17.01.2015 Views

2011 ANNUAL REPORT - Istituto Pasteur

2011 ANNUAL REPORT - Istituto Pasteur

2011 ANNUAL REPORT - Istituto Pasteur

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>2011</strong><br />

<strong>ANNUAL</strong> <strong>REPORT</strong>


2012 – <strong>Istituto</strong> <strong>Pasteur</strong>-Fondazione Cenci Bolognetti<br />

Edited by Lucia Ugo<br />

P.le Aldo Moro 5 – 00185 Roma<br />

pasteurcenci@uniroma1.it<br />

www.istitutopasteur.it


Contents<br />

Contents<br />

• Forward 7<br />

• Boards and Staff 8<br />

• Fellowships awarded 9<br />

• Conferences, Seminars and Bookshop talks 13<br />

• Scientific Reports 15<br />

Research Area 1: Molecular biology of microorganism and viruses<br />

Andrea Bellelli, Structural biology of the thiol-dependent redox systems of Schistosoma<br />

mansoni and Plasmodium falciparum 17<br />

Alberto Boffi, Bacterial globins as regulators of thiol redox equilibrium in bacteria 19<br />

Daniela De Biase, The acid resistance genes of Escherichia coli: does their complex<br />

transcriptional control hide novel biological roles 21<br />

Alberto Faggioni, Epstein-Barr virus interactions with cellular microRNAs 23<br />

Claudio Falcone, Study of RNA damage in yeast as a model for age-related degenerative<br />

diseases 25<br />

Paola Londei, From leaderless to leadered mRNAs: mRNA features modulating ribosome<br />

binding and translation initiation in Archaea 27<br />

Research Area 2: Pathogenic mechanisms of microbially associated diseases<br />

Guido Antonelli, Molecular characterization of viruses causing bronchiolitis and study of<br />

viral and host factors affecting Type I IFN antiviral response induced by respiratory viruses 31<br />

Maria Lina Bernardini, Lipopolysaccharide and peptidoglycan adaptation to host as an<br />

immune evasion strategy of gram-negative pathogens 33<br />

Bianca Colonna, Involvement of sRNA molecules in the complex regulatory circuits of<br />

virulence gene expression in Shigella flexneri and in enteroinvasive E.coli 35<br />

Francesca Cutruzzolà, Inhibition of Pseudomonas aeruginosa biofilms: new molecular<br />

strategies targeting cyclic-di-GMP metabolism 37<br />

Paolo Sarti, Defense mechanisms against oxidative and nitrosative stress in pathogenic<br />

protozoa 39<br />

Vincenzo Vullo, Immunopathogenesis of HIV infection: study of innate immunity and<br />

dendritic cells 41<br />

Research Area 3: Molecular genetics of eukaryotes<br />

Fiorentina Ascenzioni, Assembly and functional analysis of genomic context vectors<br />

containing the human CFTR locus 45<br />

Paola Ballario, Light control of filamentous fungi life cycle: from system models to<br />

applications 47<br />

Silvia Bonaccorsi, Functional analysis of morgana, a gene involved in the control of<br />

centrosome duplication 49<br />

Irene Bozzoni, RNA-RNA and RNA-protein interactions: role of small non-coding RNAs in<br />

gene expression control 51<br />

3


Contents<br />

Emanuele Cacci, Role of metalloproteinases and their tissue inhibitors in the regulation of<br />

neurogenesis and gliogenesis from neural stem/progenitor cells 53<br />

Paola Caiafa, Does PARylated PARP-1 introduce an epigenetic mark on chromatin 55<br />

Giorgio Camilloni, DNA recombination of repeated sequences and genome instability:<br />

epigenetic implications 57<br />

Antonella De Jaco, Processing of the Neuroligins proteins and autism-related mutations 59<br />

Patrizio Dimitri, Functional analysis of CG40218, a Drosophila melanogaster gene<br />

encoding a BCNT-like protein required for chromosome organization 61<br />

Maurizio Gatti, Genetic and molecular analysis of the mechanisms of Drosophila telomere<br />

protection 63<br />

Alessandro Giacomello, Role of epicardium and EMT/MET processes in cardiac progenitor<br />

cells generation and differentiation 65<br />

Alberto Gulino, Hedgehog signaling regulatory networks in brain cancer stem cells 67<br />

Marco Lucarelli, The interplay between epigenetics, cell cycle and homologous<br />

recombination in gene therapy by Small Fragment Homologous Replacement (SFHR) 69<br />

Rossella Maione, Interplay between myogenic factors and cell cycle control: regulation and<br />

role of the cdk inhibitor p57kip2 71<br />

María Elena Miranda Banos, Handling and toxicity of mutant serpins underlying human<br />

disease 73<br />

Antonio Musarò, Characterization of the muscle-nerve crosstalk in a neuromuscular<br />

disease 75<br />

Rodolfo Negri, Role of the COP9 signalosome in transcription modulation and chromatin<br />

organization in yeast and plants 77<br />

Sergio Pimpinelli, The biogenesis of piRNAs and their involvement in transposon silencing<br />

and heterochromatin formation in Drosophila 79<br />

Sabrina Sabatini, To the root of organ growth: the control of root meristem activity in<br />

Arabidopsis 81<br />

Mario Stefanini, Biological characterization and in vitro culture of spermatogonial stem cells 83<br />

Marco Tripodi, Molecular mechanisms driving liver stem cell fate 85<br />

Giuseppe Zardo, Profiling the Polycomb/Trythorax target genes in normal and leukemic<br />

hematopoiesis 87<br />

Research Area 4: Molecular recognition in biomolecules<br />

Enzo Agostinelli, Toxic effects induced by polyamine metabolites on melanoma cells: a<br />

new therapeutic approach 91<br />

Fabio Altieri, Cellular response to oxidative stress: involvement of protein disulfide<br />

isomerases 93<br />

Paolo Bianco, Role of Gsα signalling in the bone marrow hematolymphopoietic<br />

microenvironment as revealed by novel transgenic models 95<br />

Maurizio Brunori, The dark side of protein folding: denatured states and misfolded species<br />

in molecular recognition and pathological processes 97<br />

Stefano Cacchione, The role of nucleosomes in the stability of human telomeres 99<br />

Felice Cervone, Plant innate immunity: signalling and recognition of Damage-Associated<br />

Molecular Patterns (DAMPs) 101<br />

Ernesto Di Mauro, Spontaneous generation and evolution of genetic information 103<br />

Cristina Limatola, Molecular and functional approaches to investigate the neuroprotective<br />

and neuromodulatory roles of chemokines and their receptors in the central nervous<br />

system 105<br />

4


Contents<br />

Paola Paggi, Neurone response to experimental injury and lack of dystrophin: a molecular,<br />

functional and structural study in autonomic ganglia in vivo and in vitro 107<br />

Maria Rosaria Torrisi, Molecular mechanisms integrating endocytosis and signalling of<br />

fibroblast growth factor receptors 109<br />

Research area 5: Cellular and molecular immunology<br />

Vincenzo Barnaba, Interplay amongst chronic immune activation, apoptosis, crosspresentation,<br />

immune-regulation, and autoimmunity 113<br />

Rossella Paolini, Signalling events negatively regulating FceRI expression and mast cell<br />

functional responses 115<br />

Angela Santoni, Anti-tumor effector functions of NK cells in tumor microenviroment 117<br />

Isabella Screpanti, Dissection of Notch signaling-dependent pathways involved in the<br />

progression of T cell leukemia 119<br />

Rosa Sorrentino, Predisposing factors in autoimmune diseases: correlation between<br />

common genetic variations and function 121<br />

Loretta Tuosto, CD28 co-stimulatory molecule as a key regulator of NF-κB signalling<br />

pathway: role of cytoskeleton in coupling CD28 to NF-κB activation 123<br />

Elio Ziparo, Anti-tumor pathways mediated by innate immune responses 125<br />

Research area 6: New antimicrobial and antiviral agents<br />

Donatella Barra, Peptide effectors of innate immunity 129<br />

Mariangela Biava, New pyrrole derivatives of BM 212: a new class of antimycobacterial<br />

agents. Design, synthesis, biological evaluation and study of their mode of action 131<br />

Roberto Di Santo, New azole derivatives as antiprotozoal agents 133<br />

Romano Silvestri, Drug design and synthesis of non-nucleoside inhibitors of both HIV-1<br />

wild type and resistant mutant strains reverse transcriptase and Coxsackie B4 virus 135<br />

Research area 7: Biology of malaria and other vector-borne diseases<br />

Alessandra Della Torre and Vincenzo Petrarca, Genetic and phenotypic characterization<br />

of species and “molecular forms” of the Anopheles gambiae complex (Diptera: Culicidae),<br />

afrotropical malaria vectors 139<br />

David Modiano, Immune responses to malaria and autoimmune disorders: investigating<br />

common gene-regulatory networks 141<br />

Start-up program<br />

Giuseppe Lupo, Specification and maintenance of retinal stem cells 145<br />

5


Annual Report <strong>2011</strong><br />

Forward<br />

The <strong>Istituto</strong> <strong>Pasteur</strong>-Fondazione Cenci Bolognetti is a private non-profit foundation<br />

established according to the terms of the bequest of Princess Beatrice Fiorenza Cenci<br />

Bolognetti to Sapienza University of Rome for the purpose of encouraging scientific research.<br />

The funding of research projects is possible thanks to the income from real estate and, this<br />

year, thanks also to the support of BNP Paribas BNL, that we gratefully acknowledge.<br />

The <strong>Istituto</strong> <strong>Pasteur</strong>-Fondazione Cenci Bolognetti, the Italian member of the Institut<br />

<strong>Pasteur</strong> International Network, is committed to studying the mechanisms that regulate the<br />

basic processes of life, for the understanding and treatment of severe human pathologies for<br />

which there are no cures as yet, and for improving the existing ones.<br />

Researchers in the areas of biology, medicine, medicinal chemistry, bioinformatics and<br />

biotechnologies are dedicated to the study and development of novel drugs as well as<br />

cellular and gene therapies for the treatment of emerging, or re-emerging infectious diseases<br />

(influenza, herpes, tuberculosis, HIV, HCV); vector-borne diseases (malaria, Dengue fever);<br />

neurodegenerative and neoplastic diseases; disorders of the immune and haematopoietic<br />

systems (leukemia). Furthermore, the Institute has been active in promoting the study of<br />

stem cell biology and of the use of cardiac, muscle, hepatic, neuronal and male reproductive<br />

system stem cells in the repair of damaged tissues.<br />

Our Institute finances high-level research projects selected through a peer review<br />

system. We give special attention to the new generation of researchers awarding fellowship<br />

grants that support their training in highly advanced research centres abroad, upon their<br />

return to Italy and at the beginning of their career following a PhD degree. Recently, we have<br />

also activated a three-year Start-Up program for a young scientist during which he may build<br />

a solid research project and attract independent funding.<br />

Moreover, we are active in promoting scientific communication. We organize seminars,<br />

conferences and international meetings, which are important occasions for the comparison of<br />

results. We also promote a school project involving a series of meetings and practical<br />

activities with students, so they can learn to appreciate the importance of science. Lessons<br />

and courses in the fields of genetics, biotechnology, microbiology, genomics, evolution<br />

biology, ecology and neuroscience are carried out in the laboratories of the Sapienza<br />

University of Rome or, when possible, are performed directly by teachers in schools.<br />

Furthermore, we organize cultural and social events, such as bookshop talks, for the wider<br />

public.<br />

Last but not least, our financial commitment is devoted to the construction of laboratories<br />

in which established scientists of any origin may improve our research.<br />

This Annual Report documents the results obtained during this year thanks to the<br />

enthusiasm and the effort of the scientists involved. It is our pleasure to invite you to follow<br />

our activity on our website (www.istitutopasteur.it), where you may find further information.<br />

Paolo Amati<br />

President<br />

Angela Santoni<br />

Scientific Director<br />

7


Annual Report <strong>2011</strong><br />

Boards and Staff<br />

Administrative Board<br />

The Board of Administration is presided<br />

over by a President appointed by the<br />

Rector of Sapienza University of Rome<br />

chosen from a list of three names selected<br />

during a joint session of the board and the<br />

Scientific Council. Members of the board<br />

include a Scientific Director, four members<br />

of the Faculties of Pharmacy and Medicine,<br />

Natural Sciences, and Medicine and<br />

Dentistry. Other members are a legal<br />

expert designated by the University of<br />

Rome Board of Administration, three<br />

auditors nominated by the University, the<br />

Ministry of the Treasury, and the Ministry of<br />

Universities and Scientific Research.<br />

President<br />

Paolo Amati (Pharmacy and Medicine)<br />

Members<br />

Paolo Costantino (Natural Sciences),<br />

Alberto Faggioni (Medicine and<br />

Dentistry), David Modiano (Pharmacy and<br />

Medicine), Angela Santoni (Pharmacy and<br />

Medicine), Romano Silvestri (Pharmacy<br />

and Medicine)<br />

Secretary<br />

Emanuela Gloriani<br />

Administrative Expert<br />

Giuseppina Capaldo<br />

Auditors<br />

Anna Carmela Ferrante, Simona Ranalli,<br />

Carla Vassallo<br />

Scientific Council<br />

The Scientific Council is a board of seven<br />

scholars in the field of the pasteurian<br />

sciences. Members are elected to a fouryear<br />

term by the Faculties of Natural<br />

Sciences, and Pharmacy and Medicine.<br />

The Scientific Director, appointed by the<br />

Scientific Council, is ex-officio a member of<br />

the Board of Administration. The Scientific<br />

Council attends to examine and coordinate<br />

the research programs as well as the<br />

several scientific activities.<br />

Scientific Director<br />

Angela Santoni (Pharmacy and Medicine)<br />

Members<br />

Ernesto Di Mauro (Natural Sciences),<br />

Laura Frontali (Natural Sciences), Anna<br />

Teresa Palamara (Pharmacy and<br />

Medicine), Anna Tramontano (Pharmacy<br />

and Medicine), Marco Tripodi (Pharmacy<br />

and Medicine), Carlo Turano (Pharmacy<br />

and Medicine)<br />

Secretariat<br />

Sarah Gainsforth, Maria Pia Lorenzoni,<br />

Lynda Romani, Nicoletta Silvestri, Lucia<br />

Ugo<br />

Consultants<br />

Tommaso De Dominicis (legal affairs);<br />

Anna Maria Pivetti (architectural<br />

supervision); Barbara Hell (financial<br />

affairs)<br />

8


Annual Report <strong>2011</strong><br />

Fellowships awarded<br />

Fellowships awarded for two-year<br />

training in foreign laboratories<br />

Michele ARDOLINO, from Department of<br />

Molecular Medicine to Dept. of Molecular<br />

and Cell Biology, University of California,<br />

Berkeley, USA<br />

Valerio BERARDI, from Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

to State University of New York, Health<br />

Science Center, NY, USA<br />

Sonia CONI, from Department of Molecular<br />

Medicine to IBDC, Université de Nice<br />

Sophia-Antipolis, France<br />

Daniela DIMASTROGIOVANNI, from<br />

Department of Biochemical Sciences "A.<br />

Rossi Fanelli" to Department of<br />

Biochemistry, University of Cambridge,<br />

UK<br />

Michela ESPOSITO, from Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

to Institut de Génétique et Microbiologie,<br />

Université Paris XI, France<br />

Roberto GAETANI, from Department of<br />

Experimental Medicine to Department of<br />

Cardiology, University Medical Center,<br />

Utrecht, The Netherlands<br />

Francesca GASPARRINI, from<br />

Department of Molecular Medicine to<br />

Cancer Research UK, London Research<br />

Institute, UK<br />

Marta MORETTI, from Department of<br />

Experimental Medicine to Division of<br />

Immunology and Inflammation, Imperial<br />

College, London, UK<br />

Tommaso MOSCHETTI, from Department<br />

of Biochemical Sciences "A. Rossi<br />

Fanelli" to Department of Biochemistry,<br />

University of Cambridge, UK<br />

Giuseppe SCIUMÈ, from Department of<br />

Experimental Medicine to NIH/NIAMS,<br />

Bethesda, MD, USA<br />

Fellowships awarded to students who<br />

had a two-year experience abroad<br />

Daniela CECCARELLI, from Université de<br />

Sherbrooke, Québec, Canada, to<br />

Department of Biology and<br />

Biotechnologies "C. Darwin"<br />

Antonio COLUCCIA, from School of<br />

Pharmacy, University of Cardiff, UK, to<br />

Department of Medicinal Chemistry and<br />

Technologies<br />

Elvira FORTE, from Burnham Institute for<br />

Medical Research, La Jolla, CA, USA, to<br />

Department of Molecular Medicine<br />

Valentina MANGANO, from MRC Centre<br />

for Genomics and Global Health, Oxford,<br />

UK, to Department of Public Health and<br />

Infectious Diseases<br />

Fellowships awarded to students who<br />

have completed their PhD and are<br />

seeking a position<br />

Andrea BRENNA, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Cristina CAPUANO, at Department of<br />

Experimental Medicine<br />

Elisa CESARINI, at Department of Biology<br />

and Biotechnologies "C. Darwin"<br />

Isotta CHIMENTI, at Department of<br />

Experimental Medicine<br />

Laura CURCURÙ, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

9


Annual Report <strong>2011</strong><br />

Alessandra GALATI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Francesca GASPARRINI, at Department<br />

of Experimental Medicine<br />

Emanuela MICHELI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Arianna MONTANARI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Michela MUSCOLINI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Marianna PELLONI, at Department of<br />

Experimental Medicine<br />

Marialaura PETRONI, at Department of<br />

Molecular Medicine<br />

Fabrizio TESTA, at Department of<br />

Biochemical Sciences "A. Rossi Fanelli"<br />

Fellows working on the Institute<br />

research programs<br />

Georgia ABATE, at Department of Cellular<br />

Biotechnologies and Hematology<br />

Angela. A. ALAGIA, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Chiara ARDICCIONI, at Department of<br />

Biochemical Sciences "A. Rossi Fanelli"<br />

Pamela AVELLINO, at Department of<br />

Public Health and Infectious Diseases<br />

Maria Giulia BACALINI, at Department of<br />

Cellular Biotechnologies and Hematology<br />

Cecilia BATTISTELI, at Department of<br />

Cellular Biotechnologies and Hematology<br />

Orlando BORRÁS HIDALGO, at<br />

Department of Biology and<br />

Biotechnologies "C. Darwin"<br />

Anna BUSANELLO, at Department of<br />

Cellular Biotechnologies and Hematology<br />

David CACCHIARELLI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Roberta CALABRESE, at Department of<br />

Cellular Biotechnologies and Hematology<br />

Maria Rosaria CARBONE, at Department<br />

of Cellular Biotechnologies and<br />

Hematology<br />

Mariateresa CARCURO, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Nicoletta CASTIGLIONE, at Department<br />

of Biochemical Sciences "A. Rossi<br />

Fanelli"<br />

Roberta CASTRATARO, at Department of<br />

Physiology and Pharmacology "Vittorio<br />

Erspamer"<br />

Francesca CECERE, at Department of<br />

Molecular Medicine<br />

Arcangela Anna CERA, at Department of<br />

Biochemical Sciences "A. Rossi Fanelli"<br />

Sara CHIARELLA, at Department of<br />

Biochemical Sciences "A. Rossi Fanelli"<br />

Isotta CHIMENTI, at Department of<br />

Experimental Medicine<br />

Fabio CICCARONE, at Department of<br />

Cellular Biotechnologies and Hematology<br />

Raffaela CIPRIANI, at Department of<br />

Physiology and Pharmacology "Vittorio<br />

Erspamer"<br />

Ivan COLADARCI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Elisabetta DAMIA, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Federica DE ANGELIS, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Francesca DE VITO, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Enea Gino DI DOMENICO, at Department<br />

of Biology and Biotechnologies "C.<br />

Darwin"<br />

Maria Letizia DI MARTINO, at Department<br />

of Biology and Biotechnologies "C.<br />

Darwin"<br />

Antonella FARINA, at Department of<br />

Experimental Medicine<br />

Manuela FORNARA, at Department of<br />

Biochemical Sciences "A. Rossi Fanelli"<br />

10


Annual Report <strong>2011</strong><br />

Fedra FRANCOCCI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Alessandra GALATI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Francesca GARIBALDI, at Department of<br />

Cellular Biotechnologies and Hematology<br />

Francesca GASPARRINI, at Department<br />

of Experimental Medicine<br />

Leonardo GIUSTINI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Roberta GONNELLA, at Department of<br />

Experimental Medicine<br />

Gaia GRASSINI, at Department of Medical-<br />

Surgical Sciences and Biotechnologies<br />

Alfonso GRIMALDI, at Department of<br />

Physiology and Pharmacology "Vittorio<br />

Erspamer"<br />

Maria Luisa IANNITTO, at Department of<br />

Molecular Medicine<br />

Raffaella LA SCALEIA, at Department of<br />

Molecular Medicine<br />

Ramona LATTAO, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Rosalba LEPORE, at Department of<br />

Biochemical Sciences "A. Rossi Fanelli"<br />

Valerio LICURSI, at Department of Biology<br />

and Biotechnologies "C. Darwin"<br />

Carmen MARESCA, at Department of<br />

Cellular Biotechnologies and Hematology<br />

Federica MARINO, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Ramona MARRAPODI, at Department of<br />

Molecular Medicine<br />

Valentina MENGOLI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Emanuela MICHELI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Arianna MONTANARI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Linda Celeste MONTEMIGLIO, at<br />

Department of Biochemical Sciences "A.<br />

Rossi Fanelli"<br />

Angela MORRONE, at Department of<br />

Biochemical Sciences "A. Rossi Fanelli"<br />

Cassandra MOSTOCOTTO, at<br />

Department of Cellular Biotechnologies<br />

and Hematology<br />

Gaelle NOEL, at Department of Biology<br />

and Biotechnologies "C. Darwin"<br />

Ida PACIELLO, at Department of Public<br />

Health and Infectious Diseases<br />

Francesca PAGANI, at Department of<br />

Physiology and Pharmacology "Vittorio<br />

Erspamer"<br />

Valeria Luciana PALUMBO, at<br />

Department of Biology and Biotechnologies<br />

"C. Darwin"<br />

Paola PARISI, at Department of Biology<br />

and Biotechnologies "C. Darwin"<br />

Eugenia PENNACCHIETTI, at Department<br />

of Biochemical Sciences "A. Rossi<br />

Fanelli"<br />

Serena PERILLI, at Department of Biology<br />

and Biotechnologies "C. Darwin"<br />

Francesco PISCITELLI, at Department of<br />

Medicinal Chemistry and Technologies<br />

Dorina POLINARI, at Department of<br />

Cellular Biotechnologies and Hematology<br />

Biancamaria RICCI, at Department of<br />

Molecular Medicine<br />

Cinzia RIZZO, at Department of Public<br />

Health and Infectious Diseases<br />

Maddalena ROMANELLI, at Department<br />

of Molecular Medicine<br />

Valentina ROMANO, at Department of<br />

Physics<br />

Emanuele ROSCIOLI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Daniele RUNCI, at Department of<br />

Molecular Medicine<br />

11


Annual Report <strong>2011</strong><br />

Daniel SAVATIN, at Department of Biology<br />

and Biotechnologies "C. Darwin"<br />

Riccardo SILIGATO, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

Helena STABILE, at Department of<br />

Molecular Medicine<br />

Carolina UGENTI, at Department of<br />

Biology and Biotechnologies "C. Darwin"<br />

12


Annual Report <strong>2011</strong><br />

Conferences<br />

Seminars<br />

<strong>Istituto</strong> <strong>Pasteur</strong> Science Days<br />

Abbazia di Sant’Andrea in Flumine<br />

Novembre 18-19, <strong>2011</strong><br />

• Molecular mechanisms of cellular<br />

differentiation<br />

• Immunity and cellular stress response<br />

• Molecular genetics and epigenetics<br />

• Molecular biology of microorganisms viruses<br />

and parasites<br />

Institut <strong>Pasteur</strong> International Network<br />

European Regional Meeting<br />

Infection related cancer and neurodegenerative<br />

disorders<br />

Rome, October 13-15, <strong>2011</strong><br />

• Pathogen driven mucosal inflammation and<br />

cancer of Acute Respiratory Infections<br />

• Hepatitis viruses, inflammation and cancer<br />

• Viral infection and neurodegenerative<br />

disorders<br />

European Day of Immunology<br />

Un amico sorprendente: il tuo sistema<br />

immunitario<br />

Rome, April 29, <strong>2011</strong><br />

October 5 • CD28: therapeutic target for<br />

lethal cytokine storm induced by<br />

superantigen toxins and influenza virus<br />

Prof. Raymond Kaempfer<br />

The Hebrew University, Jerusalem, Israel<br />

July 15 • How not to become a systems<br />

biologist<br />

Prof. Arthur M. Lesk<br />

Dept. of Biochemistry and Molecular Biology,<br />

The Pennsylvania State University, USA<br />

June 30 • Epigenetics: the new<br />

molecular and medical genetics<br />

Prof. John C. Lucchesi<br />

Dept. of Biology and Winship Cancer Institute,<br />

Emory University Atlanta GA, USA<br />

June 9 • The Segregation Distorter<br />

system in Drosophila melanogaster:<br />

a case study of a meiotic cheater<br />

Prof. Terrence W. Lyttle<br />

Dept. Cell and Molecular Biology University of<br />

Hawaii Manoa Honolulu, USA<br />

Joint Workshop<br />

Innovative targets and drug design<br />

strategies in cancer therapy<br />

in collaboration with PhD Schools.<br />

Rome, February 18, <strong>2011</strong><br />

• Mitosis as a “druggable” target<br />

• Targeting global regulators of cellular<br />

processes<br />

• Targeting protein modifications and<br />

interactions<br />

May 16 • The mitochondrial genome in<br />

life death and disease<br />

May 17 • Revising the mitochondrial<br />

genetic code - a shift in our thinking<br />

May 18 • Can we rescue the defect<br />

associated with a pathogenic<br />

mitochondrial tRNA mutation<br />

Prof. Robert Lightowlers and Prof. Zosia<br />

Chrzanowska Lightowlers<br />

University of Newcastle upon Tyne, UK<br />

13


Annual Report <strong>2011</strong><br />

April 11 • From Aristotele to malaria<br />

control: a bioinformatic approach<br />

April 13 • Vector-borne diseases in<br />

Greece: a journey through time<br />

Prof. Kitsos Louis<br />

University of Crete, Greece<br />

Bookshop talks<br />

Virus herpes: il nemico nascosto<br />

by Anna Teresa Palamara<br />

Noi e i batteri: una lunga storia di<br />

amicizia e guerra<br />

by Maria Lina Bernardini and Daniela De<br />

Biase<br />

Le energie del futuro: la fotosintesi e le<br />

biomasse<br />

by Felice Cervone and Roberto Bassi<br />

…nunc est bibendum…<br />

Il divin teatro e la mente<br />

by Nicola Modugno, Paolo De Vita and<br />

Giovanni Mirabella. Introduction by Cristina<br />

Limatola<br />

…genere di…vino…<br />

Il genere… influenza tutto<br />

by Flavia Franconi and Letizia Gabaglio<br />

..in vino e …in vitro<br />

Cellule staminali: realtà e illusioni<br />

by Paolo Bianco and Gilberto Corbellini<br />

…In vino veritas…<br />

Il Dr. Semmelweiss e altre terribili storie<br />

Percorsi per la verità scientifica<br />

by Marco Tripodi, Angela Santoni and<br />

Mario Polsinelli<br />

Come siamo apparsi<br />

Il senso di…vino della vita<br />

by Ernesto Di Mauro, Giuditta Perozzi and<br />

Elio Ziparo<br />

14


Area 1<br />

Molecular biology of<br />

microorganisms and viruses


Area 1: Molecular biology of microorganisms and viruses<br />

Structural biology of the thiol-dependent redox systems of<br />

Schistosoma mansoni and Plasmodium falciparum<br />

Andrea Bellelli<br />

Department of Biochemical Sciences "A. Rossi Fanelli"<br />

℡: +39 06 49910955 - @: andrea.bellelli@uniroma1.it<br />

Schistosomiasis is a widespreadd tropical parasitic disease, caused by three species of<br />

the blood-fluke Schistosoma. The disease is debilitating and affects 200 million people in<br />

tropical areas; the death toll is estimated at two hundred thousands people per year.<br />

Schistosomiasis is currently treated with one drug, Praziquantel, whose precise molecular<br />

target is unknown. Several other drugs are known to kill the schistosomes in vivo and in vitro,<br />

but these are seldom employed because of toxicity, high cost, complex administration or<br />

other reasons. The improvement of known drugs or the development of entirely new ones is<br />

a desirable goal, in view of the fact that strains of Schistosoma mansoni with reduced<br />

sensitivity to Praziquantel have appeared. In this project we are exploring known or putative<br />

macromolecular targets of schistosomicidal drugs; thus we focus on the biochemistry and<br />

molecular biology of the parasite. The rationale of this approach is that drug design may<br />

become realistic if the mechanism of action of each drug were known at atomic detail, ideally<br />

as the 3D structure of the drug in complex with its target. The enzymes involved in the<br />

detoxification of reactive oxygen species (ROS) and other oxidants are potential drug targets.<br />

We have already characterized from the structural and functional point of view: Glutathione<br />

Transferase; Thioredoxin Glutathione Reductase; and Glutathione Peroxidase from either S.<br />

mansoni or S. haematobium. We have also characterized a Fatty Acid Binding Protein and a<br />

Cyclophilin, both from S. mansoni, even though these two proteins are not directly related to<br />

the ROS detoxification pathway.<br />

Two of the enzymes we characterized are known to be “druggable”, i.e. they can serve as<br />

the target of known or putative drugs: Thioredoxin Glutathione Reductase (TGR) and<br />

Glutathione Peroxidase. In the case of Thioredoxin Glutathione Reductase an effective<br />

inhibitor is available, that is known to kill not only the schistosomes, but also malarial<br />

parasites: the gold containing drug Auranofin. TGR is a NADPH-dependent flavoreductase<br />

containing a selenocysteine residue (Sec). During its enzymatic cycle thiolates and<br />

selenolates that have high affinity for transition metals are generated. Auranofin inhibits TGR<br />

(and other selenocysteine-containing flavoreductases) more effectively than non Secontaining<br />

ones (glutathione reductase); this preference was traditionally ascribed to the high<br />

affinity of selenium for gold. We solved the structure of the gold-TGR complex, and found Au<br />

combined to the sulfur of Cys residues, rather than (or in addition to) the Se of Sec. Thus our<br />

results challenge the commonly held view. Kinetic measurements have demonstrated that<br />

the relative velocity of the reaction rather than the relative affinity, depends on the presence<br />

of Sec residues, which appear to dictate AF selectivity. Indeed when an external source of<br />

17


Area 1: Molecular biology of microorganisms and viruses<br />

selenium (benzeneselenol) was added to the reaction mixture, auranofin was able to inhibit<br />

the Se-lacking reductases Glutathione Reductase and Sec->Cys mutated TGR with an<br />

efficiency similar to Se-containing ones.<br />

In order to complete the structural characterization of the enzymes belonging to the thiolmediated<br />

detoxification pathway, we solved the high-resolution crystal structure of S.<br />

mansoni Thioredoxin (SmTrx) in three states, namely: the wild-type oxidized adult enzyme<br />

and the oxidized and reduced forms of a juvenile isoform, carrying an N-terminal extension.<br />

SmTrx shows a typical thioredoxin fold, highly similar to the other components of the<br />

superfamily. SmTrx presents some functional peculiarities, e.g. the ability to reduce oxidized<br />

glutathione. Moreover it is one of the few defence proteins expressed in mature eggs and in<br />

the hatch fluid, thus confirming an important role in the parasite.<br />

Only one relevant enzyme of the pathway remains to be characterized, namely<br />

Peroxiredoxin; this protein has been heterologously expressed in E. coli, purified and<br />

crystallized and its structure is the next target of our project.<br />

Publications<br />

Angelucci A, Miele AE, Boumis G, Brunori M, Dimastrogiovanni D, Bellelli A. Macromolecular<br />

bases of antischistosomal therapy. Curr Top Med Chem <strong>2011</strong>, 11: 2012-28. doi:<br />

10.2174/156802611796575939.<br />

Boumis G, Angelucci F, Bellelli A, Brunori M, Dimastrogiovanni D, Miele AE. Structural and<br />

functional characterization of Schistosoma mansoni Thioredoxin. Protein Sci <strong>2011</strong>, 20:<br />

1069-76. doi: 10.1002/pro.634.<br />

Saccoccia F, Angelucci F, Boumis G, Brunori M, Miele AE, Williams DL, Bellelli A. On the<br />

mechanism and rate of gold incorporation into thiol-dependent flavoreductases. J Inorg<br />

Biochem <strong>2011</strong> Epub. doi: 10.1016/j.jinorgbio.<strong>2011</strong>.11.005.<br />

Research Group<br />

Adriana Erica Miele, Francesco Angelucci,<br />

researchers; Giovanna Boumis, technician,<br />

Fulvio Saccoccia, PhD student.<br />

Collaborations<br />

David L. Williams, Rush University, Chicago,<br />

IL, USA.<br />

18


Area 1: Molecular biology of microorganisms and viruses<br />

Bacterial globins as regulators of thiol redox equilibrium in bacteria<br />

Alberto Boffi<br />

Department of Biochemical Sciences "A. Rossi Fanelli"<br />

℡: +39 06 49910990 - @: alberto.boffi@uniroma1.it<br />

The specific targets of the project concern the elucidation of the mechanism of heme<br />

based thiol oxidation in bacterial truncated globins (trHbs) and the discovery of novel<br />

pathways in thiol redox homeostasis in bacteria. Spectroscopic techniques have been<br />

applied in order to characterize the properties of bacterial hemoglobins in solution as a<br />

function of standard parameters (pH, salt composition, reducing environment etc., see<br />

methods). In particular, a complete resonance Raman spectroscopic characterization of the<br />

globins from Thermobifida fusca and Bacillus subtilis has been carried out both on native<br />

proteins and in selected active-site mutants. The Raman studies brought out that these<br />

globins display the spectroscopic signature characteristics of heme peroxidases, with a<br />

strong electron donation from the heme iron to the distal ligand both in their ferrous and ferric<br />

adducts. The nature of the heme-fluoride complexes has been further explored in<br />

spectroscopic experiments that allowed for the first time the assignment of the specific ironfluoride<br />

stretching frequencies in a hemoprotein (Droghetti et al., <strong>2011</strong>). In parallel, molecular<br />

dynamics of fluoride motion inside the heme pocket has been investigated and highlighted a<br />

novel mechanism of ligand stabilization within the heme pocket, based on hydrogen bonding<br />

from a conserved tryptophan residue to the iron bound fluoride atom (Nicoletti et al., <strong>2011</strong>). A<br />

set of combinatorial mutants of the distal heme pocket has been analyzed in order to focus<br />

on the properties of distal heme cavity. The results obtained can be considered as a unique<br />

data set, because they have been obtained for a complete group of mutants where the three<br />

key amino acids of Tf-trHb (WG8, YCD1, YB10) are progressively substituted with the nonhydrogen<br />

bonding phenylalanine. The spectroscopic characterization of the fluoride<br />

complexes has unveiled a well-defined correlation between ν(Fe–F) vibrational frequencies<br />

and CT1 electronic transition energies. For the case of Tf-trHb, we have obtained a detailed<br />

picture of H-bonding in the distal cavity environment. The interpretation of the spectroscopic<br />

data is strengthened by the close relation with the observed fluoride dissociation kinetics and<br />

molecular dynamics simulations. All the techniques yield evidence that TrpG8 and TyrCD1<br />

can form strong H bonds with fluoride, whereas TyrB10 can only interact weakly. The second<br />

target of the research concerns the identification of partner proteins involved in the complex<br />

physiological network of thiol homeostasis. In the case of Bacillus subtilis the partner protein<br />

YjbH has been identified already and paralogues have been found in actinomyces, including<br />

mycobacteria, and in parasitic microorganisms, including Lehismania. On these basis, it is<br />

suggested that the globin may be able to catalyze disulfide bonds formation and thus act as a<br />

heme based peroxiredoxin. The first step towards the identification of a novel thiol redox<br />

19


Area 1: Molecular biology of microorganisms and viruses<br />

pathway in Bacillus subtilis concerns the demonstration of the postulated interaction between<br />

the globin (yjbI) and the “disulfide isomerase-like” protein (yjbH) located upstream within the<br />

same operon. In this framework, the whole operon contaning yjbI and yjbH has been<br />

expressed as a copy of the original operon from B. subtilis. Overexpression of the whole<br />

operon allowed us to single out that trHb is capable of direct oxidation of the -CxxC- motif of<br />

YjbH thus regenerating active protein. Thus, trHbs act as redox switches that are capable of<br />

oxidizing disulfide bonds of the YjbH regulator. In this framework, trHbs would be terminators<br />

of the redox stress response. The solution of these complex biological functions will be<br />

addressed in the last year of the project, by examining in detail the nature of the interactions<br />

of trHbs with their partner proteins.<br />

Publications<br />

Droghetti E, Nicoletti FP, Bonamore A, Sciamanna N, Boffi A, Feis A, Smulevich G. The<br />

optical spectra of fluoride complexes can effectively probe H-bonding interactions in the<br />

distal cavity of heme proteins. J Inorg Biochem <strong>2011</strong>, 105: 1338-43. doi: 10.1016/<br />

j.jinorgbio.<strong>2011</strong>.07.007.<br />

Nicoletti FP, Droghetti E, Boechi L, Bonamore A, Sciamanna N, Estrin DA, Feis A, Boffi A,<br />

Smulevich G. Fluoride as a probe for H-bonding interactions in the active site of heme<br />

proteins: The case of Thermobifida fusca hemoglobin. J Am Chem Soc <strong>2011</strong>, 133: 20970-<br />

80. doi: 10.1021/ja209312k.<br />

Research Group<br />

Alessandra Bonamore, Alberto Macone,<br />

research fellows; Paola Baiocco, post-doc<br />

fellow; Andrea Ilari, CNR researcher.<br />

Collaborations<br />

Giulietta Smulevich Dipartimento di Chimica,<br />

Università di Firenze; Leonardo Boechi<br />

Departamento de Química Inorgánica, Analítica<br />

y Química Física, Universidad de Buenos Aires.<br />

20


Area 1: Molecular biology of microorganisms and viruses<br />

The acid resistance genes of Escherichia coli: does their complex<br />

transcriptional control hide novel biological roles<br />

Daniela De Biase<br />

Department of Medical-Surgical Sciences and Biotechnologies<br />

℡: +39 0773 1757212 - @: daniela.debiase@uniroma1.it<br />

For successful colonization of the gut, enteric bacteria must overcome the extreme acidic<br />

stress (pH


Area 1: Molecular biology of microorganisms and viruses<br />

Structure-function relationships in GadB<br />

We performed a biochemical characterization of the GadB site-specific double mutant<br />

D86N-H465A, which was also provided to Dr G. Capitani (PSI, Villigen, Switzerland) for<br />

crystallographic analysis. We designed this mutant because D86 in the GadB active form is<br />

involved in binding of the γ-carboxylate of glutamate. We found that the Asp86Asn mutation,<br />

in association with the His465Ala mutation, which is necessary to maintain the GadB active<br />

site open, affects GadB catalytic activity by extending its pH range of activity up to pH 8.0.<br />

Because the double mutant D86N-His465A is active at pH well above neutral, we expect that<br />

it might be employed for biotechnological applications. The double mutant has been<br />

crystallized and the crystal structure is under refinement (manuscript in preparation).<br />

Role of the gadBC system in Brucella microti<br />

As part of a newly established collaboration with Dr. A. Occhialini (Montpellier, France),<br />

we constructed three acid sensitive E. coli mutants of MG1655 K12 strain, i.e. ∆gadC,<br />

∆gadA-∆gadB and ∆gadA-∆gadBC. These mutants were used for heterologous in trans<br />

complementations with the B. microti gadBC operon. All mutants recovered glutamatedependent<br />

acid resistant phenotype, were able to export GABA and to decarboxylate<br />

glutamate, thereby confirming conserved functions of the gadBC system in both bacterial<br />

species.<br />

Publications<br />

Doucette GJ, Mikulski CM, King KL, Roth PB, Wang Z, Leandro LF, Degrasse SL, White KD,<br />

De Biase D, Gillett RM, Rolland RM. Endangered North Atlantic right whales (Eubalaena<br />

glacialis) experience repeated, concurrent exposure to multiple environmental neurotoxins<br />

produced by marine algae. Environ Res <strong>2011</strong> Epub. doi: 10.1016/j.envres. <strong>2011</strong>.09.010.<br />

Yu Z, Bekker M, Tramonti A, Cook GM, van Ulsen P, Scheffers DJ, de Mattos JT, De Biase<br />

D, Luirink J. Activators of the glutamate-dependent acid resistance system alleviate<br />

deleterious effects of YidC depletion in Escherichia coli. J Bacteriol <strong>2011</strong>, 193: 1308-16.<br />

doi: 10.1128/JB.01209-10.<br />

Research Group<br />

Francesco Bossa, professor; Angela Tramonti<br />

CNR researcher; Eugenia Pennacchietti, postdoc<br />

fellow; Daniela Bastianelli, Gaia Grassini,<br />

PhD students.<br />

Collaborations<br />

Guido Capitani, Paul Scherrer Institute, Villigen<br />

PSI, Switzerland; Maurice CR Franssen<br />

Wageningen University and Research Centre,<br />

The Netherlands; Sylvie Rimsky, CNRS-ENS de<br />

Cachan, France; Alessandra Occhialini,<br />

Université Montpellier I, CPBS, UMR 5236,<br />

France; Joen Luirink, Institute of Molecular Cell<br />

Biology, Amsterdam University, The Netherlands.<br />

22


Area 1: Molecular biology of microorganisms and viruses<br />

Epstein-Barr virus interactions with cellular microRNAs<br />

Alberto Faggioni<br />

Department of Experimental Medicine<br />

℡: +39 06 4461500 - @: alberto.faggioni@uniroma1.it<br />

The discovery of microRNA (miR) represents a novel paradigm in RNA based regulation<br />

of gene expression and their dysregulation has become a hallmark of many tumors. In virally<br />

associated cancers, the host-pathogen interaction could involve alteration in miR expression.<br />

