01.07.2013 Views

chapter 2 - Bentham Science

chapter 2 - Bentham Science

chapter 2 - Bentham Science

SHOW MORE
SHOW LESS

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

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

Synthesis and Biological Applications of<br />

Glycoconjugates<br />

Editors<br />

Olivier Renaudet and Nicolas Spinelli<br />

University of Grenoble<br />

France


eBooks End User License Agreement<br />

Please read this license agreement carefully before using this eBook. Your use of this eBook/<strong>chapter</strong> constitutes your agreement<br />

to the terms and conditions set forth in this License Agreement. <strong>Bentham</strong> <strong>Science</strong> Publishers agrees to grant the user of this<br />

eBook/<strong>chapter</strong>, a non-exclusive, nontransferable license to download and use this eBook/<strong>chapter</strong> under the following terms and<br />

conditions:<br />

1. This eBook/<strong>chapter</strong> may be downloaded and used by one user on one computer. The user may make one back-up copy of this<br />

publication to avoid losing it. The user may not give copies of this publication to others, or make it available for others to copy or<br />

download. For a multi-user license contact permission@bentham.org<br />

2. All rights reserved: All content in this publication is copyrighted and <strong>Bentham</strong> <strong>Science</strong> Publishers own the copyright. You may<br />

not copy, reproduce, modify, remove, delete, augment, add to, publish, transmit, sell, resell, create derivative works from, or in<br />

any way exploit any of this publication’s content, in any form by any means, in whole or in part, without the prior written<br />

permission from <strong>Bentham</strong> <strong>Science</strong> Publishers.<br />

3. The user may print one or more copies/pages of this eBook/<strong>chapter</strong> for their personal use. The user may not print pages from<br />

this eBook/<strong>chapter</strong> or the entire printed eBook/<strong>chapter</strong> for general distribution, for promotion, for creating new works, or for<br />

resale. Specific permission must be obtained from the publisher for such requirements. Requests must be sent to the permissions<br />

department at E-mail: permission@bentham.org<br />

4. The unauthorized use or distribution of copyrighted or other proprietary content is illegal and could subject the purchaser to<br />

substantial money damages. The purchaser will be liable for any damage resulting from misuse of this publication or any<br />

violation of this License Agreement, including any infringement of copyrights or proprietary rights.<br />

Warranty Disclaimer: The publisher does not guarantee that the information in this publication is error-free, or warrants that it<br />

will meet the users’ requirements or that the operation of the publication will be uninterrupted or error-free. This publication is<br />

provided "as is" without warranty of any kind, either express or implied or statutory, including, without limitation, implied<br />

warranties of merchantability and fitness for a particular purpose. The entire risk as to the results and performance of this<br />

publication is assumed by the user. In no event will the publisher be liable for any damages, including, without limitation,<br />

incidental and consequential damages and damages for lost data or profits arising out of the use or inability to use the publication.<br />

The entire liability of the publisher shall be limited to the amount actually paid by the user for the eBook or eBook license<br />

agreement.<br />

Limitation of Liability: Under no circumstances shall <strong>Bentham</strong> <strong>Science</strong> Publishers, its staff, editors and authors, be liable for<br />

any special or consequential damages that result from the use of, or the inability to use, the materials in this site.<br />

eBook Product Disclaimer: No responsibility is assumed by <strong>Bentham</strong> <strong>Science</strong> Publishers, its staff or members of the editorial<br />

board for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any<br />

use or operation of any methods, products instruction, advertisements or ideas contained in the publication purchased or read by<br />

the user(s). Any dispute will be governed exclusively by the laws of the U.A.E. and will be settled exclusively by the competent<br />

Court at the city of Dubai, U.A.E.<br />

You (the user) acknowledge that you have read this Agreement, and agree to be bound by its terms and conditions.<br />

Permission for Use of Material and Reproduction<br />

Photocopying Information for Users Outside the USA: <strong>Bentham</strong> <strong>Science</strong> Publishers Ltd. grants authorization for individuals<br />

to photocopy copyright material for private research use, on the sole basis that requests for such use are referred directly to the<br />

requestor's local Reproduction Rights Organization (RRO). The copyright fee is US $25.00 per copy per article exclusive of any<br />

charge or fee levied. In order to contact your local RRO, please contact the International Federation of Reproduction Rights<br />

Organisations (IFRRO), Rue du Prince Royal 87, B-I050 Brussels, Belgium; Tel: +32 2 551 08 99; Fax: +32 2 551 08 95; E-mail:<br />

secretariat@ifrro.org; url: www.ifrro.org This authorization does not extend to any other kind of copying by any means, in any<br />

form, and for any purpose other than private research use.<br />

Photocopying Information for Users in the USA: Authorization to photocopy items for internal or personal use, or the internal<br />

or personal use of specific clients, is granted by <strong>Bentham</strong> <strong>Science</strong> Publishers Ltd. for libraries and other users registered with the<br />

Copyright Clearance Center (CCC) Transactional Reporting Services, provided that the appropriate fee of US $25.00 per copy<br />

per <strong>chapter</strong> is paid directly to Copyright Clearance Center, 222 Rosewood Drive, Danvers MA 01923, USA. Refer also to<br />

www.copyright.com


CONTENTS<br />

Foreword i<br />

Preface ii<br />

List of Contributors iii<br />

CHAPTERS<br />

1. Bacterial Lectins and Adhesins: Structures, Ligands and Functions 3<br />

Anne Imberty<br />

2. Ligands for FimH 12<br />

Thisbe K. Lindhorst<br />

3. Multivalent Glycocalixarenes 36<br />

Francesco Sansone, Gabriele Rispoli, Alessandro Casnati and Rocco Ungaro<br />

4. Solving Promiscuous Protein Carbohydrate Recognition Domains with Multivalent<br />

Glycofullerenes 64<br />

Yoann M. Chabre and René Roy<br />

5. Monovalent and Multivalent Inhibitors of Bacterial Toxins 78<br />

Edward D. Hayes and W. Bruce Turnbull<br />

6. Monovalent and Multivalent Glycoconjugates as High Affinity Ligands for Galectins 92<br />

Sébastien Vidal<br />

7. Combinatorial Libraires of Dendritic Glycoclusters 116<br />

Jean-Louis Reymond and Tamis Darbre<br />

8. Cyclopeptide-Based Glycoclusters 129<br />

Olivier Renaudet, Didier Boturyn and Pascal Dumy<br />

9. Oligonucleotide-Carbohydrate Conjugates 145<br />

Nicolas Spinelli and Eric Defrancq<br />

10. Glycoliposomes and Metallic Glyconanoparticles in Glycoscience 164<br />

Marco Marradi, Fabrizio Chiodo, Isabel García and Soledad Penadés<br />

11. Chemoselective Glycosylation Techniques for the Synthesis of Bioactive Neoglycoconjugates,<br />

Glyconanoparticles and Glycoarrays 203<br />

Francesco Peri<br />

12. Glycosidases in Synthesis of Glycomimetics and Unnatural Carbohydrates 226<br />

Vladimír Křen<br />

13. Diels-Alder Based Synthesis of Glycomimetics 240<br />

Cristina Nativi, Elisa Dragoni, Barbara Richichi and Stefano Roelens


14. Analytical Tools for Protein-Carbohydrate Interaction Studies 255<br />

Cédric Goyer and Pierre Labbé<br />

15. Index 267


FOREWORD<br />

Attachment of carbohydrates in all its forms is a strategy exploited by both nature and those that study nature. Since the<br />

first reviews over 70 years ago the importance of this critically modulating and even defining role has long been<br />

recognized by who those study them. The very presence of carbohydrate units in naturally occurring structures and their<br />

mimetics has a dramatic effect on their physical, chemical and biological properties. Glycoscience is by necessity broad<br />

in the range of techniques that it encompasses and I have argued for over ten years that in this context the sometimesapplied<br />

and somewhat artificial distinction between "chemical" and "biological" techniques is unhelpful.<br />

This has never been the case – we are reaching a stage in the field where remarkable techniques are able to make<br />

even some very large naturally relevant glycoconjugates. Moreover, through these methods and the design of some<br />

remarkable molecular experimental approaches, we are using these glycoconjugates to unpick the many dazzling<br />

and exciting roles that carbohydrates play in Biology and Medicine.<br />

This book highlights this superbly. Drs Renaudet and Spinelli have assembled an outstanding collection of excellent<br />

and relevant reviews written by leading protagonists in the field that complement each other superbly. Together they<br />

provide a wonderful account of the current state-of-the-art.<br />

Vital current targets illustrate what might and will be achieved through both analysis and understanding. Many<br />

pathogens exploit the display of carbohydrates on host cell surfaces to gain entry. Imberty and Lindhorst, in their<br />

<strong>chapter</strong>s, have described some of the most current and relevant examples. Integral to the approaches they have<br />

analyzed is a fundamental molecular analysis of the binding of sugars to proteins. This has been excellently<br />

amplified by the <strong>chapter</strong> of Goyer and Labbé, which describes some of the most pertinent quantitative techniques in<br />

current use, and by the several <strong>chapter</strong>s that show how this understanding can be directly related to application.<br />

These include the design of inhibitors of bacterial toxins, superbly analyzed by Turnbull and Hayes, and ligands for<br />

the important mammalian galectins, the study of which Vidal summarizes superbly.<br />

Use of well-defined scaffolds is an essential element if this study and exploitation is to be precise. Various platforms<br />

have been analyzed here: fullerenes (Roy and Chabre), dendritic clusters (Reymond and Darbre), calixarenes (Casnati et<br />

al.), cyclic peptides (Renaudet et al.) and even nucleic acids (Spinelli and Defrancq). All highlight longer-term<br />

applications, especially in applications that may allow probing of interactions in whole organisms, and these are<br />

especially elegantly drawn out in the <strong>chapter</strong> by Penadés et al. that consider particles of many relevant types.<br />

Typically syntheses of glycoconjugates adopt one of two strategies. The first strategy is the formation of the glycanaglycone<br />

link early, to form, for example, glycosylated building blocks such as glycosides, glycopeptides or<br />

glycosylated dendritic wedges, that may then be assembled. While, of course, chemical methods can create<br />

glycosidic bonds in a number of ways, the use of biocatalysts can bring with it advantages of selectivity and<br />

efficiency; Křen’s <strong>chapter</strong> highlights this utility well. The second strategy is formation of the link later on in the<br />

synthesis once the scaffold for its presentation is in place. Given the instability that may be associated with the link<br />

and the requirements for protection that need to be considered in the use of glycosylated building blocks, it is clear<br />

why the latter has often seemed the most attractive option. Peri has deftly summarized a broad range of methods for<br />

this late stage approach as applied to many useful platform types. Nativi et al. have delineated an emerging area that<br />

could allow access to glycomimetics based on the use of glycals in cycloadditions.<br />

Together, these <strong>chapter</strong>s give a powerful insight into the elucidation of the mechanism by which sugar-binding takes<br />

place, the tools used to probe into it and its consequences. This science has driven and continues to drive the<br />

synthesis and technology of glycoconjugates. These are the new tools of the glycobiology trade and I believe only<br />

the very start of an era of emerging diagnostics and therapeutics.<br />

i<br />

Prof. Ben Davis<br />

University of Oxford<br />

UK


ii<br />

PREFACE<br />

The interactions between carbohydrates and proteins are involved in major physiological and pathological events.<br />

With the recent emergence of glycomics, an increasing number of sophisticated glycosylated structures capable of<br />

mimicking the multivalent display of the cell surface glycocalix has been reported. Besides giving precious<br />

guidelines on the binding parameters that govern these complex biological processes, synthetic glycoconjugates<br />

often revealed considerable interests for diagnostic and therapeutic applications.<br />

Our main motivation to edit Synthesis and biological applications of glycoconjugates was to update the major<br />

advancements in this field through several <strong>chapter</strong>s written by renowned experts who have largely contributed to the<br />

recent progresses. Illustrated with more than 200 colour figures and including literature citations mostly from the<br />

last decade, this book covers the recent chemical methods of glycoconjugates and clearly highlights their diverse<br />

biological properties. Chapter 1 analyzes the structure of lectins from pathogenic bacteria and gives<br />

structure/function relationships that are crucial for the development of high affinity ligands. In Chapter 2<br />

carbohydrate binding by FimH is discussed and modern means for its investigation including photoaffinity labelling<br />

are described. Very recent crystallographic work is also documented that provides an explanation for shearenhanced<br />

binding of type 1 fimbriated E. coli. Chapter 3 summarizes the most convenient methodologies for the<br />

synthesis of glycocalixarenes and describes their impressive aggregation properties and many other biological<br />

applications. Chapter 4 illustrates examples of bacterial and human lectins together with bacterial toxins having<br />

varied number of carbohydrate recognition domains that necessitated multivalent glycoconjugates. It focuses on<br />

glycofullerenes that have been used to describe novel synthetic strategies and possible fit with concomitant lectins.<br />

In Chapter 5 the structures of the cholera and E. coli heat-labile toxins are described. The authors also summarize<br />

the main strategies that have led to the development of monovalent and multivalent inhibitors of these toxins and<br />

they discuss the importance of chelation and protein aggregation as mechanisms of multivalent inhibition. Chapter 6<br />

focuses on the design of high affinity ligands for galectins and provides a better knowledge of the implications of<br />

these proteins in biology. Chapter 7 demonstrates that combinatorial chemistry led to the discovery of glycopeptide<br />

dendrimers as strong ligands for lectins or drug-delivery systems and shows how the amino acid composition of the<br />

dendrimer branches can influence their biological activity. Chapter 8 focuses on the synthesis of a recent class of<br />

glycoclusters displayed on a cyclopeptide platform and highlights their promising biological properties, in particular<br />

as inhibitors or synthetic vaccines. Chapter 9 shows that the cellular delivery and bioavailability of oligonucleotides<br />

can be improved by their conjugation with carbohydrates and also describes the construction of carbohydrate<br />

biochips or glycoclusters using these conjugates. Chapter 10 focuses on glycoliposomes and covalentlyfunctionalized<br />

glyconanoparticles which make use of the “glyco-code” to address specifically pathogens or<br />

pathological-related problems. Chapter 11 focuses on the synthesis of glycoproteins, glycopeptides, glycosylated<br />

natural compounds, carbohydrate-functionalized surfaces and nanoparticles using chemoselective glycosylation. In<br />

