26.03.2013 Views

Glycobiology of Human Milk Oligosaccharides - Glycom

Glycobiology of Human Milk Oligosaccharides - Glycom

Glycobiology of Human Milk Oligosaccharides - Glycom

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

www.glycom.com<br />

The First International Conference on the<br />

<strong>Glycobiology</strong> <strong>of</strong> <strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong><br />

May 16 & 17, 2011<br />

Tivoli Hotel, Copenhagen, Denmark<br />

BOOK OF ABSTRACTS


www.glycom.com<br />

The First International Conference on the<br />

<strong>Glycobiology</strong> <strong>of</strong> <strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong><br />

May 16 & 17, 2011<br />

Tivoli Hotel, Copenhagen, Denmark<br />

Main topics<br />

Significance <strong>of</strong> Carbohydrates<br />

HMO Metabolism in <strong>Human</strong>s<br />

Microbial Colonization and HMOs metabolism by microbiota<br />

HMO Content & Composition: New Analytical Approaches<br />

Potential <strong>of</strong> HMOs & Principal Components in Health and Disease<br />

Organizing Committee<br />

Pr<strong>of</strong>essor Clemens Kunz, University <strong>of</strong> Giessen, Germany<br />

Pr<strong>of</strong>essor Sharon Donovan, University <strong>of</strong> Illinois, USA<br />

Christoph Röhrig, Judit Kovacs & Susanne Mau, <strong>Glycom</strong> A/S, Denmark<br />

Website: www.glycom.com


P 01 Contents<br />

P 03 Conference program<br />

P 05 Conference dinner<br />

CONTENTS<br />

Contents<br />

Abstracts <strong>of</strong> plenary lectures and oral communications<br />

P 07 CLEMENS KUNZ (Germany): Research on HMOs – A brief historical review<br />

P 08 HUDSON FREEZE (USA): Sweet solution: Sugars for health?<br />

P 09 PEDRO ANTONIO PRIETO et al. (Mexico): <strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong>: from synthesis to clinical tests and collateral<br />

observations<br />

P 10 TADASU URASHIMA et al. (Japan): Possible significance <strong>of</strong> the predominance <strong>of</strong> type 1 oligosaccharides, a human specific<br />

feature, in breast milk<br />

P 11 CLEMENS KUNZ et al. (Germany): In vivo 13C-labeling <strong>of</strong> milk oligosaccharides and their metabolism in infants<br />

P 12 SHARON M. DONOVAN et al. (USA): Transcriptome <strong>of</strong> the human infant intestinal ecosystem<br />

P 13 THIERRY HENNET et al. (Switzerland): The role <strong>of</strong> milk sialyllactose on intestinal bacterial colonization<br />

P 14 DAVID A. MILLS (USA): Nursing our microbiota: Interactions between bifidobacteria and milk oligosaccharides<br />

P 15 MOTOMITSU KITAOKA (Japan): Bifidobacterial enzymes involved in the metabolism <strong>of</strong> human milk oligosaccharides<br />

P 16 DAVID S. NEWBURG et al. (USA): <strong>Human</strong> milk oligosaccharides alter human faecal microbiota during in vitro<br />

fermentation<br />

P 17 DANIEL GARRIDO et al. (USA): Characterization <strong>of</strong> glycosyl hydrolases in bifidobacterium longum subsp. infantis active<br />

on human milk oligosaccharides<br />

P 18 DEVON KAVANAUGH et al. (Ireland): Increased adherence <strong>of</strong> bifidobacterium longum subsp. infantis to HT-29 cells<br />

following exposure to a predominant human milk oligosaccharide<br />

P 19 RUDOLF GEYER et al. (Germany): Strategies for screening and structural characterization <strong>of</strong> HMOs<br />

P 20 CARLITO B. LEBRILLA (USA): High throughput analysis and quantitation <strong>of</strong> free oligosaccharides<br />

P 21 JOHN S. KLASSEN et al. (Canada): Quantifying human milk oligosaccharide-protein interactions in vitro using<br />

electrospray ionization mass spectrometry<br />

P 22 DENNIS BLANK et al. (Germany): Lewis blood group detection from human milk oligosaccharide mass finger prints<br />

P 23 SHUAI WU et al. (USA): The development <strong>of</strong> an annotated structure library for human milk oligosaccharides<br />

P 24 UTE KRENGEL et al. (Norway): Blood group dependence <strong>of</strong> cholera infections<br />

P 25 NORBERT SPRENGER (Switzerland): Sialic acid utilization<br />

P 26 BING WANG (China): Nutritional significance <strong>of</strong> sialic acid in human milk: an essential nutrient for brain development<br />

and cognition<br />

P 27 LARS BODE et al. (USA): <strong>Human</strong> milk oligosaccharides in amebiasis and necrotizing enterocolitis<br />

P 28 SØRGE KELM et al. (Germany): Interactions <strong>of</strong> milk glycoconjugates with siglecs<br />

P 29 JASMINE GRINYER et al. (Australia): Protein-sugar interactions between secreted fluids and pathogens as a protective<br />

mechanism<br />

P 31 Short biographies <strong>of</strong> invited speakers<br />

1


Abstracts <strong>of</strong> posters<br />

CONTENTS<br />

Contents<br />

P 35 WAI YUEN CHEAH et al. (Australia): Role <strong>of</strong> sialic acid in innate immune protection provided by mammalian milk<br />

P 36 SHARON M. DONOVAN et al. (USA): Ascending colonic microbiota composition and SCFA patterns produced from in<br />

vitro fermentation <strong>of</strong> human milk oligosaccharides and prebiotics differ between formula-fed and sow-reared<br />

piglets<br />

P 37 VICTORIA DOTZ et al. (Germany): Oligosaccharide MALDI-MS pr<strong>of</strong>iles in milk and urine from mother-child pairs<br />

P 38 AMR EL-HAWEIT et al. (Canada): <strong>Human</strong> milk oligosaccharides as anti-adhesion candidates for clostridium difficile<br />

toxin<br />

P 39 SABRINA ETZOLD et al. (UK): Structure determination <strong>of</strong> bacterial mucus-binding proteins and their functional role<br />

in adhesion to host glycans<br />

P 40 LEONIDES FERNÁNDEZ et al. (Spain): Breast milk microbiota: is there a relationship with HMOs?<br />

P 41 SURI S. IYER (USA): Tailoring carbohydrates to capture toxins and pathogens<br />

P 42 TAKANE KATAYAMA et al. (Japan): α-L-Fucosynthase that specifically introduces Lewis a/x antigens into type-1/2<br />

chains<br />

P 43 JONATHAN A. LANE et al. (Ireland): A new methodology for screening <strong>of</strong> bacteria-carbohydrate interactions: anti-<br />

adhesive milk oligosaccharides as a case study<br />

P 44 MAGDALENA ORCZYK-PAWIŁOWICZ et al. (Poland): Sialylation and fucosylation <strong>of</strong> human milk α1-acid glycoprotein<br />

during the first two weeks <strong>of</strong> lactation<br />

P 45 MAGDALENA ORCZYK-PAWIŁOWICZ et al. (Poland): The relative amounts <strong>of</strong> fucose is<strong>of</strong>orms in oligosaccharides <strong>of</strong><br />

human milk fibronectin<br />

P 46 KRISTINE A. SCHOLAND et al. (Germany): Effects <strong>of</strong> specific milk oligosaccharides on the expression <strong>of</strong> interleukin-8<br />

and marker enzymes <strong>of</strong> intestinal cell maturation<br />

P 47 KATELYN ZAK et al. (USA): In vivo production <strong>of</strong> fucose-α1, 2-lactose<br />

P 48 BETSY YANG et al. (USA & Taiwan): Sialylated galactosides <strong>of</strong> human milk as inhibitors <strong>of</strong> enterovirus 71 and A<br />

(H1N1) 2009 influenza infections<br />

P 49 ARDYTHE L. MORROW et al. (USA): The oligosaccharide (OS) phenotype <strong>of</strong> preterm infants predicts risk: A potential<br />

indication for HMOS administration?<br />

P 50 MAKSIM NAVAKOUSKI et al. (Russia): Natural antibodies against milk oligosaccharides<br />

P 51 GER T. RIJKERS et al. (The Netherlands): Primary prevention <strong>of</strong> allergic diseases by probiotics: impact <strong>of</strong> HMOs<br />

P 52 The Sponsor: <strong>Glycom</strong><br />

P 53 List <strong>of</strong> participants<br />

2


08.00 Registration & C<strong>of</strong>fee<br />

CONFERENCE PROGRAM<br />

Conference Program<br />

Monday, May 16, 2011<br />

09.00 Welcome by John Theroux, <strong>Glycom</strong> & Clemens Kunz, University <strong>of</strong> Giessen, GERMANY<br />

09.15<br />

09.50<br />

10.25<br />

Significance <strong>of</strong> Carbohydrates<br />

Chairs: Sharon Donovan, Bing Wang<br />

Research on HMOs – A brief historical review<br />

Clemens Kunz, Ph.D. – University <strong>of</strong> Giessen, GERMANY<br />

Sweet Solution: Sugars for Health?<br />

Hudson Freeze, Ph.D. – Sanford-Burnham Medical Research Institute, La Jolla, CA, USA<br />

<strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong>: From Synthesis to Clinical Tests and Collateral Observations<br />

Pedro Antonio Prieto, Ph.D. – Tecnológico de Monterrey, Health Sciences Division, Monterrey, MEXICO<br />

10.50 Break and Poster Viewing & C<strong>of</strong>fee<br />

11.15<br />

11.50<br />

12.25<br />

13.00 Lunch<br />

14.00<br />

14.35<br />

HMO Metabolism in <strong>Human</strong>s<br />

Chairs: David Mills, Norbert Sprenger<br />

Type 1 HMO Predominance: A Specific Feature in <strong>Human</strong> <strong>Milk</strong><br />

Tadasu Urashima, Ph.D. – Obihiro University, Hokkaido, JAPAN<br />

In vivo 13 C-labeling <strong>of</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong> and Metabolism in Infants<br />

Clemens Kunz, Ph.D. – University <strong>of</strong> Giessen, GERMANY<br />

Transcriptome <strong>of</strong> the <strong>Human</strong> Infant Intestinal Ecosystem<br />

Sharon Donovan Ph.D., R.D. – University <strong>of</strong> Illinois, Urbana, USA<br />

Microbial Colonization and HMOs Metabolism by Microbiota<br />

Chairs: Hudson Freeze, Carlito Lebrilla<br />

The Role <strong>of</strong> <strong>Milk</strong> Sialyllactose on Intestinal Bacterial Colonization<br />

Thierry Hennet, Ph.D. – University <strong>of</strong> Zurich, SWITZERLAND<br />

Nursing Our Microbiota: Interactions Between Bifidobacteria and <strong>Milk</strong> <strong>Oligosaccharides</strong><br />

David Mills, Ph.D. – University <strong>of</strong> California, Davis, USA<br />

15.10 Break and Poster Viewing & C<strong>of</strong>fee<br />

15.30<br />

16. 05<br />

16.30<br />

16.45<br />

Bifidobacterial Enzymes Involved in the Metabolism <strong>of</strong> <strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong><br />

Motomitsu Kitaoka, Ph.D. – National Food Research Institute, Ibaraki, JAPAN<br />

<strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong> alter <strong>Human</strong> Fecal Microbiota During In Vitro Fermentation<br />

David S. Newburg, Ph.D. – Boston College, Chestnut Hill, MA, USA<br />

Characterization <strong>of</strong> Glycosyl Hydrolases in Bifidobacterium longum subsp. infantis active on <strong>Human</strong><br />

<strong>Milk</strong> <strong>Oligosaccharides</strong><br />

Daniel Garrido – University <strong>of</strong> California, Davis, USA<br />

Increased Adherence <strong>of</strong> Bifidobacterium longum subsp. infantis to HT-29 cells following Exposure to a<br />

Predominant <strong>Human</strong> <strong>Milk</strong> Oligosaccharide<br />

Devon Kavanaugh – Teagasc Food Research Centre, Cork, National University <strong>of</strong> Ireland, IRELAND<br />

17.00 END OF SESSION<br />

18.00 Meet in Lobby – Leave for Tivoli Gardens at 18.15<br />

19.00 Dinner at Tivoli Gardens (Reception opens at 18.30)<br />

3


08.30<br />

09.05<br />

09.40<br />

CONFERENCE PROGRAM<br />

Conference Program<br />

Tuesday, May 17, 2011<br />

HMO Content & Composition: New Analytical Approaches<br />

Chairs: Tadasu Urashima, Lars Bode<br />

Strategies for Screening and Structural Characterization <strong>of</strong> HMOs<br />

Rudolf Geyer, Ph.D. – University <strong>of</strong> Giessen, GERMANY<br />

High Throughput Analysis and Quantitation <strong>of</strong> Free <strong>Oligosaccharides</strong> in Mammalian <strong>Milk</strong><br />

Carlito Lebrilla, Ph.D. – University <strong>of</strong> California, Davis, USA<br />

Quantifying <strong>Human</strong> <strong>Milk</strong> Oligosaccharide-Protein Interactions In Vitro using Electrospray Ionization<br />

Mass Spectrometry<br />

John Klassen, Ph.D. – University <strong>of</strong> Alberta, CANADA<br />

10.05 Break and Poster Viewing & C<strong>of</strong>fee<br />

10.25<br />

10.40<br />

10.55<br />

Lewis Blood Group Detection from <strong>Human</strong> <strong>Milk</strong> Oligosaccharide Mass Finger Prints<br />

Dennis Blank – University <strong>of</strong> Giessen, GERMANY<br />

The Development <strong>of</strong> an Annotated Structure Library for <strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong><br />

Shuai Wu – University <strong>of</strong> California, Davis, USA<br />

Blood Group Dependence <strong>of</strong> Cholera Infections<br />

Ute Krengel, Ph.D. – University <strong>of</strong> Oslo, NORWAY<br />

11.10 Poster Session & C<strong>of</strong>fee<br />

13.00 Lunch<br />

14.00<br />

14.35<br />

Potential <strong>of</strong> HMOs & Principal Components in Health and Disease<br />

Chairs: Rudolf Geyer, Thierry Hennet<br />

Sialic Acid Utilization<br />

Norbert Sprenger, Ph.D. – Nestlé Research Center, Lausanne, SWITZERLAND<br />

The Nutritional Significance <strong>of</strong> Sialic Acid on Neurodevelopment and Cognition<br />

Bing Wang, Ph.D. – University <strong>of</strong> Sydney, Australia; Xiamen University & Nestlé Research Center, Beijing, P. R. CHINA<br />

15.10 C<strong>of</strong>fee break<br />

15.20<br />

15.55<br />

16.20<br />

16.35<br />

<strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong> in Amebiasis and Necrotizing Enterocolitis<br />

Lars Bode, Ph.D. – University <strong>of</strong> California, San Diego, USA<br />

Interactions <strong>of</strong> <strong>Milk</strong> Glycoconjugates with Siglecs<br />

Sørge Kelm, PhD. – University Bremen, GERMANY<br />

Protein-Sugar Interactions between Secreted Fluids and Pathogens as a Protective Mechanism<br />

Jasmine Grinyer – Macquarie University, Sydney NSW, AUSTRALIA<br />

Summary <strong>of</strong> Conference<br />

Sharon Donovan Ph.D., R.D. – University <strong>of</strong> Illinois, Urbana, USA<br />

Clemens Kunz, Ph.D. – University <strong>of</strong> Giessen, GERMANY<br />

4


CONFERENCE DINNER<br />

Conference Dinner at Tivoli Gardens<br />

The Reception for the Conference Dinner starts on Monday evening at 18.30 in the H. C. Andersen<br />

Castle inside Tivoli Gardens (① on the map). The Dinner will start at 19.00.<br />

To enter Tivoli Gardens you will require the entrance & dinner ticket that you received together with<br />

your conference material if you signed up for the dinner in time.<br />

If you stay at the Tivoli Hotel we recommend that you come to the Lobby at 18.00. To Tivoli Gardens it<br />

is approximately a 10 minute walk from the Hotel.<br />

If you stay at another location, please use the map to find Tivoli Gardens and ask then at the entrance<br />

for the H. C. Andersen Castle (Tivoli Gardens is big and there are several restaurants).<br />

5


CONFERENCE DINNER<br />

Conference Dinner at Tivoli Gardens<br />

Menu<br />

Roasted scallop with lamb’s lettuce and pesto<br />

(Ristet kammusling med vårsalat og pesto)<br />

Fillet <strong>of</strong> beef with mild rose pepper, pancetta and beans<br />

(Oksefilet med mild rosenpeber, pancetta og bønner)<br />

Chocolate cake Marie José with preserved rhubarb and créme Anglaise<br />

(Chokoladekage Marie José med syltede rabarber og creme anglaise)<br />

Wines<br />

Casa Mayor Chile, Chardonnay and Merlot<br />

6


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Abstracts <strong>of</strong> Plenary Lectures & Oral Communications<br />

Research on HMOs – A brief historical review<br />

CLEMENS KUNZ<br />

Institute <strong>of</strong> Nutritional Science, University <strong>of</strong> Giessen, Germany<br />

7<br />

Significance <strong>of</strong> Carbohydrates<br />

The early history <strong>of</strong> human milk oligosaccharides (HMOs) comprises research focusing on chemical milk<br />

composition in general, but also on physiological and nutritional aspects and chemical structure determinations.<br />

At the end <strong>of</strong> the 19th century, human milk and cow’s milk were thought to contain a mixture <strong>of</strong> lactoses<br />

differing strongly in their properties. Between the 1920s and 1950s, it was found that this lactose fraction was<br />

composed <strong>of</strong> a mixture <strong>of</strong> components called “gynolactose”. Their essential feature was the presence <strong>of</strong> nitrogen<br />

and hexosamines. Pediatricians observed that in feces <strong>of</strong> breast-fed infants compared to infants receiving<br />

formula, bifidobacteria were the predominant microorganisms; oligosaccharide fractions containing N-<br />

acetylglucosamine were growth promoting and thus named “bifidus factor”. At the same time, the structures <strong>of</strong><br />

major HMOs such as lacto-N-tetraose, neo-lacto-N-tetraose and their fucosylated derivatives, as well as sialylated<br />

and fucosylated lactose have been identified. The last 30 years have been dominated by the development <strong>of</strong><br />

powerful analytical tools to identify and characterize HMOs and by a vast number <strong>of</strong> functional studies in vitro.<br />

Today, due to the enormous biotechnological progress we are at the beginning <strong>of</strong> a new era focusing on specific<br />

effects <strong>of</strong> HMOs in animals and humans.