Interestingly EBV, besides encoding its own miRs, can also influence cellular miRs. The<br />

initial evidence that this is so came from studies showing that EBV negative and EBV positive<br />

BLs differ in their miR expression signature. Moreover, among EBV positive BLs, it was<br />

shown that the miR profile of restricted latency expressing BLs (latency I) and broader<br />

latency expressors (latency III) is diverse. Given the importance on the one hand, of LMP1<br />

and EBNA2 in EBV driven transformation and on the other, of the dysregulation of miRs in<br />

cancer, we asked how these viral proteins influence cellular miR expression.<br />

We first showed that LMP1 negatively regulates a major oncogene, TCL1, in diffuse large<br />

B-cell lymphoma (DLBCL) and BL cells. MicroRNA (miR) profiling of LMP1 transfectants<br />

showed that among others, miR-29b, is upregulated. LMP1 diminished TCL1 by inducing<br />

miR-29b through C-terminus activation region 1 (CTAR1) and CTAR2. miR-29b locked<br />

nucleic acid (LNA) antisense oligonucleotide transfection into LMP1-expressing cells reduced<br />

miR-29b expression and consequently reconstituted TCL1, suggesting that LMP1 negatively<br />

regulates TCL1 through miR-29b upregulation. The ability of LMP1 to negatively regulate<br />

TCL1 through miR-29b might underlie its B-cell lymphoma growth antagonistic property. As<br />

LMP1 is also important for B-cell transformation, we suggested that the functional dichotomy<br />

of this viral protein may depend on a combination of levels of its expression, lineage and<br />

differentiation of the target cells and regulation of miRs, which then directs the outcome of<br />

the cellular response.<br />

Next, we studied how the expression of EBNA2, a protein indispensable for the capacity<br />

of the virus to transform B cells in vitro, might affect cellular miRs. Extensive miR profiling of<br />

the virus infected and EBNA2 transfected B lymphoma cells revealed that oncomiR miR-21 is<br />

positively regulated by this viral protein. Conversely, Burkitt lymphoma (BL) cell lines infected<br />

with EBNA2 lacking P3HR1 strain did not show any increase in miR-21. EBNA2 increased<br />

phosphorylation of AKT and this was directly correlated with increased miR-21.The primary<br />

miR-21 transcripts were unaltered by EBNA2 and this suggests that the increase in mature<br />

miR-21 is due to higher post-transcriptional processing. Interestingly, miR-146a, whose<br />

expression is known to be induced by LMP1, was severely downregulated by EBNA2 in<br />

U2932 DLBCL cells. The primary miR-146a transcription and the promoter activity was<br />

hampered by EBNA2 whereas LMP1 had no such effect. Low miR-146a expression generally<br />

23


Area 1: Molecular biology of microorganisms and viruses<br />

correlates with high IRAK1 and consequently higher type I interferon level. Indeed an<br />

elevated level of IRAK1 and type I interferon was found in EBNA2 transfectants.<br />

Further studies will be required to investigate the precise molecular mechanisms how<br />

EBNA2 affects miR expression and in particular miR-21 and miR-146a. However the data<br />

suggest that EBNA2 might contribute to EBV induced B cell transformation by a positive<br />

regulation of oncomiR miR-21 and by interfering with the innate immunity through regulating<br />

a key regulator of it, namely miR-146a (Rosato et al., submitted). Additionally, this miR has<br />

been attributed with oncosuppressive functions. Our data thus suggest that EBNA2 may<br />

promote B cell transformation by simultaneously increasing an oncomiR and reducing a miR<br />

with known tumor suppressing functions.<br />

Publications<br />

Imig J, Motsch N, Zhu JY, Barth S, Okoniewski M, Reineke T, Tinguely M, Faggioni A,<br />

Trivedi P, Meister G, Renner C, Grässer FA. MicroRNA profiling in EBV-associated B-cell<br />

lymphomas. Nucleic Acid Res <strong>2011</strong>, 39: 1880-93. doi: 10.1093/nar/gkg1043.<br />

Research Group<br />

Antonio Angeloni, Pankaj Trivedi, professors;<br />

Roberta Santarelli, Mara Cirone, Antonella<br />

Farina, Roberta Gonnella, researchers; Marisa<br />

Granato, Eleni Anastasiadou, post-doc<br />

fellows; Francesco Boccellato, Paola Rosato,<br />

PhD students.<br />

24


Area 1: Molecular biology of microorganisms and viruses<br />

Study of RNA damage in yeast as a model for age-related<br />

degenerative diseases<br />

Claudio Falcone<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912278 - @: claudio.falcone@uniroma1.it<br />

The aim of the present project is to elucidate how cells can handle and respond to the<br />

accumulation of oxidized RNAs.<br />

Recently, a growing body of studies suggests that RNA oxidation is an early event in a<br />

wide variety of neurological diseases, including Alzheimer's disease (AD) and amyotrophic<br />

lateral sclerosis (ALS), and in the progressive loss of muscle mass and strength which occur<br />

during aging (sarcopenia). Uncovering on the consequences and cellular handling of the<br />

oxidatively damaged RNA may provide significant insights into the pathogenesis of<br />

neurodegenerative diseases. Due to the complexity of studying these processes in<br />

mammalian model organisms, yeast could represent, because of its easy handling and the<br />

high conservation of fundamental cell pathways, a convenient model to trace the basal root<br />

for determining the effects of RNA damage on cell physiology. We previously demonstrated<br />

that mutants in mRNA degradation, beside the accumulation of mRNAs, show premature<br />

aging, high level of intracellular ROS and apoptotic cell death features. These mutants<br />

represent a convenient tool to study the effects of mRNA modifications during aging.<br />

In preliminary experiments, we observed that in mRNA degradation mutants (Kllsm4∆1)<br />

the reversion rate of mutation hugely increased with aging, compared to the wild type. In fact,<br />

we found that after 14 days of growth, when cell viability dropped to 0,004%, about 30% of<br />

survivals were Trp+ revertants while, in the same conditions, the revertants in the wild type<br />

population were only 0,008%.<br />

High reversion rate, as that measured in mRNA degradation mutants, was also observed<br />

in extremely aged wild type cells after 20 days of cultivation, suggesting that such<br />

phenomenon is physiologically related to aging.<br />

Interestingly, we found that most of the reversions observed in both wild type and<br />

Kllsm4∆1 mutant, were not due to adaptive DNA mutations since cells, after repeated<br />

cultivation, showed again the tryptophan auxotrophy.<br />

This fact suggests that such mutations were not at the DNA level but, rather, at the RNA<br />

level (mRNA and/or tRNA). We hypothesize that the mechanism underlying the suppression<br />

of the auxotrophic mutations under selective pressure might result from translational errors<br />

induced by oxidized mRNAs.<br />

According to this, our data concerning the mutation rate might indicate that, in yeast cells,<br />

the capability to handle oxidized RNAs drops with aging. In this respect, the Kllsm4∆1<br />

mutant, which accumulates mRNAs and ages much faster that the wild type strain,<br />

25


Area 1: Molecular biology of microorganisms and viruses<br />

represents a useful tool for deciphering the molecular mechanisms of cell response to mRNA<br />

oxidation.<br />

We observed that the use of a lower glucose concentration, 0,2% instead of 2%, and the<br />

over-expression of HIR1 and PGK1 genes extended lifespan of the Kllsm4∆1 mutant. We are<br />

currently measuring the mutation rate in these conditions.<br />

Hydroxyl radicals, produced within cells, can easily modify RNA because they are highly<br />

reactive and cannot diffuse from their sites of formation. The most prevalent oxidized base in<br />

RNA is 8-hydroxyguanosine (8-OHG), which can be detect/quantified through Highperformance<br />

liquid chromatography-electrochemical detection (HPLC-ECD). To make a<br />

correlation between mutation frequency and RNA oxidation, we are developing a protocol to<br />

measure the oxidized fraction of total RNAs from wild type and Kllsm4Ä1 mutant cells during<br />

aging.<br />

Publications<br />

Mazzoni C, Falcone C. mRNA stability and control of cell proliferation. Biochem Soc Trans<br />

<strong>2011</strong>, 39: 1461-5. doi: 10.1042/BST0391461.<br />

Palermo V, Pieri L, Silvestri R, La Regina G, Falcone C, Mazzoni C. Drug-induced inhibition<br />

of tubulin polymerization induces mitochondrion-mediated apoptosis in yeast. Cell Cycle<br />

<strong>2011</strong>, 10: 3208-09. doi: 10.4161/cc.10.18.16514.<br />

Research Group<br />

Cristina Mazzoni, researcher; Vanessa<br />

Palermo, post-doc fellow; Michele Saliola,<br />

graduated technician.<br />

Collaborations<br />

Frank Madeo, Institute of Molecular Biosciences,<br />

University of Graz, Austria.<br />

26


Area 1: Molecular biology of microorganisms and viruses<br />

From leaderless to leadered mRNAs: mRNA features modulating<br />

ribosome binding and translation initiation in Archaea<br />

Paola Londei<br />

Department of Cellular Biotechnologies and Hematology<br />

℡: +39 06 4940463 - @: londei@bce.uniroma1.it<br />

Background and aims<br />

Initiation of protein synthesis is the rate-limiting step of translation and the main target of<br />

translation regulation. However, the mechanism and molecular machinery for initiation have<br />

diverged in the primary domains of life: Bacteria, Archaea and Eukarya. In Bacteria,<br />

translation initiation is relatively simple, while in Eukarya is complex and requires more<br />

components. The Archaea, despite their prokaryotic phenotype, have an unexpectedly<br />

elaborated initiation process, requiring at least six initiation factors. Archaeal mRNAs differ<br />

widely in the structure of their translation initiation regions (TIR). Only a minority of TIRs are<br />

endowed with Shine-Dalgarno (SD) sequences for ribosome binding; in a majority of cases<br />

there is no SD motif, and very often there is no 5’ untranslated region (5’UTR) at all. In the<br />

latter case the mRNA is called leaderless. Leaderless mRNAs can constitute up to 50% of all<br />

transcripts in certain archaeal species. Archaeal SD-less and leaderless mRNAs have been<br />

proposed to be translated using initiation mechanisms alternative to those employed for<br />

canonical mRNAs having SD motifs, but experimental data in support of this hypothesis are<br />

still scarce. Using the thermophilic archaeon Sulfolobus solfataricus as the model system, we<br />

will study the features of mRNA/ribosome interaction in Archaea, analyzing different types of<br />

model mRNAs endowed with different TIRs for their translational capacity, efficiency of<br />

interaction with the ribosomes and formation of 30S and 70S initiation complexes.<br />

Results and perspectives<br />

To investigate the mRNA features involved in the control of translational efficiency, we<br />

prepared a number of model constructs to be translated in vitro and in vivo. The genes of<br />

choice were a natural leaderless mRNA (lacS), a natural “quasi leaderless” mRNA (aSUI-1),<br />

whose start codon is preceded by 5 nucleotides, and an artificial leaderless mRNA (104∆L).<br />

Preliminarily, the predicted 5’ termini of aSUI1 and lacS were validated experimentally by<br />

primer extension assays. The artificial leaderless mRNA (104∆L) was derived from a<br />

leadered mRNA by removing its 5’UTR and cloning the truncated gene in the plasmid (pBS)<br />

SK+ under the control of the viral promoter T7. The aSUI1 gene and the lacS gene were also<br />

cloned in (pBS) SK+, the former from a PCR amplificate, the second by subcloning from the<br />

construct pCmalLacS (a kind gift of Sonja Albers, Marburg ).<br />

The in vitro transcripts were incubated in our well-established cell-free system for protein<br />

synthesis. The translation products were revealed by radioactive labelling, or, in the case of<br />

the lacS protein, by colorimetric detection of the products of enzymatic activity. All the<br />

27


Area 1: Molecular biology of microorganisms and viruses<br />

mRNAs were translated with sufficient efficiency. Then, we modified the TIR of each of the<br />

above three constructs. Specifically, 5’-UTR of monotonous sequence (oligo-A) and different<br />

lengths were added to the leaderless genes, while the 5-nucleotide-5’UTR of the quasileaderless<br />

aSUI1 gene was shortened progressively to 4, 3 and one nucleotide. These<br />

experiments are aimed at unravelling the features of a translatable SD-less 5’-UTR in<br />

thermophilic archaea. In particular, the use of 5’-UTRs of monotonous sequence, and, later,<br />

of 5’-UTRs capable of forming secondary structures, should enable us to discriminate<br />

between the effects of length, sequence and folding. So far, we have found that the addition<br />

of short oligo-A tracts to mRNA 104∆L drastically reduces its translational efficiency. Further<br />

experiments are in progress.<br />

In parallel, we set up to develop a system for in vivo translational analysis. In vitro<br />

translation has several limitations, for instance that it is impossible to obtain perfectly<br />

leaderless mRNAs by transcription with T7 polymerase. In vivo analysis is carried out using<br />

the PBL2025 strain of Sulfolobus solfataricus, which is uracil auxotrophic. This strain was<br />

transformed with the plasmid pCMalLacS which contains the selectable marker pyrEF and<br />

the leaderless lacS gene. A number of positive transformants have been selected on uracildeficient<br />

plates and lacS expression has been detected by the colorimetric β-galactosidase<br />

assay. The starting construct has been engineered by adding to its 5’ terminus monotonous<br />

oligo-A sequences of varying length. Translational analysis is in progress.<br />

Once completed the in vitro and in vivo analysis of TIR features, our next intent will be to<br />

determine whether an artificially increased intracellular concentration of certain translation<br />

initiation factors results in a selective advantage for the translation of leaderless mRNAs. To<br />

this end S. solfataricus PBL2025 will be co-transformed with the pCmal-IF, which carries the<br />

IF gene of interest, and the plasmid pCMalLacS encoding the leaderless lacS gene.<br />

Publications<br />

Benelli D, Londei P. Translation initiation in Archaea: conserved and domain-specific<br />

features. Biochem Soc Trans <strong>2011</strong>, 3: 89-93.<br />

Research Group<br />

Dario Benelli, researcher; Michela Pinzaglia,<br />

PhD student; Dorina Polinari, graduated yellow.<br />

Collaborations<br />

Anna La Teana, Università Politecnica delle<br />

Marche; Sonja-Verena Albers, Max Plank<br />

Institute for Terrestrial Microbiology, Marburg,<br />

Germany.<br />

28


Area 2<br />

Pathogenic mechanisms of microbially<br />

associated diseases


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

Molecular characterization of viruses causing bronchiolitis and<br />

study of viral and host factors affecting Type I IFN antiviral<br />

response induced by respiratory viruses<br />

Guido Antonelli<br />

Department of Molecular Medicine<br />

℡: +39 06 4474122 - @: guido.antonelli@uniroma1.it<br />

Bronchiolitis is the principal cause of hospitalization for infants. Several risk factors are<br />

associated with the severity of bronchiolitis, such as premature birth, immunodeficiency, and<br />

chronic disease, but the majority of the infected infants has no obvious risk factors. It is<br />

tempting to speculate that virus features (e.g. different and specific strains, viral loads, etc.),<br />

or host factors associated to an effective immune response might be also important in<br />

determining the disease severity.<br />

Hence, we proposed to characterize respiratory viruses determinants of virulence and<br />

study the role of innate immunity in antiviral response, either in patients or in cell lines.<br />

First, we characterized viral infections, with a comprehensive panel of PCR-based<br />

reactions, in respiratory samples from patients seeking medical care in Emergency<br />

Departments: no respiratory signs/symptoms could differentiate or were useful to distinguish<br />

respiratory viruses (either children or adults). Hence, a comprehensive virological diagnosis<br />

is needed to differentiate respiratory agents and their role in specific respiratory syndromes.<br />

During the first winter circulation of pandemic Influenza A H1N1 (InfA-pH1N1),<br />

respiratory syncytial virus (RSV) was the pathogen with the highest severity of infection in<br />

children, with highest intensive care rates, even though total days of hospitalization of<br />

children were independent on the type of virus found in their specimens. RSV was the major<br />

pathogen in bronchiolitis, followed by rhinovirus (RV) while metapneumovirus (hMPV) and<br />

pH1N1 were found only in few cases.<br />

Nasal washings from children positive to RSV, RV, pH1N1, Bocavirus (hBoV), and hMPV<br />

were inoculated onto tubes of different cell cultures; with the exception of the not-yetcultivated<br />

hBoV, viral stocks were obtained to characterize in vitro type I Interferon (IFN)<br />

antiviral activity. InfA pH1N1 resulted partially resistant to type I IFN with respect to a<br />

seasonal InfA and hMPV was more sensitive to IFN beta than to leukocyte IFN.<br />

We then evaluated the association between RSV load and disease severity or between<br />

different biochemical/clinical parameters and mRNA-levels of the interferon stimulated gene<br />

(ISG) 56 in respiratory samples from 132 infants hospitalized for bronchiolitis and infected<br />

with RSV as a single (90%), or as a dual infection with other respiratory viruses (10%). In the<br />

same context we also evaluated how viral load varied during the course of RSV infection.<br />

Results indicated that viral load was positively related to the clinical severity of<br />

bronchiolitis, the length of hospital stay, the levels of glycaemia and the number of ISG56-<br />

mRNA copies, whereas an inverse correlation was observed with levels of haemoglobin. We<br />

also found that the RSV load significantly decreased between the first and second sample in<br />

31


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

most single and RSV coinfections. Indeed, infants with high RSV load on hospital admission<br />

are more likely to have both more severe bronchiolitis and a higher airway activation of<br />

antiviral immune response.<br />

To clarify the important issue of RV proposed association with development of paediatric<br />

asthma, the rate of recurrent wheezing was analyzed 12 months after hospitalization for<br />

bronchiolitis: multivariate analysis identified as major independent risk factors for recurrent<br />

wheezing, rhinovirus infection and a positive family history for asthma.<br />

To tentatively relate a clinical phenotype to a RV strain, we performed RV genomic region<br />

5’ UTR and VP4/VP2 sequencing and a phylogenetic tree was constructed. We then<br />

analyzed in field isolates the close 5’ proximity of polyprotein initiation site (AUG at nt 611), a<br />

conserved 24 amino acids (aa) motif exposed during the so-called capsid breathing, the<br />

Met67-Ser68 cleavage site at the VP4-VP2 junction, the position of a putative internal cisacting<br />

replication element (cre) located within viral protein 2 (VP2). The 5’ proximity of the<br />

initiator AUG and the VP4-VP2 cleavage sites were highly conserved in patient strains of all<br />

RV species, the cre position was characteristic and conserved among RV C strains, and the<br />

24 aa motif was conserved in RV A and B (21/24 aa) but not in RV C isolates (only 9 aa out<br />

of 24). Patient data were then analyzed: age distribution and clinical data of RV A and C<br />

were not significantly different, even though there was a tendency of RV C strains to be<br />

associated with hospitalization of older children and with wheezing.<br />

Publications<br />

Pierangeli A, Scagnolari C, Selvaggi C, Verzaro S, Spina MT, Bresciani E, Antonelli G,<br />

Bertazzoni G. Rhinovirus frequently detected in elderly adults attending an emergency<br />

department. J Med Virol <strong>2011</strong>, 83: 2043-7. doi: 10.1002/jmv.22205.<br />

Scagnolari C, Trombetti S, Selvaggi C, Carbone T, Monteleone K, Spano L, Di Marco P,<br />

Pierangeli A, Maggi F, Riva E, Antonelli G. In vitro sensitivity of human metapneumovirus<br />

to type I interferons. Viral Immunol <strong>2011</strong>, 24: 159-64. doi: 10.1089/vim.2010.0073.<br />

Scagnolari C, Trombetti S, Soldà A, Selvaggi C, Monteleone K, Spano L, Pierangeli A,<br />

Clementi M, Turriziani O, Antonelli G. Pandemic 2009 H1N1 influenza virus is resistant to<br />

the antiviral activity of several interferon alpha subtypes. J Interferon Cytokine Res <strong>2011</strong>,<br />

31: 475-9. doi: 10.1089/jir.2010.0125.<br />

Research Group<br />

Ombretta Turriziani, professor; Laura<br />

Antonelli, Fabio Midulla, Paola Papoff,<br />

Alessandra Pierangeli, Carolina Scagnolari,<br />

researchers; Carla Selvaggi, research fellow;<br />

Katia Monteleone, PhD student.<br />

Collaborations<br />

Stefano Chiavelli, Carlo Concato, Ospedale<br />

Bambin Gesù, Roma; Monica Ferreri, Patrizia<br />

Bagnarelli, Ospedale Generale di Torrette,<br />

Ancona; Christina Christodoulou, Jan Richter,<br />

Molecular Virology, Cyprus Institute of Neurology<br />

& Genetics.<br />

32


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

Lipopolysaccharide and peptidoglycan adaptation to host as an<br />

immune evasion strategy of gram-negative pathogens<br />

Maria Lina Bernardini<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49917850 - @: marialina.bernardini@uniroma1.it<br />

Pathogens have evolved sophisticated strategies to finely tune the host innate immune<br />

responses and the inflammatory reaction. In fact, symptoms of infectious diseases are often<br />

based on the inflammatory reaction induced by pathogens in the site of infection.<br />

Shigella is an enteroinvasive pathogen provoking an acute and severe inflammation of<br />

colonic and rectal mucosa. Shigella is able to drive the immune sensing and response mainly<br />

by manipulating the recognition of PAMPs (Pathogens Associated Molecular Patterns) by the<br />

PRRs (Pattern Recognition Response) and by subverting the host cell-death machinery.<br />

Pseudomonas aeruginosa (PA) is an opportunistic pathogen causing acute and chronic<br />

infection in the airways of Cystic Fibrosis (CF) patients. Strains establishing life-long chronic<br />

infection are usually pathogenic variants distinguished from the initially acquired strains, often<br />

differing for the presence/absence of virulence factors and harboring PAMPs modifications,<br />

e.g. in the lypopolysaccharide (LPS) structures or in flagellin.<br />

Our research was focused on the impact of PAMPs on the host response during the<br />

invasive process of Shigella and lung colonization of PA. We also evaluated the ability of<br />

these two pathogens to hijack the host cell machinery controlling the death processes.<br />

Our findings show that Shigella proliferating within the epithelial cells is able to trigger<br />

both apoptosis and necrosis of the infected cells, depending on several parameters, including<br />

the number of intracellular bacteria and the time of infection. Apoptosis is dependent on<br />

mitochondrial dysfunction and caspase-9 activation. The stress-response molecule<br />

GADD45alfa is involved in this process. Likewise, Shigella kills dendritic cells through various<br />

mechanisms, including apoptosis and pyroptosis.<br />

PA clinical strains, isolated from a CF patient at sequential stages of lung infection,<br />

induce a pyroptotic cell death of macrophages and dendritic cells likely using different<br />

PAMPs and PRRs as elicitors. PAMPs of these PA clinical isolates display modified<br />

structures and immunopotential. LPS, Peptidoglycan (PGN) and flagellin are deeply different<br />

in the clonal strains isolated at various times of the disease. The structural modifications are<br />

reflected by the different ways of these structures to interact with their respective PRRs,<br />

leading the PA strains isolated from the chronic stage of the infection to be able of escaping<br />

from the immune system recognition. However, the PAMPs modifications make these<br />

“chronic” strains better elicitors of the inflammasome.<br />

Similarly, PAMPs of Shigella vary during infection and exhibit a dramatic influence on<br />

host cell responses. PGN is remodeled through a set of enzymes including the plasmid<br />

encoded gene SfpgdA, which acts as a PGN deacetylase producing a PGN more resistant to<br />

33


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

the lisozyme activity. PGN shedding is also a mechanism by which this pathogen dampens<br />

the immune system, especially through the sensing of the intracellular PRRs Nod1 and<br />

Nod2. Moreover, Shigella modifies LPS during infection, in this way modulating the response<br />

of different cell populations including macrophages, neutrophils and dendritic cells. The<br />

majority of the inflammatory cytokines are influenced by the LPS changes as well as the<br />

ability of neutrophils to produce factors needed to Shigella eradication.<br />

Finally, our findings suggest that, despite the different style of life, these two pathogens<br />

exploit the same strategies to establish a survival niche into the host.<br />

Publications<br />

Cigana C, Lorè NI, Bernardini ML, Bragonzi A. Dampening host sensing and avoiding<br />

recognition in Pseudomonas aeruginosa pneumonia. J Biomed Biotechnol <strong>2011</strong>, 852513.<br />

doi: 10.1155/<strong>2011</strong>/852513.<br />

Lembo Fazio L, Nigro G, Noël G, Rossi G, Chiara F, Tsilingiri K, Rescigno M, Rasola A,<br />

Bernardini ML. Gadd45α activity is the principal effector of Shigella mitochondriadependent<br />

epithelial cell death in vitro and ex vivo. Cell Death Dis <strong>2011</strong>, 2:e122. doi:<br />

10.1038/cddis.<strong>2011</strong>.4.<br />

Research Group<br />

Laura Curcurù, Luigi Lembo Fazio, post-doc<br />

fellows; Valeria Ciancarella, Gaëlle Noël, PhD<br />

students.<br />

Collaborations<br />

Abdel Allaoui, Lab. Bactériologie Moléculaire,<br />

Université Libre de Bruxelles, Belgium;<br />

Alessandra Bragonzi, Divisione di Immunologia,<br />

Trapianti e Malattie Infettive, <strong>Istituto</strong> Scientifico<br />

San Raffaele, Milano; Antonio Molinaro,<br />

Dipartimento di Chimica Organica e Biochimica,<br />

Università di Napoli Federico II; Maria Rescigno,<br />

Dipartimento di Oncologia Sperimentale, <strong>Istituto</strong><br />

Europeo di Oncologia, Milano.<br />

34


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

Involvement of sRNA molecules in the complex regulatory circuits<br />

of virulence gene expression in Shigella flexneri and in<br />

enteroinvasive E. coli<br />

Bianca Colonna<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49917582 - @: bianca.colonna@uniroma1.it<br />

Recently small noncoding RNAs (sRNA) have emerged as crucial elements in bacterial<br />

cell regulation. In particular, it is becoming increasingly evident that sRNAs, besides acting in<br />

gene regulation in general, can also play a key role in controlling the expression of virulence<br />

genes or affect adaptive stress responses which are crucial for bacteria to survive within the<br />

host.<br />

In several human bacterial pathogens sRNAs appear to be important for integrating<br />

environmental stimuli into outputs that are relevant for the pathogenicity process. Therefore<br />

we have analyzed the potential involvement of sRNA transcripts in the modulation of<br />

virulence gene expression in Shigella, a human pathogen that causes a severe enteric<br />

syndrome mainly in the developing countries. The cellular pathogenesis and the clinical<br />

presentation of shigellosis result from the complex action of a large number of bacterial<br />

virulence factors mainly located on a large virulence plasmid (pINV). By analyzing the large<br />

intergenic region separating the virA and icsA genes, we have identified the first pINV<br />

encoded sRNA of Shigella flexneri, RnaG.<br />

RnaG acts as antisense on the transcript of the icsA gene, which encodes the protein<br />

required for the intra- and intercellular spread of the bacterium inside the host cell. A detailed<br />

in vivo and in vitro characterization of the RnaG molecule and of its promoter has allowed us<br />

to understand the strategy adopted by this molecule to control icsA. As opposed to other<br />

sRNAs, which affect the ability of the target mRNA to be translated, we have found that<br />

RnaG is capable to downregulate icsA transcription by means of two, not mutually exclusive<br />

mechanisms. As for the first mechanism we have shown that silencing of the RNAG promoter<br />

results in higher activity of the convergent icsA promoter. On the other hand, RnaG can also<br />

cause a direct repression of icsA transcription by acting as antisense RNA, provoking a<br />

premature termination of the icsA messenger. In fact, binding of RnaG to the nascent icsA<br />

transcript induces the formation of a stem-loop motif at the 5’end of the mRNA, mimicking an<br />

intrinsic terminator and resulting in the synthesis of a truncated icsA transcript.<br />

Over the years evidence has built up indicating that the regulation of icsA depends on the<br />

nucleoid protein H-NS which acts as a repressor, and on the AraC-like protein VirF, the major<br />

activator of the Shigella invasivity regulon. Since the molecular details of this regulatory<br />

mechanism are still not fully understood, we have analysed the synergistic and antagonistic<br />

interplay among the regulatory proteins and the RnaG molecule in order to evaluate their<br />

contribution to the expression of the icsA gene. The results obtained show that both proteins,<br />

H-NS and VirF, recognize the icsA and RnaG promoter regions in vivo and in vitro and that<br />

35


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

the temperature-dependent expression of icsA depends on a delicate balance among H-NS<br />

repression, VirF activation and RnaG transcription.<br />

All together our data evidence the complexity of the circuits regulating virulence in<br />

Shigella and give insight into the strategy adopted by pathogenic bacteria to obtain a<br />

coordinated and efficient expression of their key virulence determinants inside the host.<br />

Publications<br />

Barbagallo M, Martino ML, Marcocci L, Pietrangeli P, Carolis ED, Casalino M, Colonna B,<br />

Prosseda G. A new piece of the Shigella pathogenicity puzzle: spermidine accumulation by<br />

silencing of the speG gene. PLoS One <strong>2011</strong>, 6:e27226. doi: 10.1371/journal.pone.<br />

0027226.<br />

De Carolis E, Posteraro B, Florio AR, Colonna B, Prosseda G, Bugli F, Lorenzetti SR,<br />

Fiscarelli E, Inzitari R, Iavarone F, Castagnola M, Fadda G, Sanguinetti M. Analysis of<br />

heat-induced changes in protein expression of Stenotrophomonas maltophilia K279a<br />

reveals a role for GroEL in the host-temperature adaptation. Int J Med Microbiol <strong>2011</strong>, 301:<br />

273-81. doi: 10.1016/j.ijmm.2010.10.001.<br />

Nicoletti M, Iacobino A, Prosseda G, Fiscarelli E, Zarilli R, De Carolis E, Petrucca A, Colonna<br />

B, Casalino M. Stenotrophomonas maltophilia isolated from cystic fibrosis patients:<br />

genotyping analysis and molecular characterization of virulence determinants. Int J Med<br />

Microbiol <strong>2011</strong>, 301: 34-43. doi: 10.1016/j.ijmm.2010.07.003.<br />

Tran CN, Giangrossi M, Prosseda G, Brandi A, Di Martino ML, Colonna B, Falconi M. A<br />

multifactor regulatory circuit involving H-NS, VirF and an antisense RNA modulates<br />

transcription of the virulence gene icsA of Shigella flexneri. Nucleic Acids Res <strong>2011</strong>, 39:<br />

8122-34. doi: 10.1093/nar/gkr521.<br />

Research Group<br />

Gianni Prosseda, researcher; Maria Letizia Di<br />

Martino, post-doc fellow; Rosaria Campilongo,<br />

PhD student.<br />

Collaborations<br />

Gioacchino Micheli, <strong>Istituto</strong> di Biologia e<br />

Patologia Molecolari, CNR, Roma; Maurizio<br />

Falconi, Dipartimento di Biologia Molecolare,<br />

Cellulare e Animale, Università di Camerino;<br />

Mariassunta Casalino, Dipartimento di Biologia,<br />

Università di Roma Tre.<br />

36


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

Inhibition of Pseudomonas aeruginosa biofilms: new molecular<br />

strategies targeting cyclic-di-GMP metabolism<br />

Francesca Cutruzzolà<br />

Department of Biochemical Sciences "A. Rossi Fanelli"<br />

℡: +39 06 49910713 - @: francesca.cutruzzola@uniroma1.it<br />

Biofilms formed by bacterial pathogens are responsible of more than 70% of all infections<br />

in developed countries and are less sensitive to treatments with antimicrobial agents. The<br />

ubiquitous second messenger 3', 5'-cyclic diguanylic acid (c-di-GMP) is used in most bacteria<br />

to control the switch to the biofilm lifestyle; c-di-GMP has no counterpart in eukaryotic cells<br />

and thus it is an ideal target to develop effective anti-biofilm strategies.<br />

The aim of this proposal is to study the metabolism of c-di-GMP in the opportunistic<br />

human pathogen Pseudomonas aeruginosa in order to find new targets for effective antibiofilm<br />

drugs. We plan to characterize strategic P. aeruginosa proteins involved in c-di-GMP<br />

metabolism (diguanylate cyclases-DGCs and phosphodiesterases-PDEs) and to study the<br />

connection between the c-diGMP pathway and other metabolic pathways relevant for<br />

pathogenesis.<br />

We developed the enzymatic and c-di-GMP binding assays using the reference DGC<br />

PleD from Caulobacter crescentus. We have measured the DGC activity of purified PleD and<br />

confirmed, by titration of PleD with c-di-GMP (employing Isothermal Titration Calorimetry-<br />

ITC), that PleD binds c-di-GMP (2 mol c-di-GMP/mol protein; Kd 0,9 µM) in agreement with<br />

literature data. We have tested putative DGC inhibitors, starting from selected compounds,<br />

previously identified in microbiological screening of a chemical library by Landini and<br />

coworkers. These compounds do not significantly inhibit PleD in vitro (up to 500 µM)<br />

suggesting that they do not inhibit c-di-GMP biosynthesis through direct interaction with DGC<br />

proteins, but likely affect the availability of nucleotide substrate(s).<br />

We have characterized putative P.aeruginosa DGCs or PDEs, focusing our attention on<br />

selected targets, i.e. the DGC PA1120 and the PDEs PA4781 and PA4108. The PA1120<br />

protein (TpbB) is a DGC linking Las-induced quorum sensing (QS) to the formation of matrix<br />

exopolysaccharide (EPS) and extracellular DNA, necessary for biofilm development and<br />

coordinated group response in P. aeruginosa. We have produced the isolated catalytic<br />

domain of TpbB and measured its catalytic activity (0,1 nmol/min/mg); PA1120 is not<br />

significantly inhibited by c-di-GMP at low GTP concentrations. Identification and<br />

characterization of inhibitors are in progress.<br />

Few biochemical data are available on PDEs containing the HD-GYP domain, despite<br />

their importance in pathogenesis and their role in controlling biofilm formation. While EALtype<br />

PDEs hydrolyze c-di-GMP to linear diguanylate (pGpG), HD-GYPs completely hydrolyze<br />

c-di-GMP to GMP in a single reaction. In P.aeruginosa, 2 HD-GYP proteins are found, which<br />

are able to decrease c-di-GMP levels in vivo; these proteins contain the PDE catalytic<br />

37


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

domain and a (putative) regulatory domain, either of the CheY-type (PA4781) or unknown<br />

(PA4108). Both proteins were purified and tested for their PDE activity in vitro, monitoring c-<br />

di-GMP hydrolysis to GMP: while the PA4108 enzyme shows c-di-GMP hydrolytic activity,<br />

PA4781 is inactive. In order to bypass a possible activation step, a truncated version of<br />

PA4781 was also analyzed (PA4781-G) and found to be inactive in vitro, but active in cell<br />

extracts or in intact E. coli cells. These results suggest that the activity of PA4781 may<br />

depend on the interaction with an intracellular partner, as previously proposed for other HD-<br />

GYP PDEs.<br />

In parallel, we have also studied Aromatic L-Aminoacid decarboxylase (DOPA<br />

decarboxylase-DDC), the human enzyme responsible for a key step in the synthesis of<br />

adrenaline, one of the eukaryotic signal molecules sensed by Gram-negative pathogens.<br />

These molecules bind to bacterial membrane receptors and activate signaling pathways<br />

involving, among others, c-diGMP, thus leading to modulation of biofilm formation. Analysis<br />

of the three-dimensional structure coupled to a kinetic study allows to identify the structural<br />

determinants of the open/close conformational change occurring upon PLP binding and<br />

thereby propose a model for the stability of Group II decarboxylases in vivo (Giardina et al.,<br />

<strong>2011</strong>).<br />

Publications<br />

Giardina G, Montioli R, Gianni S, Cellini B, Paiardini A, Voltattorni CB, Cutruzzolà F. Open<br />

conformation of human DOPA decarboxylase reveals the mechanism of PLP addition to<br />

Group II decarboxylases. Proc Natl Acad Sci USA <strong>2011</strong>, 108: 20514-9. doi: 10.1073/<br />

pnas.1111456108.<br />

Research Group<br />

Nicoletta Castiglione, Giorgio Giardina,<br />

Serena Rinaldo, post-doc fellows; Valentina<br />

Stelitano, PhD student; Manuela Caruso,<br />

technician.<br />

Collaborations<br />

Davide Antoniani, Paolo Landini, Università<br />

di Milano; Livia Leoni, Giordano Rampioni,<br />

Università Roma Tre; Loredana Cappellacci,<br />

Università di Camerino.<br />

38


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

Defense mechanisms against oxidative and nitrosative stress in<br />

pathogenic protozoa<br />

Paolo Sarti<br />

Department of Biochemical Sciences "A. Rossi Fanelli"<br />

℡: +39 06 49910944 - @: paolo.sarti@uniroma1.it<br />

Giardiasis is a well-known human infectious disease caused by the amitochondriate<br />

protozoan parasite Giardia intestinalis, whose genome has been sequenced. Giardia is an<br />

O 2 -sensitive microorganism lacking some conventional ROS-scavenging enzymes, such as<br />

catalase and superoxide dismutase. Despite that, Giardia preferentially colonizes the fairly<br />

oxygenated small intestine. Our research has been focusing on the structural and functional<br />

characterization of the enzymes playing a key role in the antioxidant defense system of this<br />

microbial pathogen.<br />

We have shown that Giardia cells express a flavodiiron protein (FDP), a superoxide<br />

reductase (SOR) and, under nitrosative stress conditions, a flavohemoglobin (FlavoHb). This<br />

is surprising because FDPs and SORs are typically absent in eukaryotes and FlavoHbs are<br />

very rarely found in protists. The three proteins were expressed in E. coli and purified. The<br />

FDP is endowed with a high H 2 O-producing O 2 -reductase activity (TN > 40 s -1 , K M,O2 ≤ 2 µM,<br />

T = 20 °C), that led us to propose that the primary function of the FDP is to protect Giardia<br />

from O 2 toxicity, a function originally attributed to NADH-oxidase. The recombinant Giardia<br />

FlavoHb aerobically metabolizes nitric oxide (NO) with high efficacy (TN = 116 ± 10 s -1 at 1<br />

µM NO, K M,O2 = 22 ± 7 µM, T = 37 °C). Consistently, we found that the NO-induced<br />

expression of FlavoHb in Giardia cells endows the parasite with the ability to degrade NO in<br />

the presence of O 2 , which likely represents a strategy to evade the host immune response<br />

during infection. In collaboration with M.Teixeira and L.M.Saraiva (New University of Lisbon,<br />

Portugal) and D.E.Cabelli (Brookhaven National Laboratory, New York, USA), more recently<br />

the recombinant Giardia SOR was also characterized. The protein, whose expression in the<br />

trophozoite was assessed by immunoblotting, shows negligible superoxide dismutase<br />

activity, but in the reduced state is highly reactive with superoxide anion, as shown by pulse<br />

radiolysis and stopped-flow spectrophotometry.<br />

These results suggest that these antioxidant enzymes favor parasite survival in the host.<br />

Publications<br />

Borisov VB, Forte E, Sarti P, Giuffrè A. Catalytic intermediates of cytochrome bd terminal<br />

oxidase at steady-state: ferryl and oxy-ferrous species dominate. Biochim Biophys Acta<br />

<strong>2011</strong>, 1807: 503-9. doi: 10.1016/j.bbabio.<strong>2011</strong>.02.007.<br />

39


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

Giuffrè A, Borisov VB, Mastronicola D, Sarti P, Forte E. Cytochrome bd oxidase and nitric<br />

oxide: from reaction mechanisms to bacterial physiology. FEBS Lett. <strong>2011</strong> Epub. doi:<br />

10.1016/j.febslet.<strong>2011</strong>.07.035.<br />

Mastronicola D, Giuffrè A, Testa F, Mura A, Forte E, Bordi E, Pucillo LP, Fiori PL, Sarti P.<br />

Giardia intestinalis escapes oxidative stress by colonizing the small intestine: a molecular<br />

hypothesis. IUBMB Life <strong>2011</strong>, 63: 21-5. doi: 10.1002/iub.409.<br />

Sarti P, Forte E, Mastronicola D, Giuffrè A, Arese M. Cytochrome c oxidase and nitric oxide<br />

in action: molecular mechanisms and pathophysiological implications. Biochim Biophys<br />

Acta <strong>2011</strong> Epub. doi: 10.1016/j.bbabio.<strong>2011</strong>.09.002.<br />

Testa F, Mastronicola D, Cabelli DE, Bordi E, Pucillo LP, Sarti P, Saraiva LM, Giuffrè A,<br />

Teixeira M. The superoxide reductase from the early diverging eukaryote Giardia<br />

intestinalis. Free Radic Biol Med <strong>2011</strong>, 51: 1567-74. doi: 10.1016/j.freeradbiomed.<br />

<strong>2011</strong>.07.017.<br />

Research Group<br />

Elena Forte, researcher, Daniela Mastronicola,<br />

Fabrizio Testa, post doc fellows, Alessandro<br />

Giuffrè, CNR researcher.<br />

Collaborations<br />

Diane E. Cabelli, Brookhaven National<br />

Laboratory, New York, USA; Pier Luigi Fiori,<br />

Università di Sassari; Brajendra K. Singh,<br />

Department of Chemistry, University of New<br />

Delhi, India; Miguel Teixera, Instituto de<br />

Tecnologia Química e Biológica, Universidade<br />

Nova de Lisboa, Portugal; João B. Vicente,<br />

Universidade de Lisboa, Portugal.<br />

40


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

Immunopathogenesis of HIV infection: study of innate immunity<br />

and dendritic cells<br />

Vincenzo Vullo<br />

Department of Public Health and Infectious Diseases<br />

℡: +39 06 49970313 - @: vincenzo.vullo@uniroma1.it<br />

The study of dendritic cells (DC) as a link among innate and specify immunity is very<br />

important for pathogenesis, clinical and therapeutic approaches in HIV infection. The<br />

chemotactic recruitment of APC at the site of infection is critical for the initiation of<br />

appropriate immune responses. There are several data suggesting that chemokine receptor<br />

