <strong>and</strong> enrichment,<br />

274–284<br />

centrifugation <strong>and</strong> ultrafiltration, 277<br />

enzymatic <strong>and</strong> chemical<br />

deglycosylation, 277–278<br />

filtration <strong>and</strong> lyophilization, 277<br />

isolation <strong>and</strong> concentration of<br />

supernatant broth, 278–280<br />

precipitation with trichloroacetic acid,<br />

methanol <strong>and</strong> chloroform, 277<br />

SDS-<strong>PAGE</strong>, 278, 282, 301<br />

two-dimensional gel electrophoresis,<br />

278, 282<br />

Fluorescence dye labeling<br />

optimization, 6–7<br />

Fluorescence resonance energy transfer<br />

(FRET), 4<br />

Foetal bovine serum (FBS), 116<br />

Formalin-fixed, paraffin-embedded<br />

(FFPE) tissues, 132<br />

sample preparation for mass<br />

spectrometry analysis, 131<br />

laser-capture microdissection, 134<br />

LC-MS/MS analysis of tryptic<br />

peptides, 133–134<br />

mass spectrometry analysis <strong>and</strong><br />

bioinformatic<br />

analysis, 136<br />

nanoflow RPLC-MS/MS analysis,<br />

135–136<br />

protein extraction <strong>and</strong> trypsin<br />

digest, 133–135<br />

tissue processing, 133, 134<br />

trypsin digest, 133<br />

trypsin-mediated 18 O-labeling, 135<br />

G<br />

Gamborg liquid medium, 190<br />

Gel pieces, reduction <strong>and</strong> alkylation,<br />

61–62<br />

Glycosyl phosphatidylinositol (GPI)-cell<br />

wall proteins, 220<br />

Gram-positive bacterial cell<br />

wall-associated proteins<br />

isolation <strong>and</strong> solubilization<br />

bacterial culture, 296, 299<br />

BCA protein assay, 300<br />

casting <strong>2D</strong> gels, 304<br />

cell wall-associated proteins<br />

extractions, 296–297, 300<br />

first dimension IEF, 303–304<br />

protein detection <strong>and</strong> gel<br />

documentation, 306<br />

SDS-polyacrylamide gel<br />

electrophoresis (SDS-<strong>PAGE</strong>),<br />

297<br />

second dimension SDS-<strong>PAGE</strong>,<br />

304–306<br />

trichloroacetic acid (TCA)<br />

precipitation, 298, 302<br />

Green plant tissue protein extraction,<br />

149–152<br />

H<br />

Hematoxylin stained histological<br />

classification, 5<br />

Herpetomonas megaseliae, 319, 320<br />

HiLoad 16/60 Superdex 75 prep grade<br />

column, 91


336 Index<br />

Hi-Trap SP equilibration<br />

buffer, 190<br />

HONE1 cells, Western blot analysis of<br />

cytoplasmatic fraction, 107<br />

Human BALF proteins, 2-D gel<br />

electrophoresis, 69<br />

Human nickel allergy, 140<br />

Human plasma<br />

albumin <strong>and</strong> immunoglobulin<br />

depletion of, 15–19<br />

proteins by 1D gel electrophoresis,<br />

20–21<br />

albumin depletion, 16–17, 18<br />

albumin-depleted plasma proteins,<br />

18–20<br />

blood sampling, 16<br />

chromatography of immunoaffinity<br />

separation of using IgY-12 high<br />

capacity LC2 column, 46<br />

1D <strong>and</strong> <strong>2D</strong> gel Electrophoresis, 17<br />

immunoglobulin depletion, 17, 20<br />

samples collection of, 17–18<br />

TCA precipitation of proteins, 17<br />

Human serum<br />

chromatogram for affinity removal of<br />

high-abundant proteins from, 31<br />

<strong>2D</strong> gel electrophoresis of, 34<br />

1D SDS gel electrophoresis of, 33<br />

high-abundant proteins,<br />

immunoaffinity depletion of,<br />

29–30<br />

immunodepleted serum <strong>and</strong> bund<br />

fraction processing, 30<br />