Chapter 12 the recent developments in glycosidase-catalyzed synthesis of unnatural semi-synthetic carbohydrate<br />

structures are presented. Chapter 13 reports the synthesis of glycomimetics and glycopeptidomimetics using hetero-<br />

Diels Alder reactions between ,’-dioxothiones and glycals. Chapter 14 aims at describing the usual methods to<br />

characterize protein-carbohydrate interactions, namely inhibition of hemagglutination assay, enzyme-linked lectin<br />

assays, isothermal titration calorimetry, surface plasmon resonance and the more recent atomic force microscopy.<br />

We believe that this Ebook will be of particular interest to a large community of graduate students, researchers and<br />

professionals in academia or industry involved in Glycoscience. We would like to express our sincerest gratitude to<br />

all of the authors who accepted to contribute to this exciting project, by sharing their strong experiences and<br />

knowledge in this research area.<br />

Olivier Renaudet<br />

Nicolas Spinelli<br />

University of Grenoble<br />

France


List of Contributors<br />

Didier Boturyn<br />

Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph Fourier, 38041<br />

Grenoble Cedex 9, France.<br />

Alessandro Casnati<br />

Dip.to di Chimica Organica e Industriale, Università degli Studi, Parco Area delle Scienze 17/A, 43124 Parma, Italy.<br />

Yoann M. Chabre<br />

Pharmaqam – Groupe de Recherche en Chimie Thérapeutique, Université du Québec à Montréal, P.O. Box 8888,<br />

Succ. Centre-ville, Montréal, Québec, Canada H3C 3P8.<br />

Fabrizio Chiodo<br />

Laboratory of GlycoNanotechnology, Biofunctional Nanomaterials Unit, CIC biomaGUNE, Parque Tecnológico de<br />

San Sebastián, Pº de Miramón 182, 20009 San Sebastián, Spain.<br />

Tamis Darbre<br />

Department of Chemistry and Biochemistry, University of Berne, Freiestrasse 3, CH-3012, Berne, Switzerland.<br />

Eric Defrancq<br />

Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph Fourier, 38041<br />

Grenoble Cedex 9, France.<br />

Elisa Dragoni<br />

ProtEra, Polo Scientifico Universita’ di Firenze, viale delle Idee, 22 I-50019 Sesto F.no (FI), Italy.<br />

Pascal Dumy<br />

Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph Fourier, 38041<br />

Grenoble Cedex 9, France.<br />

Isabel García<br />

Laboratory of GlycoNanotechnology, Biofunctional Nanomaterials Unit, CIC biomaGUNE and Biomedical<br />

Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Parque<br />

Tecnológico de San Sebastián, Pº de Miramón 182, 20009 San Sebastián, Spain.<br />

Cédric Goyer<br />

Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph Fourier, 38041<br />

Grenoble Cedex 9, France.<br />

Edward D. Hayes<br />

School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.<br />

Anne Imberty<br />

CERMAV-CNRS (affiliated to Université Joseph Fourier and ICMG), BP 53, 38041 Grenoble cedex 9, France.<br />

Vladimír Křen<br />

Institute of Microbiology, Centre of Biocatalysis and Biotransformation, Academy of <strong>Science</strong>s of the Czech<br />

Republic, Vídeňská 1083, CZ 142 20 Prague 4, Czech Republic.<br />

Pierre Labbé<br />

Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph Fourier, 38041<br />

Grenoble Cedex 9, France.<br />

iii


iv<br />

Thisbe K. Lindhorst<br />

Otto Diels Institute of Organic Chemistry, Christiana Albertina University of Kiel, D-24098 Kiel, Germany.<br />

Marco Marradi<br />

Laboratory of GlycoNanotechnology, Biofunctional Nanomaterials Unit, CIC biomaGUNE and Biomedical<br />

Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Parque<br />

Tecnológico de San Sebastián, Pº de Miramón 182, 20009 San Sebastián, Spain.<br />

Cristina Nativi<br />

Dipartimento di Chimica, Universita’ di Firenze, via della Lastruccia, 3,13 I-50019 Sesto F.no (FI), Italy.<br />

Soledad Penadés<br />

Laboratory of GlycoNanotechnology, Biofunctional Nanomaterials Unit, CIC biomaGUNE and Biomedical<br />

Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Parque<br />

Tecnológico de San Sebastián, Pº de Miramón 182, 20009 San Sebastián, Spain.<br />

Francesco Peri<br />

Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2; 20126<br />

Milano, Italy.<br />

Olivier Renaudet<br />

Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph Fourier, 38041<br />

Grenoble Cedex 9, France.<br />

Jean-Louis Reymond<br />

Department of Chemistry and Biochemistry, University of Berne, Freiestrasse 3, CH-3012, Berne, Switzerland.<br />

Barbara Richichi<br />

Dipartimento di Chimica, Universita’ di Firenze, via della Lastruccia, 3,13 I-50019 Sesto F.no (FI), Italy.<br />

Stefano Roelens<br />

Istituto Metodologie Chimiche (IMC), CNR, Dipartimento di Chimica, Universita’ di Firenze, via della Lastruccia,<br />

3,13 I-50019 Sesto F.no (FI), Italy.<br />

René Roy<br />

Pharmaqam – Groupe de Recherche en Chimie Thérapeutique, Université du Québec à Montréal, P.O. Box 8888,<br />

Succ. Centre-ville, Montréal, Québec, Canada H3C 3P8.<br />

Francesco Sansone<br />

Dip.to di Chimica Organica e Industriale, Università degli Studi, Parco Area delle Scienze 17/A, 43124 Parma, Italy.<br />

W. Bruce Turnbull<br />

School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.<br />

Nicolas Spinelli<br />

Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph Fourier, 38041<br />

Grenoble Cedex 9, France.<br />

Rocco Ungaro<br />

Dip.to di Chimica Organica e Industriale, Università degli Studi, Parco Area delle Scienze 17/A, 43124 Parma, Italy.<br />

Sébastien Vidal<br />

Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, Laboratoire de Chimie Organique 2, UMR 5246,<br />

Université Lyon 1 and CNRS, F 69622 Villeurbanne, France.


Synthesis and Biological Applications of Glycoconjugates, 2011, 3-11 3<br />

Bacterial Lectins and Adhesins: Structures, Ligands and Functions<br />

Anne Imberty *<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 1<br />

CERMAV-CNRS (affiliated to Université Joseph Fourier and ICMG), BP 53, 38041 Grenoble cedex 9, France<br />

Abstract: Infection by bacteria is often initiated by the specific recognition of host epithelial surfaces by adhesins<br />

and lectins. These glycan binding proteins (GBP) are therefore virulence factors that play a role in the first step of<br />

adhesion and invasion. The adhesins are part of organelles, they are generally located at the tip of pili or fimbriae.<br />

On the opposite, soluble lectins occur either as individual proteins, or in association with toxins or enzymes. The<br />

human targets for bacterial adhesins and lectins are mostly fucosylated human histo-blood groups and/or<br />

sialylated epitopes. The binding of lectins and adhesins to host epithelial glycoconjugates is amplified by the<br />

multivalent presentation of the binding sites. The analysis of the available crystal structures of bacterial lectins<br />

and adhesins helps deciphering the structure/function relationship for this important class of proteins. It is a<br />

prerequisite for the development of multivalent high affinity ligands in antibacterial strategies.<br />

Keywords: Lectin, adhesin, crystal structure, bacteria, infection.<br />

LECTINS: UBIQUITOUS PROTEIN WITH A TASTE FOR SUGARS<br />

Lectins are proteins of non-immune origin that bind to specific carbohydrates without modifying them [1]. These<br />

proteins are ubiquitous as they have been identified in all living organisms, from viruses to bacteria, fungi, plants<br />

and animals. As a result of their interactions with glycoproteins, glycolipids and oligosaccharides, lectins are<br />

involved in cell-cell communication; they have the crucial role of deciphering the “glycocode”. Their biological<br />

functions are very diverse. Lectins are important players in the innate immunity system of many invertebrates as<br />

they are able to differentiate between self and non-self carbohydrates. Lectins are particularly abundant in plants,<br />

where they are proposed to be involved in protection against pathogens or feeders. The biological roles of animal<br />

lectins have been more thoroughly investigated; they involve glycoprotein trafficking and clearance, development,<br />

immune defence, fertility and many others.<br />

In pathogenic micro-organims, lectins are mostly involved in host recognition and tissue adhesion [2]. They can be<br />

present as subunit parts of toxins where they target the toxic catalytic unit towards host tissues carrying some<br />

specific glycoconjugates. They can also be part of multiprotein organelles, such as bacterial fimbria (pili), flagella<br />

and viral capsids. Therefore they participate to the adhesion process at the early stage of infection. Soluble lectins<br />

are also expressed as virulence factors by opportunistic bacteria.<br />

Structural studies of lectins and of their binding sites started with the elucidation of the crystal structure of<br />

Concanavalin A, a well characterized legume lectin specific for mannose and glucose [3]. Such structural<br />

investigations yield both the detailed knowledge of the protein structure and the identification of the most important<br />

contacts that are established in the binding pocket between the polypeptide chain and the carbohydrate. This is a<br />

prerequisite for a better understanding of the biological and physical processes of recognition [4]. Three dimensional<br />

structures are also the starting points for many developments in the biotechnological and therapeutical applications<br />

of lectin/carbohydrate interactions.<br />

Lectins are relatively easy to purify due to their carbohydrate binding properties that can be exploited for affinity<br />

purification. Lectins from plants are generally abundant and can be extracted from natural sources, whereas animal<br />

lectins are nowadays generally produced by recombinant approaches. A large number of lectins has therefore been<br />

used for crystallization assays, preferably in the presence of their carbohydrate ligand(s).More than 870 threedimensional<br />

structures (corresponding to 187 different lectins) are now available and gathered in the 3D-lectin<br />

database (http://www.cermav.cnrs.fr/lectines/).<br />

*Address corresponding to Anne Imberty: CERMAV-CNRS (affiliated to Université Joseph Fourier and ICMG), BP 53, 38041 Grenoble<br />

cedex 9, France; E-mail: Anne.Imberty@cermav.cnrs.fr


4 Synthesis and Biological Applications of Glycoconjugates Anne Imberty<br />

Inspection of the database content indicates that most crystal structures have been for the time being, obtained for<br />

plant and animal lectins (Fig. 1). Nevertheless, the number of structural works dealing with viral and bacterial<br />

materials increases rapidly. Interestingly, a strong bias is observed in the nature of the architecture of the lectins. As<br />

indicated in Fig. 1, most lectins adopt a -sandwich fold. The several -sandwiches topologies adopted by lectins are<br />

very different one to each others, but some interesting structural convergences are observed. For example legume<br />

lectins and those intracellular animal lectins which are involved in quality control of glycoprotein synthesis share the<br />

same protein fold [5].<br />

Figure 1: Repartition of the 187 different lectins with structures available in the Protein data bank and accessible in the lectin<br />

database (http://www.cermav.cnrs.fr/lectines/). Lectins are classified as a function of: A) origin; B) fold.<br />

STRUCTURES OF LECTINS FROM PATHOGENIC BACTERIA<br />

Adhesins<br />

Adhesins refer to the lectin domains which are present at the tip of bacterial fimbriae (or pili). Although different<br />

types of pili are observed on bacteria, structural data have been obtained only for those lectins belonging to the twodomain<br />

adhesins (TDA)s from different pathogenic strains of Escherichia coli. These adhesins consist of a pilin<br />

domain and a lectin domain, both presenting an immunoglobulin-fold joined via a short interdomain linker [6]. The<br />

adhesins reported in Table 1 have been characterized in enteropathogenic E. coli (GafD on F17 fimbriae) and<br />

uropathogenic ones (FimH on type 1 fimbriae and PapG on P-pili).<br />

Table 1: Crystal structures representative of the different classes of bacterial adhesins and their specificity.<br />

Bacteria Adhesin Carbohydrate specificity<br />

E. coli PapG<br />

GalNAc13Gal14Gal14Glc<br />

(GbO4 oligosacch.)<br />

Structure<br />

PDB<br />

Ref.<br />

1J8R [7]<br />

E. coli GafD/F17-G GlcNAc 1O9W, 1OIO [8, 9]<br />

E. coli FimH Oligomannose 2VCO [10]<br />

Flexible F17 fimbriae of enterotoxigenic E. coli are capped with GafD adhesins specific for GlcNAc-terminated<br />

glycoconjugates. Uropathogenic E. coli adhesins bind to different parts of the human urinary tract. The long and flexible<br />

P fimbriae expose PapG adhesins specific for galabiose containing the oligosaccharide (Gal1-4Gal) motif whereas<br />

shorter type 1 pili are terminated by FimH that binds oligomannose. Despite the lack of any sequence identity, all these<br />

lectins share the same immunoglobulin-like fold of the two structural components; the corresponding pili are assembled<br />

by the chaperone-usher pathway [11]. The three fimbrial lectins, GafD, FimH and PapG share similar beta-barrel folds<br />

but display different ligand-binding regions and disulfide-bond patterns (Fig. 2).