CCR5 is involved in both positive and negative regulation of APC system by the modulation<br />

of leukocyte trafficking, cellular activation and cytokine expression. What are the in vitro and<br />

in vivo immunologic consequences of pharmacologic inhibition of CCR5 function remain to<br />

be investigated. Recently, compounds targeting CCR5 have been introduced into clinical<br />

practice for the treatment of HIV infection. In this first year of our projects study, we have<br />

investigated the direct in vitro effect of anti-HIV CCR5 antagonist maraviroc (a new<br />

antiretroviral drugs) on chemotactic activity of DCs and innate immune cells. In particular, we<br />

evaluated the in vitro effect of different concentrations of CCR5 antagonist maraviroc (MVC)<br />

on cell migration of monocytes, macrophages (MO) and monocyte-derived dendritic cells<br />

(MDC) towards peptide formyl-methionyl-leucyl-phenylalanine (fMLP) and chemokines<br />

regulated upon activation normal T cell expressed and secreted (RANTES) and<br />

CCL4/macrophage inflammatory protein-1 (MIP-1beta) and CCL2/monocyte chemotactic<br />

protein-1 (MCP-1). Results off low cytometric analysis showed that monocytes treated in vitro<br />

with MVC exhibited a significant dose-dependent reduction of chemotaxis towards MIP-1beta<br />

and MCP-1. fMLP-induced chemotactic activity decreased only at higher concentration (1<br />

mM and 10 mM of MVC). In addition, all concentrations of MVC (0·1, 1 and 10 mM) induced<br />

in vitro a significant inhibition of chemotaxis of MO and MDC in response to all tested<br />

chemoattractants. In another set of experiments, cell viability and phenotype (CD14 for<br />

monocytes, MO and CD1a for MDC) and expression of chemoattractant receptors CCR1,<br />

CCR4, CCR5 and FPR expression were investigated. We found no alteration in viability and<br />

phenotype in cells treated with MVC. Moreover, treatment with different concentrations of<br />

MVC did not modulate CCR1, CCR4, CCR5 and FPR expression in monocytes, MO and<br />

MDC. In summary our findings suggest that CCR5 antagonist MVC may have the in vitro<br />

ability of inhibiting the migration of innate immune cells by mechanism which could be<br />

independent from the pure anti-HIV effect. The drug might have a potential role in the downregulation<br />

of HIV-associated chronic inflammation by blocking the recirculation and trafficking<br />

of MO and MDC. Considering the increasing use of MVC in patients with HIV infection,<br />

further studies should be encouraged to understand the immunological consequences of<br />

41


Area 2: Pathogenetic mechanisms of microbially associated diseases<br />

CCR5 blockade in innate immune cells. Future studies are needed to understand more<br />

clearly the mechanism underlying this inhibitory phenomenon exerted in vitro by maraviroc.<br />

In a second line of investigation, we assessed also the immunologic response to<br />

tuberculosis by assessing interferon-γ release assays (IGRAs). The study of this new<br />

immunologic assay is important to understand the dynamic of immune response during<br />

tuberculosis, which is known to be an important and increasing opportunistic infections<br />

especially in HIV-infected patients. In a longitudinal study we demonstrated that the classic<br />

tuberculin skin test (TST) does not influence the outcome of subsequent IGRAs testing in<br />

individuals with negative TST results, but it can boost the IFN-g response in subjects<br />

sensitized to TB antigens and not detected by IGRA. The evaluation of the possibility that the<br />

TST administration might affect the outcome of subsequent QFT-GIT testing is important to<br />

support the adoption of the assay in immunosuppressed populations in an attempt to predict<br />

the risk of developing TB disease.<br />

Publications<br />

Rossi R, Lichtner M, De Rosa A, Sauzullo I, Mengoni F, Massetti AP, Mastroianni CM, Vullo<br />

V. In vitro effect of anti-human immunodeficiency virus CCR5 antagonist maraviroc on<br />

chemotactic activity of monocytes, macrophages and dendritic cells. Clin Exp Immunol<br />

<strong>2011</strong>, 166: 184-90. doi: 10.1111/j.1365-2249.<strong>2011</strong>.04409.x.<br />

Sauzullo I, Massetti AP, Mengoni F, Rossi R, Lichtner M, Ajassa C, Vullo V, Mastroianni CM.<br />

Influence of previous tuberculin skin test on serial IFN-γ release assays. Tuberculosis<br />

(Edinb) <strong>2011</strong>, 91: 322-26. doi: 10.1016/j.tube.<strong>2011</strong>.05.004.<br />

Research Group<br />

Claudio M. Mastroianni, professor; Miriam<br />

Lichtner, researcher; Raffaella Rossi, Ilaria<br />

Sauzullo, post-doc fellows; Gabriella d’Ettorre,<br />

Claudia D’Agostino, hospital physicians; Fabio<br />

Mengoni, graduated technician.<br />

42


Area 3<br />

Molecular genetics<br />

of eukaryotes


Area 3: Molecular genetics of eukaryotes<br />

Assembly and functional analysis of genomic context vectors<br />

containing the human CFTR locus<br />

Fiorentina Ascenzioni<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49917577 - @: fiorentina.ascenzioni@uniroma1.it<br />

We have very recently published BAC vectors containing the wt CFTR allele<br />

(polymorphisms: (GATT) 7 in intron 6a; (TG) 11 T 7 in intron 8; V470 at position 470) and<br />

including the upstream and downstream regions. Functional activity of BAC-encoded CFTR<br />

was demonstrated in model FRT cells by different approaches including RT-PCR,<br />

quantitative real-time PCR, Western blotting, indirect immunofluorescence, and<br />

electrophysiological methods. Finally, and supporting the hypothesis that CFTR functions as<br />

a receptor for Pseudomonas aeruginosa, we demonstrated that CFTR-expressing cells<br />

internalized more bacteria than parental cells that do not express CFTR.<br />

The main objectives of the project are:<br />

1) Functional caracterization of the BAC in CF cells<br />

2) CFTR-BAC engineering in order to improve handling of the molecule and to add<br />

sequences that may be relevant for CF genetic research, such as specific mutations.<br />

CF defects complementation by cCFTRD12<br />

We have recently shown, by different functional assays, that the BAC vector cCFTRD12,<br />

when transfected into rat cells, encodes a functional CFTR protein (Auriche et al., 2010]. To<br />

extend this analysis to cell models relevant to CF gene therapy cCFTRD12 we need to<br />

transfer this vector to human epithelial cells from lung origin and having mutation in the<br />

CFTR gene. Due to the lack of efficient methods to transfer such large molecules, we<br />

performed preliminary work to select the most useful transfection reagents for bronchial<br />

epithelial cells. This was motivated by the fact that lipofectamine, which was successfully<br />

used to transfer the BAC into FRT (fisher rat thyroid) cells, didn’t work well with the human<br />

bronchial cell lines (CFBE and 16HBE). Nanoparticle-mediated gene delivery may represent<br />

a valid alternative to un-structured transfection reagent. Thereby, we are testing chitosanbased<br />

nanoparticles to deliver DNA to different cell lines, including 16HBE and CFBE cells.<br />

Preliminary results obtained with HEK cells transfected with EGFP-plasmids showed that the<br />

fraction of GFP positive cells, as determined by FACS analyses, reaches about 30% 4-5<br />

days after transfection and, keeping the cells in culture, it declines slowly over 15-20 days. Of<br />

note, cells treated with control reagents (EscortV from SIGMA or Lipofectamine from<br />

Invitrogen) showed similar efficiency but the fraction of GFP+ cells picks 2 day after<br />

transfection and rapidly decrease down to 0% 4-5 days after transfection. As previously<br />

reported by other, these results suggest that chitosan nanoparticles mediate a slow release<br />

of intracellular DNA and protect it from rapid intracellular degradation. We are currently<br />

45


Area 3: Molecular genetics of eukaryotes<br />

optimizing the chitosan-nanoparticle mediated transfection protocol and extend this method<br />

to BAC vectors.<br />

CFTR-BAC engineering<br />

The most used approaches to engineer inserts cloned into BAC vectors make use of<br />

homologous recombination or site specific homologous recombination in bacterial cells. In<br />

both cases the target region remains signed by the marker gene or by the site used by the<br />

recombinase. Consequently, the engineered region would contain sequences unrelated to its<br />

function. To overcome this problem, we are setting a new procedure based on yeast<br />

homologous recombination to insert/exchange DNA fragments without using selectable<br />

markers for the targeted region. Preliminary experiments have been done with the BAC<br />

cCFTRD12 which has three selectable markers, herein identified as marker 1, 2 and 3, in the<br />

vector backbone. The BAC has been co-transfected into yeast cells with a fragment having<br />

complementary ends to either marker 1 and 2; the transformants have been selected for<br />

marker 3. All the transformants recovered have been analyzed for the presence of the unselected<br />

marker, which was detected in 10% of the clones. Phenotypic and molecular<br />

analyses of these clones are undergoing. In the same time we have designed a series of<br />

experiments in order to highlight possible preferential sites of recombination that would limit<br />

the use of this procedure.<br />

Publications<br />

Del Porto P, Cifani N, Guarnieri S, Di Domenico EG, Mariggiò MA, Spadaro F, Guglietta S,<br />

Anile M, Venuta F, Quattrucci S, Ascenzioni F Dysfunctional CFTR alters the bactericidal<br />

activity of human macrophages against Pseudomonas aeruginosa. PLoS One <strong>2011</strong>,<br />

6:e19970. doi: 10.1371/journal.pone.0019970.<br />

Mattarocci S, D'Ambrosio E, Tafaro L, Somma V, Zannino G, Marigliano V, Ascenzioni F,<br />

Cimino-Reale G. Erosion of telomeric 3'-overhangs in white blood cells of aged subjects<br />

with high frequency of very short telomeres. Mech Ageing Dev <strong>2011</strong>, 132: 27-32.<br />

http://dx.doi.org/10.1016/j.bbr.<strong>2011</strong>.03.031.<br />

Research Group<br />

Enea Gino Di Domenico, post-doc fellow,<br />

Cifani Noemi, PhD student; Paola, Parisi,<br />

graduated student; Barbara Pompili, student.<br />

Collaborations<br />

Dr. Cleofe Palocci Dipartimento di Chimica,<br />

Sapienza Università di Roma.<br />

46


Area 3: Molecular genetics of eukaryotes<br />

Light control of filamentous fungi life cycle: from system models to<br />

applications<br />

Paola Ballario<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912392 - @: paola.ballario@uniroma1.it<br />

The study of blue light transduction pathways in filamentous fungi is the main field of<br />

interest of our group. This project persecutes two short term goals. The first is to clarify the<br />

mechanism controlling the switch ON and OFF of light regulated genes. The second is the<br />

characterization of the blue light sensor, the LOV domain. Genetically encoded protein<br />

photosensors as LOV are promising tools for engineering optical control of cellular behaviour;<br />

we are only beginning to understand how to couple these light detectors to effectors of<br />

choice (D. Strickland et al., Nat Methods 2010, 7: 623-26).<br />

The first fungal blue light BL photoreceptor, White collar-1 (WC-1), was isolated in<br />

Neurospora (by us and Prof Macino group) in 1996 and identified as a phototropin-like<br />

protein previously described only in plants. The blind wc-1 mutant is a phenocopy of white<br />

collar mutant wc-2. This coincidence of phenotype suggested that the two proteins (WC-1<br />

and WC-2) were in the same signal transduction pathways and that both were necessary for<br />

light responses in Neurospora. In fact WC-1 and WC-2 constitute the WC Complex (WCC)<br />

that now we know is the most diffuse type of photoreceptor among fungi. In Neurospora<br />

crassa light induced gene expression is dependent from the presence of the heterodimeric<br />

WC-1WC-2 complex and the acetylation NGF-1-dependent (Neurospora Gcn Five)of histone<br />

H3 residue K16 at the LRR (light responsive region) of specific promoters. The acetylation of<br />

histones is light dependent and transient (Grimaldi et al., 2006).<br />

Structural and functional aspects of WC-1 suggest that it can be considered as a primitive<br />

form of Nuclear Receptor (pNR), whose incoming signals are photons. Vertebrate nuclear<br />

receptors activate the transcription of their controlled genes through the action of a<br />

Coactivator. We already demonstrated the role of NGF-1 an Histone Acetyltransferase HAT<br />

(the ortologue of S. cerevisiae Gcn5p) as coactivator for light dependent switch on of<br />

trascription in Neurospora. During this first year of the project we demonstrated that NGF-1<br />

binds to WC-1 in D and L conditions of growth, and, only upon irradiation, it becomes active<br />

in the acetylation of histone H3 in the promoter region of light-genes. The conserved LXXLL<br />

sequences, involved in vertebrate NR-coactivators interactions have been identified in WC-1<br />

and NGF-1 and their relevance tested by mutagenesis. Our analyses revealed also a stable<br />

acetylation of WC-1 that is probably necessary in order to stabilize the photoreceptor, this<br />

acetylation however is independent either by light and by NGF-1. These data (reported in a<br />

manuscript just submitted) suggest a model based on a stable pNR-Coactivator interaction<br />

that becomes enzymatically active, as chromatin modifier, only after blue light illumination.<br />

47


Area 3: Molecular genetics of eukaryotes<br />

The other aspect we have developed is the characterization of the light sensor domain<br />

(LOV for Light Oxygen and Voltage) of Tuber borchii and Tuber melanosporum. The LOV<br />

domain is a region of around 150 amino acids deriving from the less specialized proteinprotein<br />

interaction domain called PAS (stand for PER ARNT and SIM). LOV is the light<br />

sensor domain of WC-1 and of many other proteins. It is a sensor domain that shares the<br />

mixed α/β fold common to the Per-ARNT-Sim class of environmental sensors and carry a<br />

flavin chromophore (FMN), that broadly absorbs blue light (see the figure). While these<br />

cofactors are non-covalently associated with the surrounding LOV domain in the dark,<br />

photoexcitation generates a covalent adduct between the flavin C4a carbon atom and an<br />

invariant cysteine residue. There is now strong interest in characterizing LOVs from different<br />

sources or mutagenized version in order to utilize such “new” LOV domain fused to effectors<br />

domain to obtain active molecules under dark/light control. With the aim of contributing to this<br />

“collection” of LOV domains we are characterizing Tuber LOV that we postulate to have<br />

particular properties in the response to blue light deriving from its life cycle spent in the dark.<br />

We constructed a Neurospora strain<br />

containing a chimeric WC-1 with its<br />

LOV domain substituted by TBLOV<br />

under a Neurospora promoter. We<br />

are characterizing the intensity and<br />

timing of bleu light photoresponses<br />

(i.e. carotenoids biosynthesis) comparing<br />

the chimeric strain to a Wild<br />

NcWC1 LOV<br />

TbWC1 LOV<br />

Type Neurospora and to a wc-1 KO<br />

mutant.<br />

PHY3<br />

(Crosson and Moffat, 2001)<br />

Fig. 1 - NcWC-1 and TbWC-1 LOV domains<br />

structures obtained by modellization on the<br />

experimentally resolved structure of PHY3.<br />

Note the loops of random coils indicated by<br />

the arrows, this variable region should be<br />

responsible of the individual specificity of the<br />

different LOVs.<br />

Research Group<br />

Patrizia Filetici, CNR researcher; Cristina De<br />

Luca, research fellow; Andrea Brenna, postdoc<br />

fellow; Raffaele Gerace, PhD student.<br />

Collaborations<br />

Simone Ottonello, Dipartimento di Biochimica,<br />

Università di Parma; Leonardo Baciarelli<br />

Falini, Dipartimento di Biologia Vegetale e<br />

Biotecnologie Agroambientali e Zootecniche,<br />

Università di Perugia; Giuseppe Briganti,<br />

Dipartimento di Fisica, Università Sapienza<br />

Roma.<br />

48


Area 3: Molecular genetics of eukaryotes<br />

Functional analysis of morgana, a gene involved in the control of<br />

centrosome duplication<br />

Silvia Bonaccorsi<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912473 - @: silvia.bonaccorsi@uniroma1.it<br />

We have recently demonstrated that Morgana/chp1, a conserved CHORD (cysteine and<br />

histidine rich domains) containing protein, is required for proper centrosome duplication and<br />

genome stability in both Drosophila and mouse. Null mutations in Drosophila morgana (mora)<br />

cause lethality and elicit a strong mitotic phenotype. Larval brains of mora mutants exhibit a<br />

high mitotic index with cells arrested in a prophase/prometaphase-like state and a high<br />

frequency of polyploid cells. In addition, most diploid cells of mora mutants display multiple<br />

centrosomes. Notably, the fly mitotic phenotype is fully rescued by the human orthologue of<br />

mora, highlighting a conserved function of this protein. morgana -/- mice die at the<br />

perimplantation stage, while morgana +/- primary cells and mice display a high frequency of<br />

supernumerary centrosomes and increased susceptibility to neoplastic transformation. In<br />

addition, tumor tissue array histochemical analysis revealed that Morgana is underexpressed<br />

in a large fraction of breast and lung human cancers. Thus, Morgana/chp-1 appears to<br />

prevent both centrosome amplification and tumorigenesis.<br />

Our research programme is aimed at understanding the mechanism of centrosome<br />

duplication, by elucidating the role of Morgana (Mora) in the Drosophila model system. Given<br />

the striking evolutionary conservation of the Mora function, these studies will provide<br />

information that can potentially be applied to humans, specifically in the realm of cancer<br />

biology.<br />

By combining a proteomic approach with the sophisticated Drosophila genetics, we are<br />

pursuing the identification and functional characterization of Mora-interacting proteins. To this<br />

end, in the course of the first year of the project we have generated transgenic flies carrying<br />

either a mora or a mora-GFP transgene under the PUASp GAL4-responsive promoter. This<br />

promoter responds very well to the V32 GAL4 driver, causing very high overexpression levels<br />

in the female germline. Flies carrying V34 GAL4 maternal driver and the PUASp-mora<br />

transgene will be then used to overexpress wild type and GFP-tagged Mora to isolate Morainteracting<br />

proteins by co-IP experiments.<br />

We have also carried out a functional analysis of mora in the Drosophila embryo, using<br />

video time-lapse microscopy. We have performed injection experiments in wild type embryos<br />

using an Alexa 633-labeled anti-Mora antibody. In both S2 tissue culture cells and larval<br />

brains immunostained with an anti-Mora antibody, the protein had previously been shown to<br />

accumulate in the cytoplasm of dividing cells, with no specific localization. Conversely, in the<br />

microinjection experiments carried out on embryos Mora exhibits a specific enrichment at the<br />

centrosome and at the spindle microtubules (Fig.1A). In addition, injection of the antibody<br />

49


Area 3: Molecular genetics of eukaryotes<br />

caused a deformation of the embryonic spindles, strongly suggesting a role of Mora in the<br />

control of spindle structure and dynamics (Fig. 1B). We now plan to confirm the centrosomal<br />

localization of Mora also in larval brains, using flies overexpressing GFP-tagged Mora that<br />

we have recently constructed (see above).<br />

We have also generated transgenic flies carrying a rok-GFP transgene under the PUASp<br />

GAL4-responsive promoter. We are currently using these flies in immunoprecipitation<br />

experiments with anti-GFP antibodies to verify whether Rok interacts with Mora. In addition,<br />

we will determine whether V32-induced Rok overexpression leads to centrosome<br />

amplification in the embryo.<br />

Fig. 1 - Selected frames from time<br />

lapse sequences of a buffer-injected<br />

embryo (control) and embryos injected<br />

with an Alexa 633-labeled anti-Mora<br />

antibody. (A) Prometaphase cells from<br />

an anti-Mora injectd embryo, showing<br />

a clear accumulation of Mora at the<br />

centrosomes (insets). (B) Metaphase<br />

cells from two subsequent cycles of<br />

division. Note, at cycle 11, the anti-<br />

Mora staining of spindle microtubules<br />

and the presence of a few aberrant<br />

spindles (inset). As a result of spindle<br />

degeneration, at the next cycle (cycle<br />

12), the embryo contains fewer cells<br />

than the control and many dramatically<br />

defective spindles (inset).<br />

Publications<br />

Mottier-Pavie V, Cenci G, Vernì F, Gatti M, Bonaccorsi S. Phenotypic analysis of misato<br />

function reveals roles of noncentrosomal microtubules in Drosophila spindle formation. J Cell<br />

Sci <strong>2011</strong>, 124: 706-717. doi: 10.1242/jcs.072348.<br />

Research Group<br />

Maria Grazia Giansanti, CNR researcher;<br />

Valeria Palumbo, postd-doc fellow; Ramona<br />

Lattao, PhD student.<br />

Collaborations<br />

James Wakefield, College of Life and<br />

Environmental Sciences, University of Exeter,<br />

UK.<br />

50


Area 3: Molecular genetics of eukaryotes<br />

RNA-RNA and RNA-protein interactions: role of small non-coding<br />

RNAs in gene expression control<br />

Irene Bozzoni<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912202 - @: irene.bozzoni@uniroma1.it<br />

It is becoming largely accepted that the non-coding portion of the genome rather than its<br />

coding counterpart is likely to account for the greater complexity of higher eukaryotes. High<br />

throughput transcriptome analysis of the last years has disclosed that the mammalian<br />

genome is pervasively transcribed into many different complex families of RNAs: besides a<br />

large number of alternative transcriptional start sites, termination and splicing patterns, a<br />

complex collection of new antisense, intronic and intergenic transcripts was found. Moreover,<br />

almost half of the polyadenylated species resulted to be non-protein-coding RNAs. Although<br />

many studies have helped unveiling the function of many small non-coding RNAs, very little<br />

is known about the long non-coding (lncRNA) counterpart of the transcriptome.<br />

So far, a large range of functions has been attributed to lncRNAs: it is clear that ncRNA<br />

molecules associate with chromatin structures to regulate transcription, others exert their<br />

control at the post-transcriptional level and still others perform key structural or catalytic<br />

functions, hence approximating the hitherto conventional roles of proteins. Therefore, it is<br />

evident that most, if not all, cellular pathways and developmental routes are intricate<br />

networks comprising a multitude of RNA and protein molecules acting in concert to<br />

accurately convey biological signals and respond to environmental cues.<br />

In the last year we have defined the role of several small non coding RNAs (microRNA) in<br />

the translation control of target mRNAs with a relevant function in muscle differentiation and<br />

identified several new circuitries of gene expression control involving miRNA as central<br />

players.<br />

More recently, we have shown the identification of a long non-coding RNA (linc-MD1) that<br />

cross-talks with specific mRNAs by competing for miRNA binding via their miRNA recognition<br />

motifs (MRE), thereby imposing an additional level of post-transcriptional regulation. These<br />

are features described as competing endogenous RNA (ceRNA).<br />

We showed that linc-MD1 controls muscle differentiation both in mouse and human<br />

myoblasts through its ability to bind specific miRNAs and in turn to affect the translation of<br />

the corresponding targets. Downregulation or overexpression of linc-MD1 correlated with<br />

retardation or anticipation of the muscle differentiation program, respectively. We showed<br />

that linc-MD1 “sponges” miR-133 and miR-135 to regulate the expression MAML1 and<br />

MEF2C, transcription factors that activate muscle-specific gene expression. Finally, we<br />

demonstrated that linc-MD1 exerted the same control over differentiation timing in human<br />

myoblasts, and that linc-MD1 levels are strongly reduced in the Duchenne muscle cells.<br />

51


Area 3: Molecular genetics of eukaryotes<br />

Publications<br />

Cacchiarelli D, Incitti T, Martone J, Cesana M, Cazzella V, Santini T, Sthandier O, Bozzoni I.<br />

miR-31 modulates dystrophin expression: novel implications in Duchenne muscular<br />

dystrophy therapy. EMBO Rep <strong>2011</strong>, 12: 136-41. doi: 10.1038/embor.2010.208.<br />

Cacchiarelli D, Legnini I, Martone J, D’Amico A, Bertini E, Bozzoni I. miRNAs as serum<br />

biomarkers for Duchenne muscular dystrophy. EMBO Mol Med <strong>2011</strong>, 5: 258-65. doi:<br />

10.1002/emmm.<strong>2011</strong>00133.<br />

Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, Tramontano A,<br />

Bozzoni I. Key ceRNA role for the long non-coding RNA linc-MD1 in the control of muscle<br />

differentiation. Cell <strong>2011</strong>, 147: 358-69. doi: 10.1016/j.cell.<strong>2011</strong>.09.028.<br />

Ragno R, Gioia U, Laneve P, Bozzoni I, Mai A, Caffarelli E. Identification of small-molecule<br />

inhibitors of the XendoU endoribonucleases family. ChemMedChem <strong>2011</strong>, 6: 1797-805.<br />

doi: 10.1002/cmdc.<strong>2011</strong>00281.<br />

Salvatori B, Iosue I, Djodji Damas N, Mangiavacchi A, Chiaretti S, Messina M, Padula F,<br />

Guarini A, Bozzoni I, Fazi F, Fatica A. Critical role of c-Myc in acute myeloid Leukemia<br />

involving direct regulation of miR-26a and histone methyltransferase EZH2. Genes Cancer<br />

<strong>2011</strong>, 2: 585-92. doi: 10.1177/1947601911416357.<br />

Research Group<br />

Presutti Carlo, professor; Fatica Alessandro,<br />

researcher; Elisa Caffarelli, CNR researcher;<br />

Julie Martone, Tiziana Santini, Ubaldo Gioia,<br />

Beatrice Salvatori, research fellows; Davide<br />

Cacchiarelli, post-doc fellow; Cesana Marcella,<br />

PhD student; Mariangela Morlando, graduated<br />

technician.<br />

Collaborations<br />

Alberto. Gulino, Antonio. Musarò, Sapienza<br />

Università di Roma; Alberto Auricchio,<br />

TIGEM, Napoli.<br />

52


Area 3: Molecular genetics of eukaryotes<br />

Role of metalloproteinases and their tissue inhibitors in the<br />

regulation of neurogenesis and gliogenesis from neural<br />

stem/progenitor cells<br />

Emanuele Cacci<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912206 - 06 4991 2371 - @: emanuele.cacci@uniroma1.it<br />

Neural progenitor (NP) cells continuously generate new neurons and glia in the adult<br />

brain. Regulation of adult neurogenesis involves a wide spectrum of signals from the<br />

microenvironment, the so-called neurogenic niche. The identification of intrinsic and extrinsic<br />

factors implicated in the modulation of neurogenesis is crucial to define strategies to<br />

successfully manipulate the neurogenic and gliogenic potential of NPs.<br />

Microglial cells are crucial components of the niche and they exert either beneficial or<br />

detrimental effects on neurogenesis, depending on their activation state and ranges of<br />

mediators produced (Cacci et al., 2005; Cacci et al., 2008; Ajmone Cat et al., 2010). Acute<br />

exposure to the prototypical activating agent lipopolysaccharide confers antineurogenic<br />

properties upon microglial cells. We have recently shown that docosahexaenoic acid (DHA),<br />

a long-chain polyunsatured fatty acid (L-PUFA) with potent immunomodulatory properties,<br />

exerts, through the modulation of microglial function, a proneurogenic effect (Ajmone Cat et<br />

al., <strong>2011</strong>).<br />

Among extrinsic factors, matrix metalloproteinases (MMPs) and their natural tissue<br />

inhibitors (TIMPs) are emerging as contributors to neurogenesis modulation. In addition,<br />

MMPs are multifaceted contributors to neuroinflammation. Particularly MMP-2 and MMP-9<br />

have been extensively studied in the brain, where they are found expressed at low levels<br />

under physiological conditions and up-regulated in various pathological conditions. MMPs<br />

and TIMPs are expressed in NPs and other brain cell types, including microglia. Noteworthy,<br />

MMPs and TIMPs regulate cytokine production in microglia in autocrine/paracrine manner<br />

and, vice versa, cytokines appear to be potent regulators of microglial MMP production.<br />

We explored whether MMPs modulate NP properties. We found that treatment of NPs<br />

with the clinically tested broad-spectrum MMP inhibitor Marimastat profoundly affected NP<br />

differentiation fate. Marimastat treatment allowed for enrichment in neuronal cells, inducing<br />

NPs to generate a higher percentage of neurons and a lower percentage of astrocytes,<br />

possibly affecting NP commitment. Consistently with its pro-neurogenic effect, Marimastat<br />

early down-regulated the expression of Notch target genes, such as HES1 and HES5.<br />

Interestingly, a potent inhibitor of the Notch signalling (DAPT), increased NP differentiation<br />

into neurons with an efficiency comparable to that of Marimastat and its administration in<br />

combination with Marimastat increased slightly, although significantly, neuronal<br />

differentiation. These data suggest that the early Marimastat-mediated inhibition of Notch<br />

signalling supports NP differentiation.<br />

53


Area 3: Molecular genetics of eukaryotes<br />

Since MMP-2 and MMP-9 are the main MMPs in the brain, we speculated that the MMP<br />

inhibitor effects on NPs were mediated through MMP-2 and/or MMP-9 inhibition. To clarify<br />

this point we investigated the expression profile of MMP-2 and MMP-9 in NPs. MMP-2 and<br />

MMP-9 profiling on proliferating and differentiating NPs revealed that MMP-9 was not<br />

expressed, whereas MMP-2 increased in the medium as proMMP-2 during differentiation,<br />

although its active form was not detectable. MMP-2 silencing or administration of<br />

recombinant active MMP-2 demonstrated that MMP-2 does not affect NP neuronal<br />

differentiation, nor it is involved in Marimastat pro-neurogenic effect. We also found that<br />

TIMP-2 is expressed in NPs and increases during late differentiation, mainly as a<br />

consequence of astrocyte generation. Endogenous TIMP-2 did not modulate NP neurogenic<br />

potential; however the pro-neurogenic action of Marimastat was at least partially mediated by<br />

TIMP-2, as demonstrated by silencing experiments. Our data exclude a major involvement of<br />

MMP-2 and MMP-9 in the regulation of basal NP differentiation, but highlight the ability of<br />

TIMP-2 to act as a key effector of the pro-neurogenic response to an inducing stimulus, such<br />

as Marimastat.<br />

We also addressed the molecular mechanisms involved in the regulation of NP<br />

properties. We focused our attention on the RE1-silencing transcription factor (REST), a<br />

master regulator of neuronal genes that can induce neuronal differentiation in NPs.<br />

To clarify which genes are transcriptionally repressed by REST we have used an<br />

adenovirus carrying the dominant negative of REST (DN:REST), or shRNA against REST.<br />

Analysis of the gene expression profile, including the whole spectrum of MMPs and TIMPs<br />

were performed by microarray. Combination of microarray and Chip sequencing analysis<br />

identified, among many other genes, two MMPs either directly (MMP-24) or indirectly<br />

(ADAM19ts) regulated by REST. We propose these MMPs as candidate for regulating NP<br />

differentiation, possibly cooperating with TIMP-2 in mediating the pro-neurogenic effects of<br />

Marimastat.<br />

Publications<br />

Ajmone-Cat M, Salvatori M, De Simone R, Mancini M, Biagioni S, Bernardo A, Cacci E,<br />

Minghetti L. Docosahexaenoic acid modulates inflammatory and antineurogenic functions<br />

of activated microglial cells. J Neurosci Res <strong>2011</strong> Epub. doi: 10.1002/jnr.22783.<br />

Research Group<br />

Stefano Biagioni, professor; Tonino Anelli,<br />

research fellow; Pasquale Caramanica, PhD<br />

student.<br />

Collaborations<br />

Ferdinando Mannello, Gaetana A. Tonti,<br />

Università Carlo Bo, Urbino; Luisa Minghetti,<br />

Maria Antonietta Ajmone-Cat, <strong>Istituto</strong><br />

Superiore di Sanità, Roma; Bukley Noel,<br />

King’s College, London, UK.<br />

54


Area 3: Molecular genetics of eukaryotes<br />

Does PARylated PARP-1 introduce an epigenetic mark on<br />

chromatin<br />

Paola Caiafa<br />

Department of Cellular Biotechnologies and Hematology<br />

℡: +39 06 49976530 - @: caiafa@bce.uniroma1.it<br />

Poly(ADP-ribosyl)ation (PARylation), a post-translational modification catalyzed by<br />

enzymes of PARP family, leads to the covalent introduction of ADP-ribose units onto<br />

acceptor chromatin proteins and even onto PARPs themselves. Poly(ADP-ribose)<br />

glycohydrolase (PARG) degrades ADP-ribose polymers (PARs) reverting the modification by<br />

its exo and endoglycosydase activities (Hassa and Hottiger, Front Biosci 2008; Krishnakumar<br />

and Kraus, Mol Cell 2010). PARs, present on covalently PARylated proteins and<br />

heterogeneous with respect to length and the presence of branches, are themselves able to<br />

interact non covalently with other proteins that bring a 20-amino-acid PAR-binding motif<br />

(Malanga and Althaus, Biochem Cell Biol 2005) as well as well-known “macro domain”<br />

(Krishnakumar and Kraus, Mol Cell 2010). PARP1 orchestrates chromatin dynamics<br />

modulating transcription and genome organization with different mechanisms. PARylation<br />

leads to chromatin decondensation modifying PARP1 itself as well as core histones or the<br />

linker histone H1 (Altmeyer et al., Nucleic Acids Res 2009; Krishnakumar and Kraus, Mol<br />

Cell 2010, Messner et al., Nucleic Acids Res 2010) while the block of PARylation induces<br />

chromatin condensation and the presence on some nucleosomes of macro H2A, whose NHD<br />

domain recruits PARP1, mediates heterochromatic silencing (Nusinow et al., J Biol Chem<br />

2007). Concerning gene expression PARylation is also important for the maintaining of DNA<br />

methylation patterns on genome (Caiafa et al., FASEB J 2009). In fact, PARylated PARP1<br />

and Dnmt1 interact. In this complex PARs, present on PARP1 molecule, interact non<br />

covalently with Dnmt1 preventing its access to DNA and thus its DNA methyltransferase<br />

activity (Reale et al., Oncogene 2005). As a consequence, PAR depletion - due to treatment<br />

of cells with a competitive inhibitor of PARP activity or ectopic over-expression of PARG -<br />

leads to the introduction of new anomalous methyl groups onto DNA. The multifunctional<br />

factor CTCF has been identified as an important player in the mechanism that sees together<br />

PARylation and DNA methylation (Guastafierro et al., J Biol Chem 2008). In fact, CTCF is by<br />

itself able to activate PARylation of PARP1 also in absence of DNA. This, together with the<br />

fact that CTCF binds only non methylated sequences on DNA and that, as shown by in silico<br />

data, its binding sites on genome often coincide with those of PARP1, suggests that CTCF<br />

marks on the genome those regions that must be maintained non methylated.<br />

Our aim has been to verify this hypothesis and to establish if PARylated Parp1, by itself<br />

and/or following PARylation of CTCF and/or other transcription factors, introduces an<br />

epigenetic code onto chromatin by marking those DNA sequences that must be maintained<br />

in an unmethylated state in normal cells thus preventing Dnmt1 access to these sequences.<br />

55


Area 3: Molecular genetics of eukaryotes<br />

The maintaining of DNA methylation patterns in mammals is required for correct<br />

development, genome stability, and transcriptional regulation. The mechanism(s) by which<br />

DNA sequences rich in CpG dinucleotides are maintained unmethylated has still to be<br />

defined. We demonstrated this mechanism for the DMR1 region present in the mouse<br />

Igf2/H19 imprinted locus where the binding of Ctcf, Parp1, Dnmt1 and PARs within the<br />

unmethylated DMR1 depends on PARs. In cells depleted of PARs new methyl groups are<br />

introduced onto this control region and the Ctcf-Parp1 complex is released from Ctcf target<br />

sequence (Zampieri et al., Biochem J 2012). Globally, our results, together with those<br />

reported for the control of p16 gene (Witcher and Emerson, Molecular Cell 2009), support a<br />

mechanism by which Parp1 tethered and activated at specific DNA target sites by Ctcf or<br />

other transcription factors to identify, preserves their methylation-free status.<br />

Publications<br />

Bacalini MG, Di Lonardo D, Catizone A, Ciccarone F, Bruno T, Zampieri M, Guastafierro T,<br />

Calabrese R, Fanciulli M, Passananti C, Caiafa P, Reale A. Poly(ADP-ribosyl)ation affects<br />

stabilization of Che-1 protein in response to DNA damage. DNA Repair (Amst) <strong>2011</strong>, 10:<br />

380-9. doi:10.1016/j.dnarep.<strong>2011</strong>.01.002.<br />

Bacalini MG, Tavolaro S, Peragine N, Marinelli M, Santangelo S, Del Giudice I, Mauro FR, Di<br />

Maio V, Ricciardi MR, Caiafa P, Chiaretti S, Foà R, Guarini A, Reale A. A subset of chronic<br />

lymphocytic leukemia patients display reduced levels of PARP1 expression coupled with a<br />

defective irradiation-induced apoptosis. Exp Hematol <strong>2011</strong> Epub. doi: 10.1016/j.<br />

exphem.<strong>2011</strong>.11.005.<br />

Calabrese R, Zampieri M, Mechelli R, Annibali V, Guastafierro T, Ciccarone F, Coarelli G,<br />

Umeton R, Salvetti M, Caiafa P. Methylation-dependent PAD2 upregulation in multiple<br />

sclerosis peripheral blood. Mult Scler <strong>2011</strong> Epub. doi: 10.1177/1352458511421055.<br />

Research Group<br />

Anna Reale, professor, Michele Zampieri,<br />

researcher, Maria Giulia Bacalini, Tiziana<br />

Guastafierro, post-doc fellows, Fabio<br />

Ciccarone, PhD student, Roberta Calabrese,<br />

graduated student.<br />

56


Area 3: Molecular genetics of eukaryotes<br />

DNA recombination of repeated sequences and genome instability:<br />

epigenetic implications<br />

Giorgio Camilloni<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912808 - @: giorgio.camilloni@uniroma1.it<br />

This project is centered on the simple model organism Saccharomyces cerevisiae, in<br />

order to gain information concerning basic mechanisms of genome stability to be translated<br />

to mammal model systems. We focus our attention on the ribosomal DNA (rDNA) locus, a<br />

repeated region where transcription, replication and recombination meet, both physically and<br />

functionally. These basic DNA transactions must be tightly regulated to avoid rDNA instability<br />

and common regulatory mechanisms should be active.<br />

We intend to explore the role of epigenetics in connecting replication, recombination and<br />

transcription. In fact, this aspect has not been deeply investigated yet, and its detailed<br />

characterization could make epigenetics a further element to manipulate in diseases due to<br />

genome instability.<br />

During this first year of activity we identified 2 fundamental processes affecting genome<br />

stability at rDNA:<br />

i) the histone content and the nucleosome number<br />

ii) the hyperacetylation of the H4K16 residue<br />

Regulation of the histone content and nucleosome number<br />

The basic unit of genome packaging is the nucleosome, and nucleosomes have long<br />

been proposed to restrict DNA accessibility both to damage and to transcription. Nucleosome<br />

number in cells was considered fixed, but recently aging yeast and mammalian cells were<br />

shown to contain fewer nucleosomes. We have shown that mammalian cells lacking High<br />

Mobility Group Box 1 protein (HMGB1) contain a reduced amount of core, linker, and variant<br />

histones, and a correspondingly reduced number of nucleosomes, possibly because HMGB1<br />

facilitates nucleosome assembly. Yeast nhp6 mutants lacking Nhp6a and -b proteins, which<br />

are related to HMGB1, also have a reduced amount of histones and fewer nucleosomes.<br />

Nucleosome limitation in both mammalian and yeast cells increases the sensitivity of DNA to<br />

damage, increases transcription globally, and affects the relative expression of about 10% of<br />

genes. In yeast nhp6 cells, the loss of more than one nucleosome in four does not affect the<br />

location of nucleosomes and their spacing, but nucleosomal occupancy. The decrease in<br />

nucleosomal occupancy is non-uniform and can be modelled assuming that different<br />

nucleosomal sites compete for available histones. Sites with a high propensity to occupation<br />

are almost always packaged into nucleosomes both in wild type and nucleosome-depleted<br />

cells; nucleosomes on sites with low propensity to occupation are disproportionately lost in<br />

nucleosome-depleted cells. We suggest that variation in nucleosome number, by affecting<br />

57


Area 3: Molecular genetics of eukaryotes<br />

nucleosomal occupancy both genomewide and gene-specifically, constitutes a novel layer of<br />

epigenetic regulation.<br />

Hyperacetylation of the H4K16 residue<br />

Transcription-associated recombination is an important process involved in several<br />

aspects of cell physiology. In the rDNA of S. cerevisiae, RNA polymerase II transcription<br />

dependent recombination among the repeated units has been demonstrated. In our<br />

experimental results, we showed the existence of mechanisms controlling this process at the<br />

chromatin level. On the basis of a small biased screening, we found that mutants of histone<br />

deacetylases and chromatin architectural proteins alter both the amount of Pol II-dependent<br />

non coding transcripts and recombination products at rDNA, in a coordinate manner. More<br />

interestingly, ChIP analyses revealed, in the above mutants, a correspondent variation of the<br />

histone H4 acetylation along the rDNA repeat, particularly at the lysine 16. We provided<br />

evidence that a single, rapid and reversible post-translational modification, the acetylation of<br />

the H4K16 residue, represents the epigenetic basis of the coordination of transcription and<br />

recombination at rDNA.<br />

Publications<br />

Celona B, Weiner A, Di Felice F, Mancuso FM, Cesarini E, Rossi RL, Gregory L, Baban D,<br />

Rossetti G, Grianti P, Pagani M, Bonaldi T, Ragoussis J, Friedman N, Camilloni G, Bianchi<br />

ME, Agresti A. Substantial histone reduction modulates genomewide nucleosomal<br />

occupancy and global transcriptional output. PLoS Biol <strong>2011</strong>, 9:e1001086. doi:<br />

10.1371/journal.pbio.1001086.<br />

Orecchia A, Scarponi C, Di Felice F, Cesarini E, Avitabile S, Mai A, Mauro ML, Sirri V,<br />

Zambruno G, Albanesi C, Camilloni G, Failla CM. Sirtinol treatment reduces inflammation<br />

in human dermal microvascular endothelial cells. PLoS One <strong>2011</strong>, 6:e24307. doi:<br />

10.1371/journal.pone.0024307.<br />

Research Group<br />

Elisa Cesarini, Francesca Di Felice, post-doc<br />

fellows; Anna D’Alfonso, Davide Gaglio, PhD<br />

students.<br />

Collaborations<br />

Marco Bianchi, Università Vita-Salute San<br />

Raffaele, Milano; Bastia Deepak, Department<br />

of Biochemistry & Molecular Biology; University<br />

of Charleston, USA.<br />

58


Area 3: Molecular genetics of eukaryotes<br />

Processing of the Neuroligins proteins and autism-related<br />

mutations<br />

Antonella De Jaco<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912310 - @: antonella.dejaco@uniroma1.it<br />