Sweet solution: Sugars for health?<br />

HUDSON FREEZE<br />

ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Sanford Children’s Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA<br />

There’s little surprise when sugars, energy and life appear in the same sentence, but sugars are much more than nutrition and<br />

energy. Complex carbohydrates, polysaccharides, glycans, call them what you will, are seldom well known and their<br />

physiological functions are usually underappreciated. Glycan synthesis begins with activation <strong>of</strong> selected monosaccharides<br />

and their localization next to biosynthetic enzymes for assembly, remodeling and recycling in the glycan biosphere. The flat<br />

metabolic chart inadequately reflects the complexity <strong>of</strong> the cellular organization, regulation and balance inherent in these<br />

competing and complimentary homeostatic processes.<br />

The shear complexity <strong>of</strong> grappling with human health drives medicine to define and conceptualize organ systems and<br />

enumerate their pathogenic symptoms. But the fundamental rules <strong>of</strong> basic science respect no artificial constructs. Combine<br />

that fact with an under appreciation <strong>of</strong> the diversity <strong>of</strong> glycan structure and function, and it leaves the current medical system<br />

ill-prepared to deal with glycan based pathologies arising from genetic or environmental insults. Here are a few examples <strong>of</strong><br />

the consequences and complexities <strong>of</strong> faulty monosaccharide biosynthetic pathways.<br />

Congenital disorder <strong>of</strong> glycosylation (CDG) Type Ib (CDG-Ib) patients show failure to thrive, numerous gastrointestinal<br />

problems, hypoglycemia, and liver fibrosis due to a phosphomannose isomerase deficiency (MPI, Fruc-6-P→ Man-6-P).<br />

Providing oral mannose supplements reverse all symptoms except fibrosis because mannose bypasses the deficient step.<br />

Good. In mice, complete loss <strong>of</strong> MPI is embryonic lethal and additional mannose only hastens their demise due to Man-6-P<br />

accumulation and ATP depletion. Not good. Make a hypomorphic mouse line with patient-levels <strong>of</strong> residual MPI activity<br />

(15%) and embryos survive, but adult mice reach old age showing few if any pathologies; obviously not a model for CDG-Ib.<br />

But stress young mice on a high fat diet, and they gain far less weight than their wild-type brothers. That seems a good<br />

outcome. However, give a pregnant hypomorphic mouse mannose supplements in her drinking water and she aborts all the<br />

hypomorphic progeny. Reduce the amount <strong>of</strong> mannose and embryos survive, but half <strong>of</strong> the progeny are blind by 5 weeks <strong>of</strong><br />

age, because <strong>of</strong> abnormal eye development during gestation. So mannose is good, bad, and ugly in different scenarios. What<br />

does this mean for MPI-deficient patients, heterozygous parents or individuals using mannose as a therapy or prevention <strong>of</strong><br />

E. coli-based urinary tract infections? The answer is unknown, but cautious use <strong>of</strong> this simple sugar may be wise.<br />

Some patients who have a fucosylation deficiency due to the loss <strong>of</strong> a GDP-Fucose transporter in the Golgi normalize<br />

their elevated circulating neutrophils within a few days <strong>of</strong> being provided fucose. Other patients with mutations in the same<br />

gene are unresponsive. Experiments in mouse models <strong>of</strong> this disorder show that controlling leukocyte number depends on<br />

appropriate fucosylation <strong>of</strong> proteins in the critical Notch signaling pathway.<br />

Supplements <strong>of</strong> N-acetylglucosamine were reported to reverse serious intestinal pathology some among children with<br />

Crohn’s disease. These findings led to testing oral N-acetylglucosamine as a treatment for T-cell mediated autoimmunity in a<br />

mouse model with good results.<br />

Recently, a deficiency in the ER-localized glucose-6-P’ase3 (G6PC3) was found to cause ER stress and compromise<br />

glycosylation <strong>of</strong> gp91 component <strong>of</strong> the NADPH oxidase, resulting in neutropenia. Moreover, patients with glycogen storage<br />

disorder (GSD) 1b due to a glucose-6-P translocase deficiency show similar neutrophil dysfunction. These results suggest that<br />

glucose homeostasis will be important in not only nutrition, but also in the biosynthesis <strong>of</strong> both N- and O-glycans.<br />

These examples show that manipulating the metabolic flux <strong>of</strong> monosaccharides can have marked influences on protein<br />

glycosylation and health in different organ systems. Basic glycobiology underlies and interconnects multiple medical<br />

disciplines for the benefit <strong>of</strong> the most important stakeholders, the patients and their families.<br />

Supported by The Rocket Fund and a Sanford Pr<strong>of</strong>essorship in <strong>Glycobiology</strong><br />

8


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

<strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong> From Synthesis to Clinical Tests and Collateral Observations<br />

PEDRO ANTONIO PRIETO 1 AND JAMES LEACH 2<br />

1 Tecnológico de Monterrey, Health Sciences Division, Monterrey, Mexico 66250<br />

2 Abbott Nutrition, Columbus, Ohio, United States 43219<br />

During the decade <strong>of</strong> the 90s and the first years <strong>of</strong> the present century, a group <strong>of</strong> scientists and engineers<br />

sponsored by Abbott Laboratories embarked in the endeavor <strong>of</strong> synthesizing and testing human milk<br />

oligosaccharides. By the year 2000 it was possible to synthesize and test kilogram amounts <strong>of</strong> the core<br />

oligosaccharide Lacto-N-neotetraose which, in turn, allowed testing <strong>of</strong> this structure in clinical settings. In<br />

parallel fashion, we explored the production <strong>of</strong> oligosaccharides in the milk <strong>of</strong> transgenic animals, in vitro<br />

prebiotic activity <strong>of</strong> carbohydrates and an extensive analysis <strong>of</strong> human milk samples to ascertain their contents<br />

<strong>of</strong> neutral oligosaccharide structures. Some <strong>of</strong> our findings emerged from fortuitous observations such as the<br />

ability <strong>of</strong> dried yeast to drive oligosaccharide synthesis in the presence <strong>of</strong> suitable glycosyltransferases; others<br />

were predicted from previously published accounts such as the variability <strong>of</strong> oligosaccharide content in human<br />

milk samples. Yet other observations were unexpected and contradicted previously published data such as the<br />

existence <strong>of</strong> oligosaccharide pr<strong>of</strong>iles devoid <strong>of</strong> fucose-containing structures and the ability <strong>of</strong> fucosylated<br />

glycoconjugates to shot down milk production in transgenic rabbits. Taken together, the results that emerged<br />

from this project demonstrate that there are no longer obstacles for the clinical testing <strong>of</strong> certain oligosaccharide<br />

structures and that transgenic expression <strong>of</strong> secondary gene products in milk is possible in some species but not<br />

in others. The present account summarizes the evolution <strong>of</strong> a project from its inception to the evaluation <strong>of</strong><br />

scientifically relevant facts that emerged as byproducts in the quest <strong>of</strong> attaining access to free soluble<br />

carbohydrate structures from human milk.<br />

9<br />

Funded by: Abbott Nutrition, Tecnológico de Monterrey


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

10<br />

HMO Metabolism in <strong>Human</strong>s<br />

Possible significance <strong>of</strong> the predominance <strong>of</strong> type 1 oligosaccharides, a human specific feature,<br />

in breast milk<br />

TADASU URASHIMA 1, SADAKI ASAKUMA 2, MICHAEL MESSER 3, OLAV T. OFTEDAL 4<br />

1 Obihiro University <strong>of</strong> Agriculture & Veterinary Medicine, Obihiro, Hokkaido, Japan. 2 National Agriculture Research<br />

Center for Hokkaido Region, Sapporo, Japan. 3 School <strong>of</strong> Molecular and Microbial Biosciences, The University <strong>of</strong><br />

Sydney, Australia. 4 Smithsonian Environmental Research Center, Edgewater, USA.<br />

<strong>Human</strong> milk and colostrum contain 12 ~ 13 g/L and 22 ~ 24 g/L <strong>of</strong> oligosaccharides, respectively. The chemical<br />

structures <strong>of</strong> 115 human milk oligosaccharides (HMOs) have been characterized to date. We determined the<br />

concentrations <strong>of</strong> 10 neutral and 9 acidic colostrum HMOs collected during the first 3 days <strong>of</strong> lactation, using<br />

reverse phase HPLC after derivatization with 2-aminopyridine or 1-methyl-3-phenyl-5-pyrazolon. 1,2 The<br />

predominant oligosaccharides were found to be Fuc(α1-2)Gal(β1-4)Glc (2'-FL), Fuc(α1-2)Gal(β1-3)GlcNAc(β1-<br />

3)Gal(β1-4)Glc (LNFP1), Fuc(α1-2)Gal(β1-3)[Fuc(α1-4)]GlcNAc(β1-3)Gal(β1-4)Glc (LNDFH1) and Gal(β1-<br />

3)GlcNAc(β1-3)Gal(β1-4)Glc (LNT), the concentration <strong>of</strong> each <strong>of</strong> which was 1 ~ 3 g/L. As these HMOs, other than<br />

2'-FL, all contain Gal(β1-3)GlcNAc (LNB, type 1), we conclude that HMOs containing the type 1 structure<br />

predominate over other those containing Gal(β1-4)GlcNAc (LacNAc, type 2). This appears to be a feature that is<br />

specific to humans since the milk/colostrum <strong>of</strong> other species, including apes and monkeys, either contain only<br />

type 2 oligosaccharides or the type 2 predominate over the type 1. 3,4,5<br />

It was shown in another study that the relative concentration <strong>of</strong> LNT, when compared with other HMOs, is<br />

significantly lower in the feces <strong>of</strong> breast fed infants than in breast milk, suggesting that LNT is metabolized in<br />

preference to other HMOs within the infant colon. 6 In addition, specific oligosaccharides such as Gal(β1-<br />

4)GlcNAc(β1-6)Gal(β1-4)Glc or Gal(β1-4)[Fuc(α1-3)]GlcNAc(β1-6)Gal(β1-4)Glc were found in these feces. These<br />

type 2 oligosaccharides were presumably formed from LNH (Gal(β1-3)GlcNAc(β1-3)[Gal(β1-4)GlcNAc(β1-<br />

6)]Gal(β1-4)Glc) or mon<strong>of</strong>ucosyl LNH (Gal(β1-3)GlcNAc(β1-3){Gal(β1-4)[Fuc(α1-3)]GlcNAc(β1-6)}Gal(β1-<br />

4)Glc) by the release <strong>of</strong> LNB during the passage <strong>of</strong> HMOs through the infant colon, and were relatively resistant<br />

to further degradation.<br />

We hypothesize that LNB released from type 1 in preference to type 2 HMOs by Bifidobacterium bifidum<br />

lacto-N-biosidase can be utilized by other strains <strong>of</strong> bifidobacteria within the colon <strong>of</strong> fed infants. The<br />

predominance <strong>of</strong> type 1 HMOs may therefore promote the growth <strong>of</strong> beneficial colonic bifidobacteria, an effect<br />

which may be stronger than that for type 2 HMOs. Thus acquisition <strong>of</strong> the predominance <strong>of</strong> these prebiotic type 1<br />

HMOs may have had a selective advantage, in terms <strong>of</strong> survival <strong>of</strong> newborn human infants, during human<br />

evolution.<br />

1S. Asakuma et al., Biosci. Biotechnol. Biochem. 71, 1447-1451, 2007.<br />

2S. Asakuma et al., Eur. J. Clin. Nutr. 62, 488-494, 2008.<br />

3T. Urashima et al., In: Comprehensive Glycoscience (J. P. Y. Kamerling ed.), pp. 695-724, Elservier, Amserdam, 2007.<br />

4T. Urashima et al., <strong>Glycobiology</strong> 19, 499-508, 2009.<br />

5K. Goto et al., Glycoconj. J. 27, 703-715, 2010.<br />

6S. Albrecht et al., Electrophoresis 31, 1264-1273, 2010.


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

In vivo 13 C-labeling <strong>of</strong> milk oligosaccharides and their metabolism in infants<br />

CLEMENS KUNZ 1 AND SILVIA RUDLOFF 2<br />

1 Institute <strong>of</strong> Nutritional Science, University <strong>of</strong> Giessen, Wilhelmstrasse 20, 35392 Giessen, Germany<br />

2 Department <strong>of</strong> Pediatrics, University <strong>of</strong> Giessen, Feulgenstrasse 12, 35392 Giessen, Germany<br />

There is increasing evidence that HMOs may be <strong>of</strong> particular importance for the infant. Functions which are<br />

discussed include anti-adhesive and anti-inflammatory effects, an influence on brain development or preventive<br />

effects with regard to certain diseases.<br />

With regard to animal and human studies more knowledge on the metabolic fate <strong>of</strong> HMOs is needed. The use <strong>of</strong><br />

stable isotope ( 13 C)-labelled HMOs facilitates such investigations as it allows the sensitive determination <strong>of</strong> the<br />

13 C-enrichment in biological samples such as feces, blood or urine. The combination <strong>of</strong> isotope ratio mass<br />

spectrometry and other mass spectrometric methods is well suited to answer metabolic questions in humans<br />

and animals. In a previous study, we orally applied 13 C-galactose to lactating mothers investigating whether or<br />

not this monosaccharide was directly used for the biosynthesis <strong>of</strong> the large amounts <strong>of</strong> lactose (50-70 g/L) as<br />

well as <strong>of</strong> oligosaccharides (5-15 g/L) in milk without being metabolized first-pass by the liver. We hypothesized<br />

that it could be <strong>of</strong> advantage to <strong>of</strong>fer galactose instead <strong>of</strong> glucose to the lactating mammary cell because (a) milk<br />

biosynthesis can be turned on to a maximum within few minutes, (b) the amount <strong>of</strong> milk produced by a woman<br />

can reach up to several liters a day and (c) galactose but not glucose is a main component <strong>of</strong> HMOs. As the<br />

resulting in vivo- labelling <strong>of</strong> HMOs was very effective, we subsequently addressed metabolic questions in the<br />

infant by analyzing the 13 C-enrichment in faeces or in urine. The data obtained so far will be compared with the<br />

current knowledge on metabolic aspects.<br />

Interest in metabolic questions with regard to HMOs in infants started about 40 years ago when Lundblad<br />

and co-workers in Lund (Sweden) and Strecker´s group in Lille (France) observed that renal excretion <strong>of</strong><br />

oligosaccharides in an infant was partly influenced by the oligosaccharide pattern <strong>of</strong> the mother’s milk. Today,<br />

questions regarding HMOs which are <strong>of</strong> particular importance in the infant comprise (i) their potential<br />

utilization by the gut microbiota, (ii) physiological processes within the whole digestive tract (from mouth to<br />

colon), (iii) mechanisms <strong>of</strong> intestinal absorption <strong>of</strong> HMOs and (iv) their renal excretion.<br />

Supported by the German Research Foundation (DFG Ru 529/7-3 and Ku 781/8-3).<br />

11


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Transcriptome <strong>of</strong> the <strong>Human</strong> Infant Intestinal Ecosystem<br />

SHARON M. DONOVAN 1, MEI WANG 1, SHUAI WU 2, CARLITO B. LEBRILLA 2, SCOTT L. SCHWARTZ 3,4, IVAN V. IVANOV 3,4,<br />

LAURIE A. DAVIDSON 3, JENNIFER S. GOLDSBY 3, DAVID B. DAHL 5, EDWARD R. DOUGHERTY 6, IDDO FRIEDBERG 7,<br />

DAMIR HERMAN 8 AND ROBERT S. CHAPKIN 2<br />

1 Department <strong>of</strong> Food Science and <strong>Human</strong> Nutrition, University <strong>of</strong> Illinois, Urbana, IL 61801 USA. 2 Department <strong>of</strong><br />

Chemistry, University <strong>of</strong> California, Davis, CA 95616 USA. 3 Program in Integrative Nutrition, Center for<br />

Environmental & Rural Health, 4 Departments <strong>of</strong> Statistics, 5 Veterinary Physiology & Pharmacology, and 6 Electrical<br />

Engineering, Texas A&M University, College Station, TX, 77843 USA. 7 Departments <strong>of</strong> Microbiology and Computer<br />

Science & S<strong>of</strong>tware Engineering, Miami University, Oxford, OH 45056 USA. 8 Winthrop Rockefeller Cancer Institute,<br />

University <strong>of</strong> Arkansas, Little Rock, AR 72204 USA<br />

Our long-term goal is to use non-invasive approaches to define how early nutrition influences intestinal<br />

development and shapes host-microbe interactions in the intestine <strong>of</strong> breast- (BF) and formula (FF) -fed infants.<br />

<strong>Human</strong> milk contains a rich diversity <strong>of</strong> oligosaccharides (HMO) that serve as substrates for fermentation, act as<br />

prebiotics for beneficial bacteria, block the attachment <strong>of</strong> pathogens and modulate immune development <strong>of</strong> the<br />

infant. We have developed a novel molecular methodology that utilizes stool samples containing intact sloughed<br />

epithelial cells to quantify intestinal gene expression pr<strong>of</strong>iles in the developing human neonate (AJP<br />

2010;298:G582-9). Stool samples were collected from 3-month-old FF (n=10) and BF (n=12) infants and gene<br />

expression was assessed by microarray analysis. Linear Discriminant Analysis (LDA) identified single genes and<br />

the two- to three-gene combinations that distinguished the feeding groups. In addition, putative "master"<br />

regulatory genes were identified using Coefficient <strong>of</strong> Determination analysis. Recently, this database was<br />

extended to include breastmilk HMO composition determined by GC/MS, infant stool short chain fatty acids<br />

(SCFA) by GC and infant microbiota composition and gene expression (in a subset <strong>of</strong> samples, n=6/group) by<br />

Roche 454 metagenomic pyrosequencing. 2-Fucosyllactose (2’FL) was predominant in the milk <strong>of</strong> 3 mothers,<br />

whereas lacto-N-tetraose (LNT) predominated in 3 mothers. Total SCFA and propionate concentrations were<br />

greater in stool from FF vs. BF infants. Similar to host mRNA expression, bacterial DNA phylogenetic pr<strong>of</strong>iles<br />

provided strong feature sets that clearly classified FF vs. BF babies. Several quantitative approaches for<br />

integrating host cell gene expression and the bacterial phyla pr<strong>of</strong>iles were applied. First, LDA was used to predict<br />

single gene expression for each <strong>of</strong> the 16,853 host genes using the percentage <strong>of</strong> Firmicutes and Actinobacteria<br />

present in stool as covariates. For 394 genes, all model coefficients were significant (R 2 >0.7; q-values <br />

0.7, q-values


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Microbial Colonization and HMOs Metabolism by Microbiota<br />

The role <strong>of</strong> milk sialyllactose on intestinal bacterial colonization<br />

ANDREA FUHRER 1, NORBERT SPRENGER 2, EKATERINA KURAKEVICH 1, LUBOR BORSIG 1, YEN-LIN HUANG 1,<br />

CHRISTOPHE CHASSARD 3 AND THIERRY HENNET 1<br />

1 Institute <strong>of</strong> Physiology and Center for Integrative <strong>Human</strong> Physiology, University <strong>of</strong> Zurich, Switzerland<br />

2 Nestlé Research Center, Vers-chez-les-Blanc, Lausanne, Switzerland<br />

3 Laboratory <strong>of</strong> Food Biotechnology, Institute <strong>of</strong> Food, Nutrition and Health, ETH Zurich, Switzerland<br />

<strong>Milk</strong> oligosaccharides influence the composition <strong>of</strong> intestinal microbiota and thereby mucosal inflammation.<br />

Recently, we have shown that mice fed on milk deficient for sialyl(2,3)lactose were more resistant to dextran<br />

sulfate sodium (DSS)-induced colitis. By contrast, the exposure to milk containing or deficient for<br />

sialyl(2,3)lactose had no impact on the development <strong>of</strong> mucosal leukocyte populations. The resistance to DSS-<br />

induced colitis was specifically related to sialyl(2,3)lactose since mice exposed to sialyl(2,6)lactose-deficient<br />

milk did not react differently than mice exposed to normal milk. Considering the long term protection <strong>of</strong><br />

sialyl(2,3)lactose-deficient feeding, we have analysed and compared the composition <strong>of</strong> intestinal microbiota in<br />

mice exposed to normal or sialyl(2,3)lactose-deficient milk during lactation. Using temperature-gradient gel<br />

electrophoresis and real-time PCR, we did find that milk sialyl(2,3)lactose mainly affected the colonization <strong>of</strong><br />

the intestine by clostridial cluster IV bacteria. The resulting intestinal microbiota presented different metabolic<br />

properties as shown by changes at the level <strong>of</strong> short-chain fatty acid production in the caecum <strong>of</strong> mice exposed to<br />

either normal milk or sialyl(2,3)lactose-deficient milk. The relationship between intestinal microbiota and the<br />

severity <strong>of</strong> DSS-induced colitis was demonstrated by reconstituting germ-free mice with intestinal microbiota<br />

isolated from mice fed on normal milk or sialyl(2,3)lactose-deficient milk. Germ-free mice harbouring<br />

microbiota from sialyl(2,3)lactose-deficient fed mice were more resistant to DSS-induced colitis than germ-free<br />

mice reconstituted with standard intestinal microbiota. Our study shows that the contact to milk<br />

sialyl(2,3)lactose during infancy affects the bacterial colonization <strong>of</strong> the intestine, thereby influencing the<br />

susceptibility to DSS-induced colitis in adult mice.<br />

Funded by: Zurich Center for Integrative <strong>Human</strong> Physiology, Nestlé<br />

13


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Nursing our Microbiota: Interactions between Bifidobacteria and <strong>Milk</strong> <strong>Oligosaccharides</strong><br />

DAVID A. MILLS<br />

Department <strong>of</strong> Viticulture and Enology, University <strong>of</strong> California Davis, Davis, California, USA<br />

Bifidobacteria are commonly used as probiotics in dairy foods. Select bifidobacterial species are also early<br />

colonizers <strong>of</strong> the breast-fed infant colon, however the mechanism for this enrichment is unclear. We have<br />

previously shown that Bifidobacterium longum ssp. infantis is a prototypical bifidobacterial species that can<br />

readily utilize human milk oligosaccharides as a sole carbon source. Mass spectrometry-based glycopr<strong>of</strong>iling has<br />

revealed that numerous B. infantis strains preferentially consume small mass oligosaccharides, abundant in<br />

human milks. Genome sequencing revealed that B. infantis possesses a bias towards genes required to utilize<br />

mammalian-derived carbohydrates. Many <strong>of</strong> these genomic features encode enzymes that are active on milk<br />

oligosaccharides including a novel 40-kb region dedicated to oligosaccharide utilization. Biochemical and<br />

molecular characterization <strong>of</strong> the encoded glycosidases and transport proteins have further resolved the<br />

mechanism by which B. infantis selectively imports and catabolizes milk oligosaccharides. Expression studies<br />

indicate that many <strong>of</strong> these key functions are only induced during growth on milk oligosaccharides and not<br />

expressed during growth on other prebiotics. In addition, key cell surface oligosaccharide binding proteins in B.<br />

infantis bind both milk oligosaccharides and epithelial cell surface glycans. Moreover, growth on milk<br />

oligosaccharides results in significant increases in binding <strong>of</strong> B. infantis to intestinal cells in vitro. Additional<br />

sequencing <strong>of</strong> numerous B. infantis isolates has confirmed that these genomic features are common among the<br />

infantis subspecies and likely constitute a competitive colonization strategy employed by these unique<br />

bifidobacteria. By detailed characterization <strong>of</strong> the molecular mechanisms responsible, these studies provide a<br />

conceptual framework for bifidobacterial persistence and host-interaction in the infant gastrointestinal tract<br />

mediated, in part, through consumption <strong>of</strong> human milk oligosaccharides.<br />

14


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Bifidobacterial enzymes involved in the metabolism <strong>of</strong> human milk oligosaccharides<br />

MOTOMITSU KITAOKA<br />

National Food Research Institute, National Agriculture and Food Research Organization, Tsukuba, Ibaraki 305-<br />

8642, Japan<br />

Galactosyl-β1,3-N-acetylhexosamine phosphorylase (GLNBP, EC 2.4.1.211), which was first found in a cell free<br />

extract <strong>of</strong> Bifidobacterium bifidum, catalyzes the reversible phosphorolysis <strong>of</strong> galacto-N-biose (Galβ1,3GalNAc,<br />

GNB) and lacto-N-biose I (Galβ1,3GlcNAc, LNB). During the cloning <strong>of</strong> lnpA gene encoding GLNBP from<br />

Bifidobacterium longum subsp. longum, we found a gene cluster encoding the intracellular pathway specific to<br />