low-abundant proteins, reversed-phase<br />

separation of, 30, 35–36<br />

multi-component immunoaffinity<br />

subtraction <strong>and</strong> reversed-phase<br />

chromatography, 27–39<br />

proteins composition, 28<br />

I<br />

IgY-12 Microbeads regeneration, 45<br />

ImageQuant software, 11<br />

Iminodiacetic acid (IDA), 140<br />

Immobiline Dry Strip (IPG) kit, 205<br />

Immobiline Dry Strip Reswelling<br />

Tray , 205<br />

Immobiline DryStrip Kit, 118<br />

Immobilized metal ion affinity<br />

chromatography (IMAC), 139<br />

Immobilized pH gradient, 70<br />

Immunoaffinity subtraction (IAS), 94<br />

Immunoglobulin depletion, 20<br />

In-gel digestion procedure, 61–62<br />

Inter simple sequence repeat PCR<br />

(ISSR-PCR) technique, 156<br />

IPG strips, 71–72<br />

IPG-strip rehydration buffer, 22–23<br />

Isoelectric focusing (IEF) tube<br />

gels, 9<br />

K<br />

Keratin proteome in normal<br />

NLF, 84<br />

Kinematica Model PT10-35, 174<br />

Knexus program, 63<br />

L<br />

Labile CWP analysis, 191<br />

Labile /weakly bound CWP extraction by<br />

nondestructive techniques<br />

cell suspension cultures, 194<br />

liquid culture medium of seedlings,<br />

193–194<br />

Laemmli sample buffer, 17<br />

LC-MS/MS mass spectrometry, 63–64<br />

Liquid chromatography-coupled<br />

electrospray ionisation MS<br />

(LC-ESI-MS), 12<br />

Liquid Tissue -MS protein prep<br />

kit, 133<br />

Liver Infusion Trypticase Medium<br />

(LIT), 315<br />

Lowry protein assay, 80<br />

LTQ ion trap mass spectrometer, 64<br />

Lumbar puncture (LP), 54<br />

Lysis buffers, 176


Index 337<br />

M<br />

Madin-darby canine kidney (MDCK)<br />

cells sample preparation from<br />

culture medium, 113–128<br />

cell culture, 114–116, 119<br />

2-D gel electrophoresis of, 115<br />

protein quantitation by BCA assay,<br />

117, 121–122<br />

rehydration loading <strong>and</strong> isoelectric<br />

focusing, 117–118, 122–125<br />

ultracentrifugation <strong>and</strong> ultrafiltration,<br />

116–117, 119–120<br />

MALDI-TOF spectrometry, 58, 63<br />

MASCOT MS/MS ion search<br />

software, 84<br />

Mayer’s hematoxylin stain, 133<br />

Metal oxide affinity chromatography<br />

(MOAC), 140<br />

Metal-specific allergic contact dermatitis<br />

(ACD), 141<br />

Mixed bed ion exchanger resin, 118<br />

Multidimensional liquid chromatography<br />

(multi-LC), 69<br />

Multiple Affinity Removal Spin<br />

Cartridge, 91<br />

Multiple Affinity Removal System<br />

column, 30<br />

Murashige <strong>and</strong> Skoog (MS)<br />

medium, 174<br />

N<br />

Nanoflow reversed-phase liquid<br />

chromatography<br />

(nanoRPLC), 133<br />

Nasal lavage fluid (NLF) analysis<br />

data interpretation, 84<br />

by liquid chromatography <strong>and</strong> mass<br />

spectrometry, 79, 81–83<br />

preparation, 80–81<br />

acid-ethanol precipitation, 81<br />

protein digestion, 81<br />

total protein assay, 80<br />

total ion chromatogram (TIC) profile<br />

of, 82<br />

Nasal secretions<br />

nasal provocation, 79–80<br />

pharmacological treatment, 78–79<br />

preparation for proteome analysis,<br />

77–78<br />

Ni-affinity enriched proteins from human<br />

antigen, 141<br />

Nickel-nitrilotriacetic acid<br />

(Ni-NTA), 142<br />

Ni-NTA Magnetic Agarose Beads, 143<br />

Nitrilotriacetic acid (NTA), 140<br />

Nonequilibrium pH gradient gel<br />

electrophoresis (NEPHGE), 113<br />

P<br />