Bacterial Lectins and Adhesins Synthesis and Biological Applications of Glycoconjugates 5<br />

Figure 2: Crystal structure of the three bacterial adhesins obtained in complex with carbohydrate ligands. A) PapG complexed<br />

with GbO4 (code 1J8R); B) GafD complexed with GlcNAc (1O9W); C) FimH complexed with Man3GlcNAc2 (2VCO).<br />

References as in Table 1. All figures have been prepared using PyMol (DeLano Scientific).<br />

Toxin-Associated Lectins<br />

Bacteria use lectin domains to target another subunit which generally displays some enzymatic activity towards host<br />

cell types. The crystal structures that have been solved illustrate how neural or digestive tissues can be targeted by<br />

lectin domains (Table 2).<br />

Clostridia produce a large number of toxins such as neurotoxins and pore-forming toxins which are responsible for<br />

gangrenes and gastrointestinal diseases. Among these, two families of toxins contain lectin domains specific of<br />

human glycoconjugates.<br />

- Clostridia neurotoxins are well known because of the severe diseases that are caused by the<br />

corresponding bacteria: i.e. tetanus by Clostridium tetani and botulism by C. botulinum. Indeed clostridial<br />

neurotoxins are the most lethal protein toxins for humans but they are also used as therapeutic agents.<br />

These toxins have the particularity to inhibit neurotransmission. The lectin domain, at the C-term of the<br />

holotoxin, binds to complex polysialo-gangliosides present on neurons such as GT1b (Fig. 3A) [12]. After<br />

its translocation, the enzymatically active light chain specifically hydrolyses SNARE proteins thereby<br />

preventing SNARE complex assembly and consequently blocks the exocytosis of neurotransmitter [13].<br />

- The large clostridial cytotoxins (LCT) represent a family of structurally and functionally related<br />

exotoxins from different Clostridium species. It includes C. difficile which is responsible for many cases<br />

of nosocomial antibiotic-associated diarrhea [15]. Toxins A and B (TcdA and TcdB) mono-O-glucosylate<br />

(or mono-O-GlcNAcylate) contain a specific threonine residue of Rho/Ras-proteins, which is essential for<br />

the function of the molecular switches. The LCTs are single-chain protein toxins, comprising three<br />

domains involved in host-binding, translocation and catalysis. The lectin domain is located at the Cterminus<br />

of the toxin and is characterized by polypeptide repeats folds that organize in a solenoid-like<br />

structure [16]. A portion of TcdA repetitive domain has been crystallized complexed to Galα1-3Galβ1-<br />

4GlcNAc – a non human glycan (Fig. 3B) [17]. It has been reported to recognize a variety of other<br />

carbohydrate structures having a lactosamine backbone such as blood group antigens Lewis X and Lewis<br />

Y, both of which being present in human intestinal epithelium.<br />

The human pathogen Staphylococcus aureus secretes many toxins that subvert the human immune system. The<br />

family of staphylococcal superantigen-like proteins (SSLs) contains 14 proteins. Their structures are very similar to<br />

that of the superantigens (SAgs) but they do not stimulate T cells [18]. Two toxins, SSL5 and SSL11, bind to sialic<br />

acid-containing glycoconjugates present on granulocytes and monocytes. The preferred ligand is the


12 Synthesis and Biological Applications of Glycoconjugates, 2011, 12-35<br />

Ligands for FimH<br />

Thisbe K. Lindhorst *<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 2<br />

Otto Diels Institute of Organic Chemistry, Christiana Albertina University of Kiel, D-24098 Kiel, Germany<br />

Abstract: Adhesion of bacteria to glycosylated cells and surfaces is largely facilitated through adhesive organells<br />

projecting from the bacterial surface, which are called fimbriae. The most important fimbriae in Escherichia coli and<br />

in most of the other enterobacteria are the type 1 fimbriae which mediate adhesion in an -D-mannoside-specific<br />

manner. The lectin that is responsible for binding terminal -D-mannosyl residues is called FimH and is located at the<br />

tips of type 1 fimbriae. FimH is known to mediate shear-enhanced adhesion of bacteria to mannosylated cells or<br />

surfaces. Like many adhesins, FimH is a two-domain protein consisting of a pilin domain, that anchors FimH to the<br />

fimbrial shaft, and a distal, N-terminal lectin domain, harboring the carbohydrate binding pocket. A variety of natural<br />

as well as synthetic ligands for type 1 fimbriated bacteria and FimH, respectively, have been tested in order to improve<br />

our understanding of this protein-carbohydrate interaction in cellular adhesion and in an attempt to develop effective<br />

low-molecular weight compounds for an anti-adhesion therapy. Besides tailor-made ligands that have been designed<br />

based on the computer docking, various multivalent cluster mannosides have revealed interesting activities. In this<br />

review, carbohydrate binding by FimH will be discussed and modern means for its investigation including<br />

photoaffinity labelling will be described. In addition, very recent crystallographic work will be documented that<br />

provides an explanation for shear-enhanced binding of type 1 fimbriated E. coli.<br />

Keywords: Bacterial adhesion, type 1 fimbriae, FimH, mannosides, antiadhesives.<br />

INTRODUCTION<br />

In the first half of the 20th century, the term lectin was originally proposed for plant proteins that can agglutinate<br />

erythrocytes (“phytohemagglutinins”) and has later been broadened to include all sugar-specific proteins of nonimmune<br />

origin and lacking any enzymatic activity. Lectins are not only found in plants, but are ubiquitous in<br />

Nature,. Already during the first half of the 20th century it was discovered that many bacteria have the ability to<br />

agglutinate erythrocytes and these findings have prompted more systematic studies on bacterial lectins, that are also<br />

often referred to as adhesins or agglutinins [1]. It was shown that hemagglutination activity is a property that is<br />

expressed by many bacterial species, most commonly by those of the Enterobacteriaceae family. Hemagglutination<br />

activity of bacteria is almost always associated with the presence of multiple filamentous protein appendages<br />

projecting from the surface of bacteria that are called fimbriae (from the Latin word for ‘thread’) or pili (from the<br />

Latin word for ‘hair’). Gram-negative bacteria carry several hundreds of these adhesive organelles on their surface<br />

which might be of different Nature, and specificity, respectively.<br />

Studies on bacterial fimbriae have been ongoing since the 1950ies since most bacteria depend on expression of<br />

specialized adhesive organelles on the bacterial cell surface to mediate attachment to target tissues [2]. Several types of<br />

fimbriae have been identified and classified according to their carbohydrate specificity [3]. In Escherichia coli, besides<br />

type 1 fimbriae, P fimbriae (specific for Gal1,4Gal), S fimbriae (specific for Neu5Ac2,3Gal) and type G fimbriae<br />

(specific for GalNAc) are found. Oral Actinomyces express type 2 fimbriae that are specific for -galactosides. The best<br />

characterized pili, however, are type 1 fimbriae, that are one of the most abundant surface structures both in pathogenic<br />

and non-pathogenic Gram-negative bacteria and specific for terminal -mannosyl residues. Mannose specificity is<br />

mediated by a lectin called FimH forming a minor component of the fimbrial protein complex [4, 5].<br />

TYPE 1 FIMBRIAE<br />

Type 1 fimbriae serve as extremely efficient adhesion tools for bacteria inhabiting diverse environments, including<br />

biotic and abiotic surfaces [6]. They are common throughout the Enterobacteriaceae and found in Escherichia coli,<br />

*Address correspondence to Thisbe K. Lindhorst: Otto Diels Institute of Organic Chemistry, Christiana Albertina University of Kiel, D-24098<br />

Kiel, Germany; E-mail: tklind@oc.uni-kiel.de


Ligands for FimH Synthesis and Biological Applications of Glycoconjugates 13<br />

Klebsiella pneumoniae, Pseudomonas aruginosa, Salmonella spp., Serratia marcencens, and Shigella felneri. Type<br />

1 pili are present in at least 90% of all known uropathogenic strains of Escherichia coli (UPEC), which are the main<br />

cause of urinary tract infections in humans, and they have been shown to be important virulence and pathogenicity<br />

factors [7, 8]. They mediate agglutination of guinea pig red blood cells in an -mannose-inhibitable manner and are<br />

responsible for bacterial binding to a wide range of glycoproteins carrying one or more N-linked high-mannose type<br />

oligosaccharides [9].<br />

Type 1 fimbriae are uniformly distributed on the bacteria, commonly between 100 and 400 per cell. Their length<br />

varies between 0.1 to 2 micrometer, their width is ~7 nm. They are composite structures in which a short tip-fibrillar<br />

structure containing the mannose-specific adhesin FimH is joined to a rod composed predominantly of FimA<br />

subunits. At least two genetically distinct type 1 fimbriae with distinct molecular compositions and receptor binding<br />

profiles exist, the E. coli type 1 fimbriae (type 1 E ) and Salmonella type 1 fimbriae (type 1 S ) [10]. In this review, the<br />

E. coli type 1 E fimbriae will be referred to simply as type 1 fimbriae.<br />

Type 1 fimbriae are relatively rigid, highly oligomeric, proteinaceous appendages, anchored to the outer membrane<br />

of Gram-negative bacteria. Individual fimbriae can be seen extending at various angles from the cell surface in<br />

electron micrographs of type 1 fimbriated bacteria. They consist of a cylindrical rod comprising repeating<br />

immunoglobulin-like (Ig) FimA pilin subunits, and a short and stubby tip, named fibrillum [11]. They are assembled<br />

by the chaperone/usher pathway (vide infra) and in their mature form the Ig fold of every subunit is completed by an<br />

amino-terminal extension from a neighbouring subunit in a process termed “donor strand exchange” (DSE).<br />

Type 1 fimbriae can easily be visualized in the electron microscope as thin structures extending from the bacterial<br />

cell surface (Fig. 1). The rod of type 1 fimbriae is a right-handed helical structure with 27 FimA subunits in the 19.3<br />

nm helical repeat (eight turns) and a 2.1-2.5 nm-wide central axial hole. Type 1 fimbriae consist of some hundreds<br />

to thousands of FimA subunits and of a few percent minor fimbrial components including the fimbrial adhesin FimH<br />

that is responsible for receptor binding. Like the other components of type 1 fimbriae the FimH adhesin is encoded<br />

in the fim gene cluster.<br />

Figure 1: Electron micrograph of type 1 fimbriated Escherichia coli bacteria; zoomed in on the right.<br />

THE fim GENE CLUSTER<br />

Type 1 fimbriae are encoded by the fim gene cluster including all genes that are necessary for the expression and<br />

assembly of type 1 fimbriae. The fim operon contains an invertible region and 9 genes, fimA to fimI, of which four<br />

gene products, FimA, FimF, FimG, and FimH, form the fimbrial rod (Fig. 2) [12, 13]. FimA is the major subunit of<br />

type 1 fimbriae and FimH is the adhesin. Proteins FimB, FimC, FimD, and FimE are needed in pilus assembly. The<br />

function of FimI is somewhat uncertain so far, however, when lesions in fimI were created by site-directed<br />

mutagenesis a piliation-negative phenotype was obtained. This finding suggests that the product of the fimI gene is<br />

necessary for E. coli type 1 pilus biosynthesis [14].


14 Synthesis and Biological Applications of Glycoconjugates Thisbe K. Lindhorst<br />

Figure 2: The gene cluster that is required for type 1 fimbriae assembly comprises 9 genes, fimA to fimH. The genes depicted in<br />

bold encode the proteins FimA, FimF, FimG, and the adhesin FimH, which together constitute the fimbrial fibre.<br />

Each type 1 pilus is composed of 500 to 3000 copies of FimA and a short linear tip fibrillum formed by the adhesin<br />

FimH and the minor subunits FimF and FimG. Unlike all other subunits of type 1 fimbriae, FimH features an aminoterminal<br />

lectin domain, which mediates -D-mannose-specific binding to glycoprotein receptors.<br />

PILUS ASSEMBLY: THE CHAPERONE/USHER PATHWAY<br />

Attachment to host cells via adhesive surface structures is a prerequisite for the pathogenesis of many bacteria.<br />

Uropathogenic Escherichia coli assemble P and type 1 pili for attachment to the host urothelium. Assembly of these<br />

pili, like assembly of over 25 other adhesive organelles in Gram-negative bacteria, requires the conserved<br />

chaperone/usher pathway, in which a periplasmic chaperone controls the folding of pilus subunits and an outer<br />

membrane usher provides a platform for pilus assembly and secretion.<br />

Figure 3: Schematic representation of the chaperone-usher pathway for assembly of E. coli type 1 pili. Subunits are represented<br />

as oval shapes colored by subunit type. The two-domain FimH adhesin at the fimbrial tip is colored in green, FimA in light blue<br />

and the periplasmic chaperone and the OM usher are colored in yellow and blue, respectively. Blowout topology diagrams of<br />

donor-strand complemented (lower right) and donor-strand exchanged (upper right) subunits are shown. The subunit’s Nterminal<br />

extension is labeled (Nte). Reprinted from Cell, 133 / May 16, H. Remaut, C. Tang, N. S. Henderson, J. S. Pinkner, T.<br />

Wang, S. J. Hultgren, D. G. Thanassi, G. Waksman, H. Li, Fiber Formation across the Bacterial Outer Membrane by the<br />

Chaperone/Usher Pathway / 641, Copyright (2008), with permision from Elsevier.