GNB and LNB, the GNB/LNB pathway, involving N-acetylhexosamine 1-kinase, UDP-glucose-hexose/HexNAc 1-<br />

phosphate uridilyl transferase, and UDP-glucose/GlcNAc 4-epimerase as well as GLNBP. Both the galactose and<br />

N-acetylhexosamine parts <strong>of</strong> GNB and LNB are converted into the compounds to be entered the glycolytic<br />

pathway. Genes encoding the components <strong>of</strong> GNB/LNB specific transporter are placed upstream <strong>of</strong> the genes<br />

encoding these enzymes.<br />

Since oligosaccharides containing LNB in their structure (type I) are predominant in HMOs, the possession <strong>of</strong><br />

extracellular enzymes that liberate LNB from HMOs by bifidobacteria explains how HMOs promote the<br />

bifidobacterial growth. Such extracellular enzymatic system <strong>of</strong> B. bifidum has been identified including α1,2-<br />

fucosidase, α1,3/4-fucosidase, sialidase, and lacto-N-biosidase.<br />

Bifidobacterial enzymes related to their metabolism <strong>of</strong> HMOs are useful tool for preparing compounds related to<br />

HMOs. For instance LNB and GNB were produced from sucrose and GlcNAc/GalNAc in one-pot using four<br />

bifidobacterial enzyme including GLNBP. From 10 L <strong>of</strong> the reaction mixture, 1.4 kg <strong>of</strong> LNB was isolated as the<br />

crystals. The procedure does not contain any chromatographic techniques and is ready to be scaling-up.<br />

LNB promote growth <strong>of</strong> particular strains <strong>of</strong> bifidobacteria that possess GLNBP gene, but not effective to other<br />

bifidobacteria and most lactic acid bacteria. Most bifidobacterial strains that are usually isolated from feces <strong>of</strong><br />

infants possess the gene, suggesting the function <strong>of</strong> the LNB structure in HMOs for the growth <strong>of</strong> bifidobacteria in<br />

intestines <strong>of</strong> breast-fed infants. On the other hand, Bifidobacterium adolescentis, the major habitant in adult<br />

intestine, does not have GLNBP and does not utilize LNB.<br />

Funded by: The Programme for Promotion <strong>of</strong> Basic and Applied Researches for Innovations in Bio-oriented Industry<br />

15


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

<strong>Human</strong> milk oligosaccharides alter human faecal microbiota during in vitro fermentation<br />

ZHUO-TENG YU AND DAVID S. NEWBURG<br />

Higgins hall - Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA<br />

Aims:<br />

The effect <strong>of</strong> the human milk oligosaccharide fraction (HMOS) on infant fecal microbiota cultured in vitro was<br />

investigated to determine the prebiotic activity <strong>of</strong> HMOS.<br />

Methods:<br />

Slurries <strong>of</strong> infant feces were cultured in the presence or absence <strong>of</strong> HMOS. Differences in the resulting microbiota<br />

culture were measured.<br />

Results:<br />

HMOS supplemented cultures developed significantly lower pH than their unsupplemented negative controls,<br />

and even lower than fructooligosaccharide (FOS) supplemented positive controls. After fermentation, HMOS<br />

supplemented cultures contained higher lactic acid concentrations than the negative control cultures. HMOS<br />

supplemented microbiota contained significantly more Bifidobacteria and Lactobacillus sp. HMOS<br />

supplementation significantly decreased the number <strong>of</strong> E. coli, Clostridium perfringens, and Clostridium difficile. In<br />

vitro adhesion assays in the presence and absence <strong>of</strong> HMOS in the medium demonstrate that HMOS significantly<br />

reduces binding <strong>of</strong> C. perfringens to Caco-2 cells relative to untreated cells, and relative to binding by symbionts<br />

(e.g., bifidobacteria) in the presence <strong>of</strong> HMOS.<br />

Conclusions:<br />

HMOS supplementation <strong>of</strong> infant fecal bacteria in culture stimulate growth <strong>of</strong> Bifidobacteria and Lactobacilli, and<br />

inhibit the growth <strong>of</strong> clostridial pathogens. HMOS also inhibit binding by C. perfringens to intestinal epithelial<br />

cells. These changes in microbiota community composition are highly reminiscent <strong>of</strong> differences in microbiota <strong>of</strong><br />

breastfed infants relative to microbiota <strong>of</strong> prematurely weaned infants.<br />

Significance:<br />

These data suggest that the characteristic breastfed microbiome may be attributed largely to the HMOS fraction<br />

<strong>of</strong> human milk, and that the HMOS fraction <strong>of</strong> human milk is highly prebotic.<br />

Keywords:human microbiota; HMOS; prebiotics; in vitro fermentation; cell adhesion<br />

16


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Characterization <strong>of</strong> Glycosyl Hydrolases in Bifidobacterium longum subsp. infantis active on<br />

<strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong><br />

DANIEL GARRIDO 1,5,6,7, DAVID A. SELA 2,5,6,7, SHUAI WU 3,5,6, ROGELIO JIMENEZ-ESPINOZA 4,7, HYUN-JU EOM 2,5,6,7, J.<br />

BRUCE GERMAN 1,5,6,7, DAVID E. BLOCK 2,4,7, CARLITO B. LEBRILLA 3,5,6,7 AND DAVID A. MILLS 2,5,6,7<br />

Departments <strong>of</strong> 1 Food Science and Technology, 2 Viticulture and Enology, 3 Chemistry and 4 Chemical Engineering,<br />

University <strong>of</strong> California Davis, CA USA; 5 Foods for Health Institute, University <strong>of</strong> California Davis, 6 Functional<br />

<strong>Glycobiology</strong> Program, University <strong>of</strong> California Davis, 7 Robert Mondavi Institute for Wine and Food Sciences,<br />

University <strong>of</strong> California Davis, USA<br />

<strong>Human</strong> milk provides the infant with several protective mechanisms that encourage its proper development and<br />

growth. <strong>Human</strong> milk contains a high concentration <strong>of</strong> complex oligosaccharides, which putatively serve as<br />

prebiotics, favoring the growth <strong>of</strong> specific members <strong>of</strong> the infant gut microbiota. As expected, bacteria residing in<br />

this environment have evolved molecular mechanisms for the utilization <strong>of</strong> these complex substrates. This<br />

includes transport mechanisms and glycosyl hydrolases active on complex oligosaccharides. Bifidobacterium<br />

longum subsp. infantis (B. infantis) is a common member <strong>of</strong> the infant intestinal microbiota previously<br />

characterized by its ability to consume several oligosaccharides found in human milk. The genome <strong>of</strong> this<br />

microorganism revealed various enzymatic systems potentially active on complex host-derived oligosaccharides,<br />

including -fucosidases, -hexosaminidases and -galactosidases. Five genes encode for -fucosidases in B.<br />

infantis. Among these, Blon_0248, Blon_0426 and Blon_2336 were specific for 1-3-fucose linkages, while<br />

Blon_2335 was active on substrates containing 1-2 fucose. The latter two genes were induced by growth on<br />

HMO or lacto-N-tetraose, the most abundant isomer in HMO. Among five genes encoding -galactosidases,<br />

Blon_2016 was induced by LNT and was specific for Galb1-3GlcNAc linkages, abundant in type 1 HMO. On the<br />

other hand Blon_2334 was active on Galb1-4GlcNAc, motif characteristic <strong>of</strong> type 2 HMO. Both enzymes showed<br />

high Kcat values using ONPG. On the other hand, three -glucosaminidases showed activity on HMO with a<br />

general -hexosaminidase activity, but with different kinetic efficiencies. One enzyme, Blon_2355, was induced<br />

by HMO isomers such as LNT. Finally, these results provide a more complete picture <strong>of</strong> how B. infantis is able to<br />

consume host milk oligosaccharides, as well as it identifies a number <strong>of</strong> genes likely associated with<br />

consumption <strong>of</strong> host glycans.<br />

17


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Increased adherence <strong>of</strong> Bifidobacterium longum subsp. infantis to HT-29 cells following exposure<br />

to a predominant human milk oligosaccharide<br />

DEVON KAVANAUGH 1,2,3, LOKESH JOSHI 2,3, MARIAN KANE 2,3 AND RITA M. HICKEY 1,3<br />

1 Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland<br />

2 National Centre for Biomedical Engineering Science, National University <strong>of</strong> Ireland, Galway, Ireland<br />

3 Alimentary Glycoscience Research Cluster, Ireland<br />

The intestinal microbiota is vital to human health and nutrition as evident by its contributions to nutrient supply<br />

through degradation <strong>of</strong> non-digestible food components as well as through the provision <strong>of</strong> colonisation<br />

resistance to enteropathogens and modulation <strong>of</strong> mucosal immunity. Bifidobacteria are a member <strong>of</strong> the<br />

dominant microbiota and can represent up to 80% <strong>of</strong> cultivable bacteria in infants and 25% in adults.<br />

Bifidobacteria have demonstrated a capacity to digest human milk oligosaccharides (HMOs) via specific<br />

adaptations, possess the ability to influence pro-inflammatory immune responses, and have been assessed for<br />

their efficacy in the treatment and prevention <strong>of</strong> many human and animal gastrointestinal disorders.<br />

Furthermore, a recent study employing human milk, in which the fat content was removed; demonstrated that<br />

growth <strong>of</strong> a specific strain <strong>of</strong> Bifidobacterium in the treated human milk resulted in an increased genetic<br />

expression <strong>of</strong> putative type II binding fimbriae which are implicated in bacterial colonisation. As the majority <strong>of</strong><br />

oligosaccharides in breast milk are able to traverse the GI tract and reach the colon undigested, perhaps HMOs<br />

may contribute not only to selective growth <strong>of</strong> commensal bacteria, but to their specific adhesive and colonising<br />

ability, as well. In the current study, selected human-milk derived oligosaccharides and commercially available<br />

prebiotics were assayed for their ability to promote the adhesion <strong>of</strong> Bifidobacterium to the human intestinal cells.<br />

Using these human cell monolayers, we found that pre-incubation <strong>of</strong> Bifidobacterium with some HMOs<br />

significantly enhanced bacterial adhesion compared to other treatments. Putative mechanisms <strong>of</strong> how HMOs<br />

influence host-commensal interactions were investigated and will be presented in the poster.<br />

18


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

HMO Content & Composition: New Analytical Approaches<br />

Strategies for Screening and Structural Characterization <strong>of</strong> HMOs<br />

DENNIS BLANK 1, SABINE GEBHARDT 2, KAI MAASS 1, CLEMENS KUNZ 2 AND RUDOLF GEYER 1<br />

1 Institute <strong>of</strong> Biochemistry, Faculty <strong>of</strong> Medicine, Justus Liebig University <strong>of</strong> Giessen, Giessen, Germany<br />

2 Institute <strong>of</strong> Nutritional Science, Justus Liebig University <strong>of</strong> Giessen, Giessen, Germany<br />

<strong>Human</strong> milk oligosaccharides (HMOs) represent a highly heterogeneous class <strong>of</strong> carbohydrates which are widely<br />

accepted to be beneficial for human milk fed infants because <strong>of</strong> their assumed anti-inflammatory, anti-infective<br />

and immune stimulating properties. The structural heterogeneity <strong>of</strong> HMOs strongly depends on the expression <strong>of</strong><br />

specific glycosyltransferases in correlation to the mother’s Lewis blood group and secretor status. Hence,<br />

detailed information on the structural features <strong>of</strong> these carbohydrates is a prerequisite for future studies on<br />

potential biological functions <strong>of</strong> these glycans.<br />

In order to facilitate structural and/or compositional assignments we have established an experimental protocol<br />

for mass spectrometric pr<strong>of</strong>iling <strong>of</strong> HMOs starting from fifty microliters <strong>of</strong> human milk only which allows a rapid<br />

screening <strong>of</strong> milk samples from different individual donors. Moreover, diagnostically relevant fragment ions <strong>of</strong><br />

selected key compositional species could be identified by tandem mass spectrometry enabling a correlation<br />

between the respective HMO pattern and the Lewis blood group <strong>of</strong> the mother. Based on this information<br />

conclusions on the expression <strong>of</strong> specific carbohydrate epitopes for each individual milk sample can be easily<br />

drawn without the need <strong>of</strong> a blood sample.<br />

Characterization <strong>of</strong> novel HMO structures, however, still requires a different strategy and additional analytical<br />

tools. To this end glycan fractions were fluorescently tagged with 2-aminobenzamide, separated by 2-<br />

dimensional HPLC and analyzed in native as well as permethylated state by different mass spectrometric<br />

techniques. In addition, linkage positions <strong>of</strong> individual monosaccharides were assigned by digestion with specific<br />

exoglycosidases or chemical defucosylation in conjunction with GC/MS linkage analysis. Application <strong>of</strong> this<br />

approach allowed the complete structural characterization <strong>of</strong> two novel HMO isomers, i.e., a difucosylated<br />

octaose and a trifucosylated decaose.<br />

19


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

High throughput analysis and quantitation <strong>of</strong> free oligosaccharides<br />

CARLITO B. LEBRILLA<br />

University <strong>of</strong> California, Davis, CA, USA<br />

Free oligosaccharides in human milk are involved in several functions necessary for the infant’s health and<br />

development. A hallmark <strong>of</strong> these compounds is the large structural diversity, which is compounded by the<br />

complicated structures typical <strong>of</strong> oligosaccharides. For this reason research in this area has been hampered by<br />

the lack <strong>of</strong> rapid methods for structural analysis. Research in our laboratory is focused at developing methods<br />

for the rapid analysis and quantitation <strong>of</strong> milk oligosaccharides. While estimates <strong>of</strong> the number <strong>of</strong><br />

oligosaccharides range into the tens <strong>of</strong> thousands, nan<strong>of</strong>low liquid chromatography analysis <strong>of</strong> milk<br />

oligosaccharides suggests there are only a few hundred structures. A library <strong>of</strong> structure is being developed by<br />

systematically determining the structures <strong>of</strong> a number <strong>of</strong> oligosaccharides. Furthermore, nanoLC with tandem<br />

MS provides a rapid method for structural identification <strong>of</strong> known structures. In this way, hundreds <strong>of</strong><br />

structures can be monitored with complete structures <strong>of</strong> nearly 100 known. By having a library <strong>of</strong> known<br />

structures, biological questions are examined including the role <strong>of</strong> oligosaccharides as prebiotics and as<br />

pathogen blocks.<br />

20


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Quantifying human milk oligosaccharide-protein interactions in vitro using electrospray<br />

ionization mass spectrometry<br />

AMR EL-HAWIET, GLEN SHOEMAKER, ELENA N. KITOVA, RAMBOD DANESHFAR AND JOHN S. KLASSEN<br />

Department <strong>of</strong> Chemistry, University <strong>of</strong> Alberta, Edmonton, AB, Canada<br />

The direct electrospray ionization mass spectrometry (ESI-MS) assay has emerged as a powerful tool for<br />

quantifying the association constants for protein-carbohydrate interactions in vitro. The assay is based on the<br />

direct detection and quantification <strong>of</strong> free and ligand-bound protein ions by ESI-MS for solutions <strong>of</strong> known initial<br />

concentrations <strong>of</strong> protein and ligand. The technique boasts a number <strong>of</strong> strengths, including its simplicity (no<br />

labeling or immobilization required), speed (measurements can usually be completed within 1-2 min), and the<br />

unique ability to provide direct insight into stoichiometry and to study multiple binding equilibria<br />

simultaneously. Additionally, when performed using nan<strong>of</strong>low ESI, which operates at solution flow rates in the<br />

nL/min range, the ESI-MS assay affords high sensitivity, normally consuming picomoles or less <strong>of</strong> analyte per<br />

analysis. A brief overview <strong>of</strong> the ESI-MS assay will be presented, along with recent methodological advances that<br />

overcome the major sources <strong>of</strong> error in the binding measurements. Several examples illustrating the application<br />

<strong>of</strong> the assay for quantifying binding between bacterial proteins (e.g., toxins and adhesins) and human milk<br />

oligosaccharides will be given. A new high-throughput ESI-MS approach for screening carbohydrate libraries<br />

against target proteins will also be described. The “catch and release” ESI-MS assay involves incubating the<br />

target protein with the library <strong>of</strong> carbohydrates, detecting the protein-carbohydrate complexes by ESI-MS,<br />

activating the complexes to release the carbohydrate ligand, followed by fragmentation <strong>of</strong> the ligand. The<br />

identification <strong>of</strong> specific ligands is based on the measured molecular weight and the fragmentation spectrum.<br />

Collision cross section measurements also aid in ligand identification. Several examples highlighting the<br />

application <strong>of</strong> the “catch and release” ESI-MS assay for the discovery <strong>of</strong> carbohydrate ligands for a variety <strong>of</strong><br />

bacterial proteins, including Clostridium difficile toxins A and B, will be presented.<br />

21


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Lewis Blood Group Detection from <strong>Human</strong> <strong>Milk</strong> Oligosaccharide Mass Finger Prints<br />

DENNIS BLANK 1, KAI MAASS 1, VIKTORIA DOTZ 2, SABINE GEBHARDT 2, CLEMENS KUNZ 2 AND RUDOLF GEYER 1<br />

1 Institute <strong>of</strong> Biochemistry, Faculty <strong>of</strong> Medicine, Justus Liebig University <strong>of</strong> Giessen, Friedrichstrasse 24, 35392<br />

Giessen, Germany<br />

2 Institute <strong>of</strong> Nutritional Science, Justus Liebig University <strong>of</strong> Giessen, Wilhelmstrasse 20, 35392 Giessen, Germany<br />

The structural diversity <strong>of</strong> human milk oligosaccharides (HMOs) strongly corresponds with the Lewis (Le) blood<br />

group status <strong>of</strong> the individual donor. The three different Lewis blood groups, i.e., Le(a−b+), Le(a+b−) and<br />

Le(a−b−) exhibit different expression levels <strong>of</strong> fucosyltransferases which result in a great structural variety, in<br />

particular, in the case <strong>of</strong> neutral fucosylated HMO species. These differences in the oligosaccharide patterns are<br />

the basis for our mass spectrometric approach which allows us to assign a milk sample to one <strong>of</strong> the three<br />

groups. Starting from fifty microliters <strong>of</strong> human milk the established method provides a time and material saving<br />

way for a Lewis blood group classification <strong>of</strong> milk samples. The relative abundance <strong>of</strong> diagnostically relevant<br />

compositional species, such as, Hex2Fuc2, Hex3HexNAc1Fuc2 and Hex4HexNAc2Fuc3 in MS pr<strong>of</strong>ile spectra is used<br />

as a first step for the identification. In a second step MS/MS analyses <strong>of</strong> characteristic precursor ions are<br />

performed. For each Lewis blood group, specific mass pr<strong>of</strong>iles and fragment ion patterns could be identified<br />

allowing a rapid classification without the need <strong>of</strong> a blood sample. Furthermore, the outlined protocol can be<br />

used for rapid screening in clinical studies to gain detailed information about neutral as well as acidic<br />

oligosaccharide patterns <strong>of</strong> specific samples, allowing also a relative quantification <strong>of</strong> individual compositional<br />

glycan species. Moreover, the described analytical approach enables an easy quality control <strong>of</strong> milk samples<br />

acquired from milk banks.<br />

22


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

The development <strong>of</strong> an annotated structure library for human milk oligosaccharides<br />

SHUAI WU 1, NANNAN TAO 1, RUDI GRIMM 2, J. BRUCE GERMAN 3 AND CARLITO B. LEBRILLA 1<br />

1 Department <strong>of</strong> Chemistry, University <strong>of</strong> California, Davis, CA, USA . 2 Agilent Technologies, Palo Alto, CA, USA<br />

3 Department <strong>of</strong> Food Science and Technology, University <strong>of</strong> California, Davis, CA, USA<br />

Key Words:<br />

<strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong>; Structure Library; Mass Spectrometry; Exoglycosidase Digestion.<br />

Background and Significance:<br />

<strong>Human</strong> milk oligosaccharides (HMOs) are known as prebiotics stimulating the growth <strong>of</strong> beneficial intestinal bacteria, and as<br />

receptor analogs inhibiting the binding <strong>of</strong> pathogens with cell surface glycans. In addition, the development <strong>of</strong> a balanced<br />

intestinal micr<strong>of</strong>lora system may play an important role in modulating the postnatal immune system. Furthermore,<br />

HMOs involved in intestinal absorption and renal excretion may also enhance mineral absorption and promote<br />

postnatal brain development. Since the absorption, metabolism, and function <strong>of</strong> oligosaccharides have a strong correlation<br />

with their structures, a better understanding <strong>of</strong> HMO structures will provide important insights into their biological<br />

functions. Almost all HMOs have a lactose core at the reducing end, and they show heterogeneity in terms <strong>of</strong> composition,<br />

linkages, and branching resulting in a large variety <strong>of</strong> structures. Elucidating the structures <strong>of</strong> HMOs has remained a<br />

formidable challenge.<br />

Mass spectrometry provides the most sensitive and rapid method for characterizing HMOs. The combination <strong>of</strong> nano-<br />

flow liquid chromatography (nano-LC) and mass spectrometry (MS) provides orthogonal dimensions involving retention<br />

times, accurate masses, and tandem MS. A HMO structure library has been constructed and annotated using nano-<br />

LC/MS/MS system. Together with Agilent database s<strong>of</strong>tware, the HMO library serves as a novel tool for milk research as<br />

it enables users to identify oligosaccharides very easily and quickly by retention time and mass spectral data. Moreover, it<br />

provides an important reference to study the secretor and Lewis status <strong>of</strong> the mother and also the absorption and<br />

metabolism <strong>of</strong> HMOs in their <strong>of</strong>fspring.<br />

Methods and Results:<br />

The HMOs were extracted from pooled milk sample, then reduced with NaBH4 solution, and followed by desalting with solid<br />

phase extraction. Sample pr<strong>of</strong>iling was performed on the HPLC-Chip/TOF MS instrument, which separates HMOs on a nano-<br />