The α/β-hydrolase fold family is one of the largest families of proteins presenting<br />

significant structural homology with divergent functions: from catalytic hydrolysis to<br />

heterophilic cell adhesive interactions and hormone production. All the proteins of the family<br />

share a common three-dimensional core structure, containing the α/β-hydrolase fold domain<br />

that is crucial for proper protein localization and function. Mutations affecting the integrity of<br />

the α/β-hydrolase domain in conserved residues within of neuroligins, butyrylcholinesterase<br />

and thyroglobulin have been found associated with congenital disorders (De Jaco et al.,<br />

Protein Pept Lett <strong>2011</strong>). Characterization of the mutations found in the α/β-hydrolase fold<br />

domain shows that they mainly disrupt the architecture of the common structural domain,<br />

impairing trafficking along the secretory pathway and causing retention of the mutant protein<br />

in the endoplasmic reticulum (ER). This is indicating that the structural integrity of the α/βhydrolase<br />

fold domain is required for the export of the protein from the ER (De Jaco et al.,<br />

Protein Pept Lett <strong>2011</strong>, De Jaco et al., submitted). The α/β-hydrolase fold domain of the<br />

postsynaptic adhesion molecules neuroligins (NLGNs) interacts directly with presynaptic<br />

ligands belonging to the neurexins (NRXNs) family. In the last years, studying processing of<br />

the NLGNs proteins has been particularly interesting in relation to autism spectrum disorders<br />

(ASDs). Molecular genetic studies on ASDs are focusing on alterations in the functioning of<br />

synapses with the synaptic proteins playing a central role. The NLGNs-NRXNs synaptic cell<br />

adhesion complex is the best-characterized pathway implicated in ASDs. Genetic alterations<br />

in the NLGNs genes have been identified in autistic patients, suggesting that impairments in<br />

synaptic adhesion might lead to deficits in brain development (Betancur et al., Trends<br />

Neurosci 2009). We have studied an autism-associated mutation, R451C in NLGN3. We<br />

have shown that the mutation induces local protein misfolding of the α/β-hydrolase fold<br />

domain of NLGN3 that ultimately results mostly retained in the ER. The mutation slows down<br />

protein processing in the HEK-293 cell line and trafficking in neurons with a small fraction still<br />

reaching the cell surface (De Jaco et al., JBC 2010). More recently we have been<br />

investigating whether the retention of the NLGN3 R451C mutant protein in the ER is<br />

determining the activation of an ER stress response. The Unfolded Protein Response (UPR)<br />

is the most important ER signaling pathway that regulates protein folding and processing<br />

capacity of the ER (Kaufman, Genes Dev 1999). UPR is initiated by three transmembrane<br />

ER proteins, PERK (PKR-like ER kinase), IRE1 (inositol requiring kinase 1), and ATF6<br />

(activating transcription factor 6) that sense protein-misfolding and activate an ER-to-nucleus<br />

59


Area 3: Molecular genetics of eukaryotes<br />

signaling cascades in order to maintain homeostasis in the ER (Marciniak and Ron, Physiol<br />

Rev 2006). UPR activation has been detected in experimental models of neurodegenerative<br />

diseases (Matus et al., Curr Opin Cell Biol <strong>2011</strong>) but a UPR signaling pathway in ASDs has<br />

never been described. To evaluate whether the ER retention of the misfolded R451C NLGN3<br />

mutant protein was determining the activation of the UPR, we have studied the ATF6<br />

signaling pathway in the HEK-293 cell line. We have overexpressed in transient the NLGNs<br />

constructs together with a vector containing binding sites for ATF6, upstream of the<br />

luciferase reporter gene. Since these sequences are found in the promoters of most UPR<br />

target genes, the luciferase signal would indicate the activation of ATF6 and therefore and<br />

ER stress condition. Luciferase activity, measured at 24 and 48 hours after transfection<br />

shows that ATF6 activation is significantly higher for the mutant R451C NLGN3 compared to<br />

the wild type protein. We also studied IRE1 activation in response to accumulation of the<br />

R451C NLGN3 protein in the ER. Under ER stress condition, IRE1 activates the<br />

unconventional splicing of the mRNA of a transcriptional factor named XBP-1. We have<br />

studied the splicing of XBP1 mRNA by semiquantitative RT-PCR by and Real Time RT-PCR<br />

analysis, detecting both unspliced and spliced isoforms for XBP1. We have found that XBP1<br />

splicing is enhanced in the presence of the R451C mutant compared to the wild type protein<br />

(manuscript in preparation).<br />

From our results it is emerging that the autism-associated R451C mutation in NLGN3 is<br />

causing retention of the protein in the ER and this is determining an ER stress condition that<br />

results with the activation of ATF-6 and XBP-1, two of the UPR signaling pathways.<br />

Publications<br />

De Jaco A, Comoletti D, Dubi N, Camp S, Taylor P. Processing of cholinesterase-like<br />

alpha/beta-hydrolase fold proteins: alterations associated with congenital disorders. Protein<br />

Pept Lett <strong>2011</strong> Epub.<br />

Research Group<br />

Federica De Angelis, Lisa Ulbrich, post-doc<br />

fellows ; Flores Lietta Favaloro, student.<br />

Collaborations<br />

Davide Comoletti, Palmer Taylor, , University<br />

of California, San Diego, USA; Michael Lin,<br />

Stanford University, San Francisco, USA,<br />

Stefan Marciniak, University of Cambridge,<br />

UK.<br />

60


Area 3: Molecular genetics of eukaryotes<br />

Functional analysis of CG40218, a Drosophila melanogaster gene<br />

encoding a BCNT-like protein required for chromosome<br />

organization<br />

Patrizio Dimitri<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49917948 - @: patrizio.dimitri@uniroma1.it<br />

Aim of the project<br />

The GC40218 protein, also called YETI, is a member of the evolutionarily conserved<br />

family of BCNT proteins, whose function is still unclear. Using genetic and molecular<br />

analyses, as well as reverse genetics tools, we plan to delucidate the function played by<br />

YETI and CFDP1, the human ortholog of YETI.<br />

Transgenic lines expressing the YETI-tagged protein<br />

We tagged the YETI protein with GFP and 3xFLAG. Expression of the YETI-GFP and<br />

3xFLAG-YETI transgenes was induced in salivary glands and brains by the elav-GAL4 driver.<br />

Immunostaining of salivary gland chromosomes in Yeti-GFP, elav-GAL4 larvae using specific<br />

anti-GFP antibodies clearly show that the YETI-GFP fusion protein localizes in all nuclei and<br />

is recruited at hundreds of sites along the chromosome arms. Notably, the banding pattern<br />

produced by anti-GFP overlaps well with that produced by DAPI-staining. Although YETI-<br />

GFP is able to bind chromosomes, its expression driven by tub-gal4 transgene apparently<br />

does not rescue or mitigate the lethal phenotype of larvae homozygous for the LP1 null<br />

allele. We also made several transgenic lines carrying different deletions of the Yeti coding<br />

region (DYETI) tagged with GFP or 3xFLAG. The expression of such DYETI constructs<br />

under control of elavGAL4 driver will allow us to map in vivo the YETI region required for<br />

binding to chromosome.<br />

Identification of YETI interactors<br />

In order to identify the proteins that interact with YETI, we decided to perform<br />

Immunoprecipitation (IP) experiments in Drosophila S2 cells. For this purpose, a first step is<br />

to construct a pjZ4-3xFLAG-Yeti plasmid that will be transfected into S2 cells. The 3X-FLAG<br />

epitope coding region was inserted in pJZ4 and the obtained pJZ4-3XFLAG plasmid was<br />

used to clone the Yeti cDNA in frame with the epitope. Expression of such contruct in S2<br />

cells will give rise the YETI protein tagged at the N-terminal portion with the FLAG epitope. IP<br />

assay will be then performed on S2 cells containing pjZ4-3xFLAG-Yeti plasmids using the α-<br />

FLAG antibody.<br />

61


Area 3: Molecular genetics of eukaryotes<br />

Expression of the CFDP1 human protein in wild-type flies<br />

In order to investigate the effect of CFDP1 protein expression in the fly, we constructed<br />

Drosophila melanogaster transgenic lines carrying the CFDP1 human c-DNA. Expression of<br />

the CFDP1 protein in Drosophila melanogaster was performed by crossing two trangenic<br />

UAS-CFDP1 lines (15 and 16) to lines carrying ubiquitously active or tissue-specific GAL4<br />

drivers. By Western blot analysis using anti-CFDP1 monoclonal antibodies, we detected the<br />

presence of CFDP1 in protein extracts from w/w, elav-GAL4; UAS-CFDP1/+ larvae carrying<br />

the UAS-CFDP1 transgene under control of the elev-GAL4 driver. By immunostaing with anti-<br />

CFDP1, a diffuse signal was detected on polytene chromosomes and nuclei from w/w, elav-<br />

GAL4; UAS-CFDP1/+ larvae, while no staining was observed in the polytene chromosome of<br />

w/Y; UAS-CFDP1/+ control larvae. However, expression of UAS-CFDP1 transgene in wildtype<br />

flies results in lethality and phenotypic abnormalities, depending on the driver used. With<br />

elav-GAL4, lethality mainly occurs at third instar larvae, which often show reduced brains,<br />

sporadic melanotic masses in the larval hemocoel and chromosome defects; most notably, in<br />

elav-GAL4, UAS-CFDP1 larvae abnormally condensed polytene chromosomes were found.<br />

All the defects observed are clearly reminiscent of those found in Yeti mutants. In addition,<br />

when expressed simultaneously under control of elav-GAL4 driver, YETI-GFP and CFDP1<br />

protein mainly colocalize in the same polytene nuclei and their citological pattern remain<br />

unaffected. It is conceivable that the CFDP1 protein behaves as a dominant-negative and<br />

disrupts the normal function of YETI.<br />

Together, the results of our work provide evidence in favour of a conserved role of YETI<br />

and CFDP1 proteins in chromatin organization, which is extremely interesting in the light of<br />

the involvement of CFDP1 in complex developmental syndromes.<br />

Research Group<br />

Francesca Romana Mariotti, post-doc fellow,<br />

Elisabetta Damia, Giovanni Messina, PhD<br />

students, Emanuele Celauro, student.<br />

Collaborations<br />

Ruggiero Caizzi, Università di Bari; Ennio<br />

Giordano, Università di Napoli “Federico II”.<br />

62


Area 3: Molecular genetics of eukaryotes<br />

Genetic and molecular analysis of the mechanisms of Drosophila<br />

telomere protection<br />

Maurizio Gatti<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912842 - @: maurizio.gatti@uniroma1.it<br />

Telomeres are nucleoprotein complexes that protect chromosome ends from incomplete<br />

replication, degradation and fusion. In most organisms, telomeres are extended by<br />

telomerase and contain GC-rich repeats. Drosophila telomeres are elongated by<br />

transposition of 3 specialized retroelements rather than telomerase activity, and are<br />

assembled independently of the terminal DNA sequence. In organisms with telomerase,<br />

telomeres are protected by the CST and shelterin complexes, which specifically bind the<br />

telomeric DNA sequences. We have recently identified a Drosophila telomere-capping<br />

complex, terminin, which binds telomeric DNA in a sequence independent fashion and<br />

shares functional analogies with CST and shelterin. Terminin includes 4 telomere-specific<br />

proteins, HOAP, HipHop, Modigliani (Moi) and Verrocchio (Ver); Ver is structurally similar to<br />

Stn1, while HOAP, HipHop and Moi do not exhibit obvious homologies with known telomere<br />

proteins. Our research is aimed at the structural and functional characterization of terminin.<br />

We have 3 principal aims: 1) analyze the molecular interactions and the DNA binding<br />

properties of the terminin subunits using both biochemical methods and atomic force<br />

microscopy; 2) predict the three-dimensional structures of terminin subunits by bionformatic<br />

analysis, so as to design mutant proteins that will be tested for their functional properties; 3)<br />

perform genome-wide searches to identify additional proteins required for Drosophila<br />

telomere protection.<br />

In collaboration with Dr. Domenico Raimondo, we have performed a bioinformatic<br />

analysis of Moi and Ver and elaborated a structural model for Ver and a preliminary model for<br />

Moi. Based on these models we prepared several tagged (with GST or 6His) Moi and Ver<br />

truncations. We had already in hand tagged HOAP truncations fused with GST. Using these<br />

protein fragments we performed several pairwise pulldown experiments and defined the<br />

interaction domains within the terminin complex. These analyses also showed that the C-<br />

terminus of Ver contains an alpha-helix domain that mediates both DNA binding activity and<br />

formation of Ver homomultimers. These results are currently being used to produce mutant<br />

forms of HOAP, Moi and Ver, which will allow a variety of functional analyses.<br />

Our GST pulldown experiments revealed an unexpected and interesting molecular<br />

interaction. We found that Moi physically interacts with the Drosophila TAT-like (Dtl) protein,<br />

a trimethyl guanosine synthase homologous to yeast Tgs1 and human PIMT/hTGS1. Tgs1 is<br />

known to catalyze the conversion of the 7-monomethylguanosine caps of snRNAs, snoRNAs<br />

63


Area 3: Molecular genetics of eukaryotes<br />

and telomerase TLC1 RNA to 2,2,7-trimethylguanosine caps; Tgs1 has been implicated in<br />

nucleolar assembly, splicing of meiotic pre-mRNAs, and regulation of telomerase activity. Dtl<br />

is encoded by the bicistronic CG31241 locus, which produces a single transcript that<br />

contains 2 ORFs: One encodes Dtl and the other the Moi protein. These findings suggested<br />

a possible involvement of Dtl in regulation of transposition of the 3 Drosophila telomerespecific<br />

retrotransposons (HeT-A, TART and TAHRE). Preliminary results using real time RT-<br />

PCR showed that dtl mutants exhibit a 9-fold increase of TART transcripts compared to wild<br />

type. We are now exploring the possible mechanisms underlying this increase in TART<br />

transcript level.<br />

In humans, TGS1 is associated with and functionally dependent on the Survival of Motor<br />

Neuron (SMN) complex, which plays an essential role in spliceosomal snRNP assembly.<br />

Defects in human SMN1 cause spinal muscular atrophy autosomal recessive type 1 (SMA1),<br />

a neuromuscular disorder characterized by degeneration of the anterior horn cells of the<br />

spinal cord. We found that Dtl physically interacts with the Drosophila homologue of SMN1,<br />

and that hypomorphic mutations in dtl result in neurological problems in the fly. These data<br />

establish an unexpected link between the telomere and motoneuron maintenance, which will<br />

be explored in the future.<br />

Publications<br />

Mottier-Pavie V, Cenci G, Vernì F, Gatti M, Bonaccorsi S. Phenotypic analysis of misato<br />

function reveals roles of noncentrosomal microtubules in Drosophila spindle formation. J<br />

Cell Sci <strong>2011</strong>, 124: 706-717. doi: 10.1242/jcs.072348.<br />

Raffa GD, Capponi L, Cenci G, Gatti M. Terminin: a protein complex that mediatesepigenetic<br />

maintenance of Drosophila telomeres. Nucleus <strong>2011</strong>, 2: 383-91. doi: http://dx.doi.org/<br />

10.4161/ nucl.2.5.17873.<br />

Research Group<br />

Laura Ciapponi; professor; Grazia Daniela<br />

Raffa, Fiammetta Vernì, researchers; Maria<br />

Patrizia Somma, CNR researcher.<br />

Collaborations<br />

Stefano Cacchione, Dipartimento di Biologia e<br />

Biotecnologie “Charles Darwin”. Sapienza<br />

Università di Roma; Giovanni Cenci,<br />

Dipartimento di Biologia di Base ed Applicata,<br />

Università dell’Aquila; Domenico Raimondo,<br />

Dipartimento di Scienze Biochimiche “A. Rossi<br />

Fanelli”, Sapienza Università di Roma.<br />

64


Area 3: Molecular genetics of eukaryotes<br />

Role of epicardium and EMT/MET processes in cardiac progenitor<br />

cells generation and differentiation<br />

Alessandro Giacomello<br />

Department of Molecular Medicine<br />

℡: +39 06 4461481 - @: alessandro.giacomello@uniroma1.it<br />

Aim of the study. Cardiac progenitor cells (CPCs) isolated as Cardiospheres (CSps),<br />

represent a promising candidate for autologous cardiac cell therapy. CSps can be easily<br />

obtained from cells spontaneously migrating out of primary cardiac explants (explant-derived<br />

cells, EDCs) and re-create in vitro a niche-like microtissue, which seems to favor the<br />

maintenance of a “stemness” status and confers resistance to oxidative stress, thus<br />

enhancing in vivo engraftment (Messina et al., 2004, Li et al., <strong>2011</strong>). CSp-derived cells<br />

(CDCs) can be expanded in monolayers and retain the ability to form secondary<br />

cardiospheres (IICSps) when plated back in the appropriate conditions. Thus, this method<br />

yields a significant number of adult autologous CPCs suitable for clinical application, which<br />

can contribute in vivo to all the three main cell lineages of the heart. A better understanding<br />

of the mechanism underlying CSps formation and differentiative potential is required to<br />

control their fate in vivo and potentiate a cardiogenic outcome rather than a fibrogenic one,<br />

even in the hostile ischemic environment of an infarcted heart. Epithelial-to-mesenchymal<br />

transitions (EMT) play a central role in embryogenesis. Four distinct waves of EMT occur at<br />

different stages of heart morphogenesis. EMT has also been associated with the acquisition<br />

of stem cells properties both in adult tissues and cancer and with cardiac post-ischemic<br />

remodeling. The aim of this study was to analyze the possible role of EMT in CSp generation<br />

and development, by means of gene expression analysis at different culture stages and in<br />

vitro treatments with TGFβ, which is a key EMT inducer, and its antagonist SB431452.<br />

Results. Gene expression analysis, of the four different culture stages (EDCs, CSps,<br />

CDCs, IICSps), conducted using Taqman microfluidic array cards, and immunostainings<br />

reveal the involvement of EMT in CSps formation. In fact, EMT associated genes (Snail,<br />

FGFR, Slug, Twist, alk2/5, n-cadherin and smad2/3), mesenchymal markers (SMA, CX43)<br />

and mobility-associated genes (β-catenin, MMP2) are upregulated at all culture stages<br />

compared to EDCs, especially in CSps and IICSps. WT1, marker of the activated epicardium<br />

and EMT mediator, is significantly upregulated at all culture stages, following CSps<br />

formation. WT1 expression can be reactivated in adult epicardium in response to specific<br />

signals, including growth factors and ischemia. Since CSps are 3D structures, a hypoxic<br />

gradient is conceivably present from the periphery to the center, and hypoxia-mediated<br />

signals could be responsible for this re-activation. Another mediator of EMT, directly<br />

activated both by HIF1α and BMP/TGF pathway, is Notch1 involved in CPCs proliferation<br />

and maintenance. In our system it is significantly upregulated in CSps and IICSps compared<br />

65


Area 3: Molecular genetics of eukaryotes<br />

to EDCs and CDCs, similarly to Notch3 (known to control self-renewal and hypoxia survival<br />

in human mammospheres). In vitro treatment with TGFβ speeds up CSps formation and<br />

significantly increases overall CSps yield. Treatment with SB431542 induces a significant<br />

downregulation of genes related to the TGFβ pathways, and of Notch3 and 1, while BMP4 is<br />

significantly upregulated; such treatment results in a drastic inhibition of CSps formation,<br />

while it causes the spreading of pre-formed CSps.<br />

Future prospectives. In conclusion, Notch-Wt1 mediated EMT has a crucial role in CSps<br />

formation and may be involved in the acquisition of stem-cell like properties by cardiacexplant<br />

derived cells, just as observed in other adult tissues (Mani et al., 2008; Caja et al.,<br />

<strong>2011</strong>). In order to discern if Wt1 upregulation is the result of an enrichment for pre-existing<br />

Wt1+ progenitors or rather of the de-differentiation of EPDCs, transgenic Wt1-cre mice are<br />

going to be used. Based on these insights into molecular pathways involved in CSps<br />

formation and maintenance, small molecules previously screened on embryonic stem cells<br />

are going to be tested in order to enhance the in vivo cardiogenic potential of CSps.<br />

Publications<br />

Barile L, Cerisoli F, Frati G, Gaetani R, Chimenti I, Forte E, Cassinelli L, Spinardi L, Altomare<br />

C, Kizana E, Giacomello A, Messina E, Ottolenghi S, Magli MC. Bone marrow-derived cells<br />

can acquire cardiac stem cells properties in damaged heart. J Cell Mol Med <strong>2011</strong>, 15: 63-<br />

71. doi: 10.1111/j.1582-4934.2009.00968.x.<br />

Chimenti I, Rizzitelli G, Gaetani R, Angelini F, Ionta V, Forte E, Frati G, Schussler O,<br />

Barbetta A, Messina E, Dentini M, Giacomello A. Human cardiosphere-seeded gelatin or<br />

collagen scaffolds as cardiogenic engineered bioconstructs. Biomaterials <strong>2011</strong>, 32 : 9271-<br />

81. doi: 10.1016/j.biomaterials.<strong>2011</strong>.08.049.<br />

Fabrizi C, Angelini F, Chimenti I, Pompili E, Somma F, Gaetani R, Messina E, Fumagalli L,<br />

Giacomello A, Frati G. Thrombin and thrombin-derived peptides promote proliferation of<br />

cardiac progenitor cells in the form of cardiospheres without affecting their differentiation<br />

potential. J Biol Regul Homeost Agents <strong>2011</strong>, 25: S43-51.<br />

Forte E, Chimenti I, Barile L, Gaetani R, Angelini F, Ionta V, Messina E, Giacomello A.<br />

Cardiac cell therapy: the next (re)generation. Stem Cell Rev <strong>2011</strong>, 7: 1018-30. doi:<br />

10.1371/journal.pone. 0025669.<br />

Research Group<br />

Elisa Messina, medical manager; Francesco<br />

Angelini, research fellow; Isotta Chimenti,<br />

Elvira Forte, Roberto Gaetani, post-doc<br />

fellows; Vittoria Ionta, PhD student.<br />

Collaborations<br />

Luigi Aurisicchio, Takis Srl, Biogem, Rome;<br />

Mark Mercola, Sanford-Burnham Medical<br />

Research Institute, San Diego, USA; Pilar<br />

Ruiz-Lozano, Stanford School of medicine,<br />

Stanford, CA, USA.<br />

66


Area 3: Molecular genetics of eukaryotes<br />

Hedgehog signaling regulatory networks in brain cancer stem cells<br />

Alberto Gulino<br />

Department of Molecular Medicine<br />

℡: +39 06 4464021 - @: alberto.gulino@uniroma1.it<br />

Hedgehog pathway plays a pivotal role in development and tumorigenesis, processes<br />

sustained by stem cells. The transcription factor Nanog controls stemness acting as a key<br />

determinant of both embryonic stem cell self-renewal and differentiated somatic cells reprogramming<br />

to pluripotency, in concert with the loss of the oncosuppressor p53. We have<br />

shown that Nanog is highly expressed in stem cells from postnatal cerebellum and<br />

medulloblastoma, and acts as a critical mediator of Hedgehog-driven self-renewal through<br />

Gli1 and 2-dependent and p53-independent transcriptional regulation. Our data reveal a<br />

mechanism for the role of Hedgehog in the control of stemness that represents a crucial<br />

component of an integrated circuitry determining cell fate decision and involved in the<br />

maintenance of cancer stem cells.<br />

We have also observed that acetylation of Gli1 and 2 proteins (human Gli1 K518 and Gli2<br />

K757) is a transcriptional checkpoint of Hedgehog signaling, having acetylated Gli reduced<br />

transcriptional function while it is enhanced by HDAC1 and 2-dependent deacetylation.<br />

HDAC1 degradation is in turn controlled by the tumor suppressor REN KCTD11 , that is<br />

monoallelically deleted and silenced by hypermethylation in medulloblastoma. REN forms a<br />

tetrameric complex with Cul3 E3 ligase and HDAC1, leading to HDAC degradation. REN is a<br />

member of a novel family of Cul3 adaptors that includes two additional members (KCASH2<br />

and KCASH3 - KCTD Cullin3 Adaptors and Suppressors of Hedgehog). They also form<br />

homo- and heteromers of KCASH/Cul3 complexes, ubiquitinate and degrade HDAC1 and<br />

suppress in this way Hedgehog signaling. They are all downregulated in medulloblastoma<br />

and decrease tumor growth. Therefore, Hedgehog and KCASH/REN work in an opposite and<br />

coordinated fashion, by regulating HDAC1 levels and, ultimately, Gli1 acetylation, suggesting<br />

that HDAC inhibitors may be a promising and novel therapeutic strategy for<br />

medulloblastoma.<br />

Ubiquitination represents an additional Heddehog regulatory mechanism, in which we<br />

have shown that Numb activates the catalytic activity of Itch, releasing it from an inhibitory<br />

intramolecular conformation, that results in a Gli1 PPXYs and a phospho-serine/proline<br />

degron-dependent ubiquitination and degradation. Itch and Numb-insensitive Gli1 mutant<br />

determines enhanced Gli1-dependent medulloblastoma growth, migration and invasion<br />

abilities, as well as in vitro transforming activity. Our data reveal a novel mechanism of<br />

regulation of Gli1 stability and function, which influences Hedgehog/Gli1 oncogenic potential.<br />

67


Area 3: Molecular genetics of eukaryotes<br />

Publications<br />

Correale S, Pirone L, Di Marcotullio L, De Smaele E, Greco A, Mazzà D, Moretti M, Alterio V,<br />

Vitagliano L, Di Gaetano S, Gulino A, Pedone EM. Molecular organization of the cullin E3<br />

ligase adaptor KCTD11. Biochimie <strong>2011</strong>, 93: 715-24. http://dx.doi.org/10.1016/j.biochi.<br />

2010.12.014.<br />

De Smaele E, Di Marcotullio L, Moretti M, Pelloni M, Occhione MA, Infante P, Cucchi D,<br />

Greco A, Pietrosanti L, Todorovic J, Coni S, Canettieri G, Ferretti E, Bei R, Maroder M,<br />

Screpanti I, Gulino A. Identification and characterization of KCASH2 and KCASH3, 2 novel<br />

Cullin3 adaptors suppressing histone deacetylase and Hedgehog activity in<br />

medulloblastoma. Neoplasia <strong>2011</strong>, 13: 374-85. doi: 10.1593/neo.101630.<br />

Di Marcotullio L, Canettieri G, Infante P, Greco A, Gulino A. Protected from the inside:<br />

endogenous histone deacetylase inhibitors and the road to cancer. Biochim Biophys Acta<br />

Rev Cancer <strong>2011</strong>, 1815: 241-52. http://dx.doi.org/10.1016/j.bbcan.<strong>2011</strong>.01.002.<br />

Di Marcotullio L, Greco A, Mazzà D, Canettieri G, Pietrosanti L, Infante P, Coni S, Moretti M,<br />

De Smaele E, Ferretti E, Screpanti I, Gulino A. Numb activates the E3 ligase Itch to control<br />

Gli1 function through a novel degradation signal. Oncogene <strong>2011</strong>, 30: 65-76.<br />

doi:10.1038/onc.2010.394.<br />

Research Group<br />

Gianluca Canettieri, Elisabetta Ferretti<br />

professors, Enrico De Smaele, Lucia Di<br />

Marcotullio, researchers, Agnese Po, Evelina<br />

Miele, Marta Moretti, Sonia Coni, Azzura<br />

Greco, research fellows.<br />

Collaborations<br />

Alesse Edoardo, Università dell’Aquila,<br />

Pedone Carlo, Università di Napoli, Schininà<br />

Eugenia, Sapienza Università di Roma.<br />

68


Area 3: Molecular genetics of eukaryotes<br />

The interplay between epigenetics, cell cycle and homologous<br />

recombination in gene therapy by Small Fragment Homologous<br />

Replacement (SFHR)<br />

Marco Lucarelli<br />

Department of Cellular Biotechnologies and Hematology<br />

℡: +39 06 4451784 - @: marco.lucarelli@uniroma1.it<br />

The aim of this project is to clarify the molecular mechanisms underlying an in situ gene<br />

targeting approach called Small Fragment Homologous Replacement (SFHR). A small DNA<br />

fragment (SDF) is used as a corrector to stably modify a genomic sequence, restoring the<br />

wild-type gene function. Although DNA repair pathways, in particular homologous<br />

recombination (HR), seem to be involved, the molecular mechanisms of SFHR are poorly<br />

understood, with its potential currently limited by a low and variable frequency of correction.<br />

This project focuses on the scarcely studied relationships between SFHR, DNA methylation<br />

and repair, chromatin structure and cell cycle.<br />

Mouse embryonic fibroblast (MEF) clones were stably modified with a genomic<br />

integration of the wild type enhanced green fluorescent protein (wtEGFP clones) sequence,<br />

in which a non sense mutation was also introduced (mEGFP clones). In this reporter system,<br />

a corrector SDF homologous to the wtEGFP sequence can integrate in mEGFP clones and<br />

restore the fluorescence of the cells. Several molecular and cellular analyses of MEF clones<br />

confirmed the genomic correction, its persistence over time, and the absence of detectable<br />

random integration of SDF. The efficiency of SFHR is evaluated at DNA and RNA levels, as<br />

well as functionally quantified by cytofluorimetric analysis.<br />

The best experimental characteristics of corrector SDF and the best transfection<br />

conditions, in term of correction efficiency and cell viability, were selected in unsynchronized<br />

MEF clones. To determine the cell cycle influence, we evaluated gene targeting in G0/G1,<br />

G1/S and G2/M phases. In G2/M the SFHR resulted favoured, reaching 0.5% of correction,<br />

probably because of the absence of nuclear envelope, the tetraploid status of the cells and<br />

the higher activity of HR. Artificially methylated and plasmid-excised SDFs showed a lower<br />

correction efficiency than unmethylated PCR synthesized SDF, probably because of the<br />

creation of complexes between methylated SDFs and nuclear proteins that inhibit the<br />

integration. After sorting, corrected MEF gradually lost EGFP fluorescence concomitantly<br />

showing an increased site-specific methylation pattern at EGFP locus, possibly influenced by<br />

the correction event. A cause / effect relationship was evidenced by the reversion of DNA<br />

methylation and EGFP expression after treatment with the demethylating agent 5-aza-2’-<br />

deoxycytidine (aza-dC). An increment of SFHR efficiency to 2.5% was evidenced by the<br />

simultaneous inhibition of DNA methylation and poly-(ADP-ribose) polymerase, highlighting<br />

synergistic effects of these two pathways on SFHR.<br />

The quantitative expression of 84 genes involved in the response to several kinds of DNA<br />

damage, using real time expression arrays, was also investigated. Their expression levels in<br />

69


Area 3: Molecular genetics of eukaryotes<br />

MEF were heterogeneous and most of the genes affected by SFHR resulted to be low<br />

expressed genes. After an early (24h after treatment with SDF) upregulation of 70 DNA<br />

damage repair genes, there was a late (72h after treatment) downregulation with 51 genes<br />

reaching a lower expression level than untreated control. The genes with statistically<br />

significant expression differences between treatment and control resulted to be 33. On the<br />

contrary to current opinions, these genes belong to different DNA repair pathways, and not<br />

only to HR pathway. Their early upregulation may constitute the molecular basis for the<br />

correction, whereas their late downregulation may be responsible for the reduced correction<br />

efficiency, possibly as an adaptive response to cell invasion by exogenous DNA.<br />

These results contribute to the comprehension of the molecular mechanisms underlying<br />

cell invasion by exogenous DNA and its genomic integration. The selection of specific<br />

molecular targets to manipulate provides suggestions for increasing gene repair efficiency to<br />

achieve an higher correction for a practical SFHR application to ex vivo therapeutic<br />

approaches.<br />

Research Group<br />

Roberto Strom, professor; Fabrizio Ceci,<br />

Giampiero Ferraguti, researchers; Silvia<br />

Pierandrei, PhD student; Sabina Maria Bruno,<br />

graduated student.<br />

Collaborations<br />

Paola Borgiani, Annalisa Botta, Andrea<br />

Luchetti, Arianna Malgieri, Giuseppe Novelli,<br />

Federica Sangiuolo, Dip. di Biopatologia e<br />

Diagnostica per Immagini, Università di Roma<br />

Tor Vergata.<br />

70


Area 3: Molecular genetics of eukaryotes<br />

Interplay between myogenic factors and cell cycle control:<br />

regulation and role of the cdk inhibitor p57kip2<br />

Rossella Maione<br />

Department of Cellular Biotechnologies and Hematology<br />

℡: +39 06 4457737 - @: maione@bce.uniroma1.it<br />

Cell proliferation and differentiation are highly coordinated processes during<br />

development, homeostasis and regeneration of multicellular organisms. Cell cycle exit is not<br />

only critical for the formation of appropriate cell numbers, but also is a prerequisite for<br />

terminal differentiation in many cell types.<br />

The interdependence of cell proliferation and differentiation has been extensively<br />

investigated in the skeletal muscle system, in which the whole differentiation program can be<br />

initiated by the MyoD family of muscle-specific transcription factors, even when ectopically<br />

expressed in non muscle cell types. MyoD directs a complex program of gene expression<br />

through the cooperation with other transcription factors and chromatin remodeling enzymes.<br />

In addition to regulate muscle-specific genes, MyoD is also involved in coupling the onset of<br />

differentiation with cell cycle withdrawal. The regulated expression of cdk inhibitors (CKIs)<br />

plays a fundamental role in muscle differentiation.<br />

The function and the regulation of the CKI p57 kip2 (p57) are quite peculiar. p57 is the only<br />

CKI which deficiency causes severe developmental defects both in humans and mice.<br />

Moreover, compared to other CKIs, p57 shows a more restricted tissue and cell-type<br />

distribution. Finally, p57 belongs to a network of imprinted genes involved in growth<br />

regulation and is subject to a complex epigenetic control. Despite the importance of this CKI,<br />

the specific functions of p57 and the molecular mechanisms regulating its expression during<br />

differentiation processes are still poorly understood.<br />

Our research project is focused on two related topics. One concerns the specific roles<br />

that p57 plays in the establishment and in the maintenance of terminal differentiation in<br />

muscle cells. The other one concerns the transcriptional mechanisms controlling p57<br />

expression during myogenesis.<br />

During the last year, we mainly focused on the second topic. The results obtained<br />

allowed us to highlight a novel epigenetic mechanism by which MyoD regulates gene<br />

expression during myogenesis. We have shown that the induction of p57 requires MyoD<br />

binding to a long-distance element located within the imprinting control region KvDMR1 and<br />

the consequent release of a repressive chromatin loop involving p57 promoter. We have also<br />

provided evidence that differentiation-dependent regulation of p57, while involving a region<br />

implicated in the imprinting process, is distinct and hierarchically subordinated to the<br />

imprinting control. These findings highlight a novel mechanism, involving the modification of<br />

higher order chromatin structures, by which MyoD regulates gene expression. Our results<br />

also suggest that a repressive chromatin loop mediated by KvDMR1 could account for the<br />

71


Area 3: Molecular genetics of eukaryotes<br />

highly restricted expression of p57 during normal development and, possibly, for its aberrant<br />

silencing in some pathologies.<br />

The results are detailed in the following submitted manuscripts: Busanello et al., MyoD<br />

regulates p57 kip2 expression by interacting with a distant cis-element and modifying a higherorder<br />

chromatin structure and Battistelli et al., Functional interaction between MyoD and<br />

CTCF in the regulation of long-range chromatin interactions during muscle differentiation.<br />

Research Group<br />

Anna Busanello, Rosaria Carbone, post-doc<br />

fellows; Cecilia Battistelli, Agnese Ciotti,<br />

Cassandra Mostocotto, PhD students.<br />

Collaborations<br />

Maurizia Caruso, <strong>Istituto</strong> di Neurobiologia e<br />

Medicina Molecolare, CNR, Roma.<br />

72


Area 3: Molecular genetics of eukaryotes<br />

Handling and toxicity of mutant serpins underlying human disease<br />

María Elena Miranda Banos<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912310 - @: mariaelena.mirandabanos@uniroma1.it<br />

Research summary<br />

The serpinopathies are human pathologies caused by mutations that promote<br />

polymerisation and intracellular deposition of proteins of the serpin (serin protease inhibitors)<br />

superfamily, leading to toxicity and cell death. The dementia FENIB is caused by<br />

polymerisation of the neuronal serpin neuroserpin (NS). The identification of the molecular<br />

bases of polymer accumulation and toxicity using in vitro model systems is an important<br />

route for understanding these conditions and developing therapeutic interventions.<br />

Aims<br />

Our aim is to understand how NS polymers accumulate within the endoplasmic reticulum<br />

(ER) and what is the mechanism of their cellular toxicity. More specifically, we aim to:<br />

- Define the role of N-linked glycosylation on the ER handling of NS.<br />

- Characterise the cell toxicity of NS polymers in a new neuronal model of FENIB.<br />

First year results<br />

1. Role of N-linked glycosylation in the ER handling of NS. NS has three consensus<br />

motifs for N-linked glycosylation (N157, N321 and N401). We have shown that only two of<br />

them are glycosylated on wild type NS, while the fastest polymerising mutant (Gly392Glu NS)<br />

presents as two populations, with two and three glycan chains respectively. To understand<br />

the role of the extra glycan, we used direct mutagenesis to eliminate each of the three<br />

glycans in our pcDNA3.1 plasmids carrying wild type (WT) and Gly392Glu (G392E) NS. We<br />

have studied these mutants, along with the control WT and G392E plasmids, by transient<br />

transfection in COS-7 cells. Cells and culture media were analysed by SDS and nondenaturing<br />

PAGE followed by western blot for NS, luciferase (transfection efficiency control)<br />

and GAPDH (protein loading control). Our results show that: i) the N401 glycosylation site is<br />

not used in wild type NS and is additionally used in Gly392Glu NS; ii) removing the N321 site<br />

causes polymerisation of wild type NS, demonstrating that glycosylation is important for the<br />

structural stability of NS; and iii) removing the N401 glycosylation site in Gly392Glu NS<br />

causes a higher degree of mutant protein accumulation within the cells, probably by<br />

interfering with mutant protein degradation by the proteasome.<br />

2. Cell toxicity of NS polymers in a new model of FENIB. We have prepared expression<br />

constructs for constitutive, stable expression of wild type, Ser52Arg (medium degree of<br />

73


Area 3: Molecular genetics of eukaryotes<br />

polymerisation), Gly392Glu (high degree of polymerisation) and delta (a truncated version<br />

that does not polymerise) NS, by cloning these cDNAs into the pTP6 vector, suitable for high<br />

expression in neural progenitors. This vector also has an antibiotic selection marker for<br />

efficient selection of transfected cells with stable transgene integration and robust expression<br />

levels of the gene of interest. Cell lines of neural progenitors from embryonic and adult<br />

mouse brain cortex are already available for transfection through our collaboration with Dr.<br />

Giuseppe Lupo and Dr. Emanuele Cacci.<br />

Future work<br />

1. Role of N-linked glycosylation on the ER handling of NS. We will: i) assess the<br />

amounts of intra- and extracellular NS quantitatively, by using two complementary sandwich<br />

ELISA specific for total and polymerised NS respectively; ii) study the degradation of<br />

Asn401Gln-Gly392Glu NS at steady state, by using proteasome inhibitors and comparing the<br />

amounts of the glycosylation mutant with the control plasmid (Gly392Glu NS); iii) in<br />

collaboration with Prof. Lomas (Cambridge, UK), we will study the intracellular processing of<br />

our glycosylation mutants in more detail by pulse-chase analysis in COS-7 cells.<br />

2. Cell toxicity of NS polymers in a new model of FENIB. We will transfect our new<br />

constructs into neural progenitor cell lines and will generate stably transfected cells by<br />

antibiotic selection. We will assess the expression levels of each NS variant by SDS-PAGE<br />

and western blot, and their monomeric vs. polymeric state by non-denaturing PAGE and<br />

western blot. We will use sandwich ELISA to quantify intracellular vs extracellular presence<br />

of each type of neuroserpin, and immunocytochemistry to study the intracellular localisation<br />

of retained NS. After the initial evaluation, we will proceed with long-term cultures to assess<br />

the toxicity of intracellular polymers of Ser52Arg and Gly392Glu NS in this system, by looking<br />

at activation of NFkB, oxidative stress and apoptosis. This will be done in terminally<br />

differentiated neural progenitor cells, using protocols available in our lab that allow efficient<br />

terminal differentiation of neural progenitors into neurons, astrocytes or oligodendrocytes.<br />

Research Group<br />

Claudia Moriconi, post-doc fellow; Valentina<br />

Timpano, student.<br />

Collaborations<br />

Emanuele Cacci, Nicoletta Carucci,<br />

Antonella De Jaco, Giuseppe Lupo, Sapienza<br />

Università di Roma; David A. Lomas, Stefan<br />

Marciniak, University of Cambridge, UK; Juan<br />

Perez, University of Malaga, Spain; Nigel<br />

Birch, University of Auckland, New Zealand;<br />

Martino Bolognesi, Stefano Ricagno,<br />

Università di Milano, Mauro Manno, Vincenzo<br />

Martorana, Rosetta Noto, CNR, <strong>Istituto</strong> di<br />

Biofisica, Palermo; Luisa Schiaffonatti, Anna<br />

Fra, Università di Brescia.<br />

74


Area 3: Molecular genetics of eukaryotes<br />

Characterization of the muscle-nerve crosstalk in a neuromuscular<br />

disease<br />

Antonio Musarò<br />

Department of Anatomical, Histological, Forensic & Orthopaedic Sciences<br />

℡: +39 06 49766956 - @: antonio.musaro@uniroma1.it<br />

One of the crucial systems severely affected in several neuromuscular diseases,<br />

including Amyotrophic Lateral Sclerosis (ALS), Spinal Muscular Atrophy (SMA) and aging, is<br />

the loss of effective connection between muscle and nerve, leading to a pathological noncommunication<br />

between the two tissues.<br />

In a previous work (Dobrowolny et al., JCB 2005), we demonstrated that muscle<br />

expression of a local IGF-1 isoform protects motor neurons in an ALS mouse model. More<br />

recently, we showed that mIGF-1 improves muscle mass and extends the survival of severe<br />