36 Synthesis and Biological Applications of Glycoconjugates, 2011, 36-63<br />

Multivalent Glycocalixarenes<br />

Francesco Sansone, Gabriele Rispoli, Alessandro Casnati * and Rocco Ungaro<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 3<br />

Dip.to di Chimica Organica e Industriale, Università degli Studi, Parco Area delle Scienze 17/A, 43124 Parma, Italy<br />

Abstract: Calixarenes, the cyclic oligomers obtained by condensation of phenols or resorcinols with aldehydes, are<br />

ideal scaffolds for the construction of multivalent glycosylated ligands with unique properties. Thanks to the<br />

possibility that we can vary their size, valency and conformation and finely tune the topology of the saccharide units<br />

in the space, a wide variety of glycocalixarenes could have been prepared in the last 15 years. In this account, after a<br />

brief review on the basic chemistry of calixarenes, we will focus the attention on the most convenient methodologies<br />

used so far for the synthesis of glycocalixarenes and on their aggregation and biological properties. Glycocalixarenes<br />

often show, in fact, an impressive ability in the recognition and inhibition of carbohydrate binding proteins (lectins)<br />

disclosing remarkable multivalent effects. The amphiphilic character, due to the simultaneous presence of highly<br />

hydrophilic saccharide units and of a lipophilic aromatic backbone, confers to glycocalixarenes a marked tendency to<br />

self-aggregate in water solution. A quite unique peculiarity of these glycoclusters is related to their combined ability<br />

to firmly bind small organic molecules and to strongly and selectively interact with lectins, which makes them ideal<br />

candidates for tomorrow’s site-specific drug-delivery systems. Other important biological functions have been<br />

envisaged to be influenced by the multivalency of glycocalixarenes which span gene-delivery, inhibition of tumor<br />

cell migration and proliferation to stimulation of the immunogenic system.<br />

Keywords: calixarenes, multivalent effect, lectin inhibition, tumor targeting, cell transfection.<br />

INTRODUCTION<br />

Saccharides are not only fundamental chemical energy sources and structurally important elements, but also play a<br />

fundamental role in a wide range of biological processes [1]. Many important physiological and pathological events,<br />

such as intercellular communication, cell trafficking, immune response, infections by bacteria and viruses, growth and<br />

metastasis of tumour cells, occur due to the recognition of carbohydrate moieties by the corresponding cell protein<br />

receptors. These proteins, which lack immune and enzymatic activity, are known as lectins [2]. The communication<br />

between carbohydrates and lectins is so specific that it is frequently referred to as Sugar Code [3]. Interestingly, lectins<br />

are frequently characterized by the presence of equivalent binding sites for carbohydrates so that, although the affinity of<br />

a single saccharide unit for the receptor is usually low, the simultaneous presentation of several proper and identical<br />

glycoside units to lectins, ensures a strong and specific interaction. This phenomenon has been named glycoside cluster<br />

effect [4], a particular and widely spread case of multivalency [5], and the multivalent neoglycoconjugates which are<br />

synthesised to interfere with all these important phenomena are also called glycoclusters.<br />

MULTIVALENCY<br />

Multivalency is the ability of a particle (or molecule) to bind another particle (or molecule) via multiple, and<br />

simultaneous noncovalent interactions (Fig. 1) [5]. The valency is the number of ligating functionalities of the same<br />

or similar type connected to each of these entities.<br />

Even when the single interactions are rather weak (with dissociation constants Kd in the mM range), thanks to the<br />

simultaneous operation of multiple attractive interactions, multivalency is usually characterised by high<br />

thermodynamic (Kd in the subnanomolar range) and kinetic stability. Nowadays it is clearly ascertained that<br />

multivalency controls many important biological processes [1, 5]. Nature exploits multivalency to convert relatively<br />

weak interactions (e.g. carbohydrate-protein interactions) into strong and specific recognition events. It is therefore<br />

clear why, in the latest years, Supramolecular Chemistry, the chemistry of noncovalent interactions, became more<br />

*Address correspondence to Alessandro Casnati: Dip.to di Chimica Organica e Industriale, Università degli Studi, Parco Area delle Scienze<br />

17/A, 43124 Parma, Italy; E-mail: casnati@unipr.it


Multivalent Glycocalixarenes Synthesis and Biological Applications of Glycoconjugates 37<br />

and more interested in applying the multivalency concepts to the recognition of biological important molecules [6-8]<br />

and to nanotechnology [9]. Interestingly, supramolecular systems, often being simpler than the natural ones, can help<br />

the understanding and quantitative description of the multivalent effect [10]. Although multivalent ligands may<br />

remarkably differ for their topology (Fig. 2), they generally consist of a main core, called scaffold, which bears several<br />

covalent connections, linkers or spacers, to the peripheral ligating (binding) units. In principle, any multivalent scaffold<br />

can be used, from those having low valency such as benzene derivatives, monosaccharides, transition metal complexes,<br />

azamacrocycles, cyclodextrins or calixarenes to high valency ones such as dendrimers, polymers, peptoids, proteins,<br />

micelles, liposomes, and self-assembled monolayers (SAMs) on nanoparticles or surfaces (Fig. 2).<br />

Figure 1: a) Monovalent and b) multivalent complex formation.<br />

Figure 2: Different types of multivalent ligands. a) 1-D finite and linear arrangement; b) polymers/peptoids; c) 2-D<br />

cyclic/macrocyclic structures; d) 2-D self-assembled monolayers (SAM) on Au/quartz; e) 3-D cavity containing scaffolds<br />

(cyclodextrins/calixarenes); f) nanoparticles/dendrimers/liposomes.<br />

A quite useful approach to describe multivalent binding and based on the additivity of the free energies [11, 12] was<br />

proposed, where the standard binding free energy for the multivalent interaction G ° multi is<br />

G ° multi = nG ° mono + G ° interaction<br />

(eq. 1)<br />

where G ° mono is the standard binding free energy of the corresponding monovalent interaction, n is the valency of<br />

the complex and G ° interaction is the balance between favourable and unfavourable effects of tethering.<br />

A more qualitative but often rather helpful parameter, named or enhancement factor, was introduced by<br />

Whitesides [5].<br />

It is defined as = Kmulti/Kmono<br />

(eq. 2)<br />

where Kmulti and Kmono are the association constants for the multivalent and monovalent complexes, respectively.


38 Synthesis and Biological Applications of Glycoconjugates Sansone et al.<br />

A similar parameter is the relative potency (rp) often used to compare the IC50 (concentration of a<br />

substance/inhibitor needed to inhibit a given biological process by half) of a multivalent (IC50 mult ) and a monovalent<br />

(IC50 mono ) ligand. It is defined as<br />

rp = IC50 multi /IC50 mono<br />

(eq. 3)<br />

If the valency n of the cluster is known, the enhancement factor and the relative potency rp can be normalised to n,<br />

giving rise to the parameters /n and rp/n which are also quite useful to compare the efficiency of ligands having<br />

different topology, valency and linker. Ligands exhibiting high /n or rp/n values (> 1) are efficient<br />

ligands/inhibitors and show a positive multivalent effect, while low /n or rp/n values (< 1) clearly indicate a lower<br />

affinity/inhibition than the monovalent ligand and therefore are characterised by a negative multivalent effect.<br />

Kitov and Bundle [12] described a more rigorous approach to evaluate a multivalent binding process (Fig. 3). In this<br />

model, the standard free energy of the multivalent interaction, G ° avidity, is a function of three terms G ° inter (for the<br />

first intermolecular step), G ° intra (for the second intramolecular step) and a statistical term S ° avidity, namely avidity<br />

entropy, calculated on the basis of the topology and valency of the complex. The avidity entropy can increase<br />

rapidly with the valency of the complex, always favouring binding and widely counterbalancing the loss of<br />

conformational entropy which takes place during multivalent complex formation. Very important is the choice of the<br />

linker between the scaffold and the ligating units. It should be of the proper length to allow the simultaneous binding<br />

of all the ligating groups, without generating enthalpic unfavourable strains (enthalpically diminished binding).<br />

Figure 3: Inter- and intramolecular steps for the formation of a multivalent complex or of an intermolecular aggregate.<br />

CALIXARENES AS MULTIVALENT SCAFFOLDS<br />

Calixarenes recently turned out to be an important class of scaffolds for the synthesis of multivalent ligands to be<br />

used both in bioorganic chemistry and nanotechnology. The name calixarene [13], originally attributed by David C.<br />

Gutsche in 1978 to indicate the [1n]-metacyclophane (I) obtained by the condensation of para-substituted phenols<br />

and formaldehyde (Fig. 4), is due to the cup-like shape of these macrocycle which resembles that of a greek vase<br />

(Fig. 5b) named calix in latin. This name was later on associated also to other macrocyclic compounds having<br />

similar structure such as resorcinarenes (or calixresorcinarenes or resorcarenes), thiacalixarenes, oxacalixarenes,<br />

azacalixarenes and, more recently, calixpyrroles and pillarcalixarenes [6, 13-17].


64 Synthesis and Biological Applications of Glycoconjugates, 2011, 64-77<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 4<br />

Solving Promiscuous Protein Carbohydrate Recognition Domains with<br />

Multivalent Glycofullerenes<br />

Yoann M. Chabre and René Roy *<br />

Pharmaqam – Groupe de Recherche en Chimie Thérapeutique, Université du Québec à Montréal, P.O. Box 8888,<br />

Succ. Centre-ville, Montréal, Québec, Canada H3C 3P8<br />

Abstract: Studies on multivalent carbohydrate protein interactions critically depend on the nature of protein’s binding<br />

sites, their number and also their relative three dimensional orientations. Although a wide range of neoglycoconjugates<br />

including glycodendrimers has been synthesized to address this issue, no systematic rules exist yet that can predict the<br />

optimal shapes, size, and number of required exposed carbohydrate ligands. This <strong>chapter</strong> will illustrate a few examples<br />

of bacterial and human lectins together with bacterial toxins having varied number of carbohydrate recognition domains<br />

that necessitated multivalent glycoconjugates. In particular, fullerenes, because of their particular physical properties,<br />

have been used to describe novel synthetic strategies and possible fit with concomitant lectins.<br />

Keywords: glycofullerenes, multivalent inhibitors, glycosidase inhibitors, Bingel's cyclopropanation, Prato reaction,<br />

Click chemistry.<br />

INTRODUCTION<br />

The past few years have witnessed the discovery, development and, in some cases, large-scale production and<br />

manufacturing of novel materials that lie within the nanometer size scale. More particularly, the incorporation of<br />

nanotechnologies in early-stage development of new drugs, diagnostics, or therapeutics constitutes a powerful addition<br />

to the arsenal of classical macromolecular structures. Consequently, this strategy opens new avenues for the original<br />

and efficient design of adapted nanomedicines. In this context and owing to their three-dimensional architectures,<br />

fullerenes constitute promising candidates for novel nanometric constructs having biological properties and offering<br />

challenging scaffolds toward multivalent presentation of biologically relevant saccharide motifs.<br />

Fullerenes, the third allotropic form of carbon along with graphite and diamond, are an original class of spherically<br />

shaped molecules made exclusively of carbon atoms. They have generated much enthusiasm and numerous research<br />

efforts during the past few years [1]. Hence, the chemical and physical features of C60, also named<br />

Buckminsterfullerene, the most representative example among the fullerenes, have extensively been explored. Their<br />

intrinsic properties such as their size, hydrophobicity, three-dimensionality, and electronic properties have made<br />

them very promising nanostructures, offering interesting features at the interface of various scientific disciplines,<br />

ranging from material sciences [2] to biological and medicinal chemistry [3-5].<br />

In order to demonstrate and illustrate their attractive potential in the foregoing applications, an array of studies has<br />

been recorded, including cytotoxicity investigations on both unmodified and functionalized fullerenes. Initial results<br />

have indicated that these novel and fascinating architectures were not carcinogenic when applied to the skin, nor did<br />

they affect the proliferation and the viability of cells when they are internalized. Hence, despite cases of dosedependent<br />

toxicity reported for certain related derivatives, early observations point to pledge for applications in<br />

several arenas such as: DNA cleavage, photodynamic therapy, enzymatic inhibition, antiviral, antibacterial, and<br />

antiapoptotic activity [4-6]. However, the total lack of solubility in aqueous or physiological media constitutes<br />

severe drawbacks for their quick and efficient development as suitable biomolecular carriers. In order to circumvent<br />

the natural repulsion of fullerenes towards water, numerous methodologies have been adopted, including their<br />

entrapment into tailored microcapsules, suspension with the help of co-solvents, and chemical derivatization,<br />

notably their introduction onto peripheral solubilising appendages. Furthermore, it has been shown that the<br />

*Address correspondence to Rene Roy: Pharmaqam – Groupe de Recherche en Chimie Thérapeutique, Université du Québec à Montréal, P.O.<br />

Box 8888, Succ. Centre-ville, Montréal, Québec, Canada H3C 3P8; E-mail: roy.rene@uqam.ca


Solving Promiscuous Protein Carbohydrate Recognition Domains Synthesis and Biological Applications of Glycoconjugates 65<br />

multivalent presentation of polar groups around the fullerene spheres can prevent clustering phenomena in<br />

reasonably dilute solutions and consequently increase the hydrosolubility of the resulting conjugates. In this context,<br />

a variety of chemical functionalities has been utilized to increase both the hydrophilicity (with groups such as OH,<br />

CO2H, NH2, quaternary ammonium, and cyclodextrin) and to prepare novel compounds possessing valuable<br />

biological and pharmacological activity. Diversified common reactions, including oxygenations, halogenations,<br />

radical and nucleophilic additions, and cycloadditions have been utilized toward this goal [7-9].<br />

THE LOGIC OF MAKING MULTIVALENT CARBOHYDRATE LIGANDS<br />

Amongst the extensive panel of neoglycoconjugates [10], intricate fullerenoglyco-conjugates (also termed<br />

glycofullerenes) in particular display an exquisite combination of appealing properties regarding both watersolubility<br />

and biological significance. The spherical character of fullerenes has provided suitable scaffolds for<br />

multivalent carbohydrate presentation at the periphery of the molecules and valuable reactions have been adapted for<br />

their efficient and controlled covalent attachment.<br />

Since most carbohydrate binding proteins display rather low affinity and generally narrow carbohydrate recognition<br />

domains (CRDs) [11-15] involving fewer than a tetrasaccharide residue, their intrinsic specificities often reside in their<br />

valencies (number of CRDs per protein) together with their various topologies. The number of CRD in this ubiquitous<br />

family of proteins may vary as much as from one (Galectin-3) [16-18] up to fifteen (Shiga toxin) (Fig. 1) [19-21].<br />

Figure 1: Monovalent Galectin-3 with bound LacNAc (PDB 1A3K). Linear dimeric Burkholderia cenocepacia lectin (Bcla,<br />

PDB 2WRA). Rectangular tetrameric Pseudomas aeruginosa PAIL lectin bound to Gal1-3Gal1-4Glc trisaccharide (PDB<br />

2VXJ). Tetrahedral tetrameric Pseudomonas aeruginosa PA-IIL bound to trisaccharide Lewis a (PDB 1W8H). Pentameric<br />

Cholera toxin B subunit (CTB5) bound to five GM1-oligosaccharide (PDB 3CHB). Shiga-like toxin-I complexed with 15 P k<br />

trisaccharide ligands (PDB 1BOS).