LC column integrated in a micro-chip. The results generated retention times, accurate masses, monosaccharide<br />

compositions, and relative abundances for 224 HMOs. Thirty-two standard milk oligosaccharides were obtained from<br />

commercial sources and introduced into the Chip/TOF under the identical condition. The corresponding structures in the<br />

milk pool were determined by matching the retention time and accurate mass against the standards. For de novo structural<br />

analysis, the sample was separated into fractions using standard HPLC. Each fraction was analyzed by MALDI FT-ICR and<br />

Chip/TOF to identify the major oligosaccharides in the fraction. IRMPD in the FT-ICR were introduced to study the<br />

branching and connectivity <strong>of</strong> the structures. Chip/QTOF provides nano-LC separation with online MS/MS. The<br />

collision energy applied was scaled to the size <strong>of</strong> the oligosaccharides with higher collision energy for larger HMOs. The<br />

fragmentation <strong>of</strong> isomers under identical collision energy generated different tandem mass spectra. The spectra were distinct<br />

and could be used to identify the oligosaccharides in a different sample. The exact linkages were determined by employing<br />

sequential exoglycosidase digestion. So far, 75 known HMO structures yielding a total <strong>of</strong> over 200 entries are included in the<br />

library and input into Agilent database s<strong>of</strong>tware. Accurate mass, retention time, and MS/MS spectra are used to identify all<br />

the oligosaccharide entries.<br />

Funded by: NIH R0 HD061923, NIH R01 HD059127, NIH R01 GM049077<br />

23


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Blood Group Dependence <strong>of</strong> Cholera Infections<br />

ÅSA HOLMNER 1,2, ALASDAIR MACKENZIE 1 AND UTE KRENGEL 1<br />

1 Department <strong>of</strong> Chemistry, University <strong>of</strong> Oslo, Norway<br />

2 Current address: Department <strong>of</strong> Biomedical Engineering & Informatics, Västerbotten County Council, SE-901 85<br />

Umeå, Sweden<br />

The cholera toxin from Vibrio cholerae and the heat-labile enterotoxin from Escherichia coli are major virulence<br />

factors causing cholera and the milder traveler’s diarrhea, respectively. The two toxins share extensive structural<br />

and functional similarities, but they also display distinct characteristics, which find an expression in their<br />

biological function. My group is investigating these two toxins by protein crystallography and complementary<br />

methods. We are interested in elucidating the molecular determinants <strong>of</strong> these toxins, which determine their<br />

specific characteristics. A question that we have recently become especially interested in is the origin <strong>of</strong> the<br />

blood group dependence <strong>of</strong> many infectious diseases, and in particular the molecular basis <strong>of</strong> cholera blood<br />

group dependence (Holmer et al., 2010). Since human blood group antigens are very similar in structure to<br />

human milk oligosaccharides, breast feeding might have a more direct beneficiary effect by binding inhibition, in<br />

addition to stimulating immunological protection.<br />

In response to increases in temperature, sea level and precipitation variability in already exposed regions <strong>of</strong> the<br />

world, water-borne diseases are likely to become an increasing threat. The identification <strong>of</strong> individuals at<br />

particular risk for severe disease will hence become increasingly important for crisis minimization. Furthermore,<br />

understanding <strong>of</strong> the molecular basis <strong>of</strong> blood group dependence may pave the way to the development <strong>of</strong> novel<br />

intervention tools tailor-made for those groups <strong>of</strong> the population that are at highest risk.<br />

Å. Holmner, A. Mackenzie & U. Krengel (2010). Molecular basis <strong>of</strong> cholera blood-group dependence and implications for a world<br />

characterized by climate change. FEBS. Lett. 584, 2548-2555.<br />

24<br />

Funded by: University <strong>of</strong> Oslo


Sialic acid utilization<br />

NORBERT SPRENGER<br />

Nestlé Research Center, Lausanne, Switzerland<br />

ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Potential <strong>of</strong> HMOs & Principal Components in Health and Disease<br />

Postnatal mammalian development encounters milk as a key environmental variable and yet the sole nutrient<br />

source. <strong>Milk</strong> is generally considered to have co-evolved with the developmental needs <strong>of</strong> the suckling newborn.<br />

One evolutionary conserved constituent <strong>of</strong> milk is sialic acid generally displayed on glyco-conjugates and free<br />

glycans. During postnatal development high sialic acid need was proposed to be unmet by the endogenous sialic<br />

acid synthetic capacity. Hence, milk sialic acid was proposed to serve as conditional nutrient for the newborn.<br />

Using a neonatal rat model, we revisited the relationship between the sialic acid content <strong>of</strong> milk and sialic acid<br />

metabolism in gut, liver and brain, the major sites <strong>of</strong> sialic acid synthesis and display. At the other end <strong>of</strong><br />

ontogeny, reduced sialylation in brain, saliva and immune system is observed in the elderly. Thus, in analogy to<br />

the neonates the hypothesis that endogenous synthetic capacity cannot keep up with the need in this age group.<br />

Using an aged rat model, we explored sialic acid display and synthesis in gut, brain and liver, and further<br />

investigated the enteric nervous system and stimulated salivation function to assess neuronal function. The data<br />

propose a functional dietary role <strong>of</strong> sialic acid as a building block for sialylation and beyond.<br />

25


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Nutritional Significance <strong>of</strong> Sialic Acid in <strong>Human</strong> <strong>Milk</strong>: an Essential Nutrient for Brain<br />

Development and Cognition<br />

BING WANG 1,2,3<br />

1 <strong>Human</strong> Nutrition Unit, University <strong>of</strong> Sydney, NSW 2006 Australia<br />

2 School <strong>of</strong> Medicine, Xiamen University, Xiamen 361005, P. R. China<br />

3 Nestlé Research Centre Beijing, P. R. China<br />

Sialic acid, a family <strong>of</strong> 9-carbon acidic sugar molecules, are key monosaccharide units in brain gangliosides and<br />

glycoproteins, including the polysialic acid (polySia) glycotope on neural cell adhesion molecules (NCAM).<br />

<strong>Human</strong> milk is one <strong>of</strong> nature’s richest sources <strong>of</strong> sialic acid (~1 g/L) and cow’s milk based infant formulas<br />

contain very little amounts (0~0.25g/L) (1). Gangliosides and polysialylated NCAM in the brain have an<br />

important role in cell-to-cell interactions, neuronal outgrowth, modifying synaptic connectivity, and memory<br />

formation. The alpha 2,8 sialyltransferase IV (ST8SiaIV) is one <strong>of</strong> two key enzymes for synthesizing polySia on<br />

NCAM. In rodents, the level <strong>of</strong> NCAM polysialylation increases with learning behavior. The liver can synthesise<br />

sialic acid from glucose, but the activity <strong>of</strong> the limiting enzyme, UDP-N-acetylglucosamine-2-epimerase (GNE) is<br />

low during the neonatal period. An exogenous source <strong>of</strong> sialic acid may be critical under conditions <strong>of</strong> extremely<br />

rapid brain growth, particularly during the first months after birth. We have now demonstrated that dietary<br />

sialic acid supplementation increased the levels <strong>of</strong> sialic acid in neural tissues, leading to enhanced learning and<br />

memory in piglets, and to up-regulate expression <strong>of</strong> two learning related genes, Gne and ST8SiaIV (1,2). Global<br />

gene transcription pr<strong>of</strong>iling also supports dietary sialic acid supplementation has effects on brain development<br />

and cognition in piglets. We have also found that sialic acid concentration in the frontal cortex <strong>of</strong> breastfed<br />

infants is higher than the levels <strong>of</strong> formula-fed infants (1). Taken together, milk sialic acid may be a limiting<br />

nutrient in the neonatal period, facilitating optimal cognitive development in young animals. An exogenous<br />

source <strong>of</strong> sialic acid enhances brain development, providing a mechanism to explain the link between breast-<br />

feeding and higher intelligence.<br />

References:<br />

1) Wang B. Annu. Rev. Nutr 2009;29:177.<br />

2) Wang B et al. Am J Clin Nutr. 2007;85:561-9.<br />

26


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

<strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong> in Amebiasis and Necrotizing Enterocolitis<br />

EVELYN JANTSCHER-KRENN 1,2, MONICA ZHEREBTSOV 2, TINEKE LAUWAET 3, CAROLINE NISSAN 1,2, KERSTIN GOTH 4,<br />

YIGIT GUNAR 4, ANATOLY GRISHIN 4, HENRI FORD 4, SHARON REED 3, FRANCES GILLIN 3 AND LARS BODE 1,2<br />

1 Division <strong>of</strong> Neonatology, 2 Division <strong>of</strong> Gastroenterology and Nutrition, Department <strong>of</strong> Pediatrics, 3 Department <strong>of</strong><br />

Pathology, University <strong>of</strong> California, San Diego, 4 Saban Research Institute, University <strong>of</strong> Southern California, USA<br />

<strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong> (HMO) are highly abundant in human milk but not in infant formula, which<br />

triggers the questions whether and how HMO benefit the breast-fed infant. Breast-fed infants are at lower risk to<br />

develop such devastating disorders as amebiasis or necrotizing enterocolitis (NEC), and we hypothesized that<br />

HMO contribute to the beneficial effects <strong>of</strong> breast-feeding in the context <strong>of</strong> these disorders.<br />

Amebiasis, caused by the protozoan Entamoeba (E.) histolytica, is the third leading cause <strong>of</strong> death by parasitic<br />

diseases, surpassed only by malaria and schistosomiasis. Worldwide, approximately 50 million people are<br />

infected with E. histolytica, resulting in nearly 100,000 deaths annually. E. histolytica resides in the colon where<br />

it uses a specific Gal/GalNAc lectin to attach to and destroy the host’s epithelial cells. Here, we show that specific<br />

HMO but also Galactooligosaccharides (GOS) prevent E. histolytica attachment and cytotoxicity. These results<br />

suggest that HMO contribute to the lower incidence <strong>of</strong> amebiasis in breast-fed infants compared to formula-fed<br />

infants. The results also suggest the use <strong>of</strong> safe, inexpensive and readily available GOS as a novel agent to treat or<br />

even prevent amebiasis.<br />

Necrotizing Enterocolitis (NEC) is one <strong>of</strong> the most common and <strong>of</strong>ten fatal intestinal disorders in preterm<br />

infants. Almost 10% <strong>of</strong> very-low-birth-weight infants (


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Interactions <strong>of</strong> milk glycoconjugates with siglecs<br />

HENDRIK KOLIWER-BRANDL 1, NADJA SIEGERT 2, ALEXANDER TOLKACH 2, ULRICH KULOZIK 2 AND SØRGE KELM 1<br />

1 Centre for Biomolecular Interactions Bremen, Department <strong>of</strong> Biology and Chemistry, University Bremen, 28334<br />

Bremen, Germany. 2 Institute for Food Process Engineering and Dairy Science, Technische Universität München,<br />

Weihenstephaner Berg 1, 85354 Freising-Weihenstephan, Germany<br />

Siglecs are a family <strong>of</strong> sialic aid-binding immunoglobulin-like lectins occurring mainly on cells <strong>of</strong> the immune<br />

system. Their main functions are probably in the regulation <strong>of</strong> processes leading to the activation <strong>of</strong> these cells [1] .<br />

Consistent with this hypothesis is the observation that most siglecs have tyrosine-based inhibitory motifs<br />

(ITIMs) within their cytoplasmic domains. Probably their sialic acid-binding activities play important roles in<br />

these processes. Of particular interest in this context is the interplay <strong>of</strong> cis-interactions with glycoconjugates on<br />

their own cell surfaces with trans-interactions with glycoconjugates on opposing cells and in the extracellular<br />

space. Therefore, sialylated compounds like oligosaccharides, glycoproteins and glycolipids like those occurring<br />

in human and animal milk have the potential to modulate siglec-regulated activation processes in the immune<br />

system.<br />

We have used ELISA-like hapten-inhibition assays [2] to investigate the interactions <strong>of</strong> milk glycoconjugates<br />

with Siglec-2, preferentially binding to 2,6-linked sialic acids, and with Siglec-4, which binds preferentially to 2,3-<br />

linked sialic acids. As possible sources for bioactive sialylated structures we looked at complex and crude<br />

mixtures from milk fractionation processes. The complementary linkage specificities <strong>of</strong> these lectins allow the<br />

determination <strong>of</strong> both 2,3- and 2,6-linked sialic acids even in the presence <strong>of</strong> high lactose concentrations, as they<br />

are common in milk products [3] . Using these assays, the presence <strong>of</strong> bioactive sialoglycoconjugates has been<br />

determined in fractions from bovine milk.<br />

Whereas whole milk from local dairy showed no inhibition, samples from milk fractionation processes like<br />

buttermilk, skim milk, lactose-reduced serum, whey proteins as well as caseins and GMP (glycomacropeptides)<br />

were significantly inhibitory, indicating a higher Sia content in these samples. A loss <strong>of</strong> inhibitory, active<br />

sialoglycoconjugates during fractionation processes was observed for buttermilk samples treated at different<br />

temperatures. Particularly, low pH-values after heat treatment appear to be possible reasons for loss <strong>of</strong> activity.<br />

In summary, the interaction with Siglec-4 was less sensitive to temperature- and pH-treatment <strong>of</strong> the samples,<br />

whereas Siglec-2 has lost its inhibition potency completely. The loss <strong>of</strong> Siglec-2 binding activity could be caused<br />

either by a selective hydrolysis <strong>of</strong> the α2,6-linkage <strong>of</strong> sialic acids or by conformational changes in the<br />

glycoconjugate carrier molecules leading to sterically less favored presentations <strong>of</strong> sialylated glycans serving as<br />

recognition determinants.<br />

[1]Crocker PR, Paulson JC, Varki A. Siglecs and their roles in the immune system. Nat Rev Immunol. 2007 Apr;7(4):255-66.<br />

[2]Bock, N., & Kelm, S. (2006). Binding and inhibition assays for Siglecs. Methods in Molecular Biology, 347, 359–375.<br />

[3]Koliwer-Brandl, H., Siegert, N., Umnus, K., Kelm, A., Tolkach, A., Kulozik, U., Kuballa, J., Cartellieri, S., & Kelm, S. (2011). Lectin<br />

inhibition assays for the analysis <strong>of</strong> bioactive milk sialoglycoconjugates. International Dairy Journal, in press.<br />

Funding by: German Federal Ministry for Education and Research (BMBF, project BioChangePLUS 031632A), the Tönjes-Vagt<br />

28<br />

Foundation (project XXI)


ABSTRACTS OF PLENARY LECTURES & ORAL COMMUNICATIONS<br />

Protein-sugar interactions between secreted fluids and pathogens as a protective mechanism<br />

JASMINE GRINYER 1,2, WAI YUEN CHEAH 1,2, DANIEL KOLARICH 1,3 AND NICOLLE PACKER 1,2<br />

1 Department <strong>of</strong> Chemistry and Biomolecular Sciences, Macquarie University, Sydney NSW, Australia 2109<br />

2 Biomolecular Frontiers Research Centre, Macquarie University, Sydney NSW, Australia 2109<br />

3 Max Planck Institute <strong>of</strong> Colloids and Interfaces, Department <strong>of</strong> Biomolecular Systems, Glycoproteomics Group,<br />

Arnimallee 22, 14195 Berlin, Germany<br />

The oral environment, comprising the mouth, teeth, tongue and buccal cells, is home to many different bacterial<br />

species. Streptococcus gordonii is an early coloniser <strong>of</strong> tooth enamel, whereas S. mutans colonises later to cause<br />

dental caries. Bacterial lectins play a role in the initial attachment and bi<strong>of</strong>ilm formation by binding to<br />

glycoproteins on the saliva-coated enamel <strong>of</strong> teeth. The human innate immune system has several mechanisms<br />

for preventing bacterial colonisation on teeth. One such mechanism is thought to be provided by the<br />

glycoproteins in saliva and human milk by protecting the oral cavity from streptococcal species by binding to and<br />

removing these bacteria when swallowed. The removal <strong>of</strong> bacteria from the oral environment is thus in constant<br />

competition with bacterial binding and infection <strong>of</strong> the oral cavity.<br />

We have developed two techniques to monitor bacterial binding, one using fluorescent microscopy to visualise<br />

bacterial attachment to saliva-coated hydroxyapatite, and a fluorescent assay on PVDF membrane in a 96-well<br />

plate format to quantitate streptococcal binding to proteins from saliva and human milk. We found that S.<br />

gordonii binds better to saliva and milk proteins/glycoproteins than S. mutans, and S. gordonii binding is<br />

reduced after treatment with neuraminidase to remove the sialic acid residues from the glycoproteins. Structural<br />

determination <strong>of</strong> saliva and milk protein glycosylation and confirmation <strong>of</strong> the removal <strong>of</strong> sialic acids by<br />

neuraminidase was conducted using LC-ESI-MS. Free sialic acid was also shown to modify streptococcal<br />

adherence to saliva and milk. We have shown that sialic acid residues are involved in S. gordonii binding (but not<br />

S.mutans) to saliva and milk glycoproteins, indicating that these secretions can act as a decoy to protect the oral<br />

environment against infection.<br />

29<br />

Funded by: Macquarie University


NOTES<br />

30


SHORT BIOGRAPHIES OF INVITED SPEAKERS<br />

Short Biographies <strong>of</strong> Invited Speakers<br />

CLEMENS KUNZ – Institute <strong>of</strong> Nutritional Science, University <strong>of</strong> Giessen, Germany<br />

Clemens Kunz received his graduate degrees in nutrition from the University <strong>of</strong> Bonn where he did his<br />

PhD on vitamin D and metabolites in milk at the Department <strong>of</strong> Pediatrics in 1984. He then joined the<br />

group <strong>of</strong> Pr<strong>of</strong>. Heinz Egge at the Institute <strong>of</strong> Physiological Chemistry getting his first scientific contact<br />

with HMOs and has been fascinated by these unique components since then. Subsequently, he held a<br />

postdoctoral fellowship at the University <strong>of</strong> California, Davis, USA from 1986 to 1989 in Pr<strong>of</strong>. Bo<br />

Lönnerdal’s group focusing on milk proteins and glycoproteins. He then became leader <strong>of</strong> the working<br />

group Clinical Chemistry at the Research Institute for Child Nutrition in Dortmund and a Pr<strong>of</strong>essor <strong>of</strong> Physiological Chemistry at<br />

the University <strong>of</strong> Bonn, continuing his research in the field <strong>of</strong> HMOs with a focus on metabolic aspects. In 1999 he was appointed<br />

Pr<strong>of</strong>essor <strong>of</strong> <strong>Human</strong> Nutrition at the Justus-Liebig-University <strong>of</strong> Giessen. His current research focuses on structural, functional<br />

and metabolic aspects <strong>of</strong> HMOs including the use <strong>of</strong> stable isotopes and on the bioavailability and metabolism <strong>of</strong> polyphenols<br />

from berries in healthy humans. His work yielded several research awards and has continuously been funded by the German<br />

Research Foundation (DFG), the Federal Ministry <strong>of</strong> Science and Education (BMBF) and the Hessian Ministry <strong>of</strong> Science and Arts<br />

(HMWK).<br />

HUDSON FREEZE – Sanford-Burnham Medical Research Institute La Jolla, CA, USA<br />

Hudson Freeze earned his Ph.D. from the University <strong>of</strong> California at San Diego in 1976. Subsequently<br />

he held fellowships in Biology, Medicine and Neurosciences and later joined the faculty at the same<br />

institution. In 1988, Dr. Freeze was recruited to Sanford-Burnham Medical Research Institute<br />

and served as the Director <strong>of</strong> the <strong>Glycobiology</strong> and Carbohydrate Chemistry Program from 2000-<br />

2008. His work focuses on pathology resulting from faulty glycosylation, the process <strong>of</strong> adding sugar<br />

chains to proteins and lipids. Carbohydrates are required for proper secretion and targeting <strong>of</strong> thousands <strong>of</strong> proteins, an <strong>of</strong>ten<br />

overlooked fact <strong>of</strong> biology. He is driven by the search for novel therapeutics to treat patients with mutations leading to<br />

glycosylation defects called Congenital Disorders <strong>of</strong> Glycosylation or CDGs.<br />

TADASU URASHIMA – Obihiro University, Hokkaido, Japan<br />

Tadasu Urashima was born in Hiroshima at 12/3/1957. Degrees: (1) undergraduate course: Tokyo<br />

University <strong>of</strong> Agriculture and Technology, 1980. (2) master course: Tohoku University, 1982. (3) Ph.D.<br />

course: Tohoku University, 1986. Past Positions: (1) Assistant Pr<strong>of</strong>essor at Obihiro University <strong>of</strong><br />

Agriculture and Veterinary Medicine, April 1986. (2) Associate Pr<strong>of</strong>essor at Obihiro University <strong>of</strong><br />

Agriculture and Veterinary Medicine, April 1994. (3) Pr<strong>of</strong>essor at Obihiro University <strong>of</strong> Agriculture and<br />

Veterinary Medicine, August 2003. Present position: Pr<strong>of</strong>essor at Obihiro University <strong>of</strong> Agriculture and Veterinary Medicine,<br />

Graduate School <strong>of</strong> Animal and Food Hygiene. Studies: (1) 1986-present: Characterization <strong>of</strong> milk oligosaccharides <strong>of</strong> domestic<br />

farm animals. (2) 1991: Beta N-acetylglycosaminyltransferase activity in lactating mammary glands <strong>of</strong> tammar wallaby, Sydney<br />