SMA mice independent of SMN induction (Bosch-Marcé et al., Hum Mol Genet <strong>2011</strong>). Our<br />

studies suggests that muscle fibers provide appropriate factors, such as mIGF-1, for the<br />

maintenance of a functional muscle and neuron survival and that skeletal muscle is a primary<br />

target for the dominant action of inherited SOD1 mutation. This hypothesis has been<br />

validated by our recent studies, demonstrating that muscle-restricted expression of mutant<br />

SOD1 G93A was sufficient to recapitulate some of the pre-symptomatic sign of ALS<br />

(Dobrowolny et al., Cell Met 2008). The goals of this project is to define the molecular<br />

signature of the retrograde muscle-nerve interaction.<br />

In the first part of the research activity, we extended previous study with the aim to define<br />

the potential signal transduction pathways activated in response to SOD1 G93A , expressed<br />

either locally or ubiquitously, that cause muscle atrophy.<br />

Several studies have indicated the activation of apoptotic events in pathological<br />

conditions associated with muscle atrophy. The central component and the executor of the<br />

apoptotic machinery are a family of cysteine proteases called caspases. We reported that<br />

muscle atrophy in MLC/SOD1 G93A mice does not involve the activation of caspases<br />

(Dobrowolny et al., Skelet Muscle <strong>2011</strong>). Of note, the expression and activity of caspases<br />

increased in the muscle of the classical animal models of ALS, which express the SOD1 G93A<br />

gene ubiquitously, only at paralysis stage.<br />

Thus, it seems likely that muscle atrophy is a primarily events associated with the toxic<br />

effects of SOD1 G93A mutant protein, whereas motor neuron degeneration is a later event that<br />

contributes to exacerbate the atrophic phenotype, promoting paralysis and muscle wasting.<br />

The present work has provided new insights concerning the control of muscle fiber size and<br />

the mechanisms of atrophy in animal models of ALS. Moreover we started monitoring the<br />

intracellular organelles and neuromuscular junctions (NMJ). Preliminary experiments<br />

revealed significant alterations in mitochondria morphology and function and in NMJ.<br />

75


Area 3: Molecular genetics of eukaryotes<br />

Mitochondria were often clustered, abnormally shaped, and presented a translucent<br />

appearance, edematous internal matrix with abnormal and/or missing internal cristae.<br />

This initial morpho-functional alteration of skeletal muscle might be followed by NMJ<br />

destruction, distal axonopathy, astrocytosis in the spinal cord, and finally motor neuron loss.<br />

This might lead to a late exacerbation of muscle atrophy caused by caspases activation.<br />

Further work will define if and how skeletal muscle impacts the nervous system.<br />

Publications<br />

Bosch-Marcé M, Wee CD, Martinez TL, Lipkes CE, Choe DW, Kong L, Van Meerbeke JP,<br />

Musarò A, Sumner CJ. Increased IGF-1 in muscle modulates the phenotype of severe<br />

SMA mice. Hum Mol Genet <strong>2011</strong>, 20: 1844-53. doi: 10.1093/hmg/ddr067.<br />

Carosio S, Berardinelli MG, Aucello M, Musarò A. Impact of ageing on muscle cell<br />

regeneration. Ageing Res Rev <strong>2011</strong>, 10: 35-42. http://dx.doi.org/10.1016/j.arr.2009.08.001.<br />

Dobrowolny G, Aucello M, Musarò A. Muscle atrophy induced by SOD1G93A expression<br />

does not involve the activation of caspase in the absence of denervation. Skelet Muscle<br />

<strong>2011</strong>, 1: 3. doi: 10.1186/2044-5040-1-3.<br />

Forte G, Pietronave S, Nardone G, Zamperone A, Magnani E, Pagliari S, Pagliari F, Giacinti<br />

C, Nicoletti C, Musaró A, Rinaldi M, Ribezzo M, Comoglio C, Traversa E, Okano T, Minieri<br />

M, Prat M, Di Nardo P. Human cardiac progenitor cell grafts as unrestricted source of<br />

supernumerary cardiac cells in healthy murine hearts. Stem Cells <strong>2011</strong>, 29: 2051-61. doi:<br />

10.1002/stem.763.<br />

Research Group<br />

Gabriella Dobrowolny, Emanuele Rizzato,<br />

post-doc fellows; Carmine Nicoletti, technician.<br />

Collaborations<br />

Marco Sandri, Dipartimento Scienze<br />

Biomediche- VIMM, Università di Padova;<br />

Feliciano Protasi, Ce.S.I., Università degli<br />

Studi G. d’Annunzio, Chieti; Rüdiger Rudolf,<br />

Univ. of the State of Baden-Wuerttemberg and<br />

National Laboratory of the Helmholtz<br />

Association- Germany; Charlotte Sumner,<br />

Department of Neurology, Johns Hopkins<br />

Bloomberg School of Public Health, USA.<br />

76


Area 3: Molecular genetics of eukaryotes<br />

Role of the COP9 signalosome in transcription modulation and<br />

chromatin organization in yeast and plants<br />

Rodolfo Negri<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49917790 - @: rodolfo.negri@uniroma1.it<br />

Accurate regulation of transcription in time and space is critical for the establishment and<br />

maintenance of gene expression patterns in eukaryotes. In the past years, several<br />

components of the ubiquitin/proteasome system (UPS) have been shown to be necessary for<br />

the tight regulation of gene expression with possible important implications for cellular<br />

homeostasis. A key component of the UPS is the COP9 signalosome (CSN), a protein<br />

complex conserved in all eukaryotes. CSN removes the small peptide NEDD8 (an ubiquitin<br />

like modifier) from the cullin-RING family of E3 ubiquitin ligases. E3 ligases are the enzymes<br />

responsible for poly-ubiquitination of specific substrates, which can be subsequently<br />

recognized and degraded by the proteasome. The reaction catalyzed by CSN is necessary<br />

for the tight regulation of the assembly/disassembly cycles of these ligases and is essential<br />

for all higher eukaryotes, since null CSN mutants from different organisms ranging from<br />

plants to mammals are lethal at very early developmental stages.<br />

At the cellular level, CSN mutants from different organisms display de-repression and,<br />

more in general, miss-regulation of several sets of genes. This could be due simply to the<br />

fact that CSN is necessary for the proper degradation of several transcription factors.<br />

However, recent evidence suggests that CSN might perform specific tasks directly on the<br />

genomic chromatin.<br />

The CSN from budding yeast S. cerevisiae has been characterized and, like in higher<br />

eukaryotes, is responsible for the de-neddylation of cullin-RING ubiquitin ligases. However,<br />

while the CSN in most higher eukaryotes is composed of 8 subunits and is a paralog of the<br />

lid of the proteasome, the CSN from budding yeast consists of six subunits: Csn9, Csn10,<br />

Csn11/Pci8, Csn12, Csn5/Rri1 and Csi1. In addition, in contrast to other eukaryotes, all the<br />

CSN subunits from S. cerevisiae are non-essential. The non essentiality of CSN components<br />

and the availability of powerful genetic tools make S. cerevisiae a very promising model<br />

system to elucidate some aspects of the nuclear role of this complex.<br />

We performed a transcriptomic analysis of a S. cerevisiae strain deleted in CSN5 (the deneddylating<br />

subunit of the Cop9 complex), as compared with its isogenic wild type strain.<br />

Data suggest that Csn5 is involved in the modulation of several genes controlling zinc<br />

uptake and metabolism and ergosterol biosynthesis. All the modulations were confirmed by<br />

real-time RT-PCR and most of them were observed in some of the other CSN deletion<br />

mutants. We also tested the various CSN components deleted strains for phenotypic features<br />

related to defects in zinc uptake and ergosterol biosynthesis and found a good correlation<br />

with the transcriptomic pattern. ChIP on chip analysis suggests that Csn5 associates with<br />

77


Area 3: Molecular genetics of eukaryotes<br />

some of the modulated genes, suggesting a direct mode of transcriptional regulation. This<br />

mode of regulation is likely to consist in a decrease of the turnover of a transcriptional<br />

activator or in an increase of the turnover of a transcriptional repressor (Fig.1). We are now<br />

trying to identify putative targets. A promising candidate is Mot3/Rox7 which represses genes<br />

involved both in ergosterol biosynthesis and in zinc import and metabolism.<br />

Fig. 1 - Csn5 could act by increasing<br />

the turnover of a repressor or<br />

decreasing the turnover of an<br />

activator<br />

Publications<br />

Bosio MC, Negri R, Dieci G. Promoter architectures in the yeast ribosomal expression<br />

program. Transcription <strong>2011</strong>, 2: 71-77. doi: 10.4161/ trns.2.2.14486.<br />

Piccinni E, Chelstowska A, Hanus J, Widlak P, Loreti S, Tata AM, Augusti-Tocco G, Bianchi<br />

MM, Negri R. Direct interaction of Gas41 and Myc encoded by amplified genes in nervous<br />

system tumours. Acta Biochim Pol <strong>2011</strong>, 58: 529-34.<br />

Romagnoli G, Cundari E, Negri R, Crescenzi M, Farina L, Giuliani A, Bianchi MM.<br />

Synchronous protein cycling in batch cultures of the yeast Saccharomyces cerevisiae at log<br />

growth phase Exp Cell Res <strong>2011</strong>, 317: 2958-68. doi: 10.1016/j.yexcr.<strong>2011</strong>.09.007.<br />

Research Group<br />

Giovanna Serino, professor; Teresa Rinaldi,<br />

researcher; Valerio Licursi, post-doc fellow;<br />

Angela Alagia, Chiara Salvi, PhD students.<br />

Collaborations<br />

Gianni Balliano, Università degli Studi di<br />

Torino; Elah Pick, Haifa University, Israel.<br />

78


Area 3: Molecular genetics of eukaryotes<br />

The biogenesis of piRNAs and their involvement in transposon<br />

silencing and heterochromatin formation in Drosophila<br />

Sergio Pimpinelli<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912876 - @: sergio.pimpinelli@uniroma1.it<br />

The "canalization" concept (Waddington, 1959) describes the resistance of a<br />

developmental process to phenotypic variation, regardless of genetic and environmental<br />

perturbations, owing to the existence of buffering mechanisms. Severe perturbations, which<br />

overcome such buffering mechanisms, produce altered phenotypes that can be heritable and<br />

can themselves be canalized by a genetic "assimilation" process.<br />

Recent studies on Hsp90, a protein involved in several cellular processes and<br />

development pathways, seem to indicate possible molecular mechanism for canalization and<br />

genetic assimilation. In both flies and plants, mutations in the Hsp90-encoding gene induce a<br />

wide range of phenotypic abnormalities which have been interpreted as an increased<br />

sensitivity of different developmental pathways to hidden genetic variability (Rutherford and<br />

Lindquist, 1998; Queitsch, 2002). From these data, it has been proposed that Hsp90 is a<br />

capacitor of morphological evolution and buffers a pre-existing genetic variation that is not<br />

expressed and accumulates in neutral conditions. Thus, Hsp90 chaperone machinery may<br />

be an evolutionarily conserved buffering mechanism of phenotypic variance, which provides<br />

the genetic material for natural selection.<br />

Our recent study (Specchia et al., 2010) suggests an additional, perhaps alternative,<br />

explanation for proposals of a concrete mechanism underlying canalization. We show that, in<br />

Drosophila, functional alterations of Hsp90 affect the Piwi interacting RNA (piRNA; a class of<br />

germ-line-specific small RNAs) silencing mechanism leading to transposon activation and the<br />

induction of morphological mutants. This indicates that Hsp90 mutations can generate new<br />

variation by transposon-mediated ‘canonical’ mutagenesis.<br />

We propose that, in general, stress causes the activation of transposons that induce de<br />

novo gene mutations affecting development pathways. The transposon induced mutations<br />

can be expressed and fixed across subsequent generations by a process consisting in a coselection<br />

of two independent somatic and germinal events giving the same phenotype.<br />

The research activity during <strong>2011</strong> has been mainly focused on the effect of different type<br />

of stress on transposons mobilization and a screening to identify genes that are involved in<br />

heterochromatin formation and/or piRNA biogenesis in Drosophila melanogaster.<br />

1. to study the effect of stress on transposon activity, we applied discrete of continuous<br />

heat shocks to larvae and adults and analyzed, in their germ and somatic cells, the<br />

transcription and mobilization of different classes of transposons also assessing the<br />

presence of their corresponding piRNAs or endo-siRNAs. The results sofar obtained, strongly<br />

79


Area 3: Molecular genetics of eukaryotes<br />

suggest that stress activates transposons trancription and mobilization. To obtain a global<br />

view of transposons genomic redistribution, we are performing an analysis by New<br />

Generation Sequencing methodology. We are also performing a global analysis of piRNAs by<br />

cloning and deep sequencing.<br />

2. we have identified several mutations that affect piRNAs biogenesis. Their analysis is<br />

still ongoing.<br />

3. we are performing a proteomic analysis of Hsp90 complexes before and after Heat<br />

Shock. Preliminary results have indicated that Hsp90 changes some cofactors in the two<br />

different conditions.<br />

Part of these data are discussed in a paper in preparation.<br />

Research Group<br />

Laura Fanti, professor; Lucia Piacentini,<br />

researcher; Enzo Marchetti, research fellow;<br />

Marcella Marchetti, post-doc fellow.<br />

Collaborations<br />

Maria Pia Bozzetti, Università di Lecce; Maria<br />

Berloco; Università di Bari.<br />

80


Area 3: Molecular genetics of eukaryotes<br />

To the root of organ growth: the control of root meristem activity in<br />

Arabidopsis<br />

Sabrina Sabatini<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 4991 7916 - @: sabrina.sabatini@uniroma1.it<br />

A crucial aspect of development, in animals as well as plants, is how the balance<br />

between cell division and cell differentiation is maintained to ensure coherent growth of the<br />

organs and determine their correct shape and size.<br />

In plant roots, meristems ensure growth by constantly supplying fresh cells thanks to the<br />

division of stem cells; as in other developmental systems, including animal (e.g. the<br />

mammalian bone growth plate, and gut crypt), stem cell division and terminal differentiation<br />

take place in distinct and physically separated tissutal districts.<br />

We had previously shown that in the root meristem the balance between cell division and<br />

cell differentiation is maintained by the interaction between the plant hormone auxin,<br />

promoting cell division and the plant hormone cytokinin, promoting cell differentiation.<br />

In particular, we have shown that cytokinin perception at the root meristem transition<br />

zone, mediated by the histidine kinase AHK3, activates two type-B primary ARABIDOPSIS<br />

RESPONSE REGULATORS, ARR1 and ARR12 which act as transcription factors and<br />

mediate the cytokinin-dependent cell diffentiation input. The transcription factor ARR1<br />

activates the gene SHY2, a repressor of auxin signalling that negatively regulates the PIN<br />

genes that encode auxin transport facilitators. Thus, cytokinin controls the distribution of<br />

auxin, prompting cell differentiation. Conversely, auxin mediates degradation of the SHY2<br />

protein, sustaining the activity of the PIN genes and prompting cell division.<br />

Our work has therefore shown that the size of the Arabidopsis root meristem is<br />

established by a balance between the antagonistic effects of cytokinin and auxin and has<br />

provided a genetic and molecular framework - the ARR1/SHY2/PIN circuit - for this<br />

interaction.<br />

Nevertheless, major unanswered questions remained: as cells divide in the stem cell<br />

niche while cell differentiation takes place in the transition zone at the opposite end of the<br />

meristem, how the activities of these two regions of the meristem are coordinated with each<br />

other at a distance And which of the regulatory components that ensure stem cell niche<br />

maintenance mediate the coordination with the circuit active in the transition zone at the<br />

opposite end of the root meristem<br />

By means of the construction and analysis of relevant single and multiple mutants, the<br />

use of diverse reporter gene fusions, pharmacological experiments, chromatinimmunoprecipitation<br />

and quantitative RT-PCR, we provide mechanistic evidence - at the<br />

genetic and molecular level - of how the spatial coordination between cell division in the<br />

stem cell niche and cell differentiation at the meristem transition zone is achieved.<br />

81


Area 3: Molecular genetics of eukaryotes<br />

We have shown that the SCR transcription factor maintains the activity of stem cell niche<br />

by suppressing cytokinin signaling in the QC, and that it presides over the spatial<br />

coordination of stem cell division with cell differentiation in transition zone at the opposite end<br />

of the root meristem. SCR achieves this coordination by positioning the cytokinin response<br />

regulator ARR1. SCR binds directly to the promoter of ARR1 in the QC, repressing its<br />

expression and allowing stem cell division. In the transition zone, SCR controls expression of<br />

ARR1 via the DELLA protein RGA, thus controlling the rate of cell differentiation. This<br />

modulation of the cytokinin input by SCR allows a fine tuning of auxin concentration along the<br />

meristem, acting both on its synthesis via auxin biosynthesis gene ASB1, and on signalling<br />

and transport via SHY2, thus ensuring coherent root growth.<br />

Research Group<br />

Laila Moubayidin, Serena Perilli, post-doc<br />

fellows; Riccardo Di Mambro, Phd student.<br />

Collaborations<br />

Renze Heidstra, Faculty of Science, Dept of<br />

Biology, section Molecular Genetics, Utrecht<br />

University, The Netherlands.<br />

82


Area 3: Molecular genetics of eukaryotes<br />

Biological characterization and in vitro culture of spermatogonial<br />

stem cells<br />

Mario Stefanini<br />

Department of Anatomical, Histological, Forensic & Orthopaedic Sciences<br />

℡: +39 06 49766570 - @: mario.stefanini@uniroma1.it<br />

The adult human testis produces millions of sperm each day trough spermatogenesis.<br />

This highly efficient process, finely tuned by hormone and paracrine balance, relies on the<br />

biological activity of spermatogonial stem cells (SSC), the stem cells of germline. Because of<br />

the ability to transmit genes from one generation to the next, SSCs represent the only<br />

replicating, potentially totipotent, population of cells in the adult body.<br />

Surface antigenic profile of SSCs, for their prospective isolation, has been partially<br />

defined. Moreover, data from our and other laboratories support the idea that SSCs are<br />

heterogeneous in term of gene expression profile. SSC subsets may be readout of niche<br />

signals or they could represent hierarchically distinct stem/progenitor cell populations.<br />

Interestingly, we found that spermatogonial stem cell displaying SP phenotype may be<br />

generated very early during embryonic testis development (Scaldaferri et al., <strong>2011</strong>). The<br />

anatomical location and molecular components of the SSC niche are largely unknown. The<br />

molecular dissection of germline niche, will certainly shed light on SSCs maintenance and<br />

differentiation.<br />

To gain insight into lineage relationship among subsets of SSC we have developed an<br />

animal model expressing the receptor GFRA1 tagged with the enhanced green fluorescent<br />

protein (EGFP) to isolate and enrich SSCs from mice at different stage of testis development.<br />

We confirmed that expression of tagged protein is restricted to early spermatogonia. We<br />

are at the present testing the stem cell activity of sorted cell populations that have been<br />

selected based on EGFP-expression level by means of germ cell transplantation. Moreover,<br />

by video-time laps analysis we are testing the migratory activity of GFP-expressing early<br />

spermatogonia.<br />

We have also analyzed the role of the niche-derived factor GDNF upon the gene<br />

expression of SSCs. At the present GDNF is the best characterized niche-derived factor,<br />

produced by Sertoli cells the nurse cells of male germ cells. A large body of evidences show<br />

that GDNF is essential for the maintenance of SSCs both in vivo and in vitro. By in vitro<br />

short-term culture of intact seminiferous tubules and qRT-PCR we have recently found that<br />

GDNF regulates the expression of genes known to be involved in spermatogonial<br />

differentiation such as Tex14, Sohlh1 and Kit. Our data indicate that GDNF, besides its<br />

crucial role in the self-renewal of stem cells also functions in the differentiation of chained<br />

undifferentiated spermatogonia (Grasso et al., <strong>2011</strong>).<br />

Finally we were able to establish human and mouse SSC cultures. Development of SSC<br />

culture is highly relevant to the study of molecular mechanisms regulating early step of<br />

83


Area 3: Molecular genetics of eukaryotes<br />

spermatogensis, for future applications on male infertility, and to unveil molecular<br />

mechanisms underlying pluripotency.<br />

Publications<br />

Grasso M, Fuso A, Dovere L, de Rooij D, Stefanini M, Boitani C, Vicini E. Distribution of<br />

GFRA1-expressing spermatogonia in adult mouse testis. Reproduction <strong>2011</strong> Epub. doi:<br />

10.1530/REP-11-0385.<br />

Scaldaferri ML, Fera S, Grisanti L, Sanchez M, Stefanini Mario, De Felici M, Vicini E.<br />

Identification of side population cells in mouse primordial germ cells and prenatal testis. Int<br />

J Dev Biol <strong>2011</strong>, 55: 209-14. doi: 10.1387/ijdb.092977ms.<br />

Research Group<br />

Carla Boitani, professor; Elena Vicini,<br />

researcher; Lisa Dovere, Barbara Muciaccia,<br />

post-doc fellows; Dante Lamberti, PhD student;<br />

Stefania Fera, Tiziana Menna, technicians.<br />

Collaborations<br />

Dirk de Rooij, Department of Endocrinology,<br />

Utrecht University, The Netherlands; Ans Van<br />

Pelt, Center for Reproductive Medicine,<br />

Academic Medical Center, University of<br />

Amsterdam, The Netherlands.<br />

84


Area 3: Molecular genetics of eukaryotes<br />

Molecular mechanisms driving liver stem cell fate<br />

Marco Tripodi<br />

Department of Cellular Biotechnologies and Hematology<br />

℡: +39 06 4461387 - @: tripodi@bce.uniroma1.it<br />

Our work focuses on liver stem cell plasticity and differentiation mechanisms. To these<br />

aims we dispose of a number of stem cell lines named RLSCs (from Resident Liver Stem<br />

Cells) we previously stabilized and characterized. These cells spontaneously differentiate<br />

into periportal hepatocytes that, in turn, are able to switch into perivenular hepatocytes. This<br />

post-differentiative switch is triggered by Wnt signaling and converges on the HNF4a-driven<br />

transcription (Conigliaro et al., CDD 2008; Colletti et al., Gastroenterology 2009). RLSCs<br />

show a metastable molecular phenotype typical of embryonic and adult stem cells and<br />

characterized by co-expression of epithelial and mesenchymal markers as well as of a<br />

variety of chromatin remodeling genes. This highly dynamic cell state, thought to be the<br />

major determinant of stem cell fate, may be considered as the result of continue oscillations<br />

between EMT/METs.<br />

Result 1. In frame of the study of molecular mechanisms controlling EMT/MET in hepatic<br />

cells, we published a work describing the pivotal role of HNF4α in the induction and<br />

maintenance of hepatocyte identity. HNF4α was over-expressed in different hepatocyte cell<br />

lines and the resulting gene expression profile determined by qRT-PCR. HNF4α recruitment<br />

on promoters of both mesenchymal and EMT regulator genes was determined by EMSA and<br />

ChIP assays. The effect of HNF4α depletion was assessed in silenced cells and in the<br />

context of the whole liver of HNF4-KO animals. By these approaches, we identified key EMT<br />

regulators and mesenchymal genes as new targets of HNF4α. In particular HNF4α directly<br />

inhibits transcription of the EMT master gene Snail (Santangelo et al., <strong>2011</strong>). Our findings<br />

indicate that HNF4α transcriptional activation and repression of critical EMT/MET genes is<br />

pivotal for the maintenance of a stable epithelial phenotype as well as for the regulation of<br />

the dynamic process of MET. Previously, we demonstrated as Snail causes repression of<br />

Hnf4α transcription through direct promoter binding (Cicchini et al., 2006). Thus, we propose<br />

a simple cross-regulatory circuit between Snail and HNF4α in which each factor’s expression<br />

is mutually exclusive to the other due to the presence of repressor elements in each<br />

promoter. HNF4α /Snail reciprocal control may provide the molecular rationale for feed-back<br />

and reversible differentiative processes and explain, at least in part, the coherence and the<br />

reversibility of the molecular events underlying EMT/MET.<br />

Result 2. In Garibaldi et al., we described a molecular circuitry controlling RLSCs<br />

plasticity. Starting from the finding that Snail is expressed in RLSCs, we demonstrated its<br />

85


Area 3: Molecular genetics of eukaryotes<br />

positive role in stemness markers expression. This observation, unexpected considering that<br />

the transcriptional repression is the only function so far attributed to Snail, prompted us to<br />

investigate on other players integrating/mediating Snail activity. Mirror observations made in<br />

RLSCs and their derived differentiated hepatocytes RLSCdH allowed us to conclude that 1)<br />

in RLSC Snail inhibits the hepato-specific program through direct repression of HNF4 gene<br />

and of the epithelial microRNAs-200c and -34a, 2) in RLSCdH HNF4, together with a direct<br />

repression of Snail gene, directly up-regulates microRNA-200 family members (200a, b, c)<br />

and microRNA-34a transcription, thus further stabilizing the hepatocytic phenotype.<br />

Altogether these data unveiled Snail, HNF4 and microRNAs-200a, b, c and -34a as epistatic<br />

elements controlling hepatic stem cell maintenance/differentiation. (Garibaldi et al., Cell<br />

Death and Differentiation <strong>2011</strong>).<br />

Publications<br />

Garibaldi F, Cicchini C, Conigliaro A, Santangelo L, Cozzolino AM, Grassi G, Marchetti A,<br />

Tripodi M, Amicone L. An epistatic mini-circuitry between the transcription factors Snail and<br />

HNF4α controls liver stem cell and hepatocyte features exhorting opposite regulation on<br />

stemness-inhibiting microRNAs. Cell Death Differ <strong>2011</strong> Epub. doi: 10.1038/cdd.<strong>2011</strong>.175.<br />

Mancone C, Steindler C, Santangelo L, Simonte G, Vlassi C, Longo MA, D'Offizi G, Di<br />

Giacomo C, Pucillo LP, Amicone L, Tripodi M, Alonzi T. Hepatitis C virus production<br />

requires apolipoprotein A-I and affects its association with nascent low-density lipoproteins.<br />

Gut <strong>2011</strong>, 60: 378-86. doi:10.1136/gut.2010.211292.<br />

Santangelo L, Marchetti A, Cicchini C, Conigliaro A, Conti B, Mancone C, Bonzo JA,<br />

Gonzalez FJ, Alonzi T, Amicone L, Tripodi M. The stable repression of mesenchymal<br />

program is required for hepatocyte identity: a novel role for hepatocyte nuclear factor 4α.<br />

Hepatology <strong>2011</strong>, 53: 2063-74. doi: 10.1002/hep.24280.<br />

Research Group<br />

Laura Amicone, Carla Cicchini, Alessandra<br />

Marchetti, researchers, Carmine Mancone,<br />

Alice Conigliaro, post-doc fellows; Laura<br />

Santangelo, Angela Cozzolino, AIRC fellows;<br />

Valeria De Nonno, Marco De Santis Puzzonia,<br />

Viviana Costa, PhD students; Claudio<br />

Cavallari, technician.<br />

86


Area 3: Molecular genetics of eukaryotes<br />

Profiling the Polycomb/Trythorax target genes in normal and<br />

leukemic hematopoiesis<br />

Giuseppe Zardo<br />

Department of Cellular Biotechnologies and Hematology<br />

℡: +39 06 4469843 - @: zardo@bce.uniroma1.it<br />

Research aims<br />

Several studies describe the functional effects of the modifications of PcG/TrxG activities<br />

on hematopoietic stem/progenitor cell self renewal and proliferation. However, insufficient<br />

information is available about the genes, undergoing PcG/TrxG regulation in normal<br />

hematopoiesis, and deregulated during terminal differentiation, thus permitting the activation<br />

of a leukemic program. Our research aims to identify these genes and to investigate the<br />

existence of functional connections between different epigenetic mechanisms of gene<br />

regulation during normal and leukemic hematopoiesis.<br />

Results<br />

Recent findings have identified NFI-A gene as a regulator of hematopoiesis. NFI-A is a<br />

post-transcriptional miR-223 target directing human hematopoietic progenitor lineage<br />

decision. Our research demonstrates that NFI-A down-regulation, which allows normal<br />

granulopoiesis, is guaranteed also by other epigenetic events affecting NFI-A gene promoter<br />

activity and chromatin structure, including the resolution of chromatin “bivalent domains” by a<br />

modification of PcG/TrxG activities; DNA hypermethylation, recruitment of PcG-RNAi<br />

complex and by miR-223 promoter targeting activity. We found that miR-223 settles on these<br />

epigenetic events. Confocal microscopy and ChIP analyses revealed that in myeloid cells,<br />

induced to granulopoiesis by retinoic acid treatment, miR-223 localizes in the nucleus, colocalizes<br />

with PcG and RNAi proteins, and targets the NFI-A promoter region containing<br />

PcGs binding sites and miR-223 complementary DNA sequences. The knockdown of PcG<br />

and RNAi proteins, such as Suz12, Dicer1 and Ago1, and promoter activity assays, showed<br />

that both the integrity of the PcG-RNAi complex and DNA sequences, matching the seed<br />

region of miR-223, are required to induce NFI-A transcriptional silencing. PcG and RNAi<br />

Suz12, Dicer1 and Ago1 knockdown impaired the NFI-A down-regulation and granulocytic<br />

differentiation potential of HL60 cells treated with retinoic acid, as shown by NFI-A mRNA<br />

levels, myeloid cell marker CD11b levels and TGaseII expression. Ectopic miR-223<br />

expression caused heterochromatic repression of NFI-A gene and channeled granulopoiesis,<br />

while its stable knockdown produced opposite effects. Our findings indicate that endogenous<br />

miRs can affect gene expression also at the transcriptional level, functioning in a critical<br />

interface between chromatin remodelling complexes and the genome to direct fate lineage<br />

determination of hematopoietic progenitors. These results also identify PcG/TrxG functions<br />

as regulator of hematopoiesis, likely deregulated during leukemic transformation. Polycomb<br />

87


Area 3: Molecular genetics of eukaryotes<br />

protein Ezh2, which in association with Suz12 is responsible for establishment of repressive<br />

chromatin mark H3K27me3, directs DNA methylation by the recruitment of DNA<br />

methyltransferases on target genes. The presence of H3K27me3 chromatin mark at gene<br />

promoter regions in human embryonic stem cells (HESC) makes these genes sensitive to<br />

aberrant DNA methylation during tumour transformation. By RLGS we acquired the DNA<br />

methylation profiles of several human leukemic cell lines, AML patient samples and of<br />

CD34+HSC/HPCs and mature CD34- cells as control samples. We selected genes<br />

hypermethylated and derived, from literature data, the presence at their promoter region of<br />

H3K27me3 and/or H3K4me3 chromatin marks in HESC. ChIP assays showed that although<br />

the majority of these hypermethylated genes were H3K27me3 labelled in HESC,<br />

CD34+HSC/HPCs, and CD34- cells, a relevant number presented the H3K4me3 activating<br />

mark. In leukemic samples, where these genes appeared consistently hypermethylated, the<br />

H3K4me3 mark was replaced by H3K27me3 mark, thus proving that leukemic transformation<br />

is also associated with an anomalous PcG/TrxG activity.<br />

Perspectives<br />

The results of this research enable a more comprehensive knowledge of epigenetic<br />

mechanisms regulating the activity of hematopoietic master regulators and unveil new targets<br />

for leukaemia therapy.<br />

Research Group<br />

Alberto Ciolfi, post-doc fellow; Carmen<br />

Maresca, technician.<br />

Collaborations<br />

Clara Nervi, Dip. di Scienze e Biotecnologie<br />

Medico-Chirurgiche, Sapienza Università di<br />

Roma; Robin Foa’, Sabina Chiaretti, Dip. di<br />

Biotecnologie Cellulari ed Ematologia,<br />

Sapienza Università di Roma; Joseph F.<br />

Costello, Comprehensive Cancer Center,<br />

University of California, San Francisco, USA.<br />

88


Area 4<br />

Molecular recognition<br />

in biomolecules


Area 4: Molecular recognition in biomolecules<br />

Toxic effects induced by polyamine metabolites on melanoma cells:<br />

a new therapeutic approach<br />

Enzo Agostinelli<br />

Department of Biochemical Sciences "A. Rossi Fanelli"<br />

℡: +39 06 49910838 - @: enzo.agostinelli@uniroma1.it<br />

The purpose of this research is to develop a new anticancer therapy against melanoma<br />

(M14), taking advantage of the high polyamine content of tumor cells, since such polycations<br />

may become a source of cytotoxic metabolites. For their generation an enzyme, bovine<br />

serum amine oxidase (BSAO) will be employed. It is expected that by delivering BSAO into<br />

cancer cells, toxic enzymatic oxidation products could be produced intracellularly for<br />

selective killing in situ.<br />

Several studies have been performed to overcome the drug-resistant phenotype and to<br />

develop innovative chemotherapeutic strategies effective against multi-drug resistant (MDR)<br />

tumours. Our studies demonstrate that BSAO and spermine (SPM) addition to human M14<br />

cells induces cell growth inhibition and overcomes the MDR phenotype (ADR2). The<br />

oxidative reaction produces polyamine metabolites (H 2 O 2 and aldehydes) which induce<br />

oxidative stress. Cytotoxicity induced by SPM metabolites was greater in MDR cells than in<br />

wild-type ones (WT), due to an increased mitochondrial activity. To increase the induction of<br />

cell death by toxic polyamine metabolites, we are currently investigating several drug<br />

combinations. Clonogenic experiments showed that the cytotoxicity induced by BSAO/SPM<br />

addition was enhanced by the pre-treatment of tumour cells with lysosomotropic compounds,<br />

the drug chloroquine (CQ) or MDL 72527, in both WT and MDR M14 cells.<br />

The pre-treatment of M14 cells with CQ or MDL 72527 sensitized both WT and ADR2<br />

cells to the subsequent exposure to SPM metabolites. Noteworthy, the apoptotic effect was<br />

higher in ADR2 cells than in their WT counterparts. The cytotoxic effect induced by the<br />

combined treatment was characterized by cytoplasmic vacuolization and mitochondrial<br />

damage, as revealed by transmission electron microscopy (TEM) (Fig. 1). We hypothesize<br />

that the release of lysosomal enzymes produces oxidative stress and apoptosis, indicating a<br />

contribution of the lysosomotropic properties of CQ to the sensitization of M14 cells to H 2 O 2<br />

and aldehyde(s). The findings suggest that the association of BSAO/SPM with CQ or MDL<br />

72527, and mitochondrial alterations induced by SPM oxidation products, allow the design of<br />

a new therapeutic strategy based on the use of these combinations in human neoplasms,<br />

making this approach mainly attractive in treating MDR cancer patients.<br />

From a therapeutic point of view the improvement of the efficacy of in situ formation of<br />

cytotoxic polyamines metabolites is essential. Since in the present model H 2 O 2 and<br />

aldehydes are produced outside the cells, the main challenge will be to deliver BSAO into<br />

cancer cells in order to induce their selective killing by the cytotoxic factors produced<br />

intracellularly from endogenous polyamines. Therefore, strategies could be developed to find<br />

91


Area 4: Molecular recognition in biomolecules<br />

how the enzyme could be delivered in vivo. To this aim BSAO incorporates into liposomal<br />

vesicles and dedicated nano-scale drug delivery systems will be designed. Thus,<br />

endogenous polyamines present in high concentration in tumor cells could be targeted and<br />

oxidized by the enzyme.<br />

Fig. 1- Ultrastructural analysis of M14 WT ( top<br />

panels) and M14 ADR2 (lower panels) cells<br />

performed by TEM. (a and b) Untreated melanoma<br />

cells; (c and d) Cells exposed to 300 µM MDL 72527<br />

for 24 h, at 37˚C; (e and f) Cells treated for 60 min<br />

with 6.5x10-3 IU/ml BSAO and6 µM SPM at 37˚C; (g<br />

and h) Cells were first exposed for 24 h to 300 µM<br />

MDL 72527, and then treated for 60 min with<br />

6.5x10-3 IU/ml BSAO and 6 µM SPM at 37˚C.<br />

Publications<br />

Agostinelli E. Role of polyamines, their analogs and transglutaminases in biological and<br />

clinical perspectives. Amino Acids <strong>2011</strong> Epub. doi: 10.1007/s00726-011-1129-2<br />

Agostinelli E, Toninello A, Vianello F, Stevanato R. Do mammalian amine oxidases and the<br />

mitochondrial polyamine transporter have similar protein structures Amino Acids <strong>2011</strong><br />

Epub. doi: 10.1007/s00726-011-0988-x.<br />

Stevanato R, Cardillo S, Braga M, De Iuliis A, Battaglia V, Toninello A, Agostinelli E, Vianello<br />

F. Preliminary kinetic characterization of a copper amine oxidase from rat liver<br />

mitochondria matrix. Amino Acids <strong>2011</strong>, 40: 713–20. doi: 10.1007/s00726-010-0708-y.<br />

Valente S, Tomassi S, Tempera G, Saccoccio S, Agostinelli E, Mai A. Novel Reversible<br />

Monoamine Oxidase A Inhibitors: Highly Potent and Selective 3-(1H-Pyrrol-3-yl)-2-<br />

oxazolidinones. J Med Chem <strong>2011</strong>, 54: 8228-32. Epub <strong>2011</strong> Nov 7. doi: 10.1021/<br />

jm201011x.<br />

Research Group<br />

Paola Turini, professor; Giampiero Tempera,<br />

Nikenza Viceconte, Stefania Saccoccio, postdoc<br />

fellows.<br />

Collaborations<br />

Annarica Calcabrini, <strong>Istituto</strong> Superiore di<br />

Sanità, Roma; Taichi Ueshima, Wakunaga<br />

Pharmaceutical Co., Hiroshima, Japan.<br />

92


Area 4: Molecular recognition in biomolecules<br />

Cellular response to oxidative stress: involvement of protein<br />

disulfide isomerases<br />

Fabio Altieri<br />

Department of Biochemical Sciences "A. Rossi Fanelli"<br />

℡: +39 06 49910880 - @: fabio.altieri@uniroma1.it<br />

The aim of this research project is to elucidate the involvement of ERp57/PDIA3 in the<br />

cellular response to oxidative stress. Different thyroid carcinoma cell lines were subjected to<br />

oxidative stress and analyzed during the following 24 hours. We compared cell vitality, the<br />

intracellular levels of reduced and oxidized glutathione and the cellular distribution of ERp57<br />

and Ref1, STAT3 and Ku70/86, previously identified ERp57 interactors. ARO cells, which<br />

better survived to oxidative stress, showed the highest levels of ERp57 but the lowest<br />

glutathione content. A nuclear translocation of ERp57, Ref1 and Ku70/86 as a consequence<br />

of the stress treatment has been also observed, suggesting the involvement of ERp57,<br />

beside its known functions within ER, in regulatory processes at the nuclear level.<br />

We showed that in M14 cells, where STAT3 is constitutively activated, ERp57 binds with<br />

STAT3 to a subset of well-characterized STAT3-associated promoters/enhancers. The<br />

presence of ERp57 in these sites appears to be essential for the regulatory activity of STAT3<br />

on transcription, since inhibition of ERp57 expression by RNA interference is accompanied<br />

by the decrease of expression of several STAT3 dependent genes. Moreover, the addition of<br />

vancomycin and phenyl-arsenoxide, two inhibitors of ERp57 redox activity, inhibited the in<br />

vitro binding of STAT3 to DNA.<br />

ERp57-calreticulin interaction has been studied by surface plasmon resonance (SPR).<br />

ERp57 was covalently bound to the sensor surface and its binding with calreticulin (CRT)<br />

was analyzed. The estimated K D was 2.6 µM, in agreement with the observation of other<br />

authors. However, the kinetic analysis of obtained binding curves better fits with a two-step<br />

binding model where the initial protein complex undergoes to conformational changes that<br />

increase its stability. This result is in agreement with a kind of plasticity in the structure of<br />

ERp57 that allows, through specific conformational changes, to handle the interaction with so<br />

many different protein partners. SPR analysis showed that vancomycin can inhibit ERp57-<br />

CRT interaction and the K D obtained for vancomycin binding to ERp57 was similar to that<br />

observed for CRT. Vancomycin has also effect on CRT-ERp57 interaction in vivo. It has been<br />

demonstrated that cell membrane localization of calreticulin is dependent on the interaction<br />

with ERp57. Vancomycin administration to HeLa cells prevents the ERp57-mediated<br />

translocation of calreticulin to the plasma membrane. This result confirms a role for ERp57 in<br />

the trafficking of proteins from endoplasmic reticulum to plasma membrane.<br />

A joint project was started to analyze the involvement of ERp57 in EGFR activation and<br />

internalization. Breast cancer cells, where ERp57 was silenced by RNA interfering, were<br />

subjected to EGF stimulation. Preliminary studies suggest that ERp57 can interact with<br />

93


Area 4: Molecular recognition in biomolecules<br />

EGFR and that in EGF stimulated cells the silencing of ERp57 affects internalization of the<br />

receptor and its autophosphorylation, thus leading to changes in the activation of<br />

downstream pathways.<br />

The in vitro binding of ERp57 with known interactors was further analyzed. Using a thiol<br />

probe able to modify the electrophoretic mobility of reduced and oxidized forms of a protein,<br />

we confirmed the reducing activity of ERp57 on oxidized Ref1. Solid-phase binding assay<br />

and SPR experiments verified the close interaction between the two proteins with a K D in the<br />

micromolar range. Finally, we started to analyze the binding of several natural substances to<br />

ERp57 and their effects on protein activities, to find molecules able to specifically inhibit the<br />

binding between ERp57 and selected interactors and modulate its biological functions. We<br />

focused the attention on green tea catechins (GTCs) and tested their binding with ERp57 by<br />

fluorescence quenching measurements and SPR analysis. Data analysis showed that<br />

galloylated forms of GTCs better bind to ERp57 and quench the fluorescence of tryptophan<br />

residues with epigallocatechin-gallate the most active. Quenching analysis was extended to<br />

isolated redox domains of ERp57. Data analysis confirmed the binding of catechins near the<br />

ERp57 redox-active sites and this interaction requires the presence of the galloyl moiety.<br />