66 Synthesis and Biological Applications of Glycoconjugates Chabre and Roy<br />

Hence, the design of glycofullerenes possessing numerous copies and orientation of carbohydrate ligands may help<br />

decipher our understanding of challenging and promiscuous CRDs and to provide assistance in the observed mixed<br />

and matched QSAR-multivalency phenomenon.<br />

The first glycofullerene syntheses were directed toward monovalent architectures that were subsequently tailored<br />

and optimized to generate multivalent glyconanostructures based on C60 where biological properties were enhanced<br />

by taking advantage of the “glycoside cluster effect” [22].<br />

MONOVALENT C60<br />

Vasella et al. first reported the elegant synthesis of monoglycosylated fullerenes via the glycosylidene carbene<br />

precursor 1 which generated enantiomerically pure spiro C-linked glycosylated-C60 derivative 2 (Scheme 1) [23]. A<br />

similarly direct conversion of C60 with various glycosyl azides 3 provided monovalent fullerene glycoconjugates 4-8<br />

via thermal cycloaddition in boiling chlorobenzene [24]. Although this method afforded mixtures of two inseparable<br />

stereoisomers in relatively poor yields (13 to 28%), the generality of the reaction has been demonstrated with a<br />

series of useful mono- (4, D-glucopyranose and 5, D-galactopyranose), di- (6, lactose and 7, maltose), and trisaccharide<br />

(8) glycofullerenes. The adaptation of this direct cycloaddition-based strategy has also furnished<br />

interesting results under ultrasonication conditions, leading to various glycosyl C70 derivatives from various 2azidoethyl<br />

per-O-acetyl glycopyranosides [25]. However, yields for major closed [5,6]-bridged isomers remained<br />

lower than 10% and mixtures of inseparable stereoisomers were unavoidably obtained.<br />

RO RO<br />

RO RO<br />

1<br />

OR<br />

O<br />

OR<br />

OR<br />

O<br />

RO<br />

N<br />

N<br />

2<br />

PhMe<br />

r.t. or 45°C<br />

55%<br />

R 2<br />

AcO<br />

R 1<br />

R 2<br />

AcO<br />

R 1<br />

AcO<br />

OAc<br />

O<br />

AcO<br />

OAc<br />

O<br />

N<br />

C 60<br />

N 3<br />

PhCl, , 7-10h<br />

MeNHCH 2CO 2H (10)<br />

PhMe, <br />

4 R 1 = H , R 2 = OAc (28%)<br />

5 R 1 = OAc , R 2 = H (18%)<br />

6 R 1 = H , R 2 = O--Gal(OAc) 4 (13%)<br />

7 R 1 = H , R 2 = O--Glc(OAc) 4 (18%)<br />

8 R 1 = H , R 2 = O--Mal(OAc) 7 (16%)<br />

Scheme 1: Direct syntheses of monovalent glycofullerenes introduced onto C 60.<br />

3<br />

OR 1<br />

R<br />

RO<br />

2O Me<br />

N<br />

<br />

9<br />

RO<br />

OR<br />

O<br />

RO<br />

O<br />

RO<br />

11 R = R 1 = Bn , R 2 = H (14%)<br />

or<br />

R = R 2 = Bz , R 1 = H (10%)<br />

Shortly afterwards, a three-component approach involving C60, a carbohydrate aldehyde (9) and N-methylglycine 10<br />

(sarcosine) was developed leading to the formation of glycofullerenopyrrolidine monocycloadducts (11, Dgalactopyranoside<br />

or D-glucopyranoside) in low yields (10-14%). The reaction proceeded via 1,3-dipolar cycloaddition<br />

from the intermediate azomethine ylide to C60 (Prato reaction) [26]. The feasibility of this straightforward transformation<br />

was nevertheless demonstrated with the use of a family of 1-deoxy-1-C-formyl derivatives of galactopyranose,<br />

glucopyranose and mannofuranose. Additionally, this methodology has been advantageously adapted toward the<br />

syntheses of C60 derivatives containing a 6-(β-D-glycopyranosylamino)pyrimidin-4-one unit [27].<br />

Alternative approaches using the initial introduction of suitable chemical functionalities onto the C60 fullerene have<br />

been adopted by other research groups, allowing conjugation of the saccharide moieties with complementary<br />

CHO<br />

OR<br />

OR2 OR 1


78 Synthesis and Biological Applications of Glycoconjugates, 2011, 78-91<br />

Monovalent and Multivalent Inhibitors of Bacterial Toxins<br />

Edward D. Hayes and W. Bruce Turnbull*<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 5<br />

School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK<br />

Abstract: Cholera and travellers’ diarrhoea are caused by AB 5 protein toxins that bind to ganglioside GM1 at the<br />

surface of the cells lining the intestine. Inhibition of this protein-carbohydrate interaction would prevent the toxin<br />

from entering the cells, and thus prevents toxin-induced diarrhoea. In this review we will describe the structures<br />

of the cholera and E. coli heat-labile toxins, and summarize the main strategies that have led to the development<br />

of monovalent and multivalent inhibitors of these toxins. A number of key design concepts emerge from these<br />

studies including the importance of pre-organization of the sugar residues within the monovalent ligands, and also<br />

the pre-organization of monovalent ligand groups within larger multivalent ligands. The importance of chelation<br />

and protein aggregation as mechanisms of multivalent inhibition is also discussed.<br />

Keywords: bacterial toxin, multivalency, dendrimer, cholera, inhibitor.<br />

INTRODUCTION<br />

Cholera is a potentially fatal bacterial infection, with symptoms that include ‘rice water’ diarrhoea and vomiting [1]. The<br />

associated fluid loss can reach up to 30 litres per day [2], resulting in severe dehydration and, without treatment, death<br />

can occur within hours [3]. It is still a major world health issue; for example, the recent epidemic in Zimbabwe lasted<br />

almost a year with over 98,000 cases and over 4,000 deaths recorded by July 2009 [4]. In 2007, an epidemic in Angola<br />

resulted in 82,204 cases and 3,092 deaths [5]. Cholera is not only an epidemic threat to the developing world, it is also<br />

endemic in South Asia and sub-Saharan Africa [6]. World Health Organisation (WHO) Figures show that there were<br />

190,103 reported cases of cholera in 2008; an increase of 7.6% compared to 2007, and there was also a 27% increase in<br />

fatal cases [7]. It is believed that reported cases may only correspond to 5-10% of the true number [8].<br />

Cholera is caused by cholera toxin (CT) which is secreted by the Gram-negative bacterium Vibrio cholerae [9]. It is<br />

a water-borne bacterium, and infections generally occur from contaminated food and water sources. As water<br />

sources are often shared by large numbers of people, the disease can spread rapidly through whole communities [9],<br />

and across large geographical areas, thus increasing its impact. The 2008-2009 Zimbabwe outbreak affected all ten<br />

provinces in that country [7], and also spread to both Zambia and South Africa [4].<br />

A closely related disease is travellers’ diarrhoea, which is largely caused by enterotoxigenic Escherichia coli [10].<br />

These E. coli strains produce a protein toxin called heat-labile enterotoxin (LT) [11], which is very similar to cholera<br />

toxin in structure [12], mode of action [13], and in the resulting symptoms [10]. As with cholera toxin, LT causes<br />

diarrhoea, however the effects are less severe [14]. Nevertheless, heat-labile enterotoxin is believed to cause up to 1<br />

billion cases of diarrhoea a year, with 300-400 million of these cases in children under the age of five [2]. The<br />

resulting diarrhoeal dehydration is estimated to be the cause of 300,000-500,000 deaths annually [10].<br />

TOXIN STRUCTURE AND FUNCTION<br />

Cholera toxin and heat labile enterotoxin both have an AB5 structure (Fig. 1) [15, 16], and share 80% sequence<br />

identity in both the A and B-subunits [17].<br />

The A-subunit of CT (CTA) and LT (LTA) is responsible for the toxic activity of the proteins; it comprises two distinct<br />

parts A1 and A2 (Fig. 1b). The A1-component has a toxic enzyme activity, while the A2-component non-covalently<br />

links the A-subunit to the B-subunit (Fig. 1c) via an extended helix [18]. When cholera toxin binds to the cell, the whole<br />

toxin is transferred into the cell via receptor mediated endocytosis (Fig. 2). The toxin is then transported to the<br />

endoplasmic reticulum (ER), at which point the A1-subunit dissociates from the rest of the toxin assembly [19].<br />

*Address correspondence to W. Bruce Turnbull: School of Chemistry and Astbury Centre for Structural Molecular Biology, University of<br />

Leeds, Leeds LS2 9JT, UK; E-mail: w.b.turnbull@leeds.ac.uk


Monovalent and Multivalent Inhibitors of Bacterial Toxins Synthesis and Biological Applications of Glycoconjugates 79<br />

Figure 1: The crystal structure of the cholera toxin protein solved at a resolution of 1.90 Å (Protein Data Bank (PDB) accession<br />

code 1S5E). (a) Elevation view of the AB5 holotoxin (the enzymatically active A1 component is shown in blue, the A2<br />

component is green and the B-subunits are red; (b) The A-subunit showing the peptide that extends through the centre of the B 5<br />

pentamer; (c) plan view of the B5 pentamer.<br />

The process of toxin delivery to the cytosol requires that the A-subunit is first nicked by a protease, and then the<br />

disulfide group that still connects the A1- and A2-peptides must be reduced by peptide-disulfide isomerase in the ER<br />

[20, 21]. The origin of the protease differs for CT and LT. In the case of CT, proteolytic cleavage involves a<br />

protease secreted by V. cholerae [20, 22], whereas the A1-subunit of LT is cleaved by intestinal trypsin [23].<br />

Following cleavage from the toxin assembly, the A1-subunit is translocated to the cytosol, where it catalyses the<br />

NAD-dependent ADP ribosylation of arginine 201 of the stimulatory G protein (Gsα) [21]. This modification locks<br />

the Gsα in its GTP-bound conformation resulting in the continual stimulation of adenylate cyclase. The increased<br />

production of cyclic AMP causes an electrolyte imbalance and water loss into the intestine, leading to potentially<br />

fatal diarrhoea [19, 24-30].<br />

Figure 2: A cartoon depiction of the binding of cholera toxin to the cell and the subsequent transfer of the A-subunit into the cell.<br />

The B-Subunit Lectin and its Carbohydrate Ligand Ganglioside GM1<br />

The B-subunits of CT (CTB) and LT (LTB) are homo-pentamers. Each of the five monomer units comprise two<br />

alpha helices and two three-stranded beta sheets (Fig. 1c) [31, 32], which come together to form a doughnut-shaped<br />

structure, with a central pore into which the C-terminal helix of the A2-subunit extends (Fig. 1a) [33]. The natural<br />

cell surface ligand for both CT and LT is the GM1 ganglioside 1 (Fig. 3c), which comprises a branched<br />

pentasaccharide that is anchored to the surface of intestinal epithelial cells via a ceramide lipid [34]. Each B-subunit<br />

protomer has a single GM1 binding site, allowing CT and LT to bind up to five GM1 gangliosides simultaneously<br />

(Fig. 3a-b). The binding mode of GM1 1 to cholera toxin has been described as a “2-fingered grip” by Merrit et al.<br />

[35, 36]. The “thumb” is the sialic acid sugar residue (coloured red in Fig. 3c) and the “forefinger” is the terminal<br />

galactose (blue) and the N-acetylgalactosamine sugar residue (green) [21].<br />

The binding interaction between GM1 and CTB/LTB has been studied extensively by several research groups [37-<br />

43]. A detailed isothermal titration calorimetry (ITC) study by Turnbull et al. [44] determined that the GM1os ligand<br />

binds CTB with a dissociation constant (Kd) of 40 nM, making it one of the strongest protein-carbohydrate<br />

interactions known. In contrast, all of the mono- or disaccahride fragments of GM1os bind much more weakly; for


80 Synthesis and Biological Applications of Glycoconjugates Hayes and Turnbull<br />

example, the terminal galactose residue displays a 15 mM Kd, which improves only two-fold in the case of the full<br />

Gal-GalNAc “forefinger”. The sialic acid residue binds even more weakly to the protein (Kd ca. 200 mM). Of all the<br />

individual sugar residues, it is the terminal galactose sugar residue that contributes the largest proportion of free<br />

energy of binding when CTB binds to GM1 [44].<br />

Figure 3: The 1.25 Å crystal structure (PDB 3CHB) of the five B-subunits of cholera toxin with GM1 oligosaccharide (GM1os)<br />

bound (shown in blue): (a) plan view; (b) elevation view; (c) Ganglioside GM1 1: the pentasaccharide comprises two galactose<br />

residues (blue), an N-acetylgalactosamine sugar residue (green), a glucose residue (pink), and a sialic acid residue (red). The<br />

pentasaccharide is linked to the cell surface via a ceramide lipid.<br />

Structural Differences Between LT and CT<br />

The widely used abbreviation of heat labile enterotoxin (LT) is slightly ambiguous, as it is used to indentify two<br />

very similar, but distinct proteins: LTh and LTp [13, 45, 46]. LTh is the toxin produced by an E. coli strain isolated<br />

from a human sample [47, 48], while LTp originates from an E. coli strain isolated from porcine samples [49]. They<br />

both share 80% sequence identity with CT in both A- and B-subunits, and sequence analysis of the B-subunits of<br />

LTp (LTBp) and LTh (LTBh) show they have a sequence identity of 96%. From the perspective of designing CT/LT<br />

inhibitors, it is in the GM1 binding site where the most significant difference between the three proteins occurs;<br />

amino acid residue 13 is a histidine in both CT and LTh, whereas in LTp, residue 13 is an arginine (Fig. 4) [50, 51].<br />

Figure 4: Comparison of the GM1 binding sites of CTB (green, PDB 3CHB), LTBh (blue, PDB 2O2L) and LTBp (red, PDB<br />

1EFI). The difference at residue 13 is highlighted.<br />

As the GM1 binding sites of CTB and LTBh are identical, it can be assumed that inhibitors designed to inhibit the<br />

CTB-GM1 interaction will also inhibit the LTBh-GM1 interaction. There is also a high possibility that these<br />

inhibitors would also perturb the LTBp-GM1 interaction. This hypothesis is reinforced by work carried out by Fan<br />

et al. [52] who studied m-nitrophenyl α-D-galactopyranoside as an inhibitor of CTB and LTBp; the crystal structure<br />

of this ligand bound each protein was solved, and it was found that the ligand resided in identical positions in both<br />

cases (Fig. 5).