University, under Dr. Michael Messer. (3) 1995-present: Characterization <strong>of</strong> milk oligosaccharides <strong>of</strong> captured wild animals<br />

including primate species, Canoidea species, elephant, Felidae species, Cetacea species etc, with Dr. Olav Oftedal. (4) 2000-<br />

present: Determination <strong>of</strong> each human milk oligosaccharide concentration. (5) 2009-present: Determination <strong>of</strong> each milk<br />

oligosaccharide level in the broth <strong>of</strong> Bifidobacteria, with Dr. Motomitsu Kitaoka, Dr. Takane Katayama, Dr. Kenji Yamamoto and<br />

Dr. Sadaki Asakuma. (6) 1994-present: Extracellular polysaccharides produced by dairy lactic acid bacteria, with Dr. Bill Bubb<br />

and Dr. Kenji Fukuda. (7) 2006-2010: Bioactive components in bovine colostrum, with Dr. Takashi Terabayashi, Dr. Minoru<br />

Morita and Dr. Kenji Fukuda.<br />

31


SHORT BIOGRAPHIES OF INVITED SPEAKERS<br />

Short Biographies <strong>of</strong> Invited Speakers<br />

SHARON M. DONOVAN – Department <strong>of</strong> Food Science and <strong>Human</strong> Nutrition, University <strong>of</strong><br />

Illinois, Urbana, IL, USA<br />

Sharon Donovan received her B.S. and Ph.D. in Nutrition from the University <strong>of</strong> California, Davis. She is<br />

also a Registered Dietitian. After completing a post-doctoral fellowship in Pediatric Endocrinology at<br />

Stanford University School <strong>of</strong> Medicine, she accepted a faculty position at the University <strong>of</strong> Illinois,<br />

Urbana in 1991. She was promoted to Pr<strong>of</strong>essor in 2001 and, in 2003, was named the first recipient <strong>of</strong><br />

the Melissa M. Noel Endowed Chair in Nutrition and Health at the University <strong>of</strong> Illinois. Her research<br />

focuses on pediatric nutrition, with an emphasis on optimization <strong>of</strong> neonatal intestinal development. She compares the<br />

biological effects <strong>of</strong> human milk and infant formulas on intestinal function in human infants, neonatal piglets and in various<br />

models <strong>of</strong> intestinal disease. She has published over 100 peer-reviewed publications, review articles and conference proceedings.<br />

Her research is funded by NIH, USDA and private industry and foundations.<br />

THIERRY HENNET – Institute <strong>of</strong> Physiology and Center for Integrative <strong>Human</strong> Physiology,<br />

University <strong>of</strong> Zurich, Switzerland<br />

Thierry Hennet is a Pr<strong>of</strong>essor in the Institute <strong>of</strong> Physiology and Center for Integrative <strong>Human</strong><br />

Physiology at the University <strong>of</strong> Zurich. His research interest is on Defects <strong>of</strong> glycosylation that lead to<br />

diseases with variable clinical manifestations, encompassing neurological disorders, congenital<br />

muscular dystrophies, connective tissue disorders, immune deficiencies, and coagulopathie.<br />

His group investigates the roles <strong>of</strong> specific glycoconjugates and glycosyltransferase enzymes in health and disease. To this end,<br />

they combine the study <strong>of</strong> genetic animal models to biochemical assays performed in cell culture systems. In addition, he studies<br />

the molecular basis <strong>of</strong> novel types <strong>of</strong> Congenital Disorders <strong>of</strong> Glycosylation (CDG), a family <strong>of</strong> diseases characterized by abnormal<br />

biosynthesis <strong>of</strong> several classes <strong>of</strong> glycoconjugates.<br />

DAVID A. MILLS – Department <strong>of</strong> Viticulture and Enology, University <strong>of</strong> California Davis, Davis,<br />

California, USA<br />

David Mills is a Pr<strong>of</strong>essor in the Department <strong>of</strong> Viticulture and Enology in the Robert Mondavi Institute<br />

for Wine and Food Sciences at the University <strong>of</strong> California at Davis. For over 20 years Dr. Mills has<br />

studied the molecular biology <strong>of</strong> lactic acid bacteria involved in food and beverage fermentations or<br />

active as probiotics in intestinal health. In the last decade, Mills led the Lactic Acid Bacteria Genomics<br />

Consortium which resulted in a seminal comparative analysis and release <strong>of</strong> key genome sequences <strong>of</strong><br />

food-grade lactic acid bacteria and bifidobacteria. More recently with Bruce German and Carlito Lebrilla, Dr. Mills formed the UC<br />

Davis <strong>Milk</strong> Bioactives Program, a multidisciplinary effort to characterize the influence <strong>of</strong> milk glycans on intestinal health. Dr.<br />

Mills has served as a Waksman Foundation Lecturer and Chair <strong>of</strong> the Food Microbiology Division <strong>of</strong> the American Society for<br />

Microbiology. He currently serves as an associate editor for the journal Microbiology. In 2010 Dr. Mills was awarded the Cargill<br />

Flavor Systems Specialties Award from the American Dairy Science Association.<br />

Dr. Mills obtained a Bachelors <strong>of</strong> Science in Biochemistry from the University <strong>of</strong> Wisconsin-Madison. In 1991, he obtained a<br />

Masters degree in Biochemistry with Dr. Michael Flickinger at University <strong>of</strong> Minnesota, followed by a PhD in Microbiology in<br />

1995 working with Dr. Gary Dunny and Dr. Larry McKay. After postdoctoral studies at North Carolina State University with Dr.<br />

Todd Klaenhammer, Dr. Mills took a faculty position at UC Davis.<br />

32


SHORT BIOGRAPHIES OF INVITED SPEAKERS<br />

Short Biographies <strong>of</strong> Invited Speakers<br />

MOTOMITSU KITAOKA – National Food Research Institute, National Agriculture and Food<br />

Research Organization, Tsukuba, Ibaraki 305-8642, Japan<br />

Motomitsu Kitaoka is a research leader <strong>of</strong> the enzyme laboratory at the National Food Research<br />

Institute <strong>of</strong> the National Agriculture and Food Research Organization, Japan. He obtained a Ph. D. from<br />

the University <strong>of</strong> Tokyo in 1993. After completing a post-doctoral fellowship at the Iowa State<br />

University, he moved to the National Food Research Institute in 1998. He has been working on<br />

carbohydrate degrading enzymes for more than 20 years, especially on sugar phosphorylases. He has<br />

been pursuing practical procedures to produce oligosaccharides, including a large-scale preparation <strong>of</strong> lacto-N-biose I. He is also<br />

interested in rational mutation to convert glycoside hydrolytic enzymes into catalysts for the syntheses <strong>of</strong> glycosides.<br />

RUDOLF GEYER – Institute <strong>of</strong> Biochemistry, Faculty <strong>of</strong> Medicine, Justus-Liebig-University<br />

<strong>of</strong> Giessen, Friedrichstrasse 24, 35392 Giessen, Germany<br />

Rudolf Geyer studied Chemistry at the University <strong>of</strong> Darmstadt and the University <strong>of</strong> Freiburg in<br />

Germany. After graduation he worked for his Ph.D. at the Max-Planck-Institute <strong>of</strong> Immunology in<br />

Freiburg and received his degree in Biochemistry in 1977 at the University <strong>of</strong> Freiburg. He then<br />

moved to Giessen, Germany, were he worked as a research assistant and later as a university<br />

assistant at the Institute <strong>of</strong> Biochemistry <strong>of</strong> the Giessen University Medical School. In 1990 he was appointed as a pr<strong>of</strong>essor <strong>of</strong><br />

biochemistry and leader <strong>of</strong> the <strong>Glycobiology</strong> Unit at the same institute. His present research interests are mainly focused on<br />

structures and putative functions <strong>of</strong> glycoconjugates from parasitic helminths as well as on the carbohydrate structures <strong>of</strong><br />

mammalian and invertebrate glycoproteins and human milk oligosaccharides.<br />

CARLITO B. LEBRILLA – University <strong>of</strong> California, Davis, CA, USA<br />

Carlito Lebrilla is a Pr<strong>of</strong>essor at the University <strong>of</strong> California, Davis in the Department <strong>of</strong> Chemistry and<br />

Biochemistry and Molecular Medicine at the School <strong>of</strong> Medicine. He is currently the Chair <strong>of</strong> the<br />

Chemistry Department. He was born in the Philippines. He received his BS degree from the University<br />

<strong>of</strong> California, Irvine and Ph.D. from the University <strong>of</strong> California, Berkeley. He was an Alexander von<br />

Humboldt Fellow and a NSF-NATO Fellow at the Technical University in Berlin. He returned to the UC<br />

Irvine as a President’s Fellow and has been at UC Davis since 1989. His research is in Analytical<br />

Chemistry, primarily mass spectrometry with applications to clinical glycomics and bi<strong>of</strong>unctional food. He has over 225 peer-<br />

reviewed publications. He is also co-editor <strong>of</strong> Mass Spectrometry Reviews and has been on the editorial board <strong>of</strong> Mass<br />

Spectrometry Reviews, Journal <strong>of</strong> American Society for Mass Spectrometry, European Mass Spectrometry, and International<br />

Journal <strong>of</strong> Mass Spectrometry.<br />

33


SHORT BIOGRAPHIES OF INVITED SPEAKERS<br />

Short Biographies <strong>of</strong> Invited Speakers<br />

NORBERT SPRENGER – Nestlé Research Center, Lausanne, Switzerland<br />

Norbert Sprenger studied biology at the University <strong>of</strong> Basel, Switzerland, and received<br />

his B.S. in Developmental Biology and his Ph.D. in Molecular Plant Physiology with Pr<strong>of</strong>s.<br />

A. Wiemken and T. Boller for his studies on plant storage, transport and stress<br />

carbohydrates. He then took a first post-doctoral position in plant molecular physiology<br />

in the laboratory <strong>of</strong> Pr<strong>of</strong>. F. Keller at the University <strong>of</strong> Zurich, Switzerland. For a second<br />

post-doctoral fellowship, financed by grants from the Swiss National Science Foundation, Novartis and the <strong>Human</strong> Frontier<br />

Science Program Organisation, he joined the laboratory <strong>of</strong> Pr<strong>of</strong>. C. Somerville at Stanford University at the Carnegie Institute for<br />

Science department <strong>of</strong> plant biology, where he studied plant cell wall synthesis and physiology using molecular genetics<br />

approaches. He then switched to mammalian physiology accepting a position as research scientist in <strong>Glycobiology</strong> at the Nestlé<br />

Research Center in Lausanne, Switzerland. His research focuses on functional glycans for pediatric and medical nutrition, with an<br />

emphasis on intestinal development and barrier function.<br />

BING WANG – <strong>Human</strong> Nutrition Unit, University <strong>of</strong> Sydney, Australia; School <strong>of</strong> Medicine,<br />

Xiamen University, P. R. China; Nestlé Research Centre Beijing, P. R. China<br />

Bing Wang received her M.D. degree from Tianjin Medical University, China, and a Ph.D. in Science<br />

(Biochemistry) from the University <strong>of</strong> Sydney, Australia. She is a currently an Honorary Associate in the<br />

School <strong>of</strong> Molecular & Biosciences at the University <strong>of</strong> Sydney, a Min-Jiang Scholar and Adjunct<br />

Pr<strong>of</strong>essor <strong>of</strong> Molecular Medicine, School <strong>of</strong> Medicine, Xiamen University, China and a Senior Research<br />

Scientist at the Nestlé Research Centre-Beijing. She is an internationally recognized expert in<br />

nutritional glycobiology. She pioneered the development <strong>of</strong> the piglet as a model system to elucidate the molecular mechanisms<br />

underlying the role <strong>of</strong> milk sialic acid in brain development and cognition. She is also a registered Nutritionist <strong>of</strong> the Nutrition<br />

Society Australia. Her studies have been carried out at both the gene and biochemical levels on the functional effect <strong>of</strong> new food<br />

ingredients.<br />

LARS BODE – Division <strong>of</strong> Neonatology and Division <strong>of</strong> Gastroenterology and Nutrition,<br />

Department <strong>of</strong> Pediatrics, University <strong>of</strong> California, San Diego, USA<br />

Lars Bode received his M.S. and Ph.D. in Nutrition from the Justus-Liebig University, Giessen, Germany.<br />

After determining structural differences in the lipid composition <strong>of</strong> human and bovine milk gangliosides<br />

as a Master student, Lars joined Dr. Clemens Kunz’s lab as a PhD student and worked with Dr. Silvia<br />

Rudl<strong>of</strong>f to study the effects <strong>of</strong> human milk oligosaccharides on selectin-mediated cell-cell interactions in<br />

the immune system. After a predoctoral fellowship at the Institute <strong>of</strong> Child Health, University College<br />

London, Lars joint Dr. Hudson Freeze’s lab as a postdoctoral fellow in the <strong>Glycobiology</strong> and Carbohydrate Chemistry Program at<br />

the Burnham Institute for Medical Research. In 2009 the University <strong>of</strong> California, San Diego, School <strong>of</strong> Medicine recruited Lars as<br />

an Assistant Pr<strong>of</strong>essor <strong>of</strong> Pediatrics in the Division <strong>of</strong> Neonatology and the Division <strong>of</strong> Gastroenterology and Nutrition, where his<br />

lab develops a new research program to elucidate functions and biosynthesis <strong>of</strong> human milk oligosaccharides. Lars is an affiliate<br />

<strong>of</strong> the <strong>Glycobiology</strong> Research and Training Center and the Digestive Disease Research Development Center at the University <strong>of</strong><br />

California, San Diego. Lars is the Chair <strong>of</strong> the Lactation Research Interest Group <strong>of</strong> the American Society for Nutrition and on the<br />

Executive Board <strong>of</strong> the International Society for Research on <strong>Human</strong> <strong>Milk</strong> and Lactation. Lars’ research is supported by an NIH<br />

Pathway to Independence Award, and funded by NIH and private industry.<br />

34


ABSTRACTS OF POSTERS<br />

Abstracts <strong>of</strong> Posters<br />

Role <strong>of</strong> Sialic acid in Innate Immune Protection provided by Mammalian <strong>Milk</strong><br />

WAI YUEN CHEAH 1, KATHERINE WONGTRA-KUL KISH 1, DANIEL KOLARICH 2, JASMINE GRINYER 1, NICOLLE PACKER 1<br />

1 Department <strong>of</strong> Chemistry and Biomolecular Sciences, Faculty <strong>of</strong> Science, Macquarie University, Sydney, NSW,<br />

Australia<br />

2 Department <strong>of</strong> Biomolecular Systems, Max Planck Institute <strong>of</strong> Colloids and Interfaces, Berlin, Germany<br />

Sialic acid contained in mammalian milk has been reported to have an immune protective role for the infant.<br />

<strong>Human</strong> milk contains a large amount <strong>of</strong> sialic acid compared with bovine milk, with sialic acids found on<br />

glycoproteins, glycolipids and free oligosaccharides. However, the mechanism by which this monosaccharide<br />

inhibits infection remains unknown. We have investigated how milk confers innate immune protection to the<br />

infant gut through binding <strong>of</strong> gastrointestinal-associated bacteria and compare the difference in binding <strong>of</strong><br />

specific bacteria to human and bovine milk glycoconjugates.<br />

Different methods to fractionate human and bovine milk were explored to determine which fraction contains<br />

bacterial binding glycoconjugates. <strong>Milk</strong> fractions were adhered to PVDF membranes in a 96 well format.<br />

Fluorescently-labelled bacteria were added to the wells and the binding <strong>of</strong> the bacteria to the fractions was<br />

quantified by fluorescence. To determine whether the glycans were involved in this interaction, in particular<br />

sialylated glycans, the glycoconjugate structures were altered using exoglycosidases and the effect on bacterial<br />

binding was measured. Exoglycosidase activities were confirmed by glycan pr<strong>of</strong>iling <strong>of</strong> the N- and O-glycans<br />

released by PNGase F and β-elimination, respectively, <strong>of</strong> milk glycoproteins using graphitised carbon LC-ESI-<br />

MS/MS. The inhibition <strong>of</strong> binding <strong>of</strong> the bacteria to the milk glycoconjugates was tested by pre-incubation <strong>of</strong> the<br />

bacteria with free oligosaccharides.<br />

We found that specific bacteria bind differently to human and bovine milk glycoconjugates. Most bacteria had<br />

higher binding affinity to the milk protein fraction than the lipid fraction. Bacterial binding was inhibited both by<br />

removal <strong>of</strong> sialic acid with sialidase and by pre-incubation <strong>of</strong> the bacteria with sialic acid. This confirms that<br />

sialic acid can competitively inhibit bacterial adhesion to both human and bovine milk glycoconjugates. These<br />

data indicate the importance <strong>of</strong> sialylated milk glycoconjugates in binding to gastrointestinal-associated bacteria<br />

and providing innate immune protection to the infant’s gut.<br />

35<br />

Funded by: Macquarie University


ABSTRACTS OF POSTERS<br />

Ascending colonic microbiota composition and SCFA patterns produced from in vitro<br />

fermentation <strong>of</strong> human milk oligosaccharides and prebiotics differ between formula-fed and<br />

sow-reared piglets<br />

MIN LI 1, LAURA L. BAUER 2, XIN CHEN 1, MEI WANG 1, THERESA KUHLENSCHMIDT 3, MARK S. KUHLENSCHMIDT 3,<br />

GEORGE C. FAHEY JR. 2 AND SHARON M. DONOVAN 1<br />

1 Department <strong>of</strong> Food Science and <strong>Human</strong> Nutrition, 2 Department <strong>of</strong> Animal Sciences, 3 Department <strong>of</strong> Pathobiology,<br />

University <strong>of</strong> Illinois, Urbana, IL 61801, USA<br />

The objective was to compare the effect <strong>of</strong> piglet age and sow-rearing versus formula feeding on in vitro<br />

fermentation characteristics and gut microbial modulation properties <strong>of</strong> human milk oligosaccharides (HMO),<br />

lacto-N-neotetraose (LNnT), a predominant HMO, and three commonly used prebiotics: a 1:2 mixture <strong>of</strong><br />

galactooligosaccharide (GOS) and polydextrose (PDX), and short-chain fructooligosaccharides (scFOS). Ascending<br />

colon contents were obtained from four donor groups: 9 and 17 day-old formula-fed (FF9, FF17) and sow-reared<br />

(SR9, SR17) piglets. The changes in pH and gas, short chain fatty acid (SCFA) and lactate production were<br />

determined following 0, 4, 8, and 12 h <strong>of</strong> incubation. The pH change and total SCFA, acetate and propionate<br />

production were greater in the FF than SR, and in the 9- compared to the 17-day-old piglets, regardless <strong>of</strong> diet.<br />

However, SR produced higher amounts <strong>of</strong> gas, butyrate and lactate than FF piglets. For most donors, the pH<br />

change was greatest for scFOS, and least for GOS/PDX. The fermentation <strong>of</strong> LNnT produced larger amounts <strong>of</strong> gas,<br />

total SCFA, acetate, and butyrate than did the other substrates, whereas higher amounts <strong>of</strong> propionate and lactate<br />

were observed from HMO and scFOS fermentation respectively. Gut microbial populations were assessed by 16S<br />

rRNA V3 gene denaturing gradient gel electrophoresis (DGGE) analysis and group-specific real-time PCR. Global<br />

gut microbial structures differed among four piglet groups prior to fermentation. SR had higher concentration <strong>of</strong><br />

Bifidobacterium and Lactobacillus, whereas FF had greater amounts <strong>of</strong> Clostridium cluster IV and XIVa. The<br />

concentrations <strong>of</strong> these four bacteria groups were higher in 9- than 17-day-old piglets. Changes in microbial<br />

patterns during fermentation were assessed by DGGE. Bands that increased in density on most substrates were<br />

identified by sequencing as related to Bacteroides vulgatus, Collinsella aer<strong>of</strong>aciens, Anaerovibrio sp., and species<br />

belong to Clostridium cluster IV, XIVa and XVIII. The concentrations <strong>of</strong> Bifidobacterium, Clostridium cluster IV and<br />

XIVa and B. vulgatus detected by qPCR increased after 8 and 12h fermentation on most substrates. In summary,<br />

piglet diet and age affect gut microbial populations, which leads to different bacterial fermentation patterns. HMO<br />

and LNnT have potential prebiotic effects due to their ability to be fermented to SCFA and modulate microbial<br />

populations.<br />

36<br />

Funded by: NIH R01 HD061929


ABSTRACTS OF POSTERS<br />

Oligosaccharide MALDI-MS pr<strong>of</strong>iles in milk and urine from mother-child pairs<br />

VIKTORIA DOTZ 1, SILVIA RUDLOFF 1,2, DENNIS BLANK 3, SABINE GEBHARDT 1, KAI MAASS 3, RUDOLF GEYER 3 AND<br />

CLEMENS KUNZ 1<br />

1 Institute <strong>of</strong> Nutritional Science, University <strong>of</strong> Giessen, Wilhelmstrasse 20, 35392 Giessen, Germany<br />

²Department <strong>of</strong> Pediatrics, University <strong>of</strong> Giessen, Feulgenstrasse 12, 35392 Giessen, Germany<br />

³Institute <strong>of</strong> Biochemistry, Faculty <strong>of</strong> Medicine, University <strong>of</strong> Giessen, Friedrichstrasse 24, 35392 Giessen, Germany<br />