This structural feature is also responsible for the inhibition of both ERp57 reductase activity<br />

and DNA binding capability.<br />

Publications<br />

Frasconi M, Chichiarelli S, Gaucci E, Mazzei F, Grillo C, Chinazzi A, Altieri F. Interaction of<br />

ERp57 with calreticulin: Analysis of complex formation and effects of vancomycin. Biophys<br />

Chem <strong>2011</strong> Epub. doi: 10.1016/j.bpc.<strong>2011</strong>.09.003.<br />

Turano C, Gaucci E, Grillo C, Chichiarelli S. ERp57/GRP58: a protein with multiple functions.<br />

Cell Mol Biol Lett <strong>2011</strong>, 16: 539-63. doi: 10.2478/s11658-011-0022-z.<br />

Research Group<br />

Margherita Eufemi, professor; Caterina Grillo,<br />

post-doc fellow; Rossana Cocchiola, PhD<br />

student.<br />

Collaborations<br />

Silvia Chichiarelli, Daniela Ricci, Elisa<br />

Gaucci, Dipartimento di Scienze Biochimiche<br />

“A. Rossi Fanelli”, Sapienza Università di<br />

Roma.<br />

94


Area 4: Molecular recognition in biomolecules<br />

Role of Gsα signalling in the bone marrow hematolymphopoietic<br />

microenvironment as revealed by novel transgenic models<br />

Paolo Bianco<br />

Department of Molecular Medicine<br />

℡: +39 06 4441049 - @: paolo.bianco@uniroma1.it<br />

Background<br />

Cells of osteogenic lineage maintain, organize and transfer the hematopoietic<br />

microenvironment/niche in the bone marrow. In search for the specific cell type conveying<br />

these crucial physiological effects, attention over the past few years has been directed first to<br />

osteoblast proper (i.e., the mature bone forming compartment in the osteogenic lineage) and<br />

later to osteogenic progenitors (aka “mesenchymal stem cells”). Studies using osteoblasttargeted<br />

knockout have implicated Gsα signaling within cells of osteogenic lineage in the<br />

regulation of the HME/niche, particularly with respect to B lymphopoiesis. We have<br />

generated mice in which an activating missense mutation of Gsα (resulting in a severe<br />

skeletal disease in humans, Fibrous Dyplasia of bone) is either targeted to osteoblasts<br />

through the COL1A1 2.3kb promoter, or expressed widely in the bone environment as<br />

directed by costitutive promoters (PGK or EF1α). The latter model has been established both<br />

in an FVB and in a B6 background.<br />

Aim<br />

The general aim is to elucidate a) the changes in the organization of the HME/niche b)<br />

the changes in the hemato-lymphopoietic progenitor compartments brought about by<br />

overactivity of Gsα signaling.<br />

Results <strong>2011</strong><br />

1) Functional effects of mutated Gsa in different osteogenic cell compartments. Using a<br />

well standardized, timed system for monitoring osteogenic differentiation of BM-derived<br />

skeletal progenitor cells (“mesenchymal stem cells”) in vitro, we have established the<br />

differential effects of mutated Gsα on the progenitor and mature compartments of the<br />

osteogenic lineage. In the mature compartment, mutated Gsα potently downregulates SOST,<br />

a negative modulator of Wnt signaling, leading to an inappropriate excess Wnt lignaling (as<br />

monitored through Axin2 and LEF1). The latter data directly explain the high bone mass<br />

phenotype of COL1A1-GsaR201C mice, which indeed is made by these data a phenocopy of<br />

two human high bone mass disorders, Van Buchem disease and Sclerosteosis. When<br />

expressed in the progenitor compartment, mutated Gsα induces a complete replica of the<br />

human FIbrous Dysplasia phenotype. This is linked to specific cellular responses that are not<br />

not duplicated in the mature compartment, including a potent upregulation of RANKL<br />

95


Area 4: Molecular recognition in biomolecules<br />

expression, a marked blunting of Wnt signaling, and a prolonged delay of osteogenic<br />

differentiation (2-3 weeks). In vivo, this compartment corresponds to an intraosseous,<br />

perivascular population of stromal cells, noted for high expression of the Adrb2 (IHC in vivo;<br />

qPCR ex vivo), and high functional responses to adrb2 agonists ex vivo. This population is at<br />

the same time both perivascular and endosteal, i.e. is located at the two main sites where the<br />

HSC niche is thought to be.<br />

2) Functional effects of mutated Gsα expressed in the osteogenic lineage on hematolymphoid<br />

compartments. This analysis has so far revealed that detectable changes in<br />

hematolymphoid compartments do occur in mice as a result of Gsα overactivity in the<br />

osteogenic lineage. Of note, these changes do unfold in an age-dependent manner,<br />

matching the development of the bone/bone marrow structural changes. While we have not<br />

been able to detect so far clear-cut changes in the size of the HSC compartment proper<br />

(CD34- LSK), or of B, pre-B or pro-B compartments, we do detect changes in the pool of<br />

progenitors. More importantly, we do detect an age dependent activation of DNA synthesis<br />

and cell replication (BrdU; Flk-2 subset of LSK) in B6 mice with constitutive expression of<br />

mutated Gs in osteoprogenitors, so that pools of replicating HSCs approach in size, in old<br />

mutant mice, those seen in young wt.<br />

Next directions<br />

One interesting lead for developing these data further comes from combining three key<br />

observations: 1) the identification of an intraosseous, perivascular osteoprogenitor as the<br />

prime mediator of effects of mutated Gsα in vivo (leading to subversion of the HME/niche as<br />

seen in Fibrous Dysplasia); 2) the evidence that this progenitor compartment is<br />

physiologically primarily responsive to beta2-adrenergic stimulation, which in turn is a known<br />

regulator of circadian mobilization of HSCs; 3) the evidence that in conjunction with the<br />

development of FD lesions, HSCs are activated to divide. While the analysis of<br />

hematolymphoid compartments will continue in the next year, we will also pursue the<br />

hypothesis that HSCs are mobilized in mice with constitutively active Gsα in the<br />

osteoprogenitor compartment.<br />

Research Group<br />

Mara Riminucci, Isabella Saggio, professors;<br />

Alessandro Corsi, Benedetto Sacchetti,<br />

researchers; Stefania Cersosimo, Rossella<br />

Costa, Cristina Remoli, post-doc fellows;<br />

Paola Comite, Letizia Astrologo, PhD<br />

students; Emanuela Spica, technician.<br />

Collaborations<br />

Ana Cumano, <strong>Pasteur</strong> Institut, Paris<br />

96


Area 4: Molecular recognition in biomolecules<br />

The dark side of protein folding: denatured states and misfolded<br />

species in molecular recognition and pathological processes<br />

Maurizio Brunori<br />

Department of Biochemical Sciences "A. Rossi Fanelli"<br />

℡: +39 06 4991 0579 - 06 4940543 - @: maurizio.brunori@uniroma1.it<br />

The variety of chemical reactions taking place in the cell demands recognition events to<br />

be highly specific. Since specificity is achieved through complementarity between interacting<br />

species, folding is crucial to protein function. We have studied the folding and binding of<br />

selected protein systems, that are key components of important cellular processes. Particular<br />

attention has been devoted to elusive states, such as the denatured state, and to intradomain<br />

allostery, a sophisticated mechanism to modulate specificity. The two model systems<br />

are: (i) the PDZ domains, small globular domains involved in a variety of cellular processes<br />

(Model System I), and (ii) a pair of designed proteins with an extraordinarily high degree of<br />

sequence identity but different 3D structure and function (Model System II).<br />

Results<br />

Model System I. Protein-protein interactions mediated by modular protein domains are<br />

critical for cell’s life. However, given the vast number of ligands competing for binding to a<br />

limited number of domain families, it is unclear how specificity can be achieved. Such<br />

specificity may be modulated by intradomain allostery, whereby remote residues are<br />

energetically connected to the functional binding site. Whereas energetic pathways in<br />

modular domains have been predicted, there is a paucity of experimental data to validate<br />

their existence and roles. We have identified these functional energetic networks in one of<br />

the most common protein-protein interaction modules, the PDZ domain. We exploited the<br />

use of double mutant cycles, in conjunction with kinetics to capture the fine energetic details<br />

of the networks involved in peptide recognition. The analysis revealed that the allosteric<br />

pathways are dictated by the amino acid sequence rather than topology. Model System II.<br />

We have addressed the folding mechanism of two heteromorphic proteins G A 88 and G B 88,<br />

with 88% sequence identity but different structure (Fig. 1). This system offered the unique<br />

opportunity to unveil the mechanism whereby a few key residues commit the polypeptide<br />

chain to its characteristic and functionally competent native topology.<br />

An extensive characterization of the folding mechanisms of G A 88 and G B 88 by<br />

experiments and molecular dynamics (MD) simulations allowed to identify the presence of a<br />

residual structure in the denatured state of G B 88, not observed for G A 88. A signature of such<br />

a difference is the different denaturant dependence of the (un)folding rate constants.<br />

Consistently, MD simulations show that for G B 88 the non-polar solvent-accessible surface<br />

area decreases at low pH. This unexpected result suggests that the denatured state has a<br />

primary role in the folding process. Indeed, protein topology appears to be committed very<br />

97


Area 4: Molecular recognition in biomolecules<br />

early along the folding pathway, being “imprinted” in the residual structure of the denatured<br />

state; this weak, loosely defined topology is sufficient to dictate the pathway of folding.<br />

Fig. 1 - G A 88 and G B 88 structures. Sequence<br />

alignment and secondary structure are shown.<br />

The 7 residues that differ between the two<br />

proteins are shown in different colors. It may be<br />

seen that the only a-helix in G B 88 is longer<br />

compared to the topologically homologous α2 of<br />

G A 88; this is the physico-chemical feature<br />

responsible for the residual interaction in the<br />

denatured state of G B 88 which dictates the<br />

stabilization of the β-sheet fold.<br />

We have undertaken an extensive characterization of the folding mechanism of the<br />

parental protein displaying the structure of G B 88, namely GB1. We demonstrated the<br />

presence of an on-pathway folding intermediate that previously escaped detection and<br />

characterization. Elucidation of the folding mechanism of GB1 is critical for future work on the<br />

different variants with increased sequence identity with respect to the G A proteins.<br />

Publications<br />

Gianni S, Haq SR, Montemiglio LC, Jürgens MC, Engström Å, Chi CN, Brunori M, Jemth P.<br />

Sequence-specific long range networks in PSD-95/discs large/ZO-1 (PDZ) domains tune<br />

their binding selectivity. J Biol Chem <strong>2011</strong>, 286: 27167-75. doi: 10.1074/jbc.M111.239541.<br />

Morrone A, McCully ME, Bryan PN, Brunori M, Daggett V, Gianni S, Travaglini-Allocatelli C.<br />

The denatured state dictates the topology of two proteins with almost identical sequence<br />

but different native structure and function. J Biol Chem <strong>2011</strong>, 286; 3863-72. doi:<br />

10.1074/jbc.M110.155911.<br />

Morrone A, Giri R, Toofanny RD, Travaglini-Allocatelli C, Brunori M, Daggett V, Gianni S.<br />

GB1 is not a two-state folder: identification and characterization of an on-pathway<br />

intermediate”. Biophys. J <strong>2011</strong>, 101; 2053-60. doi: 10.1016/j.bpj.<strong>2011</strong>.09.013.<br />

Research Group<br />

Luca Federici, Carlo Travaglini Allocatelli,<br />

professors; Adele Di Matteo, Stefano Gianni,<br />

CNR researchers; Rajanish Giri, Angela<br />

Morrone, PhD students.<br />

Collaborations<br />

Per Jemth, University of Uppsala, Sweden;<br />

Valerie Daggett, University of Washington,<br />

Seattle, Washington, USA.<br />

98


Area 4: Molecular recognition in biomolecules<br />

The role of nucleosomes in the stability of human telomeres<br />

Stefano Cacchione<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912240 - @: stefano.cacchione@uniroma1.it<br />

Telomeres are the special nucleoprotein structures that protect chromosome ends. In<br />

humans telomeric DNA consists of TTAGGG repeats about 10 kbp long, ending in a 3’ G-rich<br />

overhang of 150-200 nucleotides. Most of telomeric DNA is organized in tightly packed<br />

nucleosomes. Two proteins, TRF1 and TRF2, compete with the histone octamer for binding<br />

to double-stranded telomeric DNA and organize a six-protein complex named shelterin,<br />

essential for telomere stability. Both telomere length maintenance and the establishment of a<br />

correct capping structure are essential for cell life and survival. Whereas the roles played by<br />

telomeric proteins in telomere protection have been widely investigated, a satisfactory<br />

description of nucleosome contribution to telomere structure and function is still lacking.<br />

Little is known also on the interplay between telomeric proteins and nucleosomes. It is not<br />

known if nucleosomes and TRF proteins occupy different domains on the telomere or<br />

whether they cooperate in telomere capping; moreover, it is still unclear if telomere<br />

nucleosomal organization is affected by abnormal expression of TRF proteins.<br />

Our goal is to characterize the nucleosomal organization of human telomeres and to<br />

elucidate the role played by nucleosomes in telomere capping structure. We addressed these<br />

issues by means of theoretical analyses, in vitro model systems and analyses on cultured<br />

cells.<br />

Theoretical analysis of the higher-order organization of telomeric chromatin<br />

Telomeric chromatin is characterized by a nucleosome repeat length about 40 bp shorter<br />

than bulk nuclear chromatin. Due to their peculiar DNA sequence features, telomeric<br />

nucleosomes occupy isoenergetic positions having the periodicity of the telomere repeat.<br />

The systematic search of the possible nucleosome packing revealed that for nucleosome<br />

repeat length of 156, 162 and 168 bp (multiples of the vertebrate telomeric repeat) different<br />

and unique structures are allowed (De Santis and Scipioni, in preparation).<br />

Nucleosome density at telomeres is regulated by TRF2<br />

To analyze whether TRF2 affects nucleosomal organization at telomeres, we altered<br />

TRF2 expression in human cells by transient transfection. We found that the density of<br />

telomeric nucleosomes was inversely proportional to the dosage of TRF2 at telomeres. This<br />

effect coincides with the end of S phase of the cell cycle. Moreover, we showed that<br />

nucleosome spacing at telomeres increased upon TRF2 overexpression. These results were<br />

confirmed by an in vitro nucleosome assembly on a telomeric DNA in the presence of purified<br />

99


Area 4: Molecular recognition in biomolecules<br />

TRF2 (Galati et al., in press). These findings raise the intriguing possibility that telomere<br />

protection is mediated, at least in part, by the TRF2-dependent regulation of nucleosome<br />

organization.<br />

Nucleosomes affect differently TRF1 and TRF2 accessibility to telomeric sequences<br />

By means of an in vitro model that mimics the binding of TRF proteins to telomeric DNA<br />

in a nucleosomal context, we found that the presence of nucleosomes close to the binding<br />

site disfavors TRF2 interaction whereas TRF1 recognizes its binding sequence with higher<br />

affinity. Our data indicate that this different behavior is mediated by histone tails and suggest<br />

a different role of TRF1 and TRF2 in the folding of telomeric chromatin (Galati et al., in<br />

preparation).<br />

Perspectives<br />

In the next future our aim is to map telomeric chromatin at a higher resolution than that<br />

present in literature. To this end, we realized a cell line containing an engineered telomere in<br />

which a strong nucleosome positioning sequence – the 601 DNA - is placed directly<br />

upstream of telomeric DNA. This positioned nucleosome will allow us to analyze whether the<br />

regular and tight nucleosome spacing is a feature of the entire telomere or whether the<br />

proximal and the distal part of the telomere are differently organized. Induction of telomere<br />

shortening by inhibition of telomerase activity will allow us to map telomeres over their entire<br />

length and to study changes of telomere chromatin organization as a function of telomere<br />

length. The possible molecular structures of telomeric chromatin will be screened adopting<br />

the theoretical model developed in our lab.<br />

Publications<br />

Scipioni A, De Santis P. Predicting nucleosome positioning in genomes: Physical and<br />

bioinformatic approaches. Biophys Chem 155: 53-64. doi: 10.1016/j.bpc.<strong>2011</strong>.03.006.<br />

Research Group<br />

Stefano Morosetti, professor; Anita Scipioni,<br />

researcher; Alessandra Galati, Emanuela<br />

Micheli, Pasqualina Punzi, post-doc fellows.<br />

Collaborations<br />

Pasquale De Santis, Armandodoriano<br />

Bianco, Dipartimento di Chimica, Sapienza<br />

Università di Roma; Daniela Rhodes, Medical<br />

Research Council, Laboratory of Molecular<br />

Biology, Cambridge, UK; Eric Gilson, Marie-<br />

Josephe Giraud-Panis, Institute for Research<br />

on Cancer and Aging, University of Nice,<br />

France.<br />

100


Area 4: Molecular recognition in biomolecules<br />

Plant innate immunity: signalling and recognition of Damage-<br />

Associated Molecular Patterns (DAMPs)<br />

Felice Cervone<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912517 - @: felice.cervone@uniroma1.it<br />

The cell wall-derived oligolacturonides (OGs) are typical damage-associated molecular<br />

patterns (DAMPs) formed during microbial infections. The molecular basis of the plant innate<br />

immunity elicited by OGs is the focus of this project. In Arabidopsis the accumulation of OGs<br />

takes place during the degradation of pectin by microbial polygalacturonases (PGs) and is<br />

favoured when PGs interact with specific plant inhibitors (PGIPs). OGs are recognized by a<br />

Wall-Associated Kinase (WAK1). We have shown that WAK1 and chimeric WAK1-derived<br />

receptors may be used to engineer resistance in crop plants by activating a signal<br />

transduction pathway that has been partially elucidated during this project (De Lorenzo et<br />

al., <strong>2011</strong>).<br />

MPK3 and MPK6 are Arabidopsis protein kinases activated by OGs. Analysis of single<br />

mapk mutants revealed that lack of MPK3 increases basal susceptibility to the fungal<br />

pathogen Botrytis cinerea but does not significantly affect elicitor-induced resistance. Instead,<br />

lack of MPK6 has no effect on basal resistance but suppresses OG-induced resistance to B.<br />

cinerea. Over-expression of the AP2C1 phosphatase leads to impaired OG-induced<br />

phosphorylation of both MPK3 and MPK6 (Galletti et al., <strong>2011</strong>).<br />

OGs inhibit adventitious root formation induced by auxin in Arabidopsis leaf explants as<br />

well as the expression of several auxin-responsive genes. The inhibition of auxin responses<br />

by OGs does not require ethylene, jasmonic acid and salicylic acid signalling and is<br />

independent of RBOHD-mediated reactive oxygen species production. OG-auxin antagonism<br />

does not involve a stabilization of auxin-response repressors or decreased levels of auxin<br />

receptor transcripts through the action of microRNAs (Savatin et al., <strong>2011</strong>).<br />

The ability of bacterial or fungal necrotrophs to produce enzymes capable of degrading<br />

pectin and producing OGs is often related to a successful initiation of the infective process.<br />

Pectin is synthesized in a methylesterified form and is subsequently de-esterified in muro by<br />

pectin methylesterase. Deesterification makes pectin more susceptible to the degradation by<br />

pectic enzymes such as PGs and pectate lyases secreted by pathogens. Pectobacterium<br />

carotovorum and B. cinerea induce in Arabidopsis a rapid expression of AtPME3 that acts as<br />

a susceptibility factor and is required for the initial colonization of the host tissue (Raiola et<br />

al., <strong>2011</strong>).<br />

The inhibitors PGIPs modulate the accumulation of OGs by interacting with microbial<br />

PGs. We have obtained a low-resolution structure of the complex formed by PG from<br />

Fusarium phyllophilum and PGIP from Phaseolus vulgaris as determined by small-angle x-<br />

ray scattering analysis. The inhibitor engages its concave surface of the leucine-rich repeat<br />

101


Area 4: Molecular recognition in biomolecules<br />

domain with the enzyme. Both sides of the enzyme active site cleft interact with the inhibitor.<br />

The structure is in agreement with previous site-directed mutagenesis data and has been<br />

further validated with structure-guided mutations and subsequent assay of the inhibitory<br />

activity (Benedetti et al., <strong>2011</strong>).<br />

Publications<br />

Benedetti M, Leggio C, Federici L, De Lorenzo G, Pavel NV, Cervone F. Structural resolution<br />

of the complex between a fungal polygalacturonase and a plant polygalacturonaseinhibiting<br />

protein by Small-Angle X-Ray Scattering. Plant Physiol <strong>2011</strong>, 157: 599-607. doi:<br />

http://dx.doi.org/10.1104/pp.111.181057.<br />

De Lorenzo G, Brutus A, Savatin DV, Sicilia F, Cervone F. Engineering plant resistance by<br />

constructing chimeric receptors that recognize damage-associated molecular patterns<br />

(DAMPs). FEBS Letters <strong>2011</strong>, 585: 1521-28. doi: 10.1016/j.febslet.<strong>2011</strong>.04.043.<br />

Galletti R, Ferrari S, De Lorenzo G. Arabidopsis MPK3 and MPK6 play different Roles in<br />

basal and oligogalacturonide- or flagellin-induced resistance against Botrytis cinerea. Plant<br />

Physiol <strong>2011</strong>, 157: 804-14. doi: 10.1104/pp.111.174003.<br />

Raiola A, Lionetti V, Elmaghraby I, Immerzeel P, Mellerowicz EJ, Salvi G, Cervone F,<br />

Bellincampi D. Pectin methylesterase is induced in Arabidopsis upon infection and is<br />

necessary for a successful colonization by necrotrophic pathogens. Mol Plant-Microbe<br />

Interact <strong>2011</strong>, 24: 432-40. doi: http://dx.doi.org/10.1094/MPMI-07-10-0157.<br />

Savatin DV, Ferrari S, Sicilia F, De Lorenzo G. Oligogalacturonide-auxin antagonism does<br />

not require posttranscriptional gene silencing or stabilization of auxin response repressors<br />

in Arabidopsis. Plant Physiol <strong>2011</strong>, 157: 1163-74. doi: http://dx.doi.org/ 10.1104/ pp.111.<br />

184663.<br />

Research Group<br />

Giulia De Lorenzo, Daniela Bellincampi,<br />

professors; Simone Ferrari, Benedetta Mattei,<br />

researchers; Giovanni Salvi, Daniela<br />

Pontiggia, Isabel Santori, research fellows;<br />

Manuel Benedetti, Claudia Fabbri, Fedra<br />

Francocci, Vincenzo Lionetti, Daniel Savatin,<br />

Francesca Sicilia, Francesco Spinelli, postdoc<br />

fellows; Federico Andreani, Elisa<br />

Bastianelli, Nora Gigli Bisceglia, Giovanna<br />

Gramegna, Matteo Gravino, Vanessa<br />

Modesti, Chiara Paparella, PhD students.<br />

102


Area 4: Molecular recognition in biomolecules<br />

Spontaneous generation and evolution of genetic information<br />

Ernesto Di Mauro<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912880 - @: ernesto.dimauro@uniroma1.it<br />

The aim of the project is to reconstruct the initial reactions that lead to the generation of<br />

genetic information. Starting from the consideration that it will not be possible to know how<br />

life started but that it is nevertheless possible to approximate its first steps, we have<br />

extended the observations that have brought us to establish three principles: 1) all the<br />

nucleic bases necessary for building extant nucleic acids can be synthesized nonenzymatically,<br />

from simple components, with high efficiency and in plausible prebiotic<br />

conditions. 2) Nucleic bases evolve, within the same prebiotic physico-chemical frame, into<br />

nucleosides and nucleotides. 3) Polymerization mechanisms exist leading from<br />

spontaneously activated precursors to long nucleic polymers.<br />

We have explored the conditions that extend the mechanisms identified so far, passing<br />

from the abiotic non-enzymatic synthesis of homogenous polymers to the generation of<br />

complex sequences. The questions asked are: 1) is it possible to spontaneously generate<br />

RNA sequence complexity, based on intrinsic properties of the RNA structure 2) Is it<br />

possible to correlate the generation of RNA complexity to the origin of the protein coding<br />

process<br />

In addition, somewhat unexpected results (see below) allow us to ask the key question: is<br />

it possible to correlate the development of (proto)genetic systems with the development of<br />

(proto)metabolic systems<br />

The data obtained provide the following answers:<br />

It is actually possible to generate sequence complementarity-driven nonenzymatic<br />

ligation of RNA. We have reported two reactions of RNA G:C sequences occurring nonenzymatically<br />

in water in the absence of any added cofactor or metal ion: (a) sequence<br />

complementarity-driven terminal ligation; (b) complementary sequence adaptor-driven<br />

multiple tandemization. The two abiotic reactions increase the chemical complexity of the<br />

resulting pool of RNA molecules and change the Shannon information of the initial population<br />

of sequences.<br />

The reaction of nonenzymatic terminal ligation of homogeneous polyA sequences<br />

previously discovered (Pino et al., J Biol Chem 2008, 283: 36494-503) has been analyzed by<br />

Atomic Force Microscopy. The products of ligation reaction of a 24 nucleotides-long PolyA<br />

RNA adsorbed on mica were analyzed by atomic force microscopy. In that study the<br />

occurrence of oligonucleotides at different degrees of polymerization was quantitatively<br />

studied before and after ligation reaction. The microscopy images at the nanoscale showed<br />

103


Area 4: Molecular recognition in biomolecules<br />

that nonenzymatic ligation of pristine RNA monomers resulted in the formation of<br />

supramolecular aggregates, with prevalence of dimers and tetramers. Analytical conditions<br />

were defined allowing the identification, the quantitative evaluation, and their distribution after<br />

ligation reaction, also providing an estimate of the degree of hydration of the objects. Such<br />

investigation provides the simplest yet model system for direct investigation of RNA reactions<br />

by advanced microscopy.<br />

Taken together, these studies show how spontaneously generated short oligonucleotides<br />

may progress along the path of generation of longer sequences. The possibility of introducing<br />

“errors” in the abiotic replication of RNA sequences has been shown, suggesting evolutionary<br />

mechanisms in the absence of complex phenotypes.<br />

The previously reported polymerization of oligonucleotides from cyclic nucleotides has<br />

been further studied and the conditions for improvement of the synthetic reactions in part<br />

published.<br />

Relevant results pertain the identification of the products of prebiotic synthesis of organic<br />

compounds from formamide in the presence of specific classes of catalysts like boroncontaining<br />

minerals and minerals from the Murchison meteorite. In addition to nucleic bases,<br />

complex population of carboxylic acids were observed, showing the possibility that in the<br />

same test-tube the precursors formed of both RNA and key metabolic cycles. The rational of<br />

these studies has been published.<br />

Publications<br />

Pino S, Costanzo G, Giorgi A, Di Mauro E. Sequence complementarity-driven nonenzymatic<br />

ligation of RNA. Biochemistry <strong>2011</strong>, 50: 2994-3003. doi: 10.1021/bi101981z.<br />

Saladino R, Crestini C, Pino S, Costanzo G, Di Mauro E. Formamide and the origin of life.<br />

Phys Life Rev <strong>2011</strong> Epub. <strong>2011</strong>. doi: 10.1016/j.plrev.<strong>2011</strong>.12.002.<br />

Research Group<br />

Giovanna Costanzo, CNR researcher,<br />

Samanta Pino, post-doc fellow; Silvia Lopizzo,<br />

technician.<br />

Collaborations<br />

Raffaele Saladino, Dip. ABAC, Università della<br />

Tuscia, Viterbo; Claudia Crestini, Università<br />

Tor Vergata, Roma; Edward N. Trifonov,<br />

University of Haifa, Israel.<br />

104


Area 4: Molecular recognition in biomolecules<br />

Molecular and functional approaches to investigate the<br />

neuroprotective and neuromodulatory roles of chemokines and<br />

their receptors in the central nervous system<br />

Cristina Limatola<br />

Department of Physiology and Pharmacology "Vittorio Erspamer"<br />

℡: +39 06 49690243 - @: cristina.limatola@uniroma1.it<br />

This research project want to investigate the action mechanism of trans-membrane<br />

chemokine fractalkine CX3CL1 in the CNS. In particular we are interested in evaluating the<br />

role of glial cells as modulators of the neuroprotective and neuromodulatory effects of this<br />

chemokine also with attention to the signaling pathways activated by adenosine receptors<br />

(ARs). In the last few years we described that microglia and the A1R are involved as<br />

mediators of the neuroprotective and neuromodulatory effects of CX3CL1. We recently<br />

extended this study to an in vivo model of cerebral ischemia in rodents and to the effects of<br />

CX3CL1 on hippocampal synaptic plasticity induced by environmental stimuli.<br />

In <strong>2011</strong> we completed the study of the neuroprotective activity of CX3CL1 in models of<br />

permanent cerebral ischemia in rats and mice. In particular, we demonstrated that CX3CL1 is<br />

neuroprotective also in vivo, reducing the cell death of cortical neurons induced by<br />

permanent mean cerebral artery (pMCAO) occlusion. The protective effect of CX3CL1 has<br />

been demonstrated: 1) by a reduction of the ischemic brain area measured with a specific<br />

vital dye, 24 hours after injury induction; 2) by an improvement of performances in sensory<br />

motor tests to evaluate the brain damage from the functional point of view. In addition, we<br />

observed that these same and other parameters (evidenced by NMR analysis) are<br />

maintained up to 50 days after pMCAO confirming and extending the importance of our<br />

results. As concern the mechanisms, we prove that in vivo the protective effect of CX3CL1 is<br />

mediated by the indirect activation of adenosine receptor type 1 (A1R). This result has been<br />

reached with two different experimental approaches: we used both a specific A1R antagonist,<br />

DPCPX, which gave a complete inhibition of the neuroprotective effect of CX3CL1 and we<br />

performed pMCAO in A1R-/- mice, treated for icv injection of CX3CL1 observing no<br />

protection from ischemic damage (Cipriani et al., <strong>2011</strong>). We also investigated another aspect<br />

of the activity of CX3CL1 in the central nervous system: in particular we have been interested<br />

in the effects on hippocampal plasticity of mice exposure for a long period to an environment<br />

“enriched” with sensory, social, motor cues, with particular attention to the changes induced<br />

in mice lacking CX3CL1 receptor (CX3CR1 GFP/GFP mice). The results demonstrated that in<br />

CX3CR1 GFP/GFP mice, hippocampal plasticity phenomena like learning in the “Water Maze”<br />

test and synaptic plasticity modulation like long-term potentiation (LTP) are increased in<br />

comparison with control wild-type (wt) mice. In contrast, while wt mice in enriched<br />

environment potentiated LTP and learning and memory ability, CX3CR1 GFP/GFP mice did not<br />

improve their performances upon “enrichment”. We also analyzed hippocampal neurogenesis<br />

105


Area 4: Molecular recognition in biomolecules<br />

in the dentate gyrus observing that environmental enrichment is equally effective in wt and<br />

CX3CR1 GFP/GFP mice in increasing the number of newborn cells, with no significant<br />

differences in the two genotypes. In basal conditions, in the absence of enrichment,<br />

CX3CR1 GFP/GFP mice have a reduced neurogenesis (Maggi et al., <strong>2011</strong>).<br />

Publications<br />

Di Angelantonio S, Piccioni A, Moriconi C, Trettel F, Cristalli G, Grassi F, Limatola C.<br />

Adenosine A2A receptor induces protein kinase A-dependent functional modulation of<br />

human 3{beta}4 nicotinic receptor. J Physiol <strong>2011</strong>, 589: 2755-66. doi:<br />

10.1113/jphysiol.<strong>2011</strong>.207282.<br />

Bernareggi A, Grilli M, Marchi M, Limatola C, Ruzzier F, Eusebi F. Characterization of<br />

GABA(A) receptors expressed in glial cell membranes of adult mouse neocortex using a<br />

Xenopus oocyte microtransplantation expression system. J Neurosci Methods <strong>2011</strong>, 198:<br />

77-83. http://dx.doi.org/10.1016/j.jneumeth.<strong>2011</strong>.03.011.<br />

Maggi L, Scianni M, Branchi I, D'Andrea I, Lauro C, Limatola C. CX3CR1 deficiency alters<br />

hippocampal-dependent plasticity phenomena blunting the effects of enriched environment.<br />

Front Cell Neurosci <strong>2011</strong>, 5-22: 1-9. doi: 10.3389/fncel.<strong>2011</strong>.00022.<br />

Cipriani R., Villa P, Chece G, Lauro C, Paladini P, Micotti E, Perego C, De Simoni MG,<br />

Fredholm BB, Eusebi F, Limatola C. CX3CL1 is neuroprotective in permanent focal<br />

cerebral ischemia in rodents. J Neurosci <strong>2011</strong>, 31: 16327–35. doi:<br />

10.1523/JNEUROSCI.3611-11.<strong>2011</strong>.<br />

Palma E, Inghilleri M, Conti L, Deflorio C, Frasca V, Manteca A, Pichiorri F, Roseti C, Torchia<br />

G, Limatola C, Grassi F, Miledi R Physiological characterization of human muscle<br />

acetylcholine receptors from ALS patients. Proc Natl Acad Sci U S A <strong>2011</strong>, 108: 20184-8.<br />

doi: 10.1073/pnas.1117975108.<br />

Research Group<br />

Francesca Grassi, Sergio Fucile, Eleonora<br />

Palma, Davide Ragozzino, professors; Myriam<br />

Catalano, Flavia Trettel, researchers; Raffaela<br />

Cipriani, Clotilde Lauro, post-doc fellows;<br />

Grimaldi Alfonso, PhD student; Giuseppina<br />

Chece, technician.<br />

Collaborations<br />

Maria Grazia De Simoni, Pia Villa, <strong>Istituto</strong><br />

Mario Negri, Milano; Letizia Antonilli,<br />

Valentina Brusadin, Dipartimento di Fisiologia<br />

e Farmacologia, Sapienza Università di Roma;<br />

Bertil Fredholm, Karolinska Institute,<br />

Stockholm, Sweden; Angelo Spinedi,<br />

Università di Roma Tor Vergata.<br />

106


Area 4: Molecular recognition in biomolecules<br />

Neurone response to experimental injury and lack of dystrophin: a<br />

molecular, functional and structural study in autonomic ganglia in<br />

vivo and in vitro<br />

Paola Paggi<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49912323 - @: paola.paggi@uniroma1.it<br />

Aim of our project is the characterisation in rodent superior cervical ganglion (SCG) of the<br />

molecular mechanisms and structural changes involved in the establishment, maintenance<br />

and plasticity of the reciprocal interactions between pre- and post-ganglionic neurones and<br />

between post-ganglionic neurones and their target organs. Damage of the ganglionic<br />

connectivity, consequent to pre- and post-ganglionic nerve crush or to the lack of dystrophin<br />

(Dp427), is used as a tool to perform our investigation. In the year <strong>2011</strong> we investigated the<br />

relevant pathological changes produced by lack of Dp427, a protein of the cortical<br />

cytoskeleton, in mdx mice, an animal model for Duchenne muscular dystrophy, investigating<br />

the following aspects: i) analysis of nicotine-evoked currents in dissociated neurones of mdx<br />

mouse SCG, in which the nicotinic acetylcholine receptors containing the α3 subunit<br />

(α3nAChRs) are drastically reduced compared with the wild-type; ii) large-scale analysis of<br />

gene expression in mdx mouse SCG to search for genes affected by the spontaneous<br />

mutation in the dystrophin gene and possibly contributing to SCG neuronal death, occurring<br />

between postnatal day 5 (P5) and 15 (P15), and to the altered reaction to axotomy and axon<br />

growth of SCG neurones, previously observed.<br />

i) In the sympathetic superior cervical ganglion (SCG), nicotinic acetylcholine receptors<br />

(nAChRs) mediate fast synaptic transmission. We previously demonstrated that in SCG<br />

neurons of mdx mice, lack of dystrophin causes a decrease, compared to the wild-type, in<br />

post-synaptic nAChRs containing the α3 subunit associated with β2 and/or β4 (α3β2/β4-<br />

nAChRs), but not of those containing the α7 subunit (α7-nAChRs). We showed, by whole cell<br />

patch-clamp recordings from cultured SCG neurons, that both nicotine- and acetylcholineevoked<br />

currents through α3β2/β4-nAChRs are significantly reduced in mdx mice compared<br />

to wild-type, while those through α7-nAChR are unaffected. This reduction associates with<br />

that of the protein levels of α3, β2 and β4 subunits. We suggest that, in mdx mouse SCG<br />

neurons, lack of dystrophin, by specifically affecting membrane stabilization of α3β2/β4-<br />

nAChRs, could determine an increase in receptor internalization and degradation, with<br />

consequent reduction in the fast intraganglionic cholinergic transmission.<br />

ii) By DNA microarray, we looked for changes in gene expression in SCG of P5, P10 and<br />

6-7 week-old mdx mice to verify whether the lack of Dp427 may affect transcript levels of<br />

specific genes, possibly giving an insight into the mechanisms involved in the structural and<br />

functional alterations we previously observed. Ontological Analysis of more than 500<br />

modulated genes showed significant differences in genetic class enrichment at each post-<br />

107


Area 4: Molecular recognition in biomolecules<br />

natal date. Up-regulated genes mainly fell in the categories of vesicular trafficking,<br />

cytoskeletal and synaptic organization, while those down-regulated were associated to axon<br />

development, growth factors, intracellular signal transduction, gene expression regulation,<br />

synapse morphogenesis and nicotinic receptor clustering. In particular, downregulation of<br />

genes related to trophic factor signaling pathways and involved in axon growth and<br />

differentiation suggested that lack of Dp427 could affect trophic factor sensitivity and axon<br />

growth dynamics.<br />

Publications<br />

Di Angelantonio S, De Stefano ME, Piccioni A, Lombardi L, Gotti C, Paggi P. Lack of<br />

dystrophin functionally affects α3β2/β4-nicotinic acethylcholine receptors in sympathetic<br />

neurons of dystrophic mdx mice. Neurobiol Dis 41: 528-537. doi:10.1016/j.nbd.2010.<br />

10.024.<br />

Research Group<br />

Maria Egle De Stefano, professor, Silvia Di<br />

Angelantonio, researcher, Valerio Licursi,<br />

Loredana Lombardi, post-doc fellows.<br />

Collaborations<br />

Tamara Petrucci, <strong>Istituto</strong> Superiore di Sanità,<br />

Laboratorio di Biologia Cellulare, Roma; Cecilia<br />

Gotti, CNR, <strong>Istituto</strong> di Neuroscienze, Centro di<br />

Farmacologia Cellulare e Molecolare, Dip. di<br />

Farmacologia Medica, Università di Milano;<br />

Irene Bozzoni, Ernesto Di Mauro, Andrea<br />

Mele, Rodolfo Negri, Alberto Oliverio,<br />

Dipartimento di Biologia e Biotecnologie “C.<br />

Darwin”, Sapienza Università di Roma; Carla<br />

Perrone Capano, Dipartimento di Scienze<br />

Biologiche, Università di Napoli “Federico II”;<br />

Casper Hoogenraad, Erasmus Medical<br />

Center, Department of Neuroscience,<br />

Rotterdam, The Netherlands; Falk Schroedl,<br />

Paracelsus University of Salzsburg, Austria.<br />

108


Area 4: Molecular recognition in biomolecules<br />

Molecular mechanisms integrating endocytosis and signalling of<br />

fibroblast growth factor receptors<br />

Maria Rosaria Torrisi<br />

Department of Clinical and Molecular Medicine<br />

℡: +39 06 33775502 - @: mara.torrisi@uniroma1.it<br />

Endocytosis of receptor tyrosine kinases (RTKs) represents one of the main mechanisms<br />

of attenuation of the signal transduction, through re-localization by internalization and<br />

subsequent degradation of signaling molecules at the plasma membrane. The endocytic<br />

machinery and the RTK signal transduction appear to be intimately linked. In contrast to most<br />

of the receptor tyrosine kinases (RTKs), the keratinocyte growth factor receptor<br />

(KGFR/FGFR2b) appears to play an unique and unusual role in epithelial tissues, controlling<br />

proliferation and differentiation and exerting a tumor suppressive function in vitro and in vivo.<br />

KGFR endocytosis, upon binding of its paracrine ligands KGF and FGF10, is clathrindependent,<br />

but once internalized the receptors may follow two distinct endocytic pathways<br />

targeting to lysosomal degradation or recycling to the plasma membrane, as a consequence<br />

of a distinct phosphorylation and ubiquitination. Therefore, FGFR2b/KGFR represent a model<br />

system to shed light on the molecular mechanisms which control the endocytic trafficking and<br />

the receptor signaling. In addition, we have proposed that the expression of E5 protein<br />

encoded by high-risk human papillomavirus (HPV) type 16 can be used as a tool to analyze<br />

the endocytic indirect recycling pathway (Belleudi et al., Oncogene <strong>2011</strong>).<br />

Since it is known that cell migration requires endocytic trafficking and polarization of<br />

adhesion molecules and RTKs to the leading edge, during this first year of the research<br />

project we investigated the role of KGFR endocytosis in the possible polarization of the<br />

receptors and their signaling and trafficking at the leading edge of migrating cells. We<br />

demonstrated the polarized localization of KGFR, upon ligand stimulation, regulated by Src.<br />

In addition, we found that the increased expression of the receptor and its polarization were<br />

able to enhance cell migration. Interestingly, both Src and the actin-binding protein cortactin<br />

appear to play a key role in the KGFR endocytosis and polarization, supporting the crucial<br />

involvement of RTK trafficking in cell motility (Belleudi et al., PLoS One <strong>2011</strong>a). Future work<br />

will be focused to characterize the recycling pathways responsible for KGFR polarization<br />

during migration. In particular, based on the present observations that KGF and FGF10 have<br />

different ability to induce Src tyrosine phosphorylation, it would be interesting to verify if the<br />

two KGFR ligands might be able to mediate the receptor polarization through its targeting to<br />

different endocytic recycling pathways. During the first year of the project we investigated<br />

also the contribution of the KGFR expression, ligand-induced activation and receptor<br />

signaling in regulating the phagocytic process. To this aim we analyzed the uptake in vitro of<br />

fluorescent latex beads and we found that KGFR ligands trigger phagocytosis. Moreover, our<br />

results obtained by KGFR depletion or overexpression of wild type or defective mutants<br />