92 Synthesis and Biological Applications of Glycoconjugates, 2011, 92-115<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 6<br />

Monovalent and Multivalent Glycoconjugates as High Affinity Ligands for<br />

Galectins<br />

Sébastien Vidal *<br />

Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, Laboratoire de Chimie Organique 2, UMR<br />

5246, Université Lyon 1 and CNRS, F 69622 Villeurbanne, France<br />

Abstract: The biological implications of lectins have prompted a large number of research projects at the interface<br />

between biology and chemistry for a better understanding of their roles. Several synthetic high affinity ligands have<br />

been designed in order to inhibit their negative effects such as bacterial or viral infections and cancer. Among these<br />

receptor proteins, galectins are galactose-binding lectins implicated in inflammation or cancer and are important<br />

biological targets for the design of treatment against cancer. The careful design of high affinity ligands for galectins<br />

has been investigated through several studies using either (1) a “medicinal chemistry” approach in which the native<br />

ligand (i.e. galactose) is modified on one or several positions or (2) based on a multivalent approach in which<br />

galactose is repeated n times at the periphery of a core scaffold. Both strategies yielded essential information about the<br />

fundamental aspects of galectin-ligand interactions and provided a better knowledge of the implications of galectins in<br />

biology. The present review with 130 references will focus particularly on the past decade and present the most recent<br />

results obtained in this field for monovalent and multivalent ligands of galectins.<br />

Keywords: Galectins, multivalency, inhibition, ligand, structure-activity relationship.<br />

INTRODUCTION<br />

Biological processes are frequently governed by the biomolecular interaction of host-guest systems either in a<br />

reaction pathway (e.g. enzymes transforming its substrate into a product) or in a recognition system between a<br />

protein and its ligand. Among these particularly important set of binding events, lectin-carbohydrate interactions are<br />

playing a major role in several pathologies such as viral or bacterial infection, cancer metastasis, cell-cell<br />

communication of even fecundation. Lectins [1] are multimeric protein capable of binding selectively to a<br />

carbohydrate motif (i.e. mono- or oligosaccharides) and do not display any enzymatic activity (e.g. hydrolysis of a<br />

glycosidic bond). They are classified in different families according to their binding properties for a specific<br />

carbohydrate. Lectins can be considered as biological targets for the design of therapeutic agents in cancer [2, 3] or<br />

in targeted drug delivery systems [4]. The galactose binding lectins are called galectins and play a prominent role in<br />

several biological pathways. The present review will cover the recent progress for the design of high affinity ligands<br />

of galectins and their potential therapeutical applications. Chemists have developed two main approaches to obtain<br />

potent ligands of these lectins through modifications of the carbohydrate scaffold in order to maximize the<br />

interaction with the lectin’s binding pocket or through multivalent glycoconjugates taking advantage of the “cluster<br />

glycoside effect” [5]. The different reviews cited in this manuscript should therefore be considered in order to reach<br />

a more general overview of this particular topic of galectins and their numerous implications in biology.<br />

GENERAL ASPECTS OF GALECTINS<br />

Different Classes of Galectins<br />

Galectins [6-11] were identified in the late 1970s and further characterized in the early 1980s [12]. They share a<br />

common sequence of roughly 130 amino acids and crystallography has been used to determine their structure [12].<br />

Galectins are proteins defined by at least one carbohydrate recognition domain (CRD) capable of binding selectively<br />

to -galactosides and presenting a minimum sequence homology [10]. They usually bind to N-acetyllactosamine<br />

(LacNAc) or lactose mainly through the recognition of the -galactose residue [13]. There are about 15 mammalian<br />

galectins identified and they can be divided into three families if they display either one or two CRD(s) with the<br />

*Address correspondence to Sebastien Vidal: Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, Laboratoire de Chimie<br />

Organique 2, UMR 5246, Université Lyon 1 and CNRS, F 69622 Villeurbanne, France; E-mail: sebastien.vidal@univ-lyon1.fr


Monovalent and Multivalent Glycoconjugates as High Affinity Synthesis and Biological Applications of Glycoconjugates 93<br />

additional chimera type [9, 14, 15]. Galectins are soluble cytosolic proteins and their quaternary structure depends<br />

on their concentration and environment [15]. Below a concentration of 10 µM, galectins are usually present as noncovalent<br />

symmetric dimers while some members may occur as trimers and tetramers (Fig. 1) [15, 16].<br />

Figure 1: Schematic representation of galectins aggregation (adapted from [16]).<br />

Galectins-1, -2, -5, -7, -10, -11, -13, -14 and -15 are characterized as monomers but their biologically active form is<br />

a monomer for galectins-5, -7 and -10 while galectins-1, -2, -11, -13, -14 and -15 require the formation of soluble<br />

homodimers (Fig. 1A). Galectin-3 is a chimera protein [16] oligomerizing in solution with a C-terminus CRD and a<br />

100-150 proline-glycine rich peptide containing phosphorylation sites important for its biological activity (Fig. 1B).<br />

Nevertheless, in the presence of a glycoconjugate displaying -galactosides, galectin-3 will create multivalent<br />

aggregates at micromolar concentrations [17] as well as for other galectins [18]. Galectins-4, -6, -8, -9 and -12 are<br />

composed of two CRDs separated by a short linker peptide of various lengths (Fig. 1C).<br />

Biological Aspects of Galectins<br />

Galectins have numerous implications in biology and can function either extracellularly with cell-surface glycolipids<br />

or glycoproteins but also intracellularly with cytoplasmic or nuclear proteins. They are therefore involved in<br />

inflammatory diseases [19, 20] or protein trafficking [21] but the main implications of galectins are in cancer as<br />

modulators of tumour progression [22] or in cell growth and apoptosis [23, 24] as for instance urological cancer<br />

[25]. Recent reviews have reported the implications of galectin-1 [26], galectin-3 [27-29], galectin-4 [30],<br />

galectin-7 [31], galectin-8 [32, 33] and galectin-9 [34] in cancer biology.<br />

Potential Applications<br />

Several galectins have been identified as targets for the development of therapeutic agents due to their role in<br />

various pathological disorders. Galectin-1 [35] is expressed in normal and pathological tissues and is implicated in<br />

cell survival and proliferation [36] as well as regulation of immune response, inflammation, allergies, metastasis or<br />

even host-pathogen interactions [37]. Galectin-2 was discovered during the cloning of galectin-1 and is thought to<br />

play a similar function to galectin-1 but in different tissues [38]. The role of galectin-5 is not well established but its<br />

large expression in erythrocytes strongly suggest an implication in these cells [38]. Galectin-6 was discovered<br />

during the study of galectin-4 and both lectins cannot be distinguished in terms of tissue expression [38]. They play<br />

a role in cell adhesion or human colon carcinoma but can also be localized at the plasma membrane and act as a<br />

stabilizing agent [38]. Galectin-7 was initially identified in keratinocytes [39] but also in the oesophagus and oral


94 Synthesis and Biological Applications of Glycoconjugates Sébastien Vidal<br />

epithelia, cornea and thymus [40]. A recent study has highlighted potential applications for the design of anticancer<br />

drugs targeting galectin-7 [41]. Galectin-9 was isolated from spleen tissue of a patient with Hodgkin’s lymphoma<br />

and 50% of patients have antibodies against this galectin [38]. Among these galactose-binding lectins, galectin-3 is<br />

probably the most intensively investigated lectin due to its unambiguously identified role in cancer apoptosis [28,<br />

42], breast cancer [43], metastasis [29] or cancer resistance [44].<br />

Design of High Affinity Ligands of Lectins<br />

The synthesis and discovery of high affinity ligands for lectins in general and galectins more particularly represents<br />

therefore a powerful and reliable approach for the design of potential therapeutic agents against several diseases<br />

such as viral of bacterial infection but also cancer as galectins are highly involved in this pathology.<br />

A first approach has been designed where small molecules can be synthesized and evaluated as ligands of lectins<br />

and their affinity will be improved by changing functional groups of the native carbohydrate recognition unit of the<br />

lectin in a medicinal chemistry approach with structure-activity relationships information. In parallel, multivalent<br />

glycoconjugates have been synthesized [45-47] and identified as powerful ligand of lectins taking advantage of the<br />

concept of multivalency [48-53] and also the so-called “cluster glycoside effect” [5] where several lectins and/or<br />

carbohydrates will come together to enhance the affinity between partners in order to reach improved avidities and<br />

specificities in comparison to monovalent interactions. In both cases, the three-dimensional description of the target<br />

lectin, ideally by crystallography, is almost required for the rational design of such high affinity ligands.<br />

The careful analysis of the binding properties for the ligands of galectins designed requires a large set of<br />

bioanalytical techniques [54]. Frontal affinity chromatography [55, 56] (FAC), hemagglutination inhibition assay<br />

[13, 57-59] (HIA), microarrays [60], flow cytometry [61] or ELISA [62] assays have been used for the<br />

determination of the binding properties of several carbohydrate derivatives towards galectins. Surface plasmon<br />

resonance (SPR) is a powerful technique for studying biomolecular interactions and determining the affinity<br />

properties of ligands [63]. Isothermal titration calorimetry (ITC) [13, 48, 64, 65] will also provide a large set of<br />

data for the comprehensive study of lectin-carbohydrate interactions. A recent study has been reported for the<br />

identification of the carbohydrate-binding preferences of eight human galectins using a carbohydrate microarray<br />

technology [66]. Seventeen oligosaccharides have been synthesized with a thiol-terminated tether at their anomeric<br />

center and then incorporated into maleimide-functionalized microarrays. This technology provides a rapid analysis<br />

and a reliable comparison technique for the determination of the binding properties of lectins in general and more<br />

particularly for the growing number of galectins identified in the recent years [6-11].<br />

Galactose-Containing Mono- and Multivalent Ligands Targeting other Lectins<br />

Even though galectins are attractive biological target for potential therapeutical applications, several galactose-based<br />

ligands have been designed and their binding has been studied towards other lectins. Modified galactosides have<br />

been designed targeting cholera [67] or shiga toxins [68]. Nevertheless, the most abundant literature can be<br />

collected for monovalent galactose derivatives as ligands for galectin-3 (see Section II). Multivalent systems have<br />

also been designed where the positioning of the carbohydrate epitopes is dynamic and can adjust itself to the<br />

targeted lectin using a poly-rotaxane system [69], vesicles [70, 71] or magnetic glycoparticles [72, 73]. Another<br />

series of multivalent macromolecules have been described incorporating galactose units on a various scaffolds such<br />

as a phthalocyanine [74], a fullerene [75], -peptides [76], -peptoids [77], oligonucleotides [78-82] or cubic<br />

shaped silsesquioxanes [83, 84]. Additional multivalent architectures have been reported including a galactose<br />

epitope (sometimes included in a lactose or LacNAc residue) and biological interactions have been studied with<br />

agglutinins (PNA [85, 86], WGA/ECA [87], RCA120 [79-82]), HIV-1 rgp120 [88], selectins [89], bacterial<br />

toxins (CTB and LTBh) [90], bacterial lectins (e.g. PA-IL [91, 92]), the asialoglycoprotein receptor (ASGP-R)<br />

[93, 94] or as gene delivery systems [95, 96]. This short list is not exhaustive but highlights some recent reports in<br />

this field and also the possible applications of high affinity ligands of lectins in bacterial or viral infections.<br />

SMALL MOLECULE INHIBITORS OF GALECTIN-3<br />

Among the several galectins identified as putative biological targets for the design of potential drugs, the most<br />

abundant literature can be found for the study of galectin-3 as a key lectin in cancer. Several studies have been


116 Synthesis and Biological Applications of Glycoconjugates, 2011, 116-128<br />

Combinatorial Libraries of Dendritic Glycoclusters<br />

Jean-Louis Reymond * and Tamis Darbre<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 7<br />

Department of Chemistry and Biochemistry, University of Berne, Freiestrasse 3, CH-3012, Berne, Switzerland<br />