<strong>Human</strong> milk contains a large variety <strong>of</strong> complex lactose-based oligosaccharides (HMO) in concentrations ranging<br />

from 10 to 20 g/L. The presence <strong>of</strong> different neutral HMO depends on the activity <strong>of</strong> specific glycosyltransferases<br />

in the mammary gland. Therefore, the presence <strong>of</strong> 1-2-, 1-3- and/or 1-4-fucosylated core oligosaccharide<br />

structures is determined by the secretor status and the Lewis (Le) phenotype <strong>of</strong> the mother (1,2). So far, little<br />

information exists about the metabolic fate <strong>of</strong> HMO in newborns. In previous studies, we found a renal excretion<br />

<strong>of</strong> about 1-2 % <strong>of</strong> the ingested LNT and LNFPII in exclusively breast-fed preterm infants (3). Here, we report on<br />

the excretion pr<strong>of</strong>ile <strong>of</strong> HMO in term infants.<br />

The aims <strong>of</strong> the study are (i) to compare the HMO pattern in milk <strong>of</strong> exclusively breastfeeding women with the<br />

urinary oligosaccharide pr<strong>of</strong>ile <strong>of</strong> their infants and (ii) to investigate whether the infants´ urinary HMO pr<strong>of</strong>ile<br />

resembles the pattern <strong>of</strong> Lewis-specific HMO in their mothers´ milk.<br />

After application <strong>of</strong> an oral 13 C-galactose bolus to 8 lactating mothers, milk at each nursing and urine <strong>of</strong> the<br />

infants was collected for 36 hours (4). For analytical purposes, 50 µL <strong>of</strong> a single milk sample and 500 µL <strong>of</strong> the<br />

infant urine sample were used for solid phase extraction prior to MALDI-MS <strong>of</strong> the non-derivatised HMO fraction.<br />

Lewis blood group determination was performed on all milk samples.<br />

Resulting from their characteristic MS pr<strong>of</strong>ile as well as MS/MS spectra, the following Lewis phenotypes were<br />

found: <strong>Milk</strong> from 6 women was Le(a−b+), one sample was Le(a+b−) and one presumably Le(a−b−) or Le(a−b+).<br />

In the corresponding urinary samples <strong>of</strong> the infants, the same characteristic MS pr<strong>of</strong>iles were observed with<br />

regard to relative HMO peak intensities. However, HMO in the lower mass range were more predominant in<br />

urine, particularly for the infant whose mother was Le(a+b−). Moreover, MS/MS data revealed that none <strong>of</strong> the<br />

Le(a−b+)-specific HMO were detected either in the milk <strong>of</strong> the Le(a+b−) mother or in the urine <strong>of</strong> her infant.<br />

MS/MS <strong>of</strong> urine from infants fed with Le(a−b+)-milk confirmed the presence <strong>of</strong> Le(a−b+)-specific HMO.<br />

Our data indicate that the HMO patterns in urine from breast-fed infants reflect those from their mothers’ milk<br />

suggesting a strong association to the mothers´ Lewis phenotype determined in milk.<br />

Literature:<br />

1) Kunz et al., Annu Rev Nutr 2000; 2) Thurl et al., Br J Nutr 2010; 3) Rudl<strong>of</strong>f et al. Acta Paediatr 1996; 4) Rudl<strong>of</strong>f<br />

et al., Glycobiol 2006<br />

Funded by: German Research Foundation Ru 529/7-3 and Ku 781/8-3<br />

37


ABSTRACTS OF POSTERS<br />

<strong>Human</strong> <strong>Milk</strong> <strong>Oligosaccharides</strong> as Anti-adhesion Candidates for Clostridium difficile Toxin<br />

AMR EL-HAWEIT 1, ELENA N. KITOVA 1, PAVEL KITOV 1, LUIZ EUGENIO 2, KENNETH K.S. NG 2, GEORGE L. MULVEY 3,<br />

TANIS C. DINGLE 3, ADAM SZPACENKO 1, GLEN D. ARMSTRONG 3 AND JOHN S. KLASSEN 1<br />

1 Department <strong>of</strong> Chemistry, University <strong>of</strong> Alberta, Edmonton, AB, Canada.<br />

2 Department <strong>of</strong> Biological Sciences, University <strong>of</strong> Calgary, Calgary, AB, Canada.<br />

3 Department <strong>of</strong> Microbiology & Infectious Diseases, University <strong>of</strong> Calgary, Calgary, AB, Canada. †Alberta Ingenuity<br />

Centre for Carbohydrate Science<br />

<strong>Human</strong> milk oligosaccharides (HMOs) play an important role in protecting neonates from pathogenic<br />

microorganism especially the enteric bacteria. This is believed to be due to their ability to act as soluble<br />

receptors that inhibit the pathogen binding to their host cell target ligands. Clostridium difficile is the leading<br />

cause <strong>of</strong> hospital acquired diarrhea and pseudomembranous colitis in Europe and North America. The key<br />

virulence determinants <strong>of</strong> C. difficile are the two exotoxins, toxin A (TcdA) and toxin B (TcdB). Those exotoxins<br />

are believed to bind to carbohydrate receptors on the intestinal epithelium. Here, we describe the first<br />

quantitative study <strong>of</strong> the binding <strong>of</strong> HMOs to the C. difficile toxins. The direct electrospray ionization mass<br />

spectrometry (ESI-MS) assay has been applied to determine the binding affinities <strong>of</strong> recombinant subfragments<br />

TcdA and TcdB <strong>of</strong> C. difficile toxins against a library <strong>of</strong> twenty one oligosaccharides representing, the most<br />

common neutral and acidic HMOs. The results <strong>of</strong> the ESI-MS measurements indicate that both toxins fragments<br />

bind specifically to several HMOs ranging in size from tri- to heptasaccharides. Notably, five <strong>of</strong> the HMOs tested<br />

bind to both toxins: Fuc(1-2)Gal(1-4)Glc, Gal(1-3)GlcNAc(1-3)Gal(1-4)Glc, Fuc(1-2)Gal(1-3)GlcNAc(1-<br />

3)Gal(1-4)Glc, Gal(1-3)[Fuc(1-4)]GlcNAc(1-3)Gal(1-4)Glc and Gal(1-4)[Fuc(1-3)]GlcNAc(1-3)Gal(1-<br />

4)Glc. Although, the binding <strong>of</strong> the HMOs is uniformly weak, with apparent affinities ≤3000 M -1 , a number <strong>of</strong><br />

HMOs bind to the toxins with a much higher affinity than the only known natural receptor,<br />

Gal(1,3)Gal(1,4)Glc, for which binding constants for both A2 and B1 were found to be ~500 M -1 . The results<br />

<strong>of</strong> the molecular docking study suggest that the general mode <strong>of</strong> carbohydrate recognition may be conserved<br />

between TcdA and TcdB, with a lactose disaccharide occupying the central portion <strong>of</strong> the carbohydrate binding<br />

site for both toxins. Analysis <strong>of</strong> the docked structures also helps to explain how the differences in distributions <strong>of</strong><br />

negatively and positively charged side chains in the binding pocket <strong>of</strong> TcdA and TcdB influence ligand binding<br />

specificity. The Verocytotoxicity neutralization assay reveals that HMO fractions extracted from human milk do<br />

not significantly inhibit the cytotoxic effects <strong>of</strong> TcdA nor TcdB. The absence <strong>of</strong> protection is attributed to the<br />

weak intrinsic affinities that the toxins exhibit towards the HMOs.<br />

Funded by: 1. Alberta Ingenuity Centre for Carbohydrate Science (AICCS)<br />

38<br />

2. Natural Sciences and Engineering Research Council (NSERC)<br />

3. University <strong>of</strong> Alberta


ABSTRACTS OF POSTERS<br />

Structure determination <strong>of</strong> bacterial mucus-binding proteins and their functional role in<br />

adhesion to host glycans<br />

SABRINA ETZOLD 1, DONALD MACKENZIE 1, LOUISE E. TAILFORD 1, ROB FIELD 2, ANDREW HEMMINGS 3 AND<br />

NATHALIE JUGE 1<br />

1 Institute <strong>of</strong> Food Research, Norwich Research Park, Norwich NR4 7UA, UK<br />

2 John Innes Centre, Norwich Research Park, NR4 7UH, UK<br />

3 University <strong>of</strong> East Anglia, Norwich NR4 7TJ, UK<br />

The mucus layer covers the epithelial cells <strong>of</strong> the gastrointestinal (GI) tract and protects the underlying mucosa<br />

from the lumen content. The main structural components <strong>of</strong> mucus are highly glycosylated mucin proteins<br />

carrying mainly O-linked glycan chains characterised by a high level <strong>of</strong> structural complexity and diversity.<br />

Protein-carbohydrate interactions are believed to play an important role in the adhesion <strong>of</strong> resident gut bacteria<br />

to the mucus layer. However, the nature <strong>of</strong> the ligands and the specificity <strong>of</strong> the interaction remain to be<br />

elucidated. Our research focuses on lactobacilli mucus-binding proteins (MUB) whose presence on the bacterial<br />

cell-surface contributes to bacterial attachment to the protective mucus layer. MUBs are composed <strong>of</strong> tandemly-<br />

arranged mucus-binding amino acid sequence repeats (Mubs). The 353 kDa MUB from the L. reuteri strain ATCC<br />

53608 consists <strong>of</strong> two types <strong>of</strong> repeats, Mub1 and Mub2, present in six and eight copies, respectively. We<br />

determined the first crystal structure <strong>of</strong> a type 2 Mub repeat (184 amino acids) at 1.8-Å, displaying high<br />

structural similarity to the repeat-unit <strong>of</strong> the Peptostreptococcus magnus Protein L (PpL), an immunoglobuling<br />

(Ig)-binding surface protein, and we showed that Mub repeats were able to interact with a number <strong>of</strong><br />

mammalian Igs in vitro. Our current work is to obtain structural information on type 1 Mub repeats and multi-<br />

repeat modules to gain further insight into the structural organisation <strong>of</strong> MUB. In addition, we focus on the<br />

characterisation <strong>of</strong> the Mub repeat interaction with mucins and mucin glycans, whose structure can be similar to<br />

oligosaccharides found in milk, using a range <strong>of</strong> biophysical methods including in vitro binding assays,<br />

carbohydrate arrays and Isothermal Titration Calorimetry. Elucidating the molecular basis <strong>of</strong> host-bacteria<br />

interaction is crucial for the understanding <strong>of</strong> host colonisation and the beneficial role <strong>of</strong> lactobacilli in the GI<br />

tract.<br />

39<br />

Funded by: BBSRC, NRP


ABSTRACTS OF POSTERS<br />

Breast milk microbiota: is there a relationship with HMOs?<br />

ANA SOTO, NIVIA CÁRDENAS, SUSANA DELGADO, JUAN MIGUEL RODRÍGUEZ AND LEONIDES FERNÁNDEZ<br />

Departamento de Nutrición, Bromatología y Tecnología de los Alimentos, Universidad Complutense de Madrid,<br />

Avda. Puerta de Hierro, Madrid, Spain<br />

Breast milk is a complex biological fluid that provides all the nutritional requirements for the first months <strong>of</strong> life<br />

but, additionally, educates the infant immune system and confers protection against pathogens. These effects<br />

result from the synergistic action <strong>of</strong> many bioactive molecules, such as cytokines, cellular components,<br />

oligosaccharides or lipids. While it is well known that breast milk is rich in oligosaccharides, it has only recently<br />

been accepted that human milk also constitutes a source <strong>of</strong> commensal and probiotic bacteria which seems to<br />

play an important role in gut colonization and modulation <strong>of</strong> the infant gut.<br />

In recent years, analyses <strong>of</strong> the bacterial diversity <strong>of</strong> human milk have revealed that this biological fluid is an<br />

important source <strong>of</strong> live staphylococci, streptococci, bifidobacteria and lactic acid bacteria to the infant gut. In<br />

contrast to other bacteria, these seem to be uniquely adapted to reside in the human digestive tract and to<br />

interact with us in symbiosis from the time we are born. Traditionally it was considered that bacteria present in<br />

breast milk were acquired by mere skin contamination. However, it has been found that lactobacilli and<br />

enterococcal isolates present in human milk are genotypically different from those isolated in the skin within the<br />

same host. Furthermore, several studies suggest the existence <strong>of</strong> a mammary microbiota during late pregnancy<br />

and lactation. Live bacteria from the maternal gut could colonize the mammary gland through an endogenous<br />

route (the so-called entero-mammary pathway), involving maternal dendritic cells and macrophages.<br />

In this context, the global objective <strong>of</strong> this work was to ascertain if there is a relationship between human milk<br />

oligosaccharides and microbiota. For that purpose, carbohydrate utilization was studied in a variety <strong>of</strong> lactic acid<br />

bacteria and bifidobacteria isolated from human milk <strong>of</strong> healthy donors, and compared with their enzymatic<br />

activity on chromogenic substrates. Genome sequences <strong>of</strong> selected bacteria were analyzed to identify possible<br />

genes involved in the metabolism <strong>of</strong> carbohydrates, especially those related to HMOs.<br />

Funded by: AGL2010-15420 (2010-2013), RM2009-00009-00-00<br />

40


ABSTRACTS OF POSTERS<br />

Tailoring carbohydrates to capture toxins and pathogens<br />

SURI S. IYER<br />

Department <strong>of</strong> Chemistry, University <strong>of</strong> Cincinnati, Cincinnati, OH 45221, USA<br />

Over the past few years, our group has been harnessing their recognition properties to develop high affinity<br />

ligands for toxins and pathogens. Carbohydrates are excellent recognition molecules and modulate essential<br />

communication processes as cell adhesion, proliferation, migration, differentiation and metastasis. Synthetic<br />

carbohydrates can be used for capturing pathogens as they are highly robust under a variety <strong>of</strong> conditions,<br />

refrigeration is not required and can be stored for extended period (over months), inexpensive, not subject to<br />

lot-to-lot variation, can be manipulated to suit any sensor platform as these are small molecules and not subject<br />

to antigenic drift because toxicity is intimately associated with the cell binding sites. However, synthetic<br />

carbohydrates have “thought” to suffer from selectivity and sensitivity issues for practical applications. We have<br />

recently demonstrated that it is possible to tailor glycoconjugates to achieve high selectivity and sensitivity<br />

towards specific analytes. We have synthesized a library <strong>of</strong> tailored carbohydrates using a novel modular<br />

strategy for the rapid production <strong>of</strong> these molecules (Bioorg. Med. Chem. Lett., 2007, 17, 2459-64). We have<br />

demonstrated that synthetic carbohydrates differentiate between closely related strains <strong>of</strong> Shiga toxin, (Angew.<br />

Chem. Int. Ed. Engl. 2008, 47, 1265-68; Highlighted in Chemical and Engineering News, 2008, 86, 42) influenza (J.<br />

Am. Chem. Soc. 2008, 130, 8169–71) and Escherichia coli. (Chembiochem, 2008, 9, 2433-42). Our other relevant<br />

publications on the molecular nature <strong>of</strong> carbohydrate-protein interactions include ChemBioChem, 2009, 10,<br />

1486-89; Biochemistry, 2010, 49, 1649-57; Medicinal Research Reviews, 2010, 30, 327-93; Bioconj. Chem., 2010,<br />

21, 1486-93 and Biochemistry, 2010, 49, 5954-67. Recently, we have developed a simple, rapid, and sensitive<br />

glycan-based magnetic relaxation switch assay for the detection <strong>of</strong> glycan binding proteins. We demonstrated<br />

that magnetic relaxation switch assays can be used to detect toxins in a complex medium such as stool and<br />

environmental samples. (Anal. Chem., 2010, 82, 7430-7435). Our efforts in this area towards the development <strong>of</strong><br />

potential diagnostics and therapeutics using natural and modified oligosaccharides will be the focus <strong>of</strong> this<br />

presentation.<br />

Funded by: NSF CAREER GRANT, NSF CHE 0845005; NIAID U01 AI075498<br />

41


ABSTRACTS OF POSTERS<br />

α-L-Fucosynthase that specifically introduces Lewis a/x antigens into type-1/2 chains<br />

TAKANE KATAYAMA 1, HARUKO SAKURAMA 1, YUJI HONDA 1, MOTOMITSU KITAOKA 2 AND KENJI YAMAMOTO 1<br />

1 Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Nonoichi, Ishikawa 921-<br />

8836, Japan<br />

2 National Food Research Institute, National Agriculture and Food Research Organization, Tsukuba, Ibaraki 305-<br />

8642, Japan<br />

Lewis a (Le a ) and Lewis x (Le x ) blood group antigens attached at the non-reducing ends <strong>of</strong> glycoconjugates<br />

control various pivotal biological events; therefore, the efficient synthesis <strong>of</strong> the antigens is important to<br />

understand their physiological roles. Here, we show that the nucleophile-mutant D703S <strong>of</strong> 1,3-1,4-α-L-fucosidase<br />

from Bifidobacterium bifidum exclusively synthesizes Le a and Le x trisaccharides when incubated with β-L-<br />

fucopyranosyl fluoride (FucF) as a donor and lacto-N-biose I (Galβ1-3GlcNAc) and N-acetyllactosamine (Galβ1-<br />

4GlcNAc) as acceptors, respectively, with yields <strong>of</strong> 56% against the added FucF. The synthase also recognized<br />

lactose, 2’-fucosyllactose and lacto-N-tetraose as acceptors, and specifically produced 3-fucosyllactose,<br />

difucosyllactose and lacto-N-fucopentaose II, respectively. In contrast, the enzyme did not accept<br />

monosaccharides, cellobiose, Glc1-4GlcNAc, N,N’-diacetylchitobiose or galacto-N-biose as substrates. The<br />

results revealed that the enzyme specifically recognizes the disaccharide structures [Galβ1-3/4GlcNAc(Glc)] at<br />

the non-reducing ends and attaches a Fuc residue via an α-(1→3/4)-linkage to the GlcNAc/Glc residue. The strict<br />

regio- and acceptor specificity <strong>of</strong> this 1,3-1,4-α-L-fucosynthase is unique and should serve as a potentially<br />

powerful tool to specifically introduce Le a and Le x epitopes in type-1 and type-2 chains <strong>of</strong> glycans, respectively.<br />

Funded by: Grant-in-Aid for Scientific Research by Young Scientists (B) 22780072 from Ministry <strong>of</strong> Education, Culture,<br />

42<br />

Sports, Science and Technology, Japan


ABSTRACTS OF POSTERS<br />

A new methodology for screening <strong>of</strong> bacteria-carbohydrate interactions: anti-adhesive milk<br />

oligosaccharides as a case study<br />

JONATHAN A. LANE 1, 2, KARINA MARIÑO 3, PAULINE M. RUDD 3, STEPHEN D. CARRINGTON 2 AND RITA M. HICKEY 1<br />

1 Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland. 2 Veterinary Science Centre, University<br />

College Dublin, Belfield, Dublin 4, Ireland. 3 Dublin Oxford <strong>Glycobiology</strong> Group, National Institute for Bioprocessing<br />

Research & Training (NIBRT), Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland<br />

Carbohydrates have been shown to inhibit the initial recognition events leading to adhesion and colonization <strong>of</strong> host<br />

tissues by pathogens and therefore have potential to prevent infection. Some <strong>of</strong> the most efficient anti-adhesion agents<br />

identified to date are present in foodstuffs - as best exemplified by human milk carbohydrates which protect newborns<br />

against infections. Such carbohydrates can display structural homology to host cell receptors or pathogenic lectins,<br />

thus functioning as receptor decoys. However, the large amounts <strong>of</strong> human milk carbohydrates which are required for<br />

intervention or clinical studies are unavailable; therefore researchers are beginning to focus their attention on sources<br />

<strong>of</strong> carbohydrates other than those from human milk. For instance, the milk from domestic animals, honey, fermented<br />

products, commensal and food-grade bacteria all contain carbohydrates which are structurally similar to human milk<br />

carbohydrates and have been in some cases shown to prevent pathogen binding to host cells. However, current<br />

screening methods for the identification <strong>of</strong> anti-adhesive oligosaccharides have limitations: they are time-consuming<br />

and require large amounts <strong>of</strong> oligosaccharides. Therefore, there is a need to develop analytical techniques that can<br />

quickly screen for, and structurally define, anti-adhesive oligosaccharides prior to using cell line models <strong>of</strong> infection.<br />

Considering this, we have developed a rapid method for screening complex oligosaccharide mixtures for potential anti-<br />

adhesive activity against bacteria. This label-free approach involves the brief and iterative exposure <strong>of</strong> a mixture <strong>of</strong><br />

free oligosaccharide to aliquots <strong>of</strong> a defined bacterial population. These organisms act as an “affinity matrix” to<br />

progressively deplete glycans with the specific capacity to bind to the surface <strong>of</strong> these microbes, permitting their<br />

identification through comparison with untreated glycan pr<strong>of</strong>iles. As a case study, the free oligosaccharides from the<br />

colostrum <strong>of</strong> Holstein Friesian cows were screened for interactions with E. coli O157:H7 cells. Reductions in<br />

oligosaccharide concentrations were determined by High pH Anion Exchange Chromatography. The use <strong>of</strong> the<br />

depletion assay showed selective bacterial interaction with lactose, 3’-sialyllactose, disialyllactose, and<br />

sialyllactosamine in a population dependent manner, in line with previous results obtained by different methodologies.<br />

The depletion assay methodology was validated by inhibition studies performed using HT-29 cells. The structures <strong>of</strong><br />

all potential anti-adhesives in the bovine mixture were confirmed in a fast and sensitive manner by HILIC-HPLC and<br />