Pancreatic adenocarcinoma cells<br />

proteome analysis, 4–5<br />

Pancreatic intraepithelial neoplasia, 1–2<br />

PanIN, Pancreatic intraepithelial<br />

neoplasia, 2<br />

Parasitic protozoa cell fractionation,<br />

313–314<br />

cell culture <strong>and</strong> st<strong>and</strong>ard equipment,<br />

315–316<br />

electron microscopy, 316<br />

hydrogenosome isolation <strong>and</strong><br />

subfractionation, 327–329<br />

isolation <strong>and</strong> subfractionation<br />

flagellum of trypanosomatids, 316<br />

hydrogenosomes, 317<br />

isolation of<br />

golgi complex, 316–317, 323–327<br />

reservosome, 316, 320–321<br />

trypanosomatid structures, 317–320<br />

Pepper seed DNA, 161–162<br />

Peptide mass fingerprinting (PMF), 12<br />

Percoll gradient solutions, 165<br />

Phenylmethylsulfonyl fluoride<br />

(PMSF), 116<br />

PIR BLAST software, 84<br />

Plant cell culture isolation of<br />

mitochondria, 163–169<br />

crude organelle pellet differential<br />

centrifugation of, 165–166


338 Index<br />

density gradient purification of<br />

mitochondria, 167<br />

materials, 164–165<br />

protoplasts disruption <strong>and</strong><br />

isolation, 165<br />

Plant cell wall poteins isolation, 191–192<br />

extraction <strong>and</strong> analysis from cell<br />

suspension cultures, 190–191<br />

labile/weakly extraction by<br />

nondestructive techniques,<br />

189–190<br />

Rosette leaves extraction <strong>and</strong> analysis,<br />

191–192<br />

Plant material high-throughput extraction<br />

device<br />

DNA electrophoresis, 156, 159–160<br />

inter simple sequence repeat PCR<br />

(ISSR-PCR), 156, 159<br />

isoelectric focusing (IEF), 155–156,<br />

158–159<br />

sample preparation, 154–155, 157–158<br />

Plant mitochondria Percoll gradient<br />

purification, 166<br />

Plant proteases, 149<br />

Plastids organelles, 171<br />

Plastoglobuli lipid-containing structures,<br />

173<br />

POROS ® beads, 17<br />

Protean IEF Cell, 70<br />

Protease inhibitor cocktail tablets, 70<br />

Protein<br />

concentration estimation kit, 176<br />

content after, sinusitis NLFs pre- <strong>and</strong><br />

post-pharmacological<br />

treatment, 82<br />

deglycosylation, 281–282<br />

with PNGase F, 282<br />

trifluoromethanesulfonic acid with,<br />

282–283<br />

Proteins<br />

analysis by fluorescence dye<br />

saturation labeling <strong>and</strong> 2-DE, 2<br />

cysteine-specific protein labeling<br />

using CyDye DIGE fluor<br />

saturation dyes, 2<br />

<strong>2D</strong>-<strong>PAGE</strong>, 3<br />

fluorescence dye labeling<br />

optimisation of, 6–7<br />

isoelectric focusing, 2–3<br />

microdissection <strong>and</strong> sample<br />

preparation, 2<br />

microdissection for proteome<br />

analysis of pancreatic<br />

adenocarcinoma cells, 4<br />

minimal number of microdissected<br />

cells determination of, 4–6<br />

protein spots micropreparation<br />

of, 12<br />

reference proteome for internal<br />

st<strong>and</strong>ardization <strong>and</strong> protein<br />

identification, 7–8<br />

two-dimensional gel<br />

electrophoresis, 9–10<br />

extraction from green plant tissue,<br />

149–150, 152<br />

materials <strong>and</strong> methods, 151<br />

G POROS column, 20<br />

G Sepharose, 58<br />

identification <strong>and</strong> reference proteome<br />

for internal st<strong>and</strong>ardization, 8–9<br />

loads for silver <strong>and</strong> Coomassie blue<br />