Abstract: Dendrimers, displaying a regularly branched tree-like structure, offer an optimal synthetic platform to<br />

explore multivalency effects, in particular to construct bioactive glycoclusters as ligands for lectins. We have<br />

used combinatorial peptide chemistry to prepare glycopeptide dendrimer libraries as glycoclusters of the general<br />

structure (sugar-XXX)4(LysXXX) 2LysXXXNH 2 (X = alpha-amino acid, Lys = branching lysine residue). Binding<br />

assay was performed on such combinatorial libraries bearing an alpha-C-fucoside endgroup, and led to the<br />

discovery of strong ligands for the fucose specific lectin LecB from Pseudomonas aeruginosa. These dendrimers<br />

are potent inhibitors of biofilms, and require a tetravalent ligand for activity. In a related approach, involving<br />

binding assay to cancer cells using galactosylated glycopeptide dendrimers led to drug-delivery dendrimers. In<br />

both cases the amino acid composition of the dendrimer branches strongly influences the biological activity of the<br />

glycoclusters in addition to the simple multivalency effects, demonstrating the utility of peptide chemistry for<br />

systematically optimizing physicochemical and biological properties of the dendrimers.<br />

Keywords: Combinatorial chemistry, peptides, solid-phase synthesis, C-glycosides, lectins, drug delivery,<br />

galactose, fucose.<br />

INTRODUCTION<br />

Dendrimers, displaying a regularly branched tree-like structure [1-3], offer an optimal synthetic platform to explore<br />

multivalency effects [4, 5]. Their potential to construct bioactive glycoclusters has been broadly recognized as<br />

reviewed in several other <strong>chapter</strong>s of this book. Glycoclusters are particularly suitable as multivalent ligands for<br />

lectins, which are proteins binding to the terminal carbohydrates of glycoproteins or glycolipids by means of<br />

generally weak but specific binding sites [6, 7]. Lectins usually possess multiple binding sites and form oligomers,<br />

which enhances binding to carbohydrate epitopes displayed in polyvalent format on cell surfaces. Glycoclusters<br />

including dendrimers generally bind strongly to lectins by virtue of such multivalency effects [8-13].<br />

In principle glycoclusters can be assembled from a variety of structures. Multivalency effects for such glycoclusters<br />

are computed by comparing their affinity to the specific lectin target with that of a reference monovalent ligand.<br />

However the higher affinity of a glycocluster might be caused not only by multivalency itself, but also by secondary<br />

binding interactions of the glycocluster with the lectin outside the carbohydrate group. Furthermore, the glycocluster<br />

backbone is likely to play a critical role in binding not only by setting the distance between individual carbohydrate<br />

epitopes, but also by determining their relative orientation, and by influencing the global physicochemical<br />

properties, in particular its charge, hydrophobicity and aqueous solubility.<br />

With the aim of addressing the optimization of these parameters in dendritic glycoclusters, we have used<br />

combinatorial peptide chemistry to prepare glycopeptide dendrimers as glycoclusters, expanding on our<br />

investigations of peptide dendrimers as synthetic protein models [14-16]. Peptide dendrimers were pioneered early<br />

on by Denkewalter and coworkers with poly-lysine dendrimers [17] and in the form of multiple antigenic peptides<br />

by Tam and coworkers [18-21]. However in these previous studies a poly-lysine tree was used for the only purpose<br />

of providing multivalency without any attempt to vary the peptide backbone for optimization. Our work performs<br />

such backbone variations in systematic manner using combinatorial solid-phase synthesis of peptide sequences<br />

featuring diamino acids branching points at every 2 nd , 3 rd or 4 th position. In this manner, peptide dendrimers are<br />

produced that can be easily varied at the core, within the branches, and at the branches’ ends (Fig. 1).<br />

*Address correspondence to Jean-Louis Reymond: Department of Chemistry and Biochemistry, University of Berne, Freiestrasse 3, CH-3012,<br />

Berne, Switzerland; E-mail: reymond@ioc.unibe.ch


Combinatorial Libraries of Dendritic Glycoclusters Synthesis and Biological Applications of Glycoconjugates 117<br />

FmocHN<br />

160 mg of Rink<br />

Amide Resin<br />

0.25 mmol/g<br />

1) piperidine, DMF<br />

2) FmocAAOH, BOP, HOBT<br />

3) repeat 1) and 2) 10 times<br />

4) 1), then Ac 2O, py<br />

5) CF 3CO 2H, then HPLC<br />

H 2N<br />

O<br />

HO<br />

X 7<br />

X 8<br />

X 7<br />

X 8<br />

X 7<br />

X 8<br />

X 7<br />

X 8<br />

L2K7 (28 mg, 10 % isolated yield)<br />

X 5<br />

X 6<br />

X 5<br />

X 6<br />

X 7<br />

X 8<br />

X 5<br />

X 6<br />

X 3<br />

X 4<br />

X 3<br />

X 4<br />

X 5<br />

X 6<br />

X 7<br />

X 8<br />

X 7 X 8<br />

X 1<br />

X 2<br />

X 7 X 8<br />

O<br />

NH<br />

HN<br />

O<br />

NH<br />

O<br />

HN H<br />

HN<br />

2N<br />

O<br />

HN<br />

H<br />

O<br />

O<br />

NH<br />

N<br />

NH<br />

NH H2N O O<br />

O NH<br />

HN O<br />

HN<br />

HN<br />

HN<br />

H<br />

O<br />

3N<br />

O NH<br />

O<br />

NH NH2 H<br />

O N<br />

O<br />

HN<br />

O NH2 HO<br />

HO<br />

NH<br />

HN<br />

HN<br />

O HN<br />

HO<br />

O<br />

O NH<br />

O<br />

NH<br />

O HN<br />

NH<br />

NH3 O<br />

NH<br />

O<br />

NH<br />

O<br />

HN<br />

O<br />

HN<br />

O<br />

HN NH<br />

O<br />

NH2 NH<br />

HN<br />

H2N NH<br />

O<br />

O<br />

HN<br />

NH2 HN NH2 H2N HN HN<br />

H<br />

NH O<br />

N<br />

O<br />

O<br />

HN<br />

NH O<br />

HN<br />

O<br />

OH<br />

NH O<br />

HN<br />

N O<br />

H<br />

NH H2N H3N 2<br />

H3N NH3 Figure 1: Synthesis and structure of the 3 rd generation peptide dendrimer L2K7.<br />

Our use of combinatorial libraries of synthetic glycoclusters in the form of peptide dendrimers to discover cytotoxic<br />

and lectin binding ligands is discussed below.<br />

COMBINATORIAL SYNTHESIS, SCREENING, AND DECODING OF DENDRITIC GLYCOCLUSTERS<br />

SPPS of Peptide Dendrimers and their Combinatorial Libraries<br />

Dendrimers are usually prepared by solution phase synthesis and convergent assembly to minimize sterical<br />

hindrance and purification problems. Our approach contradicts these principles: we use a divergent synthesis<br />

protocol on solid support under the standard conditions of solid phase peptide synthesis with Fmoc-protected amino<br />

acid building blocks [22-25] to prepare peptide dendrimers up to the fourth generation and up to 16 end groups [26-<br />

27]. The C-terminal amino acids which are coupled first to the solid support form the core of the dendrimer.<br />

Branches are grown as diamino acids are added along the sequence. A typical 11-step coupling sequence produces a<br />

dendrimer of 3 rd generation with branches two amino acids in length, which contains a total of 37 amino acids and<br />

an average molecular weight of 4,400Daltons, as shown for the aldolase dendrimer L2K7 (Fig. 1) [28].<br />

The isolated yields of peptide dendrimers obtained from Fmoc-SPPS after side-chain deprotection, cleavage from<br />

the solid support and purification by reverse-phase HPLC are comparable to the yields for linear peptides of similar<br />

size. As for linear peptides, the isolated yields are influenced by the amino acid sequence and by the physicochemical<br />

properties of the product. In particular strongly hydrophobic sequences may be difficult or impossible to<br />

purify due to their lack of solubility in any solvent. Overall, many “well-behaved” peptide dendrimers comprising a<br />

good balance of charged, polar, and hydrophobic amino acids, are readily accessible by SPPS. Peptide dendrimer<br />

synthesis is also facilitated by the commercial availability of amino acid building blocks for Fmoc-SPPS at


118 Synthesis and Biological Applications of Glycoconjugates Reymond and Darbre<br />

reasonable prices, which renders the synthesis quite affordable and potentially scalable using the existing industrial<br />

infrastructure for SPPS.<br />

The split-and-mix protocol for assembling peptide libraries by SPPS [29-33], which was a founding experiment in<br />

combinatorial chemistry, can be applied to prepare the corresponding peptide dendrimer libraries on solid support<br />

[34-36]. In these so-called “one-bead-one-compound” (OBOC) libraries, each bead of the solid support carries only<br />

one possible sequence, yet a large number of different sequences are assembled simultaneously in the course of the<br />

synthesis. The number of different sequences accessible in a split-and-mix synthesis is limited by the number of<br />

beads used in the SPPS synthesis batch. A typical synthesis batch of 500 mg of 90 micrometer diameter beads<br />

contains approximately one million beads, which imposes a limit of 100,000 sequences for the library size to ensure<br />

10-fold coverage of sequence space.<br />

Synthesis of Glycopeptide Dendrimers<br />

While the preparation of natural glycopeptides requires sophisticated chemical or enzymatic glycosylation<br />

chemistry, the decoration of synthetic macromolecules with carbohydrates is often more conveniently realized by<br />

reacting preassembled glycosidic building blocks by simpler coupling chemistries. Before its adaptation to<br />

combinatorial chemistry, we first investigated various glycosylation options for peptide dendrimers in a synthetic<br />

study aimed at the dendrimer mediated delivery of colchicine to cancer cells.<br />

Three different strategies were investigated to obtain glycopeptide dendrimers [37]. The first approach used oxime<br />

bond formation between a multivalent aldehyde-terminated peptide dendrimer and an oxyamine functionalized<br />

glycoside, as reported previously for the glycosylation of peptides [38, 39] and peptide dendrimers [40]. The second<br />

approach used reductive alkylation of the free N-terminus with lactose [41, 42], a method previously known for<br />

modifying whole proteins [43-45] and reported for the synthesis of glycodendrimers [46]. The third approach<br />

involved amide bond formation between an acetyl-protected glycosysidic carboxylic acid building block and the Ntermini<br />

of the peptide dendrimer directly on the solid support. Although this third approach required multigram<br />

quantities of carbohydrate building blocks, the synthesis was the most practical because it required only a single<br />

chromatographic purification and yielded products of excellent purity. For example the N-acetyl-glucosamine<br />

terminated peptide dendrimer colchicine conjugate A, which shows selective cytotoxicity to cancer cells, was<br />

obtained by this approach (Fig. 2).<br />

OH OH<br />

O<br />

HO<br />

AcNH<br />

O<br />

O<br />

O<br />

O<br />

HN<br />

H<br />

N<br />

O<br />

NH<br />

NH<br />

O<br />

N<br />

H<br />

N<br />

O<br />

OH<br />

O<br />

AcHN<br />

HO<br />

OH<br />

O<br />

S S<br />

N<br />

H<br />

O<br />

NH<br />

O<br />

MeO<br />

Figure 2: A cytotoxic colchicine glycopeptide dendrimer conjugate.<br />

NH<br />

OH OH<br />

O<br />

HO<br />

AcNH<br />

O<br />

HN<br />

H<br />

N<br />

N<br />

H<br />

NH<br />

O<br />

H<br />

N<br />

OMe<br />

O<br />

O<br />

OMe<br />

N<br />

O HN<br />

O<br />

O<br />

OH<br />

OMe<br />

A<br />

HN<br />

NH2 O<br />

NH<br />

O O<br />

HO<br />

AcNH<br />

O<br />

O O<br />

N<br />

H<br />

OH<br />

OH


Cyclopeptide-Based Glycoclusters<br />

Synthesis and Biological Applications of Glycoconjugates, 2011, 129-144 129<br />

Olivier Renaudet * , Didier Boturyn and Pascal Dumy<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 8<br />

Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph Fourier, 38041<br />

Grenoble Cedex 9, France<br />

Abstract: The emergence of glycomics has deeply stimulated the design of new bioactive glycoclusters over the<br />

past decade. Among the increasing number of multivalent synthetic structures reported so far, cyclopeptide-based<br />

glycoclusters (CBGs) have recently shown promising interest for diverse biological applications. This <strong>chapter</strong><br />

aims at describing the major advances in this field, with a special focus on the inhibition of carbohydrate-protein<br />

interactions and the synthetic vaccines.<br />

Keywords: Cyclopeptide, glycocluster, oxime, combinatorial chemistry, synthetic vaccine, lectin.<br />

INTRODUCTION<br />

Decoding carbohydrate/protein interactions involved in biological events [1, 2] remains crucial for the design of bioactive<br />

molecules. The “glycoside cluster effect” introduced by Lee et al. in 1995 describes that these complex recognition<br />

processes are regulated by simultaneous and cooperative contacts between cluster of glycans and multimeric proteins [3].<br />

While their kinetic and thermodynamic parameters are still not fully understood [4-6], diverse mechanisms such as<br />

chelating, proximity/statistical or clustering effects were proved to promote high selectivity and strong binding<br />

improvement, depending on the structural features of both ligand and protein [7-9]. These findings have inspired bioorganic<br />

chemists to develop diverse molecules capable of mimicking the multivalent display of carbohydrates expressed in living<br />

systems, namely glycoclusters, with the aim to study multivalency effects or to inhibit biological processes [10-12].<br />

Although impressive subnanomolar affinities were sometimes achieved [13], most of studies reported so far clearly<br />

highlight that the design of an optimal structure for an amplified biological effect remains extremely difficult to rationalise.<br />

The usual design to develop biorecognizable glycoclusters consists in grafting multiple copies of carbohydrates onto a<br />

carrier platform, so that they bind several, or ideally, every protein binding sites simultaneously. Since the biological<br />

potency of glycoclusters closely depends on the tridimensional protein structure, a large variety of synthetic structures<br />

exhibiting variable topology, flexibility, valency and sugar distribution were explored. In particular, linear (e.g. polymers<br />