<strong>of</strong>fline mass spectrometry (ESI-MS/MS). The methodology proposed here represents a novel approach for the<br />

discovery <strong>of</strong> anti-adhesive oligosaccharides in diverse feedstocks. Initial screening can be undertaken in hours,<br />

whereas conventional approaches may take days if not weeks. The depletion assay, combined with the HILIC<br />

methodology, facilitates the structural and functional characterization <strong>of</strong> animal milk oligosaccharides and could be<br />

used to detect any bacteria-oligosaccharide interactions. Indeed, our approach could be exploited for the discovery <strong>of</strong><br />

free oligosaccharides with the capacity to bind whole bacterial cells from food sources other than milk.<br />

Jonathan Lane is in receipt <strong>of</strong> a Teagasc Walsh Fellowship. This work was funded by the Department <strong>of</strong> Agriculture and Food,<br />

Ireland, under the Food Institutional Research Measure, project reference number 05/R&D/TD/368 and done in<br />

collaboration with the Alimentary Glycoscience Research Cluster (AGRC) Science Foundation Ireland under Grant No.<br />

43<br />

08/SRC/B1393


ABSTRACTS OF POSTERS<br />

Sialylation and fucosylation <strong>of</strong> human milk α1-acid glycoprotein during the first two weeks <strong>of</strong><br />

lactation<br />

MAGDALENA ORCZYK-PAWIŁOWICZ 1 AND LIDIA HIRNLE 2<br />

1 Department <strong>of</strong> Chemistry and Immunochemistry, Wrocław Medical University, Bujwida 44A, 50-345 Wrocław,<br />

Poland, 2 Department <strong>of</strong> Obstetrics and Gynaecology, Clinic <strong>of</strong> Reproduction and Obstetrics, Wrocław Medical<br />

University, Dyrekcyjna 5/7, 50-328 Wrocław, Poland<br />

The biological function <strong>of</strong> α1-acid glycoprotein (AGP), acute phase glycoprotein (40% <strong>of</strong> sugars), is not clear,<br />

although a number <strong>of</strong> activities in vitro and in vivo indicate that it may immunomodulate an inflammatory<br />

response and, at the same time, act as a plasma transport protein. <strong>Oligosaccharides</strong> <strong>of</strong> AGP are reported to<br />

influence its biological activities, and modulate cell-to-cell interactions and cellular signaling. Under pathological<br />

conditions, AGP may serve as a general protective agent in infections and against toxins by binding to toxic lectin<br />

endotoxins and bacterial lipopolysaccharide. AGP might also play anti-inflammatory and immunomodulatory<br />

roles in inflammation and cancer through its N-glycans, especially its highly branched and α1,3-fucosylated N-<br />

glycans. Van Dijk and coworkers [1998] have suggested that AGP molecules expressing sialo-Lewis x<br />

glycodeterminant may interfere with leukocyte-endothelial interactions by binding to E- or P-selectin,<br />

manifesting its anti-inflammatory properties.<br />

The aim <strong>of</strong> the present work was to study the alterations in relative amounts <strong>of</strong> sialic acids and fucoses linked by<br />

different anomeric linkages to subterminal N-glycans <strong>of</strong> human milk AGP in comparison to plasma AGP, and in<br />

relation to stages <strong>of</strong> human lactation during the first two weeks. The relative amounts <strong>of</strong> fucosyl- and sialyl-<br />

glycotopes on AGP were analyzed in human milk samples by lectin-ELISA using fucose- (LCA, Lens culinaris, LTA,<br />

Tetragonolobus purpureus and UEA-1,Ulex europaeus) and sialic acid (MAA, Maackia amurensis and SNA,<br />

Sambucus nigra)-specific biotinylated lectins.<br />

We have found that the sialylation and fucosylation patterns <strong>of</strong> human milk AGP were different than those<br />

reported for human plasma AGP. The observed changes were qualitative and quantitative. The normal plasma<br />

AGP showed very low expression <strong>of</strong> fucosylated forms. In contrast, the human milk AGP was highly decorated<br />

with fucoses. The fucosylation pattern <strong>of</strong> normal plasma AGP is limited to the innermost α1,6-fucose, whereas<br />

milk AGP contained higher relative amounts <strong>of</strong> the innermost α1,6-linked fucose and α1,2-and α1,3-linked<br />

fucoses on outer arms. However, among milk AGPs, no significant differences between samples <strong>of</strong> milk taken<br />

during the first and the second week <strong>of</strong> lactation were observed. The sialylation <strong>of</strong> human milk AGP was higher<br />

than normal plasma AGP for both types, α2,3- and α2,6-, <strong>of</strong> sialic acid linkages with a predominance <strong>of</strong> α2,6-<br />

sialylated form.<br />

We can hypothesize that terminal sugars <strong>of</strong> milk AGP are biologically active, with anti-inflammatory properties<br />

in modulation <strong>of</strong> inflammatory processes: the highly sialylated and fucosylated oligosaccharides <strong>of</strong> milk AGP<br />

could ensure homeostasis during the first weeks <strong>of</strong> lactation. However, the obtained results are preliminary, they<br />

provide the starting point for further structural and functional studies. In future, it seems to be interesting to<br />

analyze oligosaccharides <strong>of</strong> human milk AGP in inflammatory diseases, e.g. mastitis.<br />

44<br />

Funded by: partly by UDA-POKL.04.01.01-00-010/08-01


ABSTRACTS OF POSTERS<br />

The relative amounts <strong>of</strong> fucose is<strong>of</strong>orms in oligosaccharides <strong>of</strong> human milk fibronectin<br />

MAGDALENA ORCZYK-PAWIŁOWICZ 1, LIDIA HIRNLE 2 AND IWONA KĄTNIK-PRASTOWSKA 1<br />

1 Department <strong>of</strong> Chemistry and Immunochemistry, Wrocław Medical University, Bujwida 44A, 50-345 Wrocław,<br />

Poland, 2 Department <strong>of</strong> Obstetrics and Gynaecology, Clinic <strong>of</strong> Reproduction and Obstetrics, Wrocław Medical<br />

University, Dyrekcyjna 5/7, 50-328 Wrocław, Poland<br />

Fibronectin (FN), a multidomain and multifunctional large glycoprotein which is engaged in some cellular<br />

biological functions, e.g. adhesion, migration, proliferation, tissue repair, extracellular matrix remodelling. It is<br />

possible thanks FN multiple domain interactions with its natural ligands, such as fibrin, heparin, collagen, C-<br />

reactive-protein, and complement components. Moreover, FN serves binding sites to many pathogens through<br />

peptide and/or oligosaccharide sequences, thus being important for bacterial adhesion and colonization to host<br />

tissue.<br />

In human organism FN is an abundant component. FN produced by various cells (e.g. fibroblasts, chondrocytes,<br />

lymphocytes, endothelial cells) is known to be trapped into insoluble multimeric fibrills, while that present in<br />

plasma originate from hepatocyte synthesizes, and is released to blood as a compact globular dimer.<br />

<strong>Human</strong> FN contains 5-9% <strong>of</strong> oligosaccharides mainly N- type, and to a lesser degree as O-type. The extent and<br />

type <strong>of</strong> FN glycosylation varies depending on the tissue source and cell type, and human condition. The plasma<br />

and cellular FNs differ regarding the number <strong>of</strong> antennae, and the degree <strong>of</strong> sialylation and α1,6-fucosylation on<br />

the innermost GlcNAc <strong>of</strong> the chitobiose core. Plasma-derived oligosaccharides can be bi- and tri-antennary,<br />

heavily sialylated and weakly fucosylated. In contrast, cellular FN has bi-antennary glycans, largely fucosylated,<br />

but weakly sialylated. The expression <strong>of</strong> the α1,2-linked fucosylated glycotope <strong>of</strong> FN seems to be dependent on<br />

the site <strong>of</strong> FN synthesis. FN produced by hepatocytes and released into the blood lacks α1,2-linked fucose,<br />

whereas the amniotic and seminal FNs are heavily decorated by α1,2-fucoses.<br />

The N-glycosylation confers FN stability and protects against proteolytic degradation, and also may act as<br />

modulators <strong>of</strong> FN affinity to some substrates: a lack <strong>of</strong> oligosaccharides on FN markedly enhanced its ability to<br />

promote adhesion and spreading <strong>of</strong> fibroblasts, concomitantly increasing affinity to gelatin. The mucin-type O-<br />

glycans might play a significant role in segregating the neighboring domains and maintaining the topology and<br />

function <strong>of</strong> FN domains.<br />

This communication focuses on the analysis <strong>of</strong> the relative degree <strong>of</strong> α1,6, α1,3, and α1,2-linked fucoses on FN<br />

present in human milk samples during the first two weeks <strong>of</strong> lactation <strong>of</strong> healthy mothers who delivered healthy<br />

newborns. The relative amounts <strong>of</strong> exposed fucosylated glycotopes were analyzed by lectin-ELISA using LCA<br />

(Lens culinaris), LTA (Tetragonolobus purpureus), and UEA-1 (Ulex europaeus) lectins with well-defined sugar<br />

specificity. The analyses indicate the significant differences in the expositions <strong>of</strong> fucosylated glycotopes on FN<br />

present in blood plasma and human milk. It was found that the milk FN was heavily decorated with α1,6-, α1,3-,<br />

and α1,2-linked fucoses, whereas the plasma FN negligible. Since the α1,2-fucosylated glycans are known to be<br />

implicated in interaction between host and some pathogens, it can be postulated that α1,2-fucosylated FN in<br />

human milk can act as a natural inhibitor against pathogenic bacteria colonization. The α1,2-fucosylated<br />

glycotopes <strong>of</strong> soluble milk glycoproteins could constitute one <strong>of</strong> the element <strong>of</strong> an innate immune system for<br />

breastfed infants.<br />

45


ABSTRACTS OF POSTERS<br />

Effects <strong>of</strong> specific milk oligosaccharides on the expression <strong>of</strong> interleukin-8 and marker enzymes<br />

<strong>of</strong> intestinal cell maturation<br />

KRISTINE ANNA SCHOLAND 1, SABINE KUNTZ 2, CLEMENS KUNZ 2, KLAUS-PETER ZIMMER 1 AND SILVIA RUDLOFF 1,2<br />

1 Department <strong>of</strong> Pediatrics, University <strong>of</strong> Giessen, Feulgenstr. 12, D-35392 Giessen, Germany<br />

2 Institute <strong>of</strong> Nutritional Science, University <strong>of</strong> Giessen, Wilhelmstr. 20, D-35392 Giessen, Germany<br />

The microbial colonization <strong>of</strong> the neonatal gut has a major impact on the development <strong>of</strong> intestinal functions,<br />

nutrient bioavailability and the immune system. <strong>Human</strong> milk oligosaccharides (HMO) may not only be substrates<br />

for specific bacterial species, but also resemble their adhesion molecules or ligands which may enable them to<br />

directly affect gut maturation or influence inflammatory processes.<br />

Therefore, we investigated the expression <strong>of</strong> trehalase (Treh), a disaccharidase regarded as a general marker for<br />

gut maturation, and <strong>of</strong> fucosyltransferase (Fut) 1 and 2 being involved in the postnatal shift <strong>of</strong> intestinal surface<br />

glycosylation from sialylated towards more fucosylated structures. An inflammatory response was determined<br />

as interleukin (IL)-8 expression. <strong>Human</strong> intestinal cells (Caco-2, HT-29) were incubated with 2 mmol <strong>of</strong> 2’- or 3-<br />

fucosyllactose (FL), lacto-N-tetraose (LNT), neo-LNT, lacto-N-fucopentaose (LNFP) I or III, 3’- and 6’-sialyllactose<br />

(SL) or disialyl-LNT for 24 h. mRNA expression <strong>of</strong> Treh, Fut1, Fut2 and IL-8 was determined using real-time RT-<br />

PCR.<br />

In HT-29 cells, HMO did not affect the expression <strong>of</strong> Fut1 and Fut2; Treh was merely expressed in these cells<br />

confirming HT-29 as a non-differentiating cell line. In Caco-2 cells, however, Treh expression increased along<br />

with their differentiation over 14 days post confluence. Whereas HMO except for FL showed the tendency to<br />

reduce Treh expression, Fut1 expression in Caco-2 cells seemed slightly upregulated by most HMO tested; Fut2<br />

expression remained unchanged. Stimulation <strong>of</strong> Caco-2 cells with the proinflammatory cytokine tumor necrosis<br />

factor alpha (TNF-α) itself had no influence on marker enzyme expression. HMO, in particular LNT, LNFP I and III<br />

as well as 3’-SL decreased Treh expression indicating an inhibitory effect <strong>of</strong> these oligosaccharides on cell<br />

differentiation. Fut1 expression, on the other hand, was slightly enhanced by sialylated HMO in stimulated Caco-<br />

2 cells; again, there was no effect on Fut2 expression.<br />

With regard to IL-8 expression, disialyl-LNT as well as LNFP III may have the potential to modulate cytokine<br />

production. Whereas IL-8 expression seemed to be decreased in the presence <strong>of</strong> disialyl-LNT, LNFP III slightly<br />

enhanced IL-8 expression.<br />

These results confirm previous observations [1] indicating that HMO directly modulate intestinal cell<br />

differentiation. In addition, HMO may also have an impact on cytokine production.<br />

[1] Kuntz S, Rudl<strong>of</strong>f, S, Kunz C. (2008) Br J Nutr 99: 462-471<br />

46


In vivo production <strong>of</strong> fucose-α1, 2-lactose<br />

KATELYN ZAK AND PENG GEORGE WANG<br />

The Ohio State University, Columbus, Ohio, USA<br />

ABSTRACTS OF POSTERS<br />

The third largest component <strong>of</strong> human milk is complex, minimally metabolized oligosaccharides that serve to<br />

protect the infant’s health both by promoting infant intestinal microbiota and warding away harmful pathogens.<br />

The mother’s blood type cause structural variations and change HMO concentrations change lactation. While<br />

HMOs can promote growth <strong>of</strong> infant intestinal microbiota, their best characterized function is their prebiotic<br />

effects. HMOs have anti-adhesive effects prohibiting pathogens from binding to intestinal epithelial cells as well<br />

as modifying surface glycans that pathogens use to facilitate their entry. Fucosylated oligosaccharides comprise<br />

eighty percent <strong>of</strong> all HMOs <strong>of</strong> which 2-linked fucosyloligosaccharides appear the most biologically significant.<br />

The current hurdle in HMO research is the lack <strong>of</strong> large scale synthesis <strong>of</strong> HMOs to further test their biological<br />

significance. From clinical trials, fucose-α1, 2-lactose protects against diarrhea significantly better than non-2-<br />

linked fucosyloligosaccharides. Fucose-α1, 2-lactose may be produced in large scale quantities by recombinant<br />

E. coli which utilize enzymes FKP (fucokinase pyrophosphorylase) and WbsJ (α1, 2-fucoslytransferase) to<br />

enzymatically synthesize fucose-α1, 2-lactose. FKP is a bifunctional enzyme which catalyzes the reaction<br />

between fucose, ATP, and GTP to produce the expensive and hard to obtain GDP-fucose, meanwhile WbsJ is a<br />

known α1, 2-FucT which demonstrates promiscuous acceptor substrate specificity. This method is cost effective<br />

by regenerating the sugar nucleotides required for the synthesis and require only inexpensive, commercially<br />

available, fucose and lactose.<br />

47<br />

Funded by: NIH R01 HD061935


ABSTRACTS OF POSTERS<br />

Sialylated galactosides <strong>of</strong> human milk as inhibitors <strong>of</strong> enterovirus 71 and A (H1N1) 2009<br />

influenza infections<br />

BETSY YANG 1,2, HAU CHUANG 2 AND RON-FU CHEN 2<br />

1 University <strong>of</strong> North Carolina, Class <strong>of</strong> 2014, Chapel Hill, NC 27509, USA<br />

2 Department <strong>of</strong> Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan<br />

Background and specific aims:<br />

Many viruses recognize specific sugar residues, such as sialylated glycosides, as the infection receptors. <strong>Human</strong><br />

milk contains many sialylated oligoglycosides which may block viral infections to gastrointestinal tract. We<br />

postulated in the illustration below that sialic acid (SA)-linked alpha 2,3 galactosides (SA-2,3Gal) and SA<br />

2,6Gal from human milk ( ) could inhibit infection <strong>of</strong><br />

enterovirus 71 (EV71) or swine A(H1N1) influenza. We<br />

have previously shown that SA-2,6Gal and/or SA-2,3Gal<br />

from human milk specifically inhibited EV71 infection to<br />

48<br />

Flu<br />

EV71<br />

Sialylated glycans<br />

Intestinal cells<br />

DLD-1 intestinal cells (Virol J. 2009 Sep 15; 6:141). This study has been extended to investigate whether SA-<br />

2,6Gal and/or SA-2,3Gal from human milk inhibited swine A(H1N1) influenza infection to DLD-1 intestinal<br />

cells.<br />

Results:<br />

EV71 specifically infected DLD-1 intestinal cells but not K562 myeloid cells. Pretreatment <strong>of</strong> DLD-1 cells with<br />

sialidase (2mU, 2 hours) significantly reduced 20-fold EV71 replication (p


ABSTRACTS OF POSTERS<br />

The oligosaccharide (OS) phenotype <strong>of</strong> preterm infants predicts risk: A potential indication for<br />

HMOS administration?<br />

ARDYTHE L. MORROW, KURT R. SCHIBLER, JAREEN MEINZEN-DERR, DIANA TAFT AND BARBARA DAVIDSON<br />

Perinatal Institute, Cincinnati Children’s Hospital Medical Center, University <strong>of</strong> Cincinnati College <strong>of</strong> Medicine, Ohio,<br />

USA<br />

The human milk oligosaccharides (HMOS) are analogs <strong>of</strong> oligosaccharides (OS) expressed on infant mucosal<br />

surfaces and in saliva. Understanding the role <strong>of</strong> HMOs to prevent morbidity and mortality in breastfed infants<br />

also requires understanding the innately expressed OS in the infant. Using banked samples from an extant<br />

cohort, we previously reported that absent or low expression <strong>of</strong> salivary H antigen (Fuc1,2 oligosaccharide<br />

produced by transferases <strong>of</strong> the FUT2 gene) – an indirect measure <strong>of</strong> intestinal expression – significantly predicts<br />

risk <strong>of</strong> necrotizing enterocolitis or death in preterm infants (J Pediatrics, 2011).<br />

To confirm and extend that finding, we launched a prospective study <strong>of</strong> infants under 33 weeks’ gestational age<br />

enrolled in three level III neonatal intensive care units (NICUs) in Cincinnati, Ohio (study ongoing). In the first<br />

175 enrolled infants, we analyzed saliva samples at day <strong>of</strong> life 8 by enzyme-linked immunoassay to determine H<br />

antigen and other oligosaccharide epitopes as predictors <strong>of</strong> necrotizing enterocolitis or death. FUT2 genotype<br />

was determined independently, and was consistent with salivary phenotype. Using the same cut-point, we again<br />

found that low or absent H antigen significantly predicted subsequent necrotizing enterocolitis or death (odds<br />

ratio =7.7; p=0.004), controlling for low expression <strong>of</strong> Lewis a (Fuc1,4 oligosaccharide produced by<br />

transferases <strong>of</strong> the FUT3 gene), infant gestational age and birthweight.<br />

We hypothesize that low or absent H antigen is a biomarker for aberrant intestinal colonization <strong>of</strong> the preterm<br />

infant. The innate risk that we have identified in relation to low or absent H antigen expression in preterm<br />

infants suggests a prophylactic role for HMOS, which are rich in fucosylated oligosaccharide, and stimulate the<br />

growth <strong>of</strong> beneficial bacteria. Analysis <strong>of</strong> the intestinal microbiome and the role <strong>of</strong> maternal HMOS is underway<br />

in a subset <strong>of</strong> infants with longitudinal samples.<br />

Sponsored by U.S. National Institute <strong>of</strong> Child Health and <strong>Human</strong> Development, HD 13021 and HD 059140<br />

49


ABSTRACTS OF POSTERS<br />

Natural antibodies against milk oligosaccharides<br />

MAKSIM NAVAKOUSKI, NADEZHDA SHILOVA, POLINA OBUKHOVA, NAILYA KHASBIULLINA AND NICOLAI BOVIN<br />

Shemyakin & Ovchinnikov Institute <strong>of</strong> Bioorganic Chemistry RAS, Moscow, Russia<br />

Recently anti-glycan antibodies <strong>of</strong> 106 adult healthy donors’ sera were pr<strong>of</strong>iled with the help <strong>of</strong> printed<br />

glycan array (PGA) which contained about 200 glycans. It was shown that natural human anti-glycan antibodies<br />

bind at least 160 glycans [1], including milk oligosaccharides: LNT (95% <strong>of</strong> examined donors), LNnT (90%), 2`-<br />

fucosylLac (90%), Le b -Lac (80%), Lac (75%), 3`SL (60%), 6`SL (35%). The glycans were immobilized on the slide<br />

surface as N-glycine derivatives. In addition, the strong interaction between sera and some glycans closely<br />

related to milk OS was found, e.g. with SiaLe C (99%), Le C (95%), P1 (75%), Le D (50%), asialo-GM1 (100%), GA2<br />

(95%), Le Y -Lac (90%).<br />

Anti-milk OS antibodies (anti-milkOS), namely against LNT, LNnT, Le B Lac and Lac were detected in<br />

blood <strong>of</strong> 3-day infants receiving mixed feeding. These anti-milkOS belong to IgG class only and thus seem to be<br />

maternal.<br />

Currently accumulated data don’t allow us to figure out the origin <strong>of</strong> natural anti-milkOS. On one hand a<br />

reason <strong>of</strong> their appearance might be natural glycation conjugates <strong>of</strong> milk OS with proteins due to elevated<br />

concentrations <strong>of</strong> both components in colostrum; the conjugates could work as immunogens. On the other hand,<br />

the antibodies actually could be directed to different epitopes, in other words, milk OS play a role <strong>of</strong> mimetics for<br />

unknown yet antigens.<br />

1. Huflejt, M.E., et al., Anti-carbohydrate antibodies <strong>of</strong> normal sera: findings, surprises and challenges.<br />