staining, 108<br />

precipitation<br />

chloroform/methanol/trichloroacetic<br />

acid with, 280–281<br />

quantitation by BCA Assay, 117–118<br />

secretion from yeast protoplasts in<br />

active cell wall<br />

regeneration, 241<br />

alkaline phosphatase assay, 247,<br />

250–254<br />

collection of proteins, 245–249<br />

concentration of proteins secreted<br />

from regenerating protoplasts,<br />

247, 250


Index 339<br />

preparation of yeast<br />

protoplasts, 249<br />

recovery of proteins secreted from<br />

regenerating protoplasts,<br />

246, 250<br />

Protoplast formation efficiency of<br />

evaluation, 259<br />

R<br />

Recombinant His-tagged proteins, 140<br />

Reverse-phase C18 column, 79<br />

Rosette leaves<br />

labile CWP analysis by 2-DE, 196<br />

proteins extraction, 194–196<br />

vacuum-infiltration, 195<br />

S<br />

S. cerevisiae <strong>and</strong> C. albicans, cell wall,<br />

218, 221–222<br />

S. pyogenes strain 5448 cell wall<br />

proteome, 305<br />

SEQUEST, 64<br />

Silver staining procedures, 60<br />

Size exclusion chromatography<br />

(SEC), 91<br />

Spectra/Por ® cellulose ester, 190<br />

Speed Vac centrifugation, 73<br />

Streptococcus mutans, silver stained 2-D<br />

gel, 290<br />

T<br />

TCA/Acetone precipitation, 50, 120<br />

Terminator device, 154–155<br />

Triacyglycerol (TAG) biosynthesis, 203<br />

Trichomonad structures isolation, 325<br />

Trifluoromethanesulfonic acid, chemical<br />

protein deglycosylation with,<br />

281–282<br />

Tritrichomonas foetus, 315<br />

Trypanosoma cruzi, 314<br />

Trypsin-mediated incorporation of<br />

O 18 , 137<br />

Trypticase Yeast Maltose Medium<br />

(TYM), 315<br />

U<br />

Ultraflex MALDI-TOF spectrometer, 143<br />

Urinary protein concentrates, size<br />

exclusion chromatography,<br />

90, 93<br />

Urine samples preparation for proteomic<br />

analysis, 89<br />

final sample preparation for proteomic<br />

analysis, 91<br />

immunoaffinity subtraction of<br />

proteins, 91<br />

size exclusion chromatography of<br />

urinary protein concentrates, 90,<br />

92–94<br />

urinary protein concentrate<br />

preparation, 90, 91<br />

V<br />

Vivapure Anti-HSA/IgG removal kit, 91<br />

Vmax ® microplate reader, 117<br />

Voyager DE-Pro mass spectrometer, 63<br />

W<br />

Weakly bound CWP analysis,<br />

191–192, 196<br />

Weakly bound CWP extraction<br />

destructive techniques, 192,<br />

196–197<br />

cell wall preparation, 192,<br />

196–197<br />

protein extraction <strong>and</strong> separation,<br />

192, 197<br />

Western blot assembly, 106<br />

Y<br />

Yeast <strong>and</strong> fungal cell walls protein<br />

solubilization<br />

-1,3 glucanase treatment,<br />

224–225, 229<br />

detergents <strong>and</strong> reducing agents,<br />

224, 228


340 Index<br />

<strong>and</strong> exochitinase treatment, 225,<br />

229–230<br />

under mild alkali conditions, 224,<br />

228–229<br />

Yeast <strong>and</strong> fungal proteomics cell wall<br />

fractionation, 217–237<br />

cell wall isolation from yeasts <strong>and</strong><br />

filamentous fungi, 223–228<br />

protein precipitation, 225, 230<br />

protein solubilization from isolated<br />

yeast <strong>and</strong> fungal cell walls,<br />

224–225, 228<br />

Yeast proteome 2-D electrophoresis,<br />

269–270<br />

Yeast-Peptone-D-glucose<br />

(YPD), 222

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!