109


Area 4: Molecular recognition in biomolecules<br />

demonstrated the promoting effect exerted by the activated receptor on the phagocytic<br />

ingestion and indicated the crucial role of the PLCγ signaling pathway in this KGFR-mediated<br />

mechanism of uptake. In addition, while the block of the receptor kinase activity and signaling<br />

inhibits both phagocytosis and receptor endocytosis, the block of the receptor clathrinmediated<br />

endocytosis by RNA interference does not impair the KGF activity in promoting the<br />

phagocytosis, unequivocally demonstrating that the receptor signaling and activation of the<br />

PLCγ downstream pathway occurs from the plasma membrane (Belleudi et al., FASEB J<br />

<strong>2011</strong>). Finally, to identify the possible receptor-mediated signaling pathways involved in the<br />

earlier induction of differentiation, we modulated in vitro the receptor expression and we<br />

induced a synchronous wave of differentiation, demonstrating that KGFR overexpression,<br />

activation and signalling increase the early differentiation, while receptor depletion reduces it.<br />

We found also that the PI3K/Akt signaling pathway is involved in the control of KGFRmediated<br />

keratinocyte differentiation. This in vitro experimental model indicates that<br />

FGFR2b/KGFR expression represents a key event regulating keratinocyte early<br />

differentiation during the switch from undifferentiated to differentiating cells (Belleudi et al.,<br />

PLoS One 2001b)<br />

Publications<br />

Belleudi F, Purpura V, Scrofani C, Persechino F, Leone L, Torrisi MR. Expression and<br />

signaling of the tyrosine kinase FGFR2b/KGFR regulates phagocytosis and melanosome<br />

uptake in human keratinocytes. FASEB J <strong>2011</strong>, 25: 170-81. doi: 10.1096/fj.10-162156.<br />

Belleudi F, Leone L, Purpura V, Cannella F, Scrofani C, Torrisi MR. HPV16 E5 affects the<br />

KGFR/FGFR2b-mediated epithelial growth through alteration of the receptor expression,<br />

signaling and endocytic traffic. Oncogene <strong>2011</strong>, 30: 4963-76. doi: 10.1038/onc.<strong>2011</strong>.203.<br />

Belleudi F, Scrofani C, Torrisi MR, Mancini P. Polarized endocytosis of the keratinocyte<br />

growth factor receptor in migrating cells: role of Src-signaling and cortactin. PLoS One<br />

<strong>2011</strong>, 6:e29159. doi: 10.1371/journal.pone.0029159.<br />

Belleudi F, Purpura V, Torrisi MR. The receptor tyrosine kinase FGFR2b/KGFR controls<br />

early differentiation of human keratinocytes. PLoS One <strong>2011</strong>, 6:e24194. doi:<br />

10.1371/journal.pone.0024194.<br />

Research Group<br />

Patrizia Mancini, Maurizio Alimandi, Lavinia<br />

Lotti, Vincenzo Visco, professors, Francesca<br />

Belleudi, Salvatore Raffa, researchers; Valeria<br />

Purpura, Danilo Ranieri, PhD students;<br />

Antonio Sabatucci, technician.<br />

110


Area 5<br />

Cellular and molecular<br />

immunology


Area 5: Cellular and molecular immunology<br />

Interplay amongst chronic immune activation, apoptosis, crosspresentation,<br />

immune-regulation, and autoimmunity<br />

Vincenzo Barnaba<br />

Department of Internal Medicine and Medical Specialties<br />

℡: +39 06 491268 - @: vincenzo.barnaba@uniroma1.it<br />

In previous studies, we found that the proteome of apoptotic T cells includes prominent<br />

caspase-cleaved cellular proteins and that a high proportion of distinct epitopes in these<br />

fragments (apoptotic epitopes) can be cross-presented by DCs to autoreactive CD8 + T cells<br />

(Moroni-Rawson et al., Nat Med 2007). In chronic HIV infection, these autoreactive CD8 + T<br />

cells correlate with the proportion of apoptotic CD4 + T cells in vivo and are involved in<br />

establishing polyclonal T cell activation that in the long run results in generalized T cell<br />

dysfunction/depletion. In addition, our previous report showed that apoptotic cells derived<br />

from activated T cells (in contrast to those derived from resting T cells or from non-lymphoid<br />

cells) retain the expression of CD40 ligand (L) and can then condition CD40 + DCs to acquire<br />

high capacities to prime or cross-prime autoreactive T cells (Propato et al., Nat Med 2001).<br />

This mechanism is consistent with the evidence that the signals provided by CD40L +<br />

apoptotic cells and not those provided by conventional apoptotic cells facilitate the<br />

emergence of autoreactive T cell responses to apoptotic self-antigens.<br />

Here we used the hepatitis C virus (HCV) infection as a human model of acute infection<br />

that generally undergoes chronic progression to verify whether CD8 + T cells that are specific<br />

for apoptotic self-epitopes have a distinct effector type-1, -2, or -17 phenotype, to distinguish<br />

which of them may participate in determining the fate of a viral infection (recovery versus<br />

chronicity), and to ascertain the mechanisms whereby these responses are induced and<br />

maintained. We demonstrated for the first time that the emergence of mixed polyfunctional<br />

(type-1, -2, -17) CD8 + T EM cell responses to apoptotic self-epitopes is related to the chronic<br />

evolution of acute HCV infection. The responses were directly correlated with the plasma<br />

viral load or the serum ALT levels, and were then sustained over time in relation to the viral<br />

persistence. These results suggest that, in conditions in which HCV has been able to evade<br />

the virus-specific immunity, strong CD8 + T cell responses against apoptotic self-epitopes are<br />

maintained and may contribute to the liver immunopathology through the production of high<br />

levels of inflammatory cytokines. This hypothesis is further emphasized by our parallel study<br />

indicating that similar autoreactive CD8 + T cell responses in chronically infected patients are<br />

recruited in the inflamed livers, are related with the signs of hepatic damage, and decrease in<br />

relation with the decline or the disappearance of the viral load upon antiviral therapy<br />

(interferon plus ribavirin @ ).<br />

We also demonstrated that cross-presentation of apoptotic T cells by DCs promptly<br />

activates CD8 + T EM cells specific for caspase-cleaved apoptotic self-epitopes ex vivo<br />

indicating that this mechanism might be operative in the induction of the resulting<br />

113


Area 5: Cellular and molecular immunology<br />

polyfunctional autoreactive responses in patients with acute HCV infection who are<br />

experiencing chronic progression. The strong production of IFN-γ and IL-17 may favor the<br />

triggering of recruitment of inflammatory cells to the liver, which contribute to liver pathology.<br />

An important facet of our findings is that they demonstrate a link between the TCR avidity<br />

of autoreactive CD8 + T cells and the difference in the responsiveness of apoptotic epitopespecific<br />

CD8 + T cells exhibited by patients experiencing chronic infection and those<br />

undergoing infection resolution. Our results suggest that autoreactive CD8 + T cells with<br />

higher avidity for apoptotic self-epitopes are sustained over time and correlate with the<br />

progression toward chronic infection. The selection of the autoreactive CD8 + T cells with<br />

higher avidity likely occurs because of a sustained stimulation by apoptotic antigens. By<br />

contrast, lower avidity CD8 + T cells in the presence of weaker stimuli would undergo rapid<br />

contraction, as seen in the peripheral blood of patients with self-limited HCV infection.<br />

Publications<br />

Oliviero B, Cerino A, Varchetta S, Paudice E, Pai S, Ludovisi S, Zaramella M, Michelone G,<br />

Pugnale P, Negro F, Barnaba V, Mondelli MU. Enhanced B-cell differentiation and reduced<br />

proliferative capacity in chronic hepatitis C and chronic hepatitis B virus infections. J<br />

Hepatol <strong>2011</strong>, 55: 53-60. doi.org/10.1016/j.jhep.2010.10.016.<br />

Spadaro F, Lapenta C, Donati S, Abalsamo L, Barnaba V, Belardelli F, Santini SM, Ferrantini<br />

M. Interferon-alpha enhances cross-presentation in human dendritic cells by modulating<br />

antigen survival, endocytic routing and processing. Blood <strong>2011</strong> Epub. doi: 10.1182/blood-<br />

<strong>2011</strong>-06-363564.<br />

Research Group<br />

Daniele Accapezzato, Silvia Piconese,<br />

researcher; Alessandra Citro, Helen Martini,<br />

Valeria Schinzari, post-doc fellows; Carmela<br />

Martire, Alessandra Proia, Eleonora Timperi,<br />

PhD students.<br />

114


Area 5: Cellular and molecular immunology<br />

Signalling events negatively regulating FcεRI expression and mast<br />

cell functional responses<br />

Rossella Paolini<br />

Department of Molecular Medicine<br />

℡: +39 06 4468448 - @: rossella.paolini@uniroma1.it<br />

Mast cells are key effectors in allergic diseases: upon ligation of the high affinity receptor<br />

for IgE (FcεRI) they release preformed and newly synthesized pro-inflammatory mediators.<br />

FcεRI is composed of an IgE-binding α chain, and the ITAM-containing β and γ subunits.<br />

Upon FcεRI cross-linking, the β chain-associated Src family PTK Lyn, phosphorylates β and<br />

γ-chain ITAMs allowing the recruitment and consequent activation of Syk, responsible for<br />

signal propagation (1). Notably, the full activation of FcεRI requires the recruitment of<br />

aggregated receptors into lipid rafts, where engaged receptors are concentrated (2) and can<br />

more easily interact with signaling molecules, such as active Lyn (3), favouring<br />

phosphorylation events.<br />

Besides these positive signals, FcεRI engagement has recently been understood to<br />

generate negative intracellular signals capable of limiting the rate and the extent of mast cell<br />

functional responses (4). Relevant to this, others and we have demonstrated that the Cbl<br />

family proteins control the amplitude of FcεRI-generated signals by specific ubiquitin<br />

modification of activated receptor subunits and associated protein tyrosine kinases (5,6).<br />

Concurrently, engaged receptors trigger their own endocytosis to extinguish signaling<br />

through removal of activated FcεRI complexes from the cell surface and delivery to<br />

lysosomes for degradation (7).<br />

The present study was aimed at identifying the molecular mechanisms ensuring the<br />

clearance of antigen-stimulated FcεRI complexes from the cell surface, thus contributing to<br />

the termination of mast cell functional program.<br />

We initially demonstrated that FcεRI subunits are monoubiquitinated by c-Cbl at multiple<br />

sites upon stimulation, and provided evidence for a role of ubiquitin as a signal regulating the<br />

initial step of internalization: endocytosis of engaged FcεRI complexes is dramatically<br />

affected in conditions of impaired receptor ubiquitination and requires the integrity of lipid<br />

rafts (8).<br />

We have further investigated the involvement of adaptor proteins harboring ubiquitin<br />

interacting motifs, namely Eps15, Eps15R, epsin and Hrs, in coupling ubiquitinated FcεRI to<br />

the endocytic machinery. Eps15, Eps15R and Epsin display similar functions: they control the<br />

early steps of the endocytic route coupling ubiquitinated receptors with components of the<br />

budding vesicles (9,10). The key role of Hrs is, instead, the delivery of ubiquitinated proteins<br />

to the outer membrane of the late endosomes and the sorting of the cargo into internal<br />

vesicles of multivesicular bodies for lysosomal degradation (11).<br />

Although we failed to observe a significant decrease of FcεRI entry in early endosomes<br />

upon individual depletion of Eps15, Eps15R or Epsin, the simultaneous depletion of all of<br />

115


Area 5: Cellular and molecular immunology<br />

them impaired ligand-induced receptor endocytosis, suggesting a partial overlapping function<br />

of these adapters in ubiquitinated FcεRI uptake. Notably, Hrs depletion retains ubiquitinated<br />

receptors into early endosomes and partially prevents their sorting into lysosomes supporting<br />

a critical role for Hrs in controlling the fate of internalized receptor complexes (8).<br />

In summary, our data demonstrate a key role for the ubiquitin pathway to ensure proper<br />

endocytic trafficking of antigen-stimulated FcεRI to a lysosomal compartment where<br />

degradation of the complexes can take place (8,12).<br />

The outcome of our proposal would be a better comprehension of the molecular<br />

mechanisms that functionally regulate the endocytic adaptor Hrs. In this respect, our<br />

preliminary results demonstrate that Hrs is subjected to antigen-dependent tyrosine<br />

phosphorylation and monoubiquitination, and that Syk is the main kinase regulating both Hrs<br />

covalent modifications (Gasparrini F. et al., unpublished observations). At present we are<br />

investigating whether and how Syk-induced modifications of Hrs impact on its endosomal<br />

localization, thus regulating Hrs function.<br />

References<br />

1) Nadler MJ, Matthews SA, Turner H, Kinet JP. Adv Immunol, 76:325-355, 2000.<br />

2) Field KA, Holowka D, Baird B. J Biol Chem, 272:4276-4280, 1997.<br />

3) Sheets ED, Holowka D, Baird B. J Cell Biol, 145:877-887,1999.<br />

4) Molfetta R, Peruzzi G, Santoni A, Paolini R. Arch Immunol Ther Exp, 55:219-229, 2007.<br />

5) Paolini R, Molfetta R, Beitz LO, Zhang J, Scharenberg AM, Piccoli M, Frati L, Siraganian R,<br />

Santoni A. J Biol Chem, 277:36940-36947, 2002.<br />

6) Kyo S, Sada K, Qu X, Maeno K, Miah SM, Kawauchi-Kamata K, Yamamura H. Genes Cells,<br />

8:825-836, 2003.<br />

7) Molfetta R, Belleudi F, Peruzzi G, Morrone S, Leone L, Dikic I, Piccoli M, Frati L, Torrisi M R,<br />

Santoni A, Paolini R. J Immunol, 175: 4208-4216, 2005.<br />

8) Molfetta R, Gasparrini F, Peruzzi G, Vian L, Piccoli M, Frati L, Santoni A, Paolini R. PLoS One,<br />

4(5):e5604, 2009.<br />

9) Carbone R, Fré S, Cannolo G, Belleudi F, Mancini P, Pelicci PG, Torrisi M, Di Fiore PP. Cancer<br />

Res, 57:5498-5504, 1997.<br />

10) Chen H, Fre S, Slepnev VI, Capua MR, Takei K, Butler MH, Di Fiore PP, De Camilli P. Nature<br />

394:793-797, 1998.<br />

11) Bache K., Brech A, Mehlum A, Stenmark H. J Cell Biol, 162:435-442, 2003.<br />

12) Molfetta R, Gasparrini F, Santoni A, Paolini R. Mol Immunol, 47:2427-2434, 2010.<br />

Publications<br />

Gasparrini F, Molfetta R, Santoni A, Paolini R. Cbl family proteins: balancing FcεRI-mediated<br />

mast cell and basophil activation. Int Arch Allergy Immunol <strong>2011</strong>; 156: 16-26. doi:<br />

10.1159/000322236.<br />

Research Group<br />

Rosa Molfetta, researcher; Alessandra Porzia;<br />

post-doc fellow; Linda Quatrini, PhD student.<br />

116


Area 5: Cellular and molecular immunology<br />

Anti-tumor effector functions of NK cells in tumor microenviroment<br />

Angela Santoni<br />

Department of Molecular Medicine<br />

℡: +39 06 44340632 - @: angela.santoni@uniroma1.it<br />

Our research proposal is aimed at identifying the molecular mechanisms underlying the<br />

NK cell recognition of tumor cells, and the defects of NK cell maturation and functional<br />

competence promoted by tumor microenvironment. To this end, a tumor type particularly<br />

suitable is the multiple myeloma (MM), a clonal B cell malignancy characterized by an excess<br />

of mature plasma cells mainly in the BM where NK cell maturation occurs. We have<br />

previously shown that treatment of MM cells with low doses of drugs largely used in the<br />

therapy of MM, such as doxorubicin, melphalan and bortezomib, up-regulate DNAM-1 and<br />

NKG2D ligands and is associated with increased NK cell degranulation. Drug-induced<br />

DNAM-1 and NKG2D ligand expression was abolished following treatment with the<br />

pharmacological inhibitors (caffeine and KU-55933) of the DNA Damage Response (DDR)-<br />

initiating kinases ATM/ATR, and was preferentially observed on senescent cells arrested in<br />

the G2 phase of the cell cycle (Soriani et al., Blood 2009).<br />

More recently, we have investigated the signaling pathways underlying NK cell<br />

recognition of drug-induced senescent MM cells with particular attention given to the role<br />

played by changes in the cellular redox state. We initially found that low doses of doxorubicin<br />

and melphalan can trigger the DNA damage response as indicated by chk1/2 checkpoint<br />

kinase activation, phosphorylation of histone H2AX and p53 serine 15 phosphorylation in MM<br />

cells. Thus, we tested the effect of pharmacological inhibitors targeting chk1/chk2<br />

(SB218078, UCN-01) and p53 (pifitrin-a) on the expression of NKG2D and DNAM-1 ligands.<br />

We found that MICA, MICB and PVR up-regulation was dependent on chk1/chk2 activation,<br />

whereas it was not affected upon treatment with pifitrin-a, thus excluding the involvement of<br />

p53 in this event. To evaluate the role of changes in the cellular redox state in ligand upregulation<br />

by doxorubicin and melphalan known to induce ROS generation, we performed<br />

experiments in the presence NAC (N-acetyl-L-cysteine), a potent scavenger of ROS in SKO-<br />

007(J3) MM cell line or in MM patients’ CD138 + malignant PCs. We observed that druginduced<br />

up-regulation of NKG2D and DNAM-1 ligand protein and mRNA expression was<br />

reduced in the presence of NAC. NAC-mediated inhibition of ligand up-regulation was<br />

associated with impaired ATM and chk1/chk2 activation and senescent cell associated-cell<br />

cycle G2/M arrest.<br />

We have also analyzed whether drug treatment of MM cell lines could also result in MICA<br />

and MICB release. Our preliminary findings show that basal levels of soluble MICB were<br />

strongly up-regulated after 24h-treatment and peaked at 72h, whereas soluble MICA was<br />

barely detected. In an effort to elucidate the molecular mechanisms underlying MICB<br />

117


Area 5: Cellular and molecular immunology<br />

shedding, we analyzed the ability of different inhibitors to affect MICB shedding on drugtreated<br />

cells. Our results indicate that the metalloproteinase inhibitor Marimastat dramatically<br />

decreased the shedding of MICB from drug-treated cells and this decrease was<br />

accompanied by a concomitant increase of cell surface MICB. Similar results were obtained<br />

using the dithiobisnitrobenzoic acid that inhibits protein disulfide isomerase activity. Finally,<br />

we demonstrated that cell surface MICB but not MICA is associated with the membrane raft<br />

compartment, further suggesting that different mechanisms control MICA and MICB shedding<br />

in MM cell lines in response to therapeutic agents.<br />

Overall, our findings show that low doses of chemotherapeutic drugs up-regulate NKG2D<br />

and DNAM-1 ligands on MM cells in a ATM/Chk1/2- dependent and p53-independent<br />

manner, and this event is triggered by ROS-dependent activation of DNA Damage<br />

Response. In addition, we suggest a model in which the senescence program promotes<br />

tumor cell recognition and elimination by NK cells.<br />

Publications<br />

La Regina G, Bai R, Rensen W, Coluccia A, Piscitelli F, Gatti V, Bolognesi A, Lavecchia A,<br />

Granata I, Porta A, Maresca B, Soriani A, Iannitto ML, Mariani M, Santoni A, Brancale A,<br />

Ferlini C, Dondio G, Varasi M, Mercurio C, Hamel E, Lavia P, Novellino E, Silvestri R.<br />

Design and Synthesis of 2-Heterocyclyl-3-arylthio-1H-indoles as Potent Tubulin<br />

Polymerization and Cell Growth Inhibitors with Improved Metabolic Stability. J Med Chem<br />

<strong>2011</strong>, 54: 8394-406. doi: 10.1021/jm2012886.<br />

Gasparrini F, Molfetta R, Santoni A, Paolini R. Cbl family proteins: balancing FcεRI-mediated<br />

mast cell and basophil activation. Int Arch Allergy Immunol <strong>2011</strong>, 15: 16-26. doi:<br />

10.1159/000322236.<br />

Ardolino M, Zingoni A, Cerboni C, Cecere F, Soriani A, Iannitto ML, Santoni A. DNAM-1<br />

ligand expression on Ag-stimulated T lymphocytes is mediated by ROS-dependent<br />

activation of DNA-damage response: relevance for NK-T cell interaction. Blood <strong>2011</strong>;117:<br />

4778-86. doi: 10.1182/blood-2010-08-300954<br />

Sciumè G, De Angelis G, Benigni G, Ponzetta A, Morrone S, Santoni A, Bernardini G.<br />

CX3CR1 expression defines 2 KLRG1+ mouse NK-cell subsets with distinct functional<br />

properties and positioning in the bone marrow. Blood <strong>2011</strong>, 117: 4467-75. doi:<br />

10.1182/blood-2010-07-297101.<br />

Research Group<br />

Marco Cippitelli, Angela Gismondi,<br />

professors; Giovanni Bernardini, Cristina<br />

Cerboni, Cinzia Fionda, Alessandra Soriani,<br />

Helena Stabile, Alessandra Zingani,<br />

researchers; Maria Luisa Iannitto, PhD student.<br />

118


Area 5: Cellular and molecular immunology<br />

Dissection of Notch signaling-dependent pathways involved in the<br />

progression of T cell leukemia<br />

Isabella Screpanti<br />

Department of Molecular Medicine<br />

℡: +39 06 44700816 - @: isabella.screpanti@uniroma1.it<br />

The role of Notch signaling in the development of T cell leukemia is well established,<br />

whereas its role in promoting and sustaining leukemia progression remains undefined. We<br />

previously showed that the triggering of NF-kB canonical pathway, sustained by the<br />

constitutive expression of preTCR, inducing in turn the activation of several anti-apoptotic<br />

and pro-proliferative signals (Bcl2-A1, IL2, Cyclin D1), appears to exert an essential role<br />

during the expansion phase of the disease.<br />

However, although it is widely known the role of Notch intracellular domain as a<br />

transcriptional activator, a lot remains to be clarified on the mechanisms regulating the<br />

switch-on/off of the signalling. This is particularly important with respect to the possibility to<br />

pharmacologically intervene on the constitutive activation of Notch signaling in T cell<br />

leukemia. We recently identified a specific novel property of Notch3, known to be<br />

overexpressed in T-ALL, that is acetylated and deacetylated by p300 and HDAC1,<br />

respectively. Notch3 acetylation primes ubiquitination and proteasomal-mediated degradation<br />

of the protein. As a consequence, its transcriptional activity is decreased, thus resulting in the<br />

impairment of downstream signaling as well as in vitro T cell proliferation and in vivo growth<br />

of Notch3-induced T cell leukemia in transgenic mice.<br />

Although the acetylatable lysine residues in the RAM domain are evolutionarily<br />

conserved among almost all the Notch proteins from various species, whether the<br />

mechanism, as well as the functional consequences, we described might be extended to<br />

other members of the Notch family and to different cell contexts remain to be further<br />

investigated. Indeed, the Sirt1 loss of function-induced Notch1 acetylation was shown to<br />

sustain its stabilization and to play a positive role in endothelial cells and in in vivo models of<br />

vascular maturation/differentiation, while in our study HDAC inhibition-induced Notch3<br />

acetylation, resulting in its degradation, is inhibitory in human and mouse T-ALL cell lines and<br />

in a in vivo mouse model of T-ALL.<br />

Overall this observations, together with ours, representing the first evidence highlighting<br />

the involvement of a reversible acetylation mechanism in Notch3 protein stability and<br />

function, suggest two possible scenarios: either the cell context determines the acetylationdependent<br />

output of the Notch signaling or the overall Notch signaling is mediated by the<br />

integration of the differential roles of Notch1 and Notch3 acetylation.<br />

We previously reported that, besides its oncogenic/leukemogenic role, Notch3, in<br />

cooperation with the pTalpha/preTCR pathway, positively regulates the generation and<br />

function of naturally occurring regulatory T cells, which have been suggested to facilitate<br />

119


Area 5: Cellular and molecular immunology<br />

tumor development by suppressing protective antitumor immune responses. More recently,<br />

we showed that Notch3 triggers the trans-activation of Foxp3 promoter depending on the T<br />

cell developmental stage. Moreover, we discovered a novel CSL/NF-kB overlaping binding<br />

site within the FoxP3 promoter and demonstrate that the activation of NF-kB, mainly<br />

represented by p65-dependent canonical pathway, plays a positive role in Notch3-dependent<br />

regulation of Foxp3 transcription. Accordingly, the deletion of PKCtheta, which mediates<br />

canonical NF-kB activation, markedly reduces regulatory T cell number and per cell Foxp3<br />

expression in transgenic mice with a constitutive activation of Notch3 signalling.<br />

Collectively, our data indicate that the cooperation among Notch3, PKCtheta and p65/NFkB<br />

subunit modulates Foxp3 expression, adding new insights in the understanding of the<br />

molecular mechanisms involved in regulatory T cell homeostasis and function.<br />

Publications<br />

Palermo R, Checquolo S, Giovenco A, Grazioli P, Kumar V, Campese AF, Giorgi A,<br />

Napolitano M, Canettieri G, Ferrara G, Schininà ME, Maroder M, Frati L, Gulino A, Vacca<br />

A, Screpanti I. Acetylation controls Notch3 stability and function in T-cell leukemia.<br />

Oncogene <strong>2011</strong> Epub. doi: 10.1038/onc.<strong>2011</strong>.533.<br />

Barbarulo A, Grazioli P, Campese AF, Bellavia D, Di Mario G, Pelullo M, Ciuffetta A,<br />

Colantoni S, Vacca A, Frati L, Gulino A, Felli MP, Screpanti I. Notch3 and canonical NFkappaB<br />

signaling pathways cooperatively regulate Foxp3 transcription. J Immunol <strong>2011</strong>,<br />

186: 6199-206. doi: 10.4049/jimmunol.1002136.<br />

De Smaele E, Di Marcotullio L, Moretti M, Pelloni M, Occhione MA, Infante P, Cucchi D,<br />

Greco A, Pietrosanti L, Todorovic J, Coni S, Canettieri G, Ferretti E, Bei R, Maroder M,<br />

Screpanti I, Gulino A. Identification and characterization of KCASH2 and KCASH3, 2 novel<br />

Cullin3 adaptors suppressing histone deacetylase and Hedgehog activity in<br />

medulloblastoma. Neoplasia <strong>2011</strong>, 13: 374-85. doi: 10.1593/neo.101630.<br />

Research Group<br />

Maria Pia Felli, professor; Diana Bellavia,<br />

Antonio F. Campese, researchers; Paola<br />

Grazioli, Gaia Scafetta, post-doc fellows; Maria<br />

Pelullo, Roberta Quaranta, PhD students.<br />

Collaborations<br />

Guido Franzoso, Department of Immunology,<br />

Imperial College of London, UK; Antony J.<br />

Capobianco, Molecular Oncology Research<br />

Program, Division of Surgical Oncology,<br />

University of Miami, USA.<br />

120


Area 5: Cellular and molecular immunology<br />

Predisposing factors in autoimmune diseases: correlation between<br />

common genetic variations and function<br />

Rosa Sorrentino<br />

Department of Biology and Biotechnologies "Charles Darwin"<br />

℡: +39 06 49917706 - @: rosa.sorrentino@uniroma1.it<br />

Many complex disorders share a common ground with the autoimmune diseases that is<br />

the dysregulation of the network controlling the inflammatory response. The main interest of<br />

our research group is the correlation between common genetic polymorphisms and their<br />

functional outcome in models of inflammatory diseases, with a particular emphasis on the<br />

role played by the HLA-B27 molecules in conferring susceptibility to Ankylosing Spondylitis<br />

(AS).<br />

In order to establish the effect of the polymorphisms in the HLA-B27 molecules on the<br />

antigen presentation properties, much work has been done by us and much is still ongoing.<br />

In particular, our research has been firstly focused on the functional differences between two<br />

HLA-B27 alleles differing for a single amino acid (D116H) in the antigen presenting groove<br />

but differentially associated with AS. In this context, a further association between AS and a<br />

gene mapping in the HLA-region has been pinpointed by us. This SNP, rs 1264457, is a<br />

common polymorphism in the HLA-E gene mapping close to the HLA-B locus. This prompted<br />

us to investigate in more details how the expression of the HLA-E molecules is modulated.<br />

These molecules, present on the surface of many cell types, are the only known ligands for<br />

NKG2A and NKG2C that are inhibitory and activating receptors, respectively, present on<br />

Natural Killer and cytotoxic T cells. The engagement of one or the other receptor induces<br />

opposite signals that halt or trigger the killing by these cells. We have firstly analyzed the<br />

expression of the HLA-E molecules in cells of the immune system such as B cells and<br />

monocytes. We have found that their expression is modulated along with cell differentiation.<br />

We are now exploring whether this might have an impact on the defense of differentiated<br />

cells from the killing by cytotoxic T cells.<br />

Meanwhile, along this line of research, we have taken part to a project carried out by<br />

many groups around the world showing that the reported association of a common<br />

polymorphism in the ERAP1, a gene involved in antigen presentation, with the AS, occurs<br />

only in the presence of HLA-B27 since patients who are HLA-B27 negative do not show this<br />

association (Evans et al., Nat Genet <strong>2011</strong>).<br />

Monocytes and the plasticity of their effector functions plays a major role in tuning the<br />

inflammatory response. These aspects have become the focus of a leading interest in our<br />

laboratory as also shown by a recent publication (Paladini et al., Toxicol Sci <strong>2011</strong>). We<br />

pointed our attention on the Vasoactive Intestinal Peptide and its receptors as modulators of<br />

the inflammatory response in monocytes. We have first analyzed the distribution of<br />

121


Area 5: Cellular and molecular immunology<br />

polymorphisms mapping in this gene and found, in agreement with other groups, an<br />

association with several diseases such as Rheumatoid Arthritis, Ankylosing Spondylitis,<br />

Idiopathic Achalasia and, finally, type II diabetes (Paladini et al., Gene Epub <strong>2011</strong>).<br />

Moreover, we have identified a microRNA, 525-5p, as negative regulator of the expression of<br />

this receptor (Cocco et al., PLoS One 2010). Interestingly, this microRNA is upregulated by<br />

LPS and, in turn, down-regulates the expression of VPAC1, tuning down its<br />

counterinflammatory functions when monocytes are exposed to bacterial infections. Work is<br />

in progress to establish the effect of SNPs mapping in the VPAC1 3’UTR region and in the<br />

microRNA 525-5p on the expression of this receptor that plays a major role in the<br />

neuroinflammatory network and is considered an interesting therapeutic target in several<br />

complex diseases.<br />

Publications<br />

Paladini F, Adinolfi V, Cocco E, Ciociola E, Tamburrano G, Cascino I, Lucantoni F, Morano<br />

S, Sorrentino R. Gender-dependent association of type 2 diabetes with the vasoactive<br />

intestinal peptide receptor 1. Gene <strong>2011</strong> Epub. doi: 10.1016/j.bbr.<strong>2011</strong>.03.031.<br />

Paladini F, Cocco E, Potolicchio I, Fazekasova H, Lombardi G, Fiorillo MT, Sorrentino R<br />

Divergent effect of cobalt and Beryllium salts on the fate of peripheral blood monocytes and<br />

T lymphocytes. Toxicol Sci <strong>2011</strong>, 119: 257-69. doi: 10.1093/toxsci/kfq328.<br />

Research Group<br />

Maria Teresa Fiorillo, researcher; Fabiana<br />

Paladini, post-doc fellow; Giorgio Camilli,<br />

Elisa Cocco, Sinem Tuncer, PhD student,<br />

Valentina Tedeschi, student.<br />

Collaborations<br />

Isabella Cascino, CNR, Roma; Alessandro<br />

Mathieu, Università di Cagliari; Andreas<br />

Ziegler, Charité, Freie Universität, Berlin,<br />

Germany; Giovanna Lombardi, King’s<br />

College, London, UK; Rainer A. Bockmann,<br />

University of Erlangen-Nurnberg, Erlangen,<br />

Germany.<br />

122


Area 5: Cellular and molecular immunology<br />

CD28 co-stimulatory molecule as a key regulator of NF-κB<br />

signalling pathway: role of cytoskeleton in coupling CD28 to NF-κB<br />

activation<br />

Loretta Tuosto<br />

Department of Biology and Biotechnology "Charles Darwin"<br />

℡: +39 06 49917595 - @: loretta.tuosto@uniroma1.it<br />

CD28 is an important co-stimulatory receptor for T lymphocytes that following<br />

engagement by its natural ligand B7 may act as a TCR-independent signalling units and<br />

activate a non-canonical NF-κB2-like cascade and the selective transcription of both proinflammatory<br />

and survival genes. We recently found that the actin binding protein filamin A<br />

(FLNa), a large cytoplasmic protein that crosslinks cortical actin, constitutively binds NIK, a<br />

MAP-3 kinase involved in the activation of IKKα and NF-κB, and that both FLNa and NIK are<br />

recruited to the the C-terminal proline rich motif (Y 206 QP 208 YAPP) of CD28, an important<br />

domain also involved in the recruitment of Vav-1.<br />

Starting from these data the aim of the present project was to identify:<br />

1) The molecular basis of FLNa/NIK association and its role in the activation of IKKα and<br />

NF-κB<br />

2) The mechanisms and molecules involved in the recruitment of Vav-1 to the C-terminal<br />

Y 206 QP 208 YAPP motif and their role in CD28 signalling<br />

Role of FLNa/NIK association in the activation of IKKα and NF-κB in CD28 stimulated cells<br />

The activation of IKKα and the non-canonical NF-κB pathway requires NIK. We found<br />

that in both Jurkat T cell lines and primary T cells NIK associated with IKKα and CD28<br />

engagement by B7 significantly induced the kinase activity of NIK-bound IKKα. Mutation of<br />

proline residues in the C-terminal Y 206 QP 208 YAPP motif of CD28 (CD28-3A) failed to induce<br />

NIK-associated IKKα kinase activity.<br />

Altogether, these data evidenced that the C-terminal proline-rich motif of CD28 regulates<br />

the recruitment of both FLNa/NIK complexes, thus leading to NIK/IKKα activation and to the<br />

induction of the non-canonical NF-κB2 pathway.<br />

Identification of a novel interaction between Vav-1 and the type I phosphatidylinositol-4-<br />

phosphate 5-kinase (PIP5KI)<br />

In searching for specific molecules coupling CD28 to Vav-1, we identify PIP5KIα as a<br />

novel binding partner of Vav-1. PIP5KIα belongs to a family of phosphatidylinositol<br />

phosphate kinases, which phosphorylate phosphatidylinositol-4-phosphate in the D5 position,<br />

thus generating phosphatidylinositol 4,5-biphosphate (PIP2) a key lipid that serves as a<br />

substrate of PLC-γ and regulates both the Ca 2+ -calcineurin/NF-AT and PKCθ/NF- κB<br />

123


Area 5: Cellular and molecular immunology<br />

signalling cascades. Vav-1 constitutively associated with PIP5KIα in un-stimulated cells and<br />

also PIP5KIα was recruited to the C-<br />

terminal Y 206 QP 208 YAPP of CD28<br />

following stimulation with B7 (Fig. 1). We<br />

also found that CD28 stimulation by both<br />

B7 and anti-CD28 antibodies induces<br />

PIP5KIα kinase activity.<br />

Fig. 1 - Confocal Analysis of PIP5KIa and actin<br />

recruitment in Jurkat cells (JCH7C17) expressing<br />

CD28WT or CD28Y 191 F or CD28-3A mutants.<br />

The future prospectives will be aimed to characterize the nature of Vav-1/PIP5KIα<br />

interaction and identify the functional role of this association in the context of CD28-mediated<br />

regulation of both Ca 2+ -calcineurin/NF-AT and PKCθ/NF-κB signalling cascades.<br />

Publications<br />

Muscolini M, Montagni E, Palermo V, Di Agostino S, Gu W, Abdelmoula-Soussi S, Mazzoni<br />

C, Blandino G, Tuosto L. The cancer-associated K351N mutation affects the ubiquitination<br />

and the translocation to mitochondria of p53 protein. J Biol Chem <strong>2011</strong>, 286: 39693-702.<br />

doi: 10.1074/jbc.M111.279539.<br />

Muscolini M, Sajeva A, Caristi S, Tuosto L. A novel association between Filamin A and NFkB<br />

Inducing Kinase couples CD28 to Inhibitor of NF-kB kinase a and NF-κB activation.<br />

Immunol Lett <strong>2011</strong>,136: 203-12. doi:10.1016/j.imlet.<strong>2011</strong>.01.011.<br />

Tuosto L. NF-κB family of transcription factors: Biochemical players of CD28 co-stimulation.<br />

Immunol Lett <strong>2011</strong>, 135: 1-9. doi:10.1016/j.imlet.2010.09.005.<br />

Research Group<br />

Cristina Camperio, research yellow; Michela<br />

Muscolini, post-doc fellow; Laura Muzi, PhD<br />

student; Petra Tomassini, graduate student;<br />

Silvana Caristi, technician.<br />

Collaborations<br />

Balbino Alarcón, Centro de Biología Molecular<br />

Severo Ochoa, Universidad Autónoma de<br />

Madrid, Spain; Giovanni Blandino, Laboratorio<br />

di Oncogenomica Traslazionale, Roma;<br />

Ricciarda Galandrini, Dipartimento di<br />

Medicina Sperimentale, Sapienza Università di<br />

Roma.<br />

124


Area 5: Cellular and molecular immunology<br />

Anti-tumor pathways mediated by innate immune responses<br />

Elio Ziparo<br />

Department of Anatomical, Histological, Forensic & Orthopaedic Sciences<br />

℡: +39 06 49766586 - @: elio.ziparo@uniroma1.it<br />

Toll-like receptors (TLRs), membrane-bound pattern recognition receptors (PRRs) that<br />

play a central role in responses to pathogens, have been recently implied in the control of<br />

tumor progression.<br />

Our previous data demonstrated that the synthetic TLR-3 agonist poly(I:C) induces<br />

apoptosis (Paone et al., 2008), accompanied by NF-κB activation and parallel induction of<br />

cytokines and chemokines (Galli et al., 2010), in the prostate cancer cell line LNCaP.<br />

Poly(I:C), by exerting both a direct pro-apoptotic effect on LNCaP cells and an immunemediated<br />

effect due to the recruitment of NK and cytotoxic CD8 cells, might therefore be a<br />

potentially valid therapeutic agent in prostate cancer. However, we observed that TLR3<br />

activation provokes anti-tumour signaling leading to apoptosis of prostate cancer cells<br />

LNCaP and PC3 with much more efficiency in the former than in the second more aggressive<br />

line (Paone et al., 2008). Since PC3 cells represent an androgen-independent cell line, it<br />

would be crucial to render it sensitive to cytotoxic agents. A family of cytosolic PRRs, such as<br />

RNA helicases (retinoic acid inducible gene protein 1, RIG-1 and melanoma differentiationassociated<br />

gene-5, MDA5) (Inohara et al., 2005) and the RNA-dependent protein kinase<br />

PKR, use the mitochondrial-bound MAVS as adapter (Kawai, 2005) and downstream activate<br />

the same protein kinases and transcription factors as TLR-3. It has been shown that the<br />

complex of poly(I:C) and cationic liposome (PIC-liposome) induces tumor growth inhibition<br />

(Fujimura, 2006) or apoptosis (Besch, 2009) and the anti-tumoral effect is mediated by RIG-1<br />

and MDA-5.<br />

An aim of this project is to increase the apoptotic rate of poly (I:C) in the more aggressive<br />

cell line PC3. To this purpose, we transfected poly(I:C) by means of lipofectamine reagent<br />

into PC3 cells to stimulate cytosolic PRR and then evaluated apoptosis rate and the<br />

cytoplasmic molecules involved. We found that the transfection of poly (I:C) at 2 µg/ml in PC3<br />

cells for 24 h induced a cell viability inhibition higher than 80%, (evaluated by MTT assay)<br />

respect to the untreated cells and a parallel increase of PC3 apoptosis up to 40% after<br />

poly(I:C) transfection compared to 10% of apoptosis after extracellularly delivered poly(I:C)<br />

(evaluated by PI staining). Moreover, cleaved form of caspase-3 was clearly upregulated and<br />

caspase-3 inhibitor completely reverted apoptosis caused by transfection of poly(I:C). To<br />

clarify the nature of apoptosis, we used specific inhibitors of caspase-9 and caspase-8, the<br />

mediators of intrinsic and extrinsic apoptosis pathways, respectively. Both inhibitors blocked<br />

PC3 apoptosis, suggesting the involvement of the two apoptotic pathways that now we are<br />

further investigating.<br />

125


Area 5: Cellular and molecular immunology<br />

Thus, we analysed the expression of cytosolic PRR by RT-PCR and Western blot.<br />

Although the constitutive expression of MDA-5 was relatively low in PC3 cells, it is<br />

significantly up-regulated when exposed to PIC-liposome, whereas RIG-1 was absent and<br />

the functional phosphorylated form of PKR was similarly expressed in PC3 cells treated with<br />

poly(I:C)/liposome or not. These data strongly suggest that MDA-5 could play a key role in<br />

anti-tumor effect of poly(I:C)/liposome in PC3 cells and we are performing siRNA-mediated<br />

ablation of MDA-5 to definitely determine its function. To test the involvement of TLR3, we<br />

are going to investigate this issue by the analysis of apoptosis and signalling molecules after<br />

transfection of poly(I:C) in a stable TLR-3 dominant negative PC3 cell line.<br />

Publications<br />

D'Alessio A, Esposito B, Giampietri C, Ziparo E, Pober JS, Filippini A. Plasma membrane<br />

micro domains regulate TACE-dependent TNFR1 shedding in human endothelial cells. J<br />

Cell Mol Med <strong>2011</strong> Epub. doi: 10.1111/j.1582-4934.<strong>2011</strong>.01353.x.<br />

Esposito B, Gambara G, Lewis AM, Palombi F, D'Alessio A, Taylor LX, Genazzani AA,<br />