Mol Immunol, 2009. 46(15): p. 3037-49.<br />

Funded by: European Commission’s Marie Curie program (the EuroGlycoArrays ITN)<br />

50


ABSTRACTS OF POSTERS<br />

Primary prevention <strong>of</strong> allergic diseases by probiotics: impact <strong>of</strong> HMOs<br />

GER T. RIJKERS 1,2, TITIA NIERS 1, NICOLE RUTTEN 2, SASKIA VAN HEMERT 3, MAARTEN HOEKSTRA 1, ARINE VLIEGER 2<br />

1 University Medical Center Utrecht, 2 St. Antonius Hospital, Nieuwegein, 3 Winclove Bioindustries, Amsterdam, The<br />

Netherlands<br />

Modification <strong>of</strong> the intestinal microbiota by administration <strong>of</strong> probiotic bacteria may be a potential approach to<br />

prevent allergic disease. We aimed to study primary prevention <strong>of</strong> allergic disease in high-risk children by<br />

perinatal supplementation <strong>of</strong> selected probiotic bacteria. In a double-blind, randomized, placebo-controlled trial,<br />

a mixture <strong>of</strong> probiotic bacteria selected by in vitro screening (Bifidobacterium bifidum, Bifidobacterium lactis,<br />

and Lactococcus lactis, as Ecologic®Panda) was prenatally administered to mothers <strong>of</strong> high-risk children<br />

(positive family history <strong>of</strong> allergic disease) and to their <strong>of</strong>fspring for the first 12 months <strong>of</strong> life. Probiotics were<br />

administered daily in a dose <strong>of</strong> 1 x 109 cfu per strain. The placebo-group received an equivalent amount <strong>of</strong><br />

carrier material (rice starch plus maltodextran) daily. Up to 80% <strong>of</strong> mothers breast fed their babies for an<br />

average <strong>of</strong> 7.1 months. Parental-reported eczema during the first 3 months <strong>of</strong> life was significantly lower in the<br />

intervention group compared with placebo, 6/50 vs. 15/52 (P=0.035), a relative risk reduction <strong>of</strong> 58%. The<br />

number needed to treat was 5.9 at age 3 and 12 months and 6.7 at age 2 years. Similarly, physician-confirmed<br />

eczema during the first 3 months was 3/50 and 11/52 in the intervention and control groups, respectively<br />

(P=0.021). The intervention group was significantly more frequently colonized with higher numbers <strong>of</strong> Lc. lactis.<br />

In 30% <strong>of</strong> babies in the placebo group Lc. Lactis was found, which may a consequence <strong>of</strong> breast feeding. At age 3<br />

months, in vitro production <strong>of</strong> IL-5 in anti-CD2/CD28 mAb-stimulated whole blood was decreased in the<br />

probiotic group compared with placebo (P=0.04). In conclusion, Ecologic®Panda reduced the incidence <strong>of</strong><br />

eczema in high-risk children at 3 months <strong>of</strong> life, an effect which seems to be sustained throughout the first 2<br />

years. Additional studies at 5-6 years assessing lung function and asthma development are ongoing.<br />

We are planning to study the potential synergistic effect <strong>of</strong> HMOs on immunomodulation by probiotics. To that<br />

end we will first determine in in vitro studies which HMOs show the highest potential to modulate the neonatal<br />

immune system for induction <strong>of</strong> Th1 and Treg cells.<br />

Funded by: The Wilhelmina Children's Hospital and the Ministery <strong>of</strong> Economic Affairs<br />

51


THE SPONSOR<br />

<strong>Glycom</strong> A/S<br />

www.glycom.com<br />

<strong>Glycom</strong> is a privately held company founded in 2005 in Copenhagen, Denmark whose focus is to synthesize and<br />

commercialize human milk oligosaccharides (HMOs) along with their precursors and intermediates.<br />

HMOs have been the object <strong>of</strong> scientific curiosity for over 40 years. Developed over millions <strong>of</strong> years <strong>of</strong> human<br />

evolution, HMOs are the 3rd largest component <strong>of</strong> mother’s milk and are attributed with many <strong>of</strong> its wonderful<br />

health effects. Until now, study <strong>of</strong> these natural biopharmaceuticals has been limited and commercialization has<br />

been blocked by lack <strong>of</strong> available material and high costs – with virtually none available from extractive sources<br />

and only a few synthesized in extremely low quantities and extremely high costs.<br />

<strong>Glycom</strong>’s breakthrough synthetic chemistry has changed this equation permanently. We have developed efficient<br />

chemical and enzymatic technologies for synthesis <strong>of</strong> HMOs representing over 50% <strong>of</strong> total HMO concentration in<br />

typical samples <strong>of</strong> mother’s milk. The leading HMOs in the <strong>Glycom</strong> pipeline have established robust scale up<br />

technology in industrial scales and GMP conditions, with glycosylations in multi cubic meter reactors and<br />

production in ton scale and in pure form suitable for human consumption. The opportunity for large scale,<br />

widespread study and commercialization <strong>of</strong> HMOs has arrived as a result <strong>of</strong> our efforts and the support <strong>of</strong> our<br />

partners and investors.<br />

In order to efficiently develop HMOs, <strong>Glycom</strong> has also developed breakthrough synthesis <strong>of</strong> the four mono and<br />

disaccharide precursors/building blocks <strong>of</strong> HMOs. Two <strong>of</strong> these precursors – sialic acid and L-fucose – are among the<br />

handful <strong>of</strong> basic natural human sugars. None <strong>of</strong> them are currently available in sufficient quantities and costs for<br />

high volume commercial applications – including HMO synthesis. As with the HMOs themselves, the <strong>Glycom</strong><br />

technology for the first wave precursors has been scaled up to industrial conditions. As a result, <strong>Glycom</strong> is already<br />

the world’s largest producer <strong>of</strong> L-fucose and has already achieved cost levels that are a small fraction <strong>of</strong> its current<br />

market price. Other precursors are in line for similar breakthroughs.<br />

52


ALBERMANN, CHRISTOPH<br />

Institute <strong>of</strong> Microbiology, University <strong>of</strong> Stuttgart, Germany<br />

christoph.albermann@imb.uni-stuttgart.de<br />

BEAUPREZ, JOERI<br />

Ghent University, Belgium<br />

joeri.beauprez@ugent.be<br />

BERTELSEN, HANS<br />

Arla Foods, Videbæk, Denmark<br />

hans.bertelsen@arlafoods.com<br />

BODE, LARS<br />

University <strong>of</strong> California, San Diego, USA<br />

lbode@ucsd.edu<br />

BØTTKJÆR, KIM<br />

<strong>Glycom</strong> A/S, Kongens Lyngby, Denmark<br />

kib@fihpartners.com<br />

BØJSTRUP, MARIE<br />

Carlsberg Laboratory, Copenhagen, Denmark<br />

mab@crc.dk<br />

CILIEBORG, MALENE<br />

University <strong>of</strong> Copenhagen, Denmark<br />

macilie@life.ku.dk<br />

CONTRACTOR, NIKHAT<br />

Pfizer Nutrition, Collegeville, PA, USA<br />

nikhat.contractor@pfizer.com<br />

DONOVAN, SHARON M.<br />

University <strong>of</strong> Illinois, Urbana, USA<br />

sdonovan@illinois.edu<br />

DEKANY, GYULA<br />

<strong>Glycom</strong> A/S, Kongens Lyngby, Denmark<br />

gyula.dekany@glycom.com<br />

DROUILLON, MARGRIET<br />

University <strong>of</strong> Ghent, Ghent, Belgium<br />

Margriet.Drouillon@UGent.be<br />

EL-HAWIET, AMR<br />

University <strong>of</strong> Alberta, Canada<br />

elhawiet@ualberta.ca<br />

FERNANDEZ, LEONIDES<br />

Dept. Food Science & Tech, Universidad Complutense de Madrid, Spain<br />

leonides@vet.ucm.es<br />

FISCHER, LUTZ<br />

University <strong>of</strong> Hohenheim, Germany<br />

lutz.fischer@uni-hohenheim.de<br />

GARRIDO, DANIEL<br />

University <strong>of</strong> California, Davis, USA<br />

dagarrido@ucdavis.edu<br />

GRINYER, JASMINE<br />

Faculty <strong>of</strong> Science, Macquarie University, Australia<br />

jasmine.grinyer@mq.edu.au<br />

HENNET, THIERRY<br />

Universität Zürich, Switzerland<br />

thennet@access.uzh.ch<br />

LIST OF PARTICIPANTS<br />

List <strong>of</strong> Participants<br />

53<br />

AUSTIN, SEAN<br />

Nestlé Research Center, Lausanne, Switzerland<br />

sean.austin@rdls.nestle.com<br />

BERING, STINE BRANDT<br />

Department <strong>of</strong> <strong>Human</strong> Nutrition, Copenhagen University, Denmark<br />

sbs@life.ku.dk<br />

BLANK, DENNIS<br />

Justus-Liebig University, Giessen, Germany<br />

dennis.blank@chemie.uni-giessen.de<br />

BOYLE, ROBERT<br />

Department <strong>of</strong> Paediatrics Imperial College, London, UK<br />

r.boyle@nhs.net<br />

BRASSART, DOMINIQUE<br />

Nestec Ltd., Vevey, Switzerland<br />

dominique.brassart@nestle.com<br />

CHEAH, WAI YUEN<br />

Faculty <strong>of</strong> Science, Macquarie University, Australia<br />

wai.cheah@mq.edu.au<br />

COPPA, GIOVANNI V.<br />

Polytechnic University <strong>of</strong> Marche, Ancona, Italy<br />

g.v.coppa@univpm.it<br />

CRISÀ, ALESSANDRA<br />

C.R.A., Monterotondo, Italy<br />

alessandra.crisa@entecra.it<br />

DAVIS, STEVEN<br />

Abbott Nutrition, Columbus, OH, USA<br />

steven_davis@abbott.com<br />

DOTZ, VIKTORIA<br />

Justus-Liebig University <strong>of</strong> Giessen, Germany<br />

viktoria.dotz@uni-giessen.de<br />

DUNCAN, PETER<br />

Nestlé Research Center, Lausanne, Switzerland<br />

monique.unternaehrer@rdls.nestle.com<br />

ETZOLD, SABRINA<br />

Norwich Research Park, Institute <strong>of</strong> Food Research, Norwich, UK<br />

sabrina.etzold@bbsrc.ac.uk<br />

FICHOT, MARIE-CLAIRE<br />

Nestec Ltd., Vevey, Switzerland<br />

marie-claire.fichot@nestle.com<br />

FREEZE, HUDSON<br />

Sanford-Burnham Medical Research Institute, La Jolla, USA<br />

Hudson@sandfordburnham.org<br />

GEYER, RUDOLF<br />

Justus Liebig Universität Giessen, Germany<br />

rudolf.geyer@biochemie.med.uni.giessen.de<br />

HALTRICH, DIETMAR<br />

University <strong>of</strong> Natural Resources and Life Sciences, Vienna, Austria<br />

dietmar.haltrich@boku.ac.at<br />

HESTER, SHELLY<br />

University <strong>of</strong> Illinois, Urbana, USA<br />

sndavis2@illinois.edu


HINDSGAUL, OLE<br />

Carlsberg Laboratory, Copenhagen, Denmark<br />

hindsgaul@crc.dk<br />

HORLACHER, PETER<br />

Cognis, Illertissen, Germany<br />

Peter.Horlacher@cognis.com<br />

JØRGENSEN, JULIE BØCK<br />

University <strong>of</strong> Copenhagen, Denmark<br />

julie_boeck@hotmail.com<br />

KATAYAMA, TAKANE<br />

Ishikawa Prefectural University, Nonoichi, Japan<br />

takane@ishikawa-pu.ac.jp<br />

KELM, SØRGE<br />

University Bremen, Germany<br />

skelm@uni-bremen.de<br />

KLARENBEEK, BERT<br />

Friesland Campina, Beilen, The Netherlands<br />

bert.klarenbeek@frieslandcampina.com<br />

Kovacs, JUDIT<br />

<strong>Glycom</strong> A/S, Kongens Lyngby, Denmark<br />

judit.kovacs@glycom.com<br />

KRISTENSEN, METTE BACH<br />

Arla Foods, Viby J., Denmark<br />

mekre@arlafoods.com<br />

KVISTGAARD, ANNE STAUDT<br />

Arla Foods, Viby J., Denmark<br />

askv@arlafoods.com<br />

LEBRILLA, CARLITO<br />

University <strong>of</strong> California, Davis, USA<br />

cblebrilla@ucdavis.edu<br />

MANURUNG, SARMAULI<br />

Danish Technical University, Veterinary Institute, Denmark<br />

saem@vet.dtu.dk<br />

MAU, SUSANNE<br />

<strong>Glycom</strong> A/S, Kongens Lyngby, Denmark<br />

susanne.mau@glycom.com<br />

MILLS, DAVID A.<br />

University <strong>of</strong> California, Davis, USA<br />

damills@ucdavis.edu<br />

MUNBLIT, DANIEL<br />

Imperial College, London Clinical Medicine, London, UK<br />

daniel.munblit08@imperial.ac.uk<br />

MØRKBAK, ANNE LOUISE<br />

Arla Foods, Viby J., Denmark<br />

anmor@arlafoods.com<br />

NEWBURG, DAVID S.<br />

Boston College, MA, USA<br />

david.newburg@bc.edu<br />

ORCZYK-PAWILOXICZ, MAGDALENA<br />

Dept. Chemistry & Immunology, Wroclaw Medical University, Poland<br />

morczyk@immchem.am.wroc.pl<br />

PETERBAUER, CLEMENS<br />

University <strong>of</strong> Natural Resources and Life Sciences, Vienna, Austria<br />

clemens.peterbauer@boku.ac.at<br />

LIST OF PARTICIPANTS<br />

54<br />

HOLSCHER, HANNAH D.<br />

University <strong>of</strong> Illinois, Urbana, USA<br />

hholsche@illinois.edu<br />

IYER, SURI S.<br />

University <strong>of</strong> Cincinnati, Ohio, USA<br />

suri.iyer@uc.edu<br />

KAMERLING, JOHANNIS P.<br />

University <strong>of</strong> Utrecht, Utrecht, The Netherlands<br />

j.p.kamerling@uu.nl<br />

KAVANAUGH, DEVON<br />

Teagasc Food Research Centre, Cork, Ireland<br />

devon.kavanaugh@teagasc.ie<br />

KITAOKA, MOTOMITSU<br />

National Food Research Institute, Tsukuba, Japan<br />

mkitaoka@affrc.go.jp<br />

KLASSEN, JOHN S.<br />

University <strong>of</strong> Alberta, Canada<br />

john.klassen@ualberta.ca<br />

KRENGEL, UTE<br />

Department <strong>of</strong> Chemistry, University <strong>of</strong> Oslo, Norway<br />

ute.krengel@kjemi.uio.no<br />

KUNZ, CLEMENS<br />

Justus Liebig Universität Giessen, Germany<br />

clemens.kunz@ernaehrung.uni-giessen.de<br />

LANE, JONATHAN<br />

Moorepark Food Research Centre, Cork, Ireland<br />

Jonathan.Lane@teagasc.ie<br />

LI, YANQI<br />

Department <strong>of</strong> <strong>Human</strong> Nutrition, University <strong>of</strong> Copenhagen, Denmark<br />

yli@life.ku.dk<br />

MARIOTTA, MARIAROSARIA<br />

Teagasc Food Research Centre, Cork, Ireland<br />

mariarosaria.marotta@teagasc.ie<br />

MIKLUS, MICHAEL<br />

PBM Nutritional, Georgia, USA<br />

mmiklus@pbmnutritionals.com<br />

MORROW, ARDYTHE L.<br />

Cincinnati Children's Hospital, Ohio, USA<br />

ardythe.morrow@cchmc.org<br />

MUTUNGI, GISELLA<br />

Pfizer Nutrition, Collegeville, PA, USA<br />

gisella.mutungi@pfizer.com<br />

NAVAKOUSKI, MAKSIM<br />

Institute <strong>of</strong> Bioorganic Chemistry, Moscow, Russia<br />

maxushob@gmail.com<br />

NIELSEN, DENNIS S.<br />

Department <strong>of</strong> Food Science, University <strong>of</strong> Copenhagen, Denmark<br />

dn@life.ku.dk<br />

PERONI, DIEGO<br />

Chorus S.p.A., Clinica Pediatrica, Verona, Italy<br />

r.cosenza@lachorus.com<br />

PRIETO, PEDRO ANTONIO<br />

Technologico de Monterrey, Mexico<br />

paprieto@itesm.mx


PUTZE, JOHANNES<br />

Children's University Hospital Mannheim, Germany<br />

johannes.putze@medma.uni-heidelberg.de<br />

RÖHRIG, CHRISTOPH H.<br />

<strong>Glycom</strong> A/S, Kongens Lyngby, Denmark<br />

christoph.roehrig@glycom.com<br />

SANGILD, PER TORP<br />

University <strong>of</strong> Copenhagen, Denmark<br />

psa@life.ku.dk<br />

SCHOLAND, KRISTINE ANNA<br />

University Giessen, Germany<br />

kristine.a.scholand@paediat.med.uni-giessen.de<br />

SKALKAM, MARIA<br />

Oxie, Sweden<br />

maria.lena.skalkam@post.au.dk<br />

SPRENGER, NORBERT<br />

Nestlé Research Center, Lausanne, Switzerland<br />

norbert.sprenger@rdls.nestle.com<br />

STAUDACHER, ERIKA<br />

University Vienna, Austria<br />

erika.staudacher@boku.ac.at<br />

TAPPENDEN, KELLY<br />

University <strong>of</strong> Illinois, Urbana, USA<br />

tappende@illinois.edu<br />

THIEM, JOACHIM<br />

University <strong>of</strong> Hamburg, Germany<br />

thiem@chemie.uni-hamburg.de<br />

URASHIMA, TADASU<br />

Obihiro University <strong>of</strong> Agriculture & Veterinary Medicin, Japan<br />

urashima@obihiro.ac.jp<br />

VAN LEEUWEN, SANDER<br />

University <strong>of</strong> Groningen, Groningen, The Netherlands<br />

s.s.van.leeuwen@rug.nl<br />

WANG, BING<br />

University <strong>of</strong> Sydney, Australia. Xiamen University & Nestlé Research<br />

Center, Beijing, P. R. China<br />

bing.wang@sydney.edu.au<br />

WELMAN, ALAN<br />

Fonterra Coop. Group, Palmerston, New Zealand<br />

alan.welman@fonterra.com<br />

YANG, KUENDER D.<br />

Department <strong>of</strong> Medical Research, Taiwan<br />

yangkd.yeh@hotmail.com<br />

YEH, SHI-HUI<br />

Chang Gung Institute <strong>of</strong> Technology, Taiwan<br />

yangkd.yeh@msa.hinet.net<br />

ZAK, KATELYN<br />

Ohio State University, Columbus, USA<br />

zak.27@osu.edu<br />

LIST OF PARTICIPANTS<br />

55<br />

RIJKERS, GER T.<br />

St. Antonius Hospital, Dept. Medical Microbiology, The Netherlands<br />

g.rijkers@antoniusziekenhuis.nl<br />

SALCEDO DOMÍNGUEZ, JAIME<br />

Facultad de Farmacia, Universidad de Valencia, Spain<br />

jaime.salcedo@uv.es<br />

SCHOEMAKER, RUUD<br />

Friesland Campina, Deventer, The Netherlands<br />

ruud.schoemaker@frieslandcampina.com<br />

SCHWAB, CLARISSA<br />

University <strong>of</strong> Vienna, Austria<br />

clarissa.schwab@univie.ac.at<br />

SOETAERT, WIM<br />

Ghent University, Belgium<br />

wim.soetaert@ugent.be<br />

STAHL, BERND<br />

Danone Research, Friedrichsdorf, Germany<br />

bernd.stahl@danone.com<br />

TANAKA, HIDENORI<br />

Carlsberg Laboratory, Copenhagen, Denmark<br />

tanaka@crc.dk<br />

THEROUX, JOHN<br />

<strong>Glycom</strong> A/S, Kongens Lyngby, Denmark<br />

jt@glycom.com<br />

THURL, STEPHAN<br />

Fulda University <strong>of</strong> Applied Sciences, Fulda, Germany<br />

stephan.thurl@lt.hs-fulda.de<br />

VAN LEUSEN, ELLEN<br />

Friesland Campina, Beilen, The Netherlands<br />

ellen.vanleusen@frieslandcampina.com<br />

WALKER, CAREY D.<br />

Mead Johnson Nutrition, Evansville, IN, USA<br />

carey.walker@mjn.com<br />

WEICHERT, STEFAN<br />

Children's University Hospital, Mannheim, Germany<br />

stefan.weichert@medma.uni-heidelberg.de<br />

WU, SHUAI<br />

University <strong>of</strong> Davis, CA, USA<br />

shuwu@ucdavis.edu<br />

YANG, BETSY YAH-HAN<br />

University <strong>of</strong> North Carolina, Durham, USA<br />

yaha727@gmail.com<br />

YUE, KE<br />

Justus-Liebig University <strong>of</strong> Giessen, Germany<br />

yueke1983@yahoo.com.cn<br />

ØSTERGAARD, METTE VIBERG<br />

University <strong>of</strong> Copenhagen, Department <strong>of</strong> <strong>Human</strong> Nutrition, Denmark<br />

mevo@life.ku.dk


NOTES


NOTES

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!