Ziparo E, Galione A, Churchill GC, Filippini A. NAADP links histamine H1 receptors to<br />

secretion of von Willebrand factor in human endothelial cells. Blood <strong>2011</strong>, 117: 4968-77.<br />

doi: 10.1182/blood-2010-02-266338.<br />

Research Group<br />

Antonio Filippini, professor; Anna Riccioli,<br />

researcher; Claudia Giampietri, post-doc<br />

fellow; Sara Palchetti, PdD student; Fabrizio<br />

Padula, Simonetta Petrungaro, Donatella<br />

Starace, technicians.<br />

Collaborations<br />

Paola De Cesaris, Dipartimento di Medicina<br />

Sperimentale, Università de L’Aquila.<br />

126


Area 6<br />

New antimicrobial and<br />

antiviral agents


Area 6: New antimicrobial and antiviral agents<br />

Peptide effectors of innate immunity<br />

Donatella Barra<br />

Department of Biochemical Sciences "A. Rossi Fanelli"<br />

℡: +39 06 4456663 - @: donatella.barra@uniroma1.it<br />

Peptides with diverse biological activities are particularly abundant in skin secretions of<br />

amphibians. Many of these compounds are pivotal components of the innate immune<br />

response, while others are related to mammalian hormones or neurotransmitters.<br />

We have focused the attention on the peptide components of amphibian skin secretions,<br />

discovering a number of molecules displaying antimicrobial activity, as well as small proteins<br />

with different properties, among which the most interesting is Bv8. Mammalian homologues<br />

of Bv8 have been described also in humans (prokineticin 1/EG-VEGF and prokineticin<br />

2/human Bv8), and found to display chemokine-like activities. These two classes of<br />

molecules can be considered as the effectors of the evolutionary ancient immune system,<br />

having overlapping functions: inactivate invading bacterial, fungal, or viral pathogens and<br />

recruit and activate leukocytes.<br />

Referring to the first issue, we have studied the mechanism of synergism between<br />

temporins, a property described in a previous publication (Mangoni et al., J Biol Chem 2008).<br />

Using NMR and fluorescence spectroscopy, the structures and interactions of temporins in<br />

LPS micelles were investigated providing the first structural insight into the mode of action<br />

and synergism of antimicrobial peptides at the level of the LPS-outer membrane.<br />

On temporin A, to obtain a peptide with a better therapeutic index, we used alanine<br />

scanning analogs to elucidate the contribution of the side chains of each amino acid residue<br />

to the peptide's antimicrobial and hemolytic activity, thus providing the basis for optimizing<br />

the design of temporin-based lead structures for the production of new anti-infective agents.<br />

On temporin L, which displays the strongest antimicrobial activity, but also a high toxicity<br />

on human erythrocytes, we studied the structure-activity relationships of a library of temporin<br />

L derivatives, focusing on the correlation between the α-helix content of the peptides, the<br />

nature of their cationic residues, and their antibacterial/antiyeast/hemolytic activities.<br />

Studies on bombinins, specifically the H2/H4 pair, revealed interesting relationships<br />

between the presence of a single D-amino acid in the sequence of an antimicrobial peptide<br />

and its target microbial cell selectivity/membrane-perturbing activity.<br />

Dr. Mangoni has been invited to act as guest Editor for an issue of Cellular and Molecular<br />

Life Science on “Host-defense peptides: from biology to therapeutic strategies”.<br />

Concerning Bv8/prokineticins, we have studied their selective interaction with the G<br />

protein-coupled receptors, PKR1 and PKR2. Defects in the signalling pathways lead to<br />

various pathologies, in particular the Kallmann syndrome and carcinogenesis. We have<br />

demonstrated the dimerization of the PK2 receptor through in vivo experiments on a modified<br />

yeast cells strain that does not express PK receptors.<br />

129


Area 6: New antimicrobial and antiviral agents<br />

Publications<br />

Bhunia A, Saravanan R, Mohanram H, Mangoni ML, Bhattacharjya S. NMR structures and<br />

interactions of temporin-1Tl and temporin-1Tb with lipopolysaccharide micelles:<br />

mechanistic insights into outer membrane permeabilization and synergistic activity. J Biol<br />

Chem <strong>2011</strong>, 286: 24394-406. doi: 10.1074/jbc.M110.189662.<br />

Coccia C, Rinaldi AC, Luca V, Barra D, Bozzi A, Di Giulio A, Veerman EC, Mangoni ML.<br />

Membrane interaction and antibacterial properties of two mildly cationic peptide<br />

diastereomers, bombinins H2 and H4, isolated from Bombina skin. Eur Biophys J <strong>2011</strong>, 40:<br />

577-88. doi: 10.1007/s00249-011-0681-8.<br />

Grieco P, Luca V, Auriemma L, Carotenuto A, Saviello MR, Campiglia P, Barra D, Novellino<br />

E, Mangoni ML. Alanine scanning analysis and structure-function relationships of the frogskin<br />

antimicrobial peptide temporin-1Ta. J Pept Sci <strong>2011</strong>, 17: 358-65. doi: 10.1002/<br />

psc.1350.<br />

Mangoni ML, Carotenuto A, Auriemma L, Saviello MR, Campiglia P, Gomez-Monterrey I,<br />

Malfi S, Marcellini L, Barra D, Novellino E, Grieco P. Structure-activity relationship,<br />

conformational and biological studies of temporin L analogues. J Med Chem <strong>2011</strong>, 54:<br />

1298-307. doi: 10.1021/jm1012853.<br />

Mangoni ML, Shai Y. Short native antimicrobial peptides and engineered ultrashort<br />

lipopeptides: similarities and differences in cell specificities and modes of action. Cell Mol<br />

Life Sci <strong>2011</strong>, 68: 2267-80. doi: 10.1007/s00018-011-0718-2.<br />

Marsango S, Bonaccorsi di Patti MC, Barra D, Miele R. Evidence that prokineticin receptor 2<br />

exists as a dimer in vivo. Cell Mol Life Sci <strong>2011</strong>, 68: 2919-29. doi: 10.1007/s00018-010-<br />

0601-6, doi: 10.1007/s00018-010-0601-6.<br />

Research Group<br />

Maurizio Simmaco, Giuseppina Mignogna,<br />

professors; M. Luisa Mangoni, Rossella Miele,<br />

Alessandro Paiardini, researchers; Sara<br />

Marsango, Vincenzo Luca, PhD students;<br />

Alessandra Franco, Alessandra Giorgi,<br />

technicians.<br />

Collaborations<br />

Günther Kreil, Institute of Molecular Biology,<br />

Austrian Academy od Sciences, Salzburg,<br />

Austria; Yechiel Shai, Department of Biological<br />

Chemistry, Weizmann Institute, Rehovot,<br />

Israele; Lorenzo Stella, Dipartimento di<br />

Scienze e Tecnologie Chimiche, Università di<br />

Roma Tor Vergata; Lucia Negri, Dipartimento<br />

di Fisiologia e Farmacologia “V. Erspamer”,<br />

Sapienza Università di Roma; Paolo Grieco,<br />

Dipartimento di Chimica Farmaceutica e<br />

Tossicologica, Università di Napoli “Federico II”.<br />

130


Area 6: New antimicrobial and antiviral agents<br />

New pyrrole derivatives of BM 212: a new class of antimycobacterial<br />

agents. Design, synthesis, biological evaluation and study of their<br />

mode of action<br />

Mariangela Biava<br />

Department of Medicinal Chemistry and Technologies<br />

℡: +39 06 49913812 - @: mariangela.biava@uniroma1.it<br />

In 2010, the World Health Organization estimated that one-third of the global population<br />

has been infected with M. tuberculosis (MTB) 1 , thus Tuberculosis still represents a major<br />

challenge toward which research efforts are needed.<br />

We identified a new chemical class of 1,5-diphenyl pyrrole derivatives endowed with potent<br />

antimycobacterial activity whose hit compounds are BM212 (MIC=0.7µg/mL; CC 50 =4µg/mL;<br />

PI(CC 50 /MIC)=5.6) and BM521 (MIC=0.25µg/mL; CC 50 =64µg/mL; PI=256µg/mL). This class of<br />

small molecules exhibited a very good biological profile, better than that shown by streptomycin<br />

and rifampin 2 .<br />

This project was devoted to the synthesis and characterization of new derivatives,<br />

starting from the chemical structures of BM212 and BM521, in the attempt to further improve<br />

both their biological and ADME profile. The syntheses of three chemical classes of BM<br />

derivatives were planned, following both literature and molecular modelling suggestions.<br />

Noteworthy, these 1,5-diphenyl pyrrole analogues were synthesized employing a very easy<br />

synthetic pathway comprised of three steps, that were optimized by us, allowing us to obtain<br />

them quickly and in good yields.<br />

Compounds synthesized till now and belonging to the first and second classes were<br />

tested as previously done for BM 212 and BM 521 and many of them proved to be very<br />

active against MTB.<br />

In particular, BM579, exhibited a very good activity with a MIC= 0.125µg/mL, CC 50 =<br />

>128µg/mL, PI= >1000, higher than that of BM212, BM521 and rifampin, and comparable to<br />

that of Isoniazid.<br />

N NCH 3<br />

N<br />

S<br />

N<br />

S<br />

N<br />

N<br />

N<br />

Cl<br />

H 3 C<br />

H 3 CS<br />

Cl<br />

BM212 BM 521<br />

F<br />

F<br />

BM 579<br />

The last year of the project has been focused on the synthesis of the third class of<br />

compounds, designed according to the results previously obtained. Among them, several<br />

derivatives proved to be very active. Data-sets are, however, still incomplete. In particular<br />

131


Area 6: New antimicrobial and antiviral agents<br />

BM708 (MIC=0.016µg/mL, CC 50 =18.89µg/mL, PI=1180.62) and BM720 (MIC=0.125µg/mL,<br />

CC 50 = 179.26µg/mL, PI=1434.08) proved to be the best ones.<br />

Pursuing the hit-to-lead optimization process, the best compounds of the three classes<br />

were further evaluated for better defining their microbiological profile as well as their ADME<br />

profile. In particular, the LORA (low-oxygen recovery assay) test, running in oxygen deprived<br />

conditions simulating the not-replicating persistent –TB phenotype environment (NRP-TB),<br />

has been used for evaluating the activity of the selected compounds towards persistent<br />

strains, that represent one of the most important problem with respect to current therapies.<br />

All selected derivatives showed good activity with a MIC ranging from 5.7 µg/ml to 7.7 µg/ml 3 .<br />

They also showed both good microsomal stability and good activity against intramacrophagic<br />

mycobacteria 4 . Pharmacokinetic studies for the best derivatives have been planned, and they<br />

are still in progress. Finally, during this last year of project the molecular target for BM212<br />

has been identified 5 , and different approaches for shedding light on the direct interaction of<br />

BM212 with the molecular receptor as well as for elucidating the mechanism of action have<br />

been planned.<br />

In conclusion, we have currently reached all our preset goals on our way toward the<br />

development of a promising class of antimycobacterial compounds.<br />

References<br />

1) WHO Report <strong>2011</strong>.<br />

2) Biava M. et al., J Med Chem. 2008, 51: 3644-3648,.<br />

3) Biava M. et al., manuscript in preparation.<br />

4) a) Biava M. et al., Eur. J. Med. Chem. 2009, 44: 4734–4738; b) Biava M. et al. Bioorg. Med.<br />

Chem. 2010, 18: 8076–8084; c) Biava M. et al. ChemMedChem <strong>2011</strong>, 6: 593-599.<br />

5) La Rosa V. et al., Antimicrob. Agents Chemother. 2012, 56: 324–331.<br />

Publications<br />

Biava M, Porretta GC, Poce G, Battilocchio C, Alfonso S, de Logu A, Manetti F, Botta M.<br />

Developing pyrrole-derived antimycobacterial agents: a rational lead optimization<br />

approach. ChemMedChem <strong>2011</strong>, 6: 593-9. doi: 10.1002/cmdc.201000526.<br />

Research Group<br />

Giulio Cesare Porretta, professor, Giovanna<br />

Poce, post-doc fellow; Claudio Battilocchio,<br />

Salvatore Alfonso, PhD students, Roberto<br />

Torri, Technician.<br />

Collaborations<br />

Raffaello Pompei, Alessandro De Logu, Dip.<br />

di Scienze e Tecnologie Biomediche, Università<br />

di Cagliari; Maurizio Botta, Fabrizio Manetti,<br />

Dip. Farmaco Chimico Tecnologico, Università<br />

di Siena; Edda De Rossi, Dip. di Genetica e<br />

Microbiologia, Università di Pavia; Scott G.<br />

Franzblau, Institute for Tuberculosis Research,<br />

College of Pharmacy, University of Illinois at<br />

Chicago, USA; Eric Rubin, Dep. of<br />

Immunology and Infectious Diseases, Harvard<br />

School of Public Health, Boston, USA.<br />

132


Area 6: New antimicrobial and antiviral agents<br />

New azole derivatives as antiprotozoal agents<br />

Roberto Di Santo<br />

Department of Medicinal Chemistry and Technologies<br />

℡: +39 06 49913150 - @: roberto.disanto@uniroma1.it<br />

Chronic infection with Trypanosoma cruzi is a major cause of morbidity and mortality in<br />

Latin America with as many as 200000 new cases of infection (Chagas disease) occurring<br />

per year. Parasites of the genus Leishmania give rise to a number of different clinical<br />

manifestations. The most severe is visceral leishmaniasis (VL), which is almost always fatal<br />

unless treated. Estimated numbers of new VL cases are 500,000 per annum, with<br />

approximately 50,000 deaths each year, although VL is often not recognized or reported.<br />

Limitations of current drugs useful against Chagas and VL include significant toxicity, high<br />

cost and long treatment courses as well as a lack of efficacy due to the insorgence of drug<br />

resistance. Moreover, no vaccines are available against the above pathogens, up to day.<br />

A series of compounds that were determined to be highly potent inhibitors of the T. cruzi<br />

lanosterol 14α-demethylase (L14DM) enzyme are in preclinical development for Chagas<br />

disease. The same target is also present in Leishmania donovani and has potential to be<br />

exploited for treatment of VLs.<br />

Fig. 1 - Hit compound found as antiprotozoal agent.<br />

A few years ago we started studies on new antifungal agents characterized by imidazole<br />

and pyrrole moieties, exemplified by the structure of RDS 2394 (Fig. 1), that displayed EC 50 s<br />

5-10 nM against C. albicans and Candida spp. The target of these azole antifungal agents is<br />

the L14DM of fungi. Thus, taking account of the presence of L14DMs in protozoa and our<br />

availability of an in house library of azole antifungal compounds, we decided to test RDS<br />

2394 and a few derivatives against a panel of protozoa. RDS 2394 resulted the best hit and<br />

have been proven to bind L14DM of T. cruzi by monitoring the spectral shift of the heme-iron<br />

Soret band.<br />

Main results. During the first year of the project about the research on new azole<br />

derivatives as anti-protozoal agents, we have designed and synthesized a series of RDS<br />

2394 analogues. The first round of compounds was designed based on information coming<br />

form the binding mode of RDS 2394 with T. cruzi L14DM, that was found by the means of<br />

133


Area 6: New antimicrobial and antiviral agents<br />

molecular modelling. As a parallel approach we decided to try to validate the binding mode of<br />

the RDS 2394, by the means of the design of further azole derivative characterized by the<br />

lack of one or more features depicted by the above binding mode. In general, the newly<br />

synthesized compounds were good antiprotozoal agents. The potency of these compounds<br />

is high against T. cruzi, good against P. falciparum, medium against L. donovani, low against<br />

T. brucei. A number of compounds were more potent than the reference compounds, and a<br />

few derivatives were active at nanomolar concentrations.<br />

Nome<br />

T. b. rhod. T. cruzi L. don. axen. P. falciparum<br />

IC 50 (µg/mL) IC 50 (µg/mL) IC 50 (µg/mL) IC 50 (µg/mL)<br />

RC 184 5.56 5.59 5.03 2.12<br />

RC 190 13.9 0.068 6.65 0.046<br />

RC 195 15.3 0.090 6.77 0.026<br />

RC 201 4.03 0.013 1.35 0.133<br />

RC 230 5.98 0.007 2.46 0.131<br />

RC 247 6.89 0.020 2.02 0.136<br />

RDS 416 6.98 0.014 1.52 0.064<br />

RDS 2037 14.9 0.002 2.15 0.072<br />

RDS 2060 34.8 0.153 3.72 0.076<br />

RDS 2087 13.6 0.027 0.89 0.035<br />

RDS 2056 2.72 0.001 1.11 0.289<br />

RDS 2128 15.8 0.001 0.88 0.258<br />

RDS 2137 15.3 0.001 2.18 0.539<br />

RDS 2394 8.92 0.026 0.20 0.119<br />

Melarsoprol 0.03<br />

Benznidazole 0.435<br />

Miltefosine 0.149<br />

Chloroquine 0.105<br />

Table 1 - In vitro potency of the newly synthesized azoles of RDS and RC series<br />

against T. cruzi Tulahuen C4 amastigotes, L. donovani MHOM-ET-67/L82<br />

amastigotes, as well as other protozoa like T. brucei rhodiesiense STIB 900<br />

trypomastygotes and Plasmodium falciparum K1 IEF, Reference compounds are<br />

melarsoprol, benznidazole, miltefosine and RDS 2394.<br />

Perspective. The activity showed by these new azole derivatives led us to hypothesize<br />

that further targets in addition to L14DM could be involved in inhibition of the growth of<br />

protozoa. Efforts will be devoted to determine if farnesyl transferase enzyme is involved as a<br />

further target. Thus the main perspective of our work will be the development of structureactivity<br />

relationships in these series of compounds to better elucidate information useful to<br />

define the mechanism of action of these azoles as well as to further improve the potency<br />

against protozoa of this compounds.<br />

Research Group<br />

Roberta Costi, professor; Giuliana Cuzzucoli<br />

Crucitti, Luca Pescatori, Gaetano Miele,<br />

Alberto Iacovo, post-doc fellows; Federica<br />

Rosi, PhD student.<br />

Collaborations<br />

Luciana Marinelli, Ettore Novellino,<br />

Università di Napoli “Federico II”; Michael<br />

Gelb, Christophe Verlinde, Washington<br />

University, Seattle, USA.<br />

134


Area 6: New antimicrobial and antiviral agents<br />

Drug design and synthesis of non-nucleoside inhibitors of both<br />

HIV-1 wild type and resistant mutant strains reverse transcriptase,<br />

and Coxsackie B4 virus<br />

Romano Silvestri<br />

Department of Drug Chemistry and Technologies<br />

℡: +39 06 49913800 - @: romano.silvestri@uniroma1.it<br />

Acquired immunodeficiency syndrome (AIDS) causes over 5700 deaths every year, and<br />

at the same time over 6800 people become infected with human immunodeficiency virus<br />

(HIV). We obtained new potent non-nucleoside inhibitors of the HIV-1 reverse transcriptase<br />

(NNRTIs) having the indolylarylsulfone (IAS) scaffold by coupling the indolo-2-carboxamide<br />

functionality with natural and unnatural aminoacids and introducing different electronwithdrawing<br />

substituents at position 4 and 5 of the indole nucleus. The new IASs inhibited the<br />

HIV-1 replication in human T-lymphocyte (CEM) cells at low/subnanomolar concentration and<br />

were weakly cytostatic. Against the mutant L100I, K103N, and Y181C RT HIV-1 strains in<br />

CEM cells, these sulfones were comparable to EFV. Most importantly, these new IASs were<br />

inhibitors to Coxsackie B4 virus at low micromolar concentration, that is 25 times lower than<br />

required for ribavirin. The compounds were evaluated against Coxsackie B4 virus in Vero cell<br />

cultures. The antiviral potency was in general less pronounced in Vero than in HeLa cell<br />

cultures. Five compounds showed anti-Coxsackie B4 virus activity in both HeLa and Vero cell<br />

cultures, and for two derivatives the inhibitory concentrations against respiratory syncytial<br />

virus were ~4 µM. The activity against Coxsackie B4 virus seemed to be positively affected<br />

by the presence of the bromine atom at position 5 of the indole and an amino acid unit<br />

bearing a methyl group. The antivirally active compounds showed selectivity index against<br />

Coxsackie B4 that was several orders of magnitude lower than the selectivity index for HIV.<br />

We questioned whether the observed antiviral activity is due to a direct anti-Coxsackie virus<br />

effect or caused by the cytotoxic activity of the compounds. Several substituted<br />

benzimidazole-based structures have been identified in the past to be endowed with anti-<br />

Coxsackie virus activity in cell culture. One of these compounds (TBZE-029) was, like the<br />

present structures, initially discovered as an NNRTI active against HIV- 1. TBZE-029 is active<br />

at an EC 50 value of 1.2 µg/mL, a concentration close to the EC 50 of the most inhibitory IAS<br />

derivatives. The TBZE-029 compound affects viral RNA synthesis by targeting the<br />

nonstructural protein 2C. The most active indole derivatives are under investigation for their<br />

ability to target virus-specific protein to reveal a potential viral target for this novel class of<br />

compounds. Therefore, we believe that this study allowed disclosure of a new lead class of<br />

Coxsackie B4 virus and respiratory syncytial virus inhibitors. Coxsackie B virus may be the<br />

causative agent of viral polymyositis and rhabdomyolysis, a pathological condition that may<br />

be associated with immunodepressed conditions.Such agents may provide an useful starting<br />

point for the development of new effective inhibitors of both HIV-1 RT and Coxsackie B4<br />

virus.<br />

135


Area 6: New antimicrobial and antiviral agents<br />

Recently, a new concept of binding interaction with the HIV-1 drug-resistant mutant<br />

strains emerged: the “flexibility hypothesis”. According to this idea, a powerful inhibition of the<br />

mutant variants of the HIV-1 should be correlated to the ability of the molecule to adopt<br />

variable binding conformations which would be not affected by the interchanges of the amino<br />

acids residues into the NNBS of the mutant RT. Having in mind the newer “horseshoe-like”<br />

model, we synthesized new IAS derivatives bearing cyclic substituents at the indole-2-<br />

carboxamide linked through a methylene ethylene spacer. The new IASs were potent<br />

inhibitors of HIV-1 WT replication in CEM cells and showed inhibitory concentrations in the<br />

low nanomolar range. Against the mutant L100I and K103N RT HIV-1 strains, these<br />

compounds showed antiviral potency superior to that of NVP and EFV and were equipotent<br />

to ETV. The compounds proved to inhibit effectively different HIV clades in PBMC with EC 50<br />

values in the higher picomolar or lower nanomolar range of concentrations. These IASs may<br />

be useful in EFV-based HIV-1 therapies that show the emergence of the L100I and K103N<br />

mutations.<br />

Publications<br />

La Regina G, Gatti V, Piscitelli F, Silvestri R. Open vessel and cooling while heating<br />

microwave-assisted synthesis of pyridinyl N-aryl hydrazones. ACS Comb Sci <strong>2011</strong>, 13: 2-6.<br />

doi: 10.1021/co100015b.<br />

La Regina G, Coluccia A, Brancale A, Piscitelli F, Gatti V, Maga G, Samuele A,<br />

Pannecouque C, Schols D, Balzarini J, Novellino E, Silvestri R. Indolylarylsulfones as HIV-1<br />

non-nucleoside reverse transcriptase inhibitors. New cyclic substituents at the indole-2-<br />

carboxamide. J Med Chem <strong>2011</strong>, 54, 1587-1598. doi: 10.1021/jm101614j.<br />

Samuele A, Bisi S, Kataropoulou A, La Regina G, Piscitelli F, Gatti V, Silvestri R, Maga G.<br />

Mechanism of interaction of novel indolyl arylsulfone derivatives with K103N and Y181I<br />

mutant HIV-1 reverse transcriptase in complex with its substrates. Antiviral Chem Chemoth<br />

<strong>2011</strong>, 22, 107-118, doi: 10.3851/IMP1855.<br />

Research Group<br />

Giuseppe La Regina, researcher; Cesare<br />

Giordano, CNR researcher; Antonio Coluccia,<br />

post-doc fellow; Valerio Gatti, PhD student.<br />

Collaborations<br />

Giovanni Maga, CNR, <strong>Istituto</strong> di Genetica<br />

Molecolare, Pavia; Jan Balzarini, Rega<br />

Institute for Medical Research, K.U. Leuven,<br />

Belgium; Andrea Brancale, Welsh School of<br />

Pharmacy, Cardiff University, UK; José A.<br />

Esté, IrsiCaixa, Hospital Universitari Germans<br />

Trias i Pujol, Universitat Autonoma de<br />

Barcelona, Badalona, Spain.<br />

136


Area 7<br />

Biology of malaria and other<br />

vector-borne diseases


Area 7: Biology of malaria and other vector-borne diseases<br />

Genetic and phenotypic characterization of species and “molecular<br />

forms” of the Anopheles gambiae complex (Diptera: Culicidae),<br />

afrotropical malaria vectors<br />

della Torre Alessandra - Vincenzo Petrarca<br />

Dept of Public Health and Infectious Diseases - Dept of Biology and Biotechnologies “C. Darwin”<br />

℡: +39 06 49694268 - +39 06 49914932<br />

@: ale.dellatorre@uniroma1.it - vincenzo.petrarca@uniroma1.it<br />

In <strong>2011</strong>, we focused on 2 major lines of research, as follows:<br />

1. Analysis of the genetic and ecological differentiation of Anopheles gambiae M and S<br />

molecular forms in areas of high hybridization. M and S populations from the western edge<br />

of the species, where hybridization rates have been show to be much higher than in eastern<br />

regions (>20%), offer the opportunity to clarify several aspects of the speciation process<br />

ongoing within the major Afrotropical malaria vector species. We applied recently developed<br />

diagnostic assays for detection of M- and S-specific pericentromeric SNPs to samples<br />

collected in The Gambia and in Guinea Bissau, and revealed a level of admixture not<br />

observed in the rest of the range (Caputo et al., <strong>2011</strong>). In particular, we found: i)<br />

heterozygous genotypes at each marker, although at frequencies lower than expected under<br />

panmixia; ii) virtually all possible genotypic combinations between markers on different<br />

chromosomes, although genetic association was nevertheless detected; iii) discordant M and<br />

S genotypes at two X-linked markers close to the centromere, suggestive of introgression<br />

and inter-locus recombination. We hypothesized that this geographic area may represent a<br />

secondary contact zone between M and S.<br />

2. Analysis of differentiation of genes involved in post-mating behaviour in the An.<br />

gambiae complex. Proteins involved in reproduction evolve rapidly due to positive selection<br />

resulting from intersexual interaction and sexual conflict. In Drosophila, fast evolution driven<br />

by positive selection has been detected in proteins expressed both in the male accessory<br />

glands (MAGs) and in the female lower reproductive tract (LRT). In A. gambiae, MAGproducts<br />

are transferred to females as a solid mating plug that induces a series of<br />

physiological post-mating responses in females. We have analyzed the molecular evolution<br />

of two clusters of 3 MAG- and 3 LRT- specific genes potentially involved in post-mating<br />

mechanisms in five members of the A. gambiae complex. Two MAG-specific paralog genes<br />

were shown to be highly conserved among species, although one replacement fixed along<br />

the whole geographical distribution of A. arabiensis was found. Moreover, we identified at<br />

third gene clustering with the former two, which is not annotated in the A. gambiae genome,<br />

and showed a high homology with one of the previous genes. Intriguingly, this gene was<br />

found to be highly differentiated among A. melas, A. merus and A. quadriannulatus, due to<br />

positive selective pressure and purifying selection maintaining lineage-specific products<br />

(Mancini et al., <strong>2011</strong>a).The three LRT-specific genes analyzed encode serine-proteases that<br />

are down-regulated after mating, two of which are expressed in the atrium and interact with<br />

the mating plug and one in the spermatheca. The analysis of polymorphisms revealed a high<br />

139


Area 7: Biology of malaria and other vector-borne diseases<br />

level of replacement polymorphisms consistent with relaxed evolutionary constraints of<br />

duplicated genes, allowing to rapidly fix novel replacements to perform new or more specific<br />

functions. Adaptive evolution was detected in several codons of the 3 genes and evidences<br />

of episodic selection were also found. In addition, the structural modelling of these proteases<br />

highlighted some important differences in their substrate specificity, and provided evidence<br />

that a number of sites evolving under selective pressures lie relatively close to the catalytic<br />

triad and/or on the edge of the specificity pocket, known to be involved in substrate<br />

recognition or binding. The observed patterns suggest that these proteases may interact with<br />

factors transferred by males during mating and that differently evolved in independent A.<br />

gambiae lineages (Mancini et al., <strong>2011</strong>b).<br />

Publications<br />

Caputo B, Santolamazza F, Vicente JL, Nwakanma DC, Jawara M, Palsson K, Jaenson T,<br />

White BJ, Mancini E, Petrarca V, Conway DJ, Besansky NJ, Pinto J, della Torre A. The<br />

“Far-West” of Anopheles gambiae molecular forms. PLoS One <strong>2011</strong>, 6:e16415. doi:<br />

10.1371/journal. pone.0016415.<br />

Mancini E, Baldini F, Tammaro F, Calzetta M, Serrao A, George P, Morlais I, Masiga D,<br />

Sharakhov IV, Rogers DW, Catteruccia F, della Torre A. Molecular characterization and<br />

evolution of a gene family encoding for male-specific reproductive proteins in the African<br />

malaria vector Anopheles gambiae. BMC Evol Biol <strong>2011</strong>a, 11: 292. doi: 10.1186/1471-<br />

2148-11-292.<br />

Mancini E, Tammaro F, Baldini F, Via A, Raimondo D, George P, Audisio P, Sharakhov IV,<br />

Tramontano A, Catteruccia F, della Torre A. Molecular evolution of a gene cluster of serine<br />

proteases expressed in the Anopheles gambiae female reproductive tract. BMC Evol Biol<br />

<strong>2011</strong>b, 11: 72. doi: 10.1186/1471-2148-11-72.<br />

Research Group<br />

Marco Pombi, researcher, Beniamino Caputo,<br />

Emiliano Mancini, Federica Santolamazza,<br />

post-doc fellows, Maria Calzetta, research fellow,<br />

Federica Tammaro, PhD student.<br />

Collaborations<br />

Nora J Besansky, University of Notre Dame,<br />

USA; Flaminia Catteruccia, Imperial College,<br />

London, UK; David Conway, MRC, Banjul, The<br />

Gambia; Elena Levashina, CNRS, Strasburg,<br />

France; Paolo Pelosi, Università di Pisa; Joao<br />

Pinto, Universidad de Nova Lisboa, Portugal;<br />

Hilary Ranson, Liverpool School of Tropical<br />

Medicine, UK; N’Fale Sagnon, CNRFP,<br />

Ouagadougou, Burkina Faso; Igor Sharakhov,<br />

Virginia Tech, Blacksburg, USA; Fred Simard,<br />

IRD, Montpellier, France; Anna Tramontano,<br />

Sapienza Università di Roma.<br />

140


Area 7: Biology of malaria and other vector-borne diseases<br />

Immune responses to malaria and autoimmune disorders:<br />

investigating common gene-regulatory networks<br />

David Modiano<br />

Department of Public Health and Infectious Diseases<br />

℡: +39 06 49914933 - @: david.modiano@uniroma1.it<br />

Background<br />

The Fulani mount stronger immune responses to P. falciparum and are less susceptible<br />

to malaria than sympatric populations (Modiano et al., PNAS 1996). The Fulani also show<br />

higher responses to other pathogens, and both their Th1 and Th2 responses are enhanced,<br />

suggesting that their resistance to malaria could result from a generally stronger immune<br />

activation. Indeed, key genes related to T reg cell function are down-regulated in the Fulani<br />

(Torcia et al., PNAS 2008). This disorder of immune homeostasis could be driven by genetic<br />

factors positively selected by P. falciparum and may underlie the higher susceptibility of the<br />

Fulani to diseases with autoimmune pathogenesis. To investigate this hypothesis, we are<br />

conducting a large-scale epidemiological and immunogenetic study supported by the Malaria<br />

Genomic Epidemiology Network (MalariaGEN, Nature 2008).<br />

Genotype data<br />

We genotyped 363 SNPs on 2186 DNA samples from Fulani, Mossi and Rimaibé using<br />

the Sequenom MassArray System. SNPs included polymorphisms involved in susceptibility<br />

and immune response to malaria, as well as SNPs at autoimmunity loci.<br />

Population genetics analysis<br />

Principal component analysis revealed that Mossi and Rimaibé are not genetically distinct<br />

among themselves, whereas the Fulani are a distinct group, in agreement with data on HLA<br />

class I and class II alleles (Modiano et al., Tissue Antigens 2001; Lulli et al., Hum Immunol<br />

2009)<br />

Genetic association analysis<br />

We conducted association analysis with P. falciparum infection as phenotype using<br />

logistic regression adjusted for age, gender and village, both within each ethnic group and in<br />

the all population stratifying by ethnicity. When conducting the analysis in Fulani and Non-<br />

Fulani separately the first observation was that in both populations the strongest signals are<br />

shown by the 5q31 region, which has been linked to P. falciparum blood infection levels<br />

(Rihet et al., AJHG 1998) and actually lie in the IRF1 gene, encoding Interferon Regulatory<br />

Factor-1. These results confirm previous evidence that IRF1 polymorphisms are associated<br />

with the control of P. falciparum infection (Mangano et al., Genes Immun 2008). Among<br />

genes involved in resistance to severe malaria, we noticed association with the ABO locus,<br />

141


Area 7: Biology of malaria and other vector-borne diseases<br />

encoding for the blood group system ABO. Children with blood group O have been reported<br />

to have a lower risk of severe malaria (Fry et al., Hum Mol Genet 2008). Also the DDC locus<br />

shows signals of association, among the Non-Fulani only. The DDC locus encodes for dopa<br />

decarboxylase, which is involved in dopamine and serotonin synthesis and has been recently<br />

associated with severe malaria in a GWAS in The Gambia (Jallow et al., Nat Genet 2009) as<br />

well as linked to malaria refractoriness in mosquitoes (Romans et al., Am J Trop Med 1999).<br />

Among genes related to T regulatory cell function and/or involved in autoimmunity the most<br />

interesting association signals were shown by IL10, CD25 and TGFBR3 SNPs. IL10<br />

polymorphisms have been associated with SLE and IBD (Franke et al., Nat Genet 2008;<br />

Nath et al., Hum Genet 2005) while CD25 polymorphisms have been associated with AIT,<br />

MS, T1D (Xavier et al., Nat Rev Immunol 2008). On the other hand, it has been shown that<br />

the expression of TGFBR3 is lower in T regulatory cells from Fulani than Non-Fulani (Torcia<br />

et al., PNAS 2008). Most of the protective factors against P. falciparum infection that we<br />

identified have a higher frequency in the Fulani, similar to that of Europeans, who have a<br />

higher prevalance of autoimmune diseases.<br />

Publications<br />

Paganotti GM, Gallo BC, Verra F, Sirima BS, Nebié I, Diarra A, Coluzzi M, Modiano D.<br />

Human genetic variation is associated with Plasmodium falciparum drug resistance. J<br />

Infect Dis <strong>2011</strong>, 20411: 1772-8. doi: 10.1093/infdis/jir629.<br />

Rizzo C, Ronca R, Fiorentino G, Mangano VD, Sirima SB, Nèbiè I, Petrarca V, Modiano D,<br />

Arcà B. Wide cross-reactivity between Anopheles gambiae and Anopheles funestus SG6<br />

salivary proteins supports exploitation of gSG6 as a marker of human exposure to major<br />

malaria vectors in tropical Africa. Malar J <strong>2011</strong>, 10: 206. doi: 10.1186/1475-2875-10-206.<br />

Rizzo C, Ronca R, Fiorentino G, Verra F, Mangano V, Poinsignon A, Sirima SB, Nèbiè I,<br />

Lombardo F, Remoue F, Coluzzi M, Petrarca V, Modiano D, Arcà B. Humoral response to<br />

the Anopheles gambiae salivary protein gSG6: a serological indicator of exposure to<br />

Afrotropical malaria vectors. PLoS One <strong>2011</strong>, 6:e17980. doi: 10.1371/journal.pone.<br />

0017980.<br />

Research Group<br />

Pamela Avellino, Valentina Mangano, Cinzia<br />

Rizzo, post-doc fellows.<br />

Collaborations<br />

Dominic Kwiatkowski, Wellcome Trust Center<br />

for Human Genetics, Oxford, UK; Eleanor<br />

Riley, London School of Tropical Medicine and<br />

Hygiene, London, UK; Bienvenu Sirima,<br />

Centre National de Recherche et Formation sur<br />

le Paludisme, Ouagadougou, Burkina Faso.<br />

142


Start up program<br />

Specification and maintenance of<br />

retinal stem cells


Start-up Program<br />

Specification and maintenance of retinal stem cells<br />

Giuseppe Lupo<br />

Department of Biology and Biotechnology "Charles Darwin"<br />

℡: +39 06 49912206 - @: giuseppe.lupo@uniroma1.it<br />

Our work is focused on the molecular mechanisms controlling specification and<br />

maintenance of regional identity in progenitor/stem cells of the mammalian central nervous<br />

system (CNS), with particular interest in retinal and forebrain cell fates. We are using<br />

embryonic stem cells (ESCs) to model the extrinsic signalling systems directing the early<br />

patterning of the developing CNS, and neural stem cells (NSCs) isolated from specific<br />

regions of the foetal CNS to model the intrinsic regulatory systems that maintain positional<br />

identities of neural progenitors following their initial specification.<br />

While several studies have described the production of various neural cell types from<br />

ESCs, there have been few systematic studies of how different regional identities are<br />

established. Using chemically-defined culture conditions, we have found that ESCs can be<br />

efficiently converted into neuroectoderm by Nodal pathway inhibition along with either Bone<br />

Morphogenetic Protein (BMP) inhibition or treatment with Fibroblast Growth Factor (FGF).<br />

However, in the presence of Nodal inhibition, BMP inhibition allows specification towards<br />

anterior neural fates (forebrain/midbrain), while FGF promotes predominantly posterior fates<br />

(hindbrain/spinal cord). Upregulation of endogenous Wnt signalling during differentiation also<br />

contributes to neuroectoderm posteriorization. Based on these results, we have obtained<br />

efficient forebrain specification by inhibition of Nodal, BMP and Wnt pathways and<br />

minimization FGF signalling. Notably, these condition are not permissive for the specification<br />

of retinal progenitor fates. We are currently addressing the signals needed to stir early<br />

forebrain progenitors to retinal fates and our available evidences point to a requirement for<br />

functional Nodal and BMP pathways.<br />

We have also found that NSCs derived from different foetal CNS regions maintain in vitro<br />

transcriptional profiles consistent with their region of derivation. For example, NSCs derived<br />

from the foetal cortex and striatum retain expression of forebrain, but not spinal cord<br />

markers, while the opposite pattern is detectable in spinal cord-derived NSCs. To address<br />

whether these regional identities are irreversibly specified or can be influenced by molecular<br />

cues involved in CNS patterning, region-specific NSCs were treated with Retinoic Acid (RA)<br />

or FGF. We have found that both pathways were functional and elicited transcriptional<br />

responses in different NSC lines. Remarkably, however, each NSC line showed a unique<br />

transcriptional output coherent with its positional character. These results indicate that the<br />

regional identity of NSCs is specified at an early phase of neural development through<br />

mechanisms that can be maintained in vitro even after exposure to exogenous patterning<br />

signals. Experiments are underway in order to elucidate the intrinsic mechanisms controlling<br />

the differential response of region-specific NSCs to RA and FGF.<br />

145


Start-up Program<br />

In collaboration with Prof. Ohta at Kumamoto University, Japan, we have also<br />

investigated the signals supporting proliferation of RSCs located at the periphery of the<br />

mouse retina. Our work has shown a crucial role for the secreted molecule Tsukuchi (TSK),<br />

belonging to the small leucine-rich proteoglycan (SLRP) family, in regulating RSC<br />

proliferation. TSK is expressed in the peripheral retina from embryonic stages to adulthood<br />

and TSK knock-out mice show increased proliferation in this region. Activation of the Wnt<br />

pathway also stimulates RSC proliferation and we found that TSK inhibits Wnt signalling by<br />

competing for Wnt ligands to receptor binding. These results suggest that RSC proliferation<br />

in the peripheral retina is modulated by the antagonistic interaction between TSK and Wnt<br />

ligands and they also raise the interesting possibility that TSK activity may be responsible for<br />

the limited or absent RSC self-renewal in the adult peripheral retina. In order to gain insight<br />

into the molecular mechanisms of TSK action of cell proliferation, we are studying its<br />

expression pattern and function in NSCs derived from the foetal mouse brain, where TSK is<br />

also expressed in vivo. Preliminary real time PCR analyses showed that TSK is transcribed<br />

in NSCs from both the embryonic cortex and striatum and that its expression levels<br />

significantly increase during cell cycle exit and differentiation induced by growth factor<br />

withdrawal, in agreement with TSK role in inhibiting progenitor proliferation. We are currently<br />

analysing expression of TSK protein in proliferating and differentiating NSCs. Furthermore,<br />

we are establishing stable NSC lines where TSK expression is abrogated or enhanced, in<br />

order to determine whether it is necessary and/or sufficient to restrain proliferation of neural<br />

progenitors.<br />

Publications<br />

Ohta K, Ito A, Kuriyama S, Lupo G, Kosaka M, Ohnuma S, Nakagawa S, Tanaka H.<br />

Tsukushi functions as a Wnt signaling inhibitor by competing with Wnt2b for binding to<br />

transmembrane protein Frizzled4. Proc Natl Acad Sci USA <strong>2011</strong>, 108: 14962-7. doi:<br />

10.1073/pnas.1100513108.<br />

Research Group<br />

Nicoletta Carucci, post-doc fellow; Valentina<br />

Taschetta, undergraduate student.<br />

Collaborations<br />

Emanuele Cacci, Maria Elena Miranda<br />

Banos, Dipartimento di Biologia e<br />

Biotecnologie “C. Darwin”, Sapienza Università<br />

di Roma; William Harris, Department of<br />

Physiology, Development and Neuroscience,<br />

Univ. of Cambridge, UK; Roger Pedersen,<br />

Anne McLaren Laboratory for Regenerative<br />

Medicine, Univ. ofCambridge, UK; Kunimasa<br />

Ohta, Dept. of Developmental Neurobiology,<br />

Kumamoto University, Japan.<br />

146


<strong>Istituto</strong> <strong>Pasteur</strong> – Fondazione Cenci Bolognetti<br />

P.le Aldo Moro 5, 00185 Roma - Tel: +39 06 49255625/6/7/8 - Fax: +39 06 49255629<br />

www.istitutopasteur.it - pasteurcenci@uniroma1.it

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!