[7, 14], oligonucleotides [15-17]), cyclic (e.g. calixarenes [18, 19], cyclodextrins [20], fullerenes [21, 22]) and branched<br />

platforms (peptide dendrimers [23-25], poly(amidoamine) dendrimers [26]), liposomes or nanoparticles [27, 28] have been<br />

intensively investigated in different biological contexts. The use of cyclopeptides as carrier platforms for carbohydrates<br />

was more rarely described until recent studies have considerably renewed their interests [29]. This <strong>chapter</strong> focuses on the<br />

synthesis of several cyclopeptide-based glycoclusters (CBGs) that revealed promising biological properties.<br />

CYCLOPEPTIDE PLATFORM<br />

Pioneering studies by Mutter et al. on protein mimics have introduced an original concept that addresses crucial<br />

questions on peptide assembly, protein folding and function [30, 31]. For this purpose, conformationally restricted<br />

cyclopeptides were utilized as topological platforms to assemble and stabilize the secondary structure of peptide building<br />

blocks in a native-like folding [32]. Following a similar concept, other sophisticated platforms, such as the<br />

Regioselectively Addressable Functionalized Templates (RAFT) [33], were further used for the construction of<br />

biorecognizable compounds [29, 34].<br />

As illustrated in Fig. 1, a typical cyclopeptide platform is composed of amide bonds in the trans configuration and<br />

contains generally two -turn inducers i.e. L-proline-glycine (Pro-Gly in green, Fig. 1A) that constrained the<br />

structure into an antiparallel -sheet (yellow rubber, Fig. 1B).<br />

*Address correspondence to Olivier Renaudet: Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph<br />

Fourier, 38041 Grenoble Cedex 9, France; E-mail: olivier.renaudet@ujf-grenoble.fr


130 Synthesis and Biological Applications of Glycoconjugates Renaudet et al.<br />

A recognition domain<br />

B<br />

R 1<br />

R 1<br />

R 1<br />

R 1<br />

Gly Lys<br />

Lys Lys Pro<br />

turn turn<br />

Pro Lys<br />

Lys Lys Gly<br />

R 2<br />

R 2<br />

effector domain<br />

Figure 1: A) Chemical drawing and B) molecular model of an example of cyclopeptide scaffold. This platform is composed of<br />

ten amino acids, including two Gly-Pro β-turns and six Lys residues which define two addressable faces, namely “recognition<br />

domain” (upper, R 1 = carbohydrates, peptides) and “effector domain” (lower, R 2 = peptide, oligonucleotide, surface, drug).<br />

Alternating intramolecular hydrogen bonds stabilize the backbone into a locked and rigid cyclic conformation. The<br />

other amino acids into the peptide sequence may be selected to provide regioselective anchoring sites (e.g. lysine<br />

[Lys] as shown in Fig. 1, or aspartic acid [Asp], glutamic acid [Glu], aparagine [Asn], glutamine [Gln], threonine<br />

[Thr] in other cases) pointing to opposite faces of the template backbone. Thus these platforms display two<br />

multivalent addressable faces, defined as “recognition” and “effector” domains once decorated with binding and/or<br />

functional building blocks (Fig. 1B).<br />

Besides their topology and constrained conformation, the utilization of cyclopeptides offers other advantages. Their<br />

preparation indeed benefits from a plethora of technical improvements developed for peptide chemistry (e.g. solid<br />

supports, automation, non-racemizing coupling reagents…) that facilitate the rapid synthesis of their linear peptide<br />

precursor in high scale. More importantly, the large variety of commercial building blocks with orthogonal<br />

protecting groups provides both synthetic versatility and modularity. This ensures the stepwise construction and the<br />

structural integrity of complex macromolecules with tailor-made properties. Finally, it has to be mentioned that<br />

cyclic structures were proved to prevent proteolytic degradation in vivo compared to linear peptides [35], which<br />

strengthen their interest as addressable platforms for biological applications.<br />

CBGs AS CARBOHYDRATE-BINDING PROTEINS LIGANDS<br />

Selectins<br />

In 1996, the group of Kunz used for the first time a cyclopeptide for the multivalent presentation of biologically relevant<br />

carbohydrates [36]. Cycloheptapeptides displaying sialyl-Le x moiety were prepared using a convergent approach from<br />

the linear peptide sequence 1 which was synthesized by solid-phase peptide synthesis (Fig. 2). It contains a D-Ala<br />

residue as a -turn inducer and several protected Asp as anchoring site for the incorporation of sugars. After the<br />

intramolecular cyclization under high dilution then deprotection of the peptide by acidolysis with trifluoroacetic acid<br />

(TFA), a benzylated tetrasaccharide was conjugated to three Asp side chains in 48% yield using the Lansbury<br />

aspartylation reaction. The trivalent asparagine-linked glycoconjugate 2 was finally obtained after final deprotection.<br />

Selectins are transmembrane glycoproteins involved in cell-adhesion processes that are mediated by selective<br />

multivalent binding to Lewis-containing ligands [37]. To evaluate the inhibitory potency of the trivalent sialyl-Le X<br />

glycopeptide 2, cell adhesion assays were performed with recombinant human fusion proteins (E-selectinimmunoglobulin)<br />

in competition to tumor cells displaying the same carbohydrate moiety (cell line HL60). The<br />

synthetic compound 2 was found to inhibit the adhesion of tumor cells 3-fold more efficiently (0.35-0.6 mM) than<br />

the monomeric sialyl-Le X [36]. This modest binding has prompted the same group to design new ligands containing<br />

a flexible tetraethyleneglycol spacer between the sialyl-Le X moiety and the peptide platform [38]. However the<br />

biological evaluation of the divalent cyclopeptide 3 did not show a significant inhibition improvement on the<br />

adhesion of murine neutrophiles to E-selectin.


Cyclopeptide-Based Glycoclusters Synthesis and Biological Applications of Glycoconjugates 131<br />

Figure 2: Synthesis of sialyl-Le x containing cycloheptapeptide conjugates.<br />

Plant Lectins<br />

To overcome the synthetic difficulties related to the synthesis of glycoconjugates, our group developed an oximebased<br />

chemoselective procedure that revealed extremely efficient for the construction of CBGs [39]. This strategy<br />

requires the prior incorporation of mutually reactive aldehyde and oxyamine groups into the molecules to be<br />

conjugated. Only few procedures (i.e. Mitsunobu coupling [40], stereoselective glycosylation using phase transfer<br />

catalysis [41]) were reported to introduce an aminooxy function to the anomeric position of glycans. We developed<br />

a synthetic alternative to afford alpha and beta aminooxy glycosyls starting from glycosyl fluoride donors and Nhydroxyphthalimide<br />

(PhthOH) in the presence of BF3.Et2O as promoter [42] (Fig. 3). This strategy was used<br />

successfully to synthesize biologically relevant carbohydrates (e.g. lactose 4, mannose 7 and fucose 8 [43]) or<br />

tumor-associated carbohydrate antigens (e.g. Tn 9 and Thomsen-Friedenreich (TF) 10 [44]).<br />

Figure 3: Synthesis of aminooxy glycosyls.<br />

With these aminooxy glycosyls in hands, glycoclusters with one or two addressable domains were next designed.<br />

Typically, linear peptides containing a various number of orthogonally protected Lys (e.g. Alloc, Boc, Dde) were<br />

prepared on solid-phase using acido labile resin (e.g. Sasrin TM ) then cyclised in solution through the C-terminal<br />

glycine to prevent epimerization. To design one domain-containing compounds, four Lys pointing above the<br />

platform were deprotected to incorporate serine residues. Successive treatment with sodium periodate generate<br />

glycoxyl aldehyde groups [45]. Fully deprotected aminooxy glycosyls were finally conjugated under mild aqueous


Synthesis and Biological Applications of Glycoconjugates, 2011, 145-163 145<br />

Oligonucleotide-Carbohydrate Conjugates<br />

Nicolas Spinelli * and Eric Defrancq<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 9<br />

Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph Fourier, 38041<br />

Grenoble Cedex 9, France<br />

Abstract: Nucleic acids have been widely investigated as biological tools for various applications including gene<br />

silencing, DNA chips and drug targets. Interactions involving nucleic acids are generally well known making<br />

their rationalization straightforward. However, they generally suffer from a lack of bioavailability. One of the<br />

main strategies to overcome these limitations is the conjugation of oligonucleotides with an appropriate reporter.<br />

As carbohydrates are involved in a huge variety of biological processes including molecular and cell surface<br />

recognitions, oligonucleotide glycoconjugates are perhaps one of the most promising approaches to improving<br />

cellular delivery of oligonucleotides. Other applications of these conjugates have emerged like the use of DNAdirected<br />

immobilization to prepare carbohydrate biochips or the construction of glycoclusters by self organization<br />

of oligonucleotides. This <strong>chapter</strong> summarizes the applications of these conjugates and approaches to prepare them<br />

through some recent examples.<br />

Keywords: Nucleic acids, conjugation, solid-supported synthesis, chemoselectivity.<br />

INTRODUCTION<br />

Nucleic acids have been widely investigated as biological tools especially for gene silencing. They can be used for<br />

targeting mRNA by either antisens [1-3] or RNAi [4] process or double strand DNA by antigen [5] strategy.<br />

However their applications are not just restricted to genetics [6]. They can be used as aptamers [7], for diagnostic<br />

applications [8] including DNA biosensors [9] and aptasensors [10], for designing of nanostructures [11-13] and for<br />

DNA mediated reactions or catalysis [14, 15]. However, their biological applications they suffer from a lack of<br />

bioavailability principally due to their susceptibility to nuclease degradation and their poor cellular uptake. One of<br />

the main strategy to overcome those limitations is the use of oligonucleotide-conjugates [16, 17].<br />

Carbohydrates are involved in various biological recognition events [18-20] such as cell-cell adhesion, fertilization,<br />

immune defense and degradation of blood clots. In particular, study of carbohydrate-protein (lectin) interactions [21]<br />

has received a great interest toward the enhancement of targeting of drugs and gene delivery [22] via receptor<br />

mediated endocytosis. Lectin-carbohydrate interactions are generally very specific, but affinities are generally weak<br />

for monovalent interactions (millimolar range) [23]. These weak affinities are usually overcome by preparing<br />

glycoclusters on various scaffold (see the other <strong>chapter</strong>s) for mimicking the “glycoside cluster effect” [24].<br />

As a consequence, oligonucleotide glyconjugates are perhaps one of the most promising approaches for improving<br />

cellular delivery of oligonucleotides [25]. In addition, other applications of these conjugates involving the<br />

hybridization of oligonucleotides for studying carbohydrate-aromatic stacking, preparation of carbohydrate biochips<br />

or elaboration of glycoclusters have emerged. Preparation and applications of oligonucleotides-glyconjugates has<br />

been reviewed in 2004 [26], so this <strong>chapter</strong> deals with recent advancements concerning methodologies for synthesis<br />

of these compounds through some examples. The different synthetic approaches and corresponding applications are<br />

described in the following sections.<br />

ON-SUPPORT SYNTHESIS<br />

Since most of the protecting groups (e.g. acetyl) used for protection of carbohydrates are also compatible with the<br />

synthetic protocols of oligonucleotide (phosphoramidite or H-phosphonate), the approach consisting in the<br />

*Address correspondence to Nicolas Spinelli: Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph<br />

Fourier, 38041 Grenoble Cedex 9, France; E-mail: nicolas.spinelli@ujf-grenoble.fr


146 Synthesis and Biological Applications of Glycoconjugates Spinelli and Defrancq<br />

incorporation of phosphorus glycosides during oligonucleotide synthesis is perhaps the more straightforward. Solidphase<br />

conjugation of oligonucleotides and solid phase synthesis of glycoconjugates have been reviewed [27, 28].<br />

Several carbohydrate derivatives have been prepared for 5’, in-sequence or 3’ conjugation depending on the purpose<br />

of the conjugate.<br />

Use of Carbohydrate Phosphoramites for 5’-Conjugation<br />

Carbohydrate phosphoramidites can be incorporated at the 5’-end of oligonucleotide. For example, some of them<br />

have been developed in order to elaborate a model system to conduct a systematic study of the carbohydratearomatic<br />

stacking [29]. Models were designed as follows: a self-complementary sequence ( 5’ Carbohydrate-<br />

XCGCGCG 3’ ) carrying benzene nucleotide (X) and a carbohydrate at 5’-end (Fig. 1).<br />

Figure 1: a) Use of DNA base pairing for studying carbohydrate-aromatic stacking. b) Carbohydrate phosphoramidites for the<br />

synthesis of 5’-glycoconjugates.<br />

Sugars were incorporated as the corresponding phosphoramidite synthon; phosphorus function is linked to the sugar<br />

through the anomeric position via an ethylene glycol linker. According to this strategy, several conjugates were<br />

synthesized including -glucose 1, -galactose 2, -2-deoxyglucose 3,-2-deoxyglucose 4 or -L-fucose 5.<br />

Sugar-benzene interactions were estimated measuring thermodynamic parameters of self-complementary duplexes<br />

and geometry of carbohydrate-benzene stacking was estimated by NMR. This study has shown that dispersion<br />

forces and non-conventional hydrogen bonding seems to take place between sugar and benzene ring.<br />

Synthesis of 5’-multivalent glycoconjugates, i.e. oligonucleotides bearing several carbohydrates motif has been<br />

performed with this strategy using branched phosphoramidites [30] (Fig. 2). In a recent work, the influence of the<br />

presentation of carbohydrates has been evaluated in terms of the cellular uptake and cell surface binding via glucose<br />

receptors [31]. Several conjugates were synthesized bearing monovalent glucose 6, maltose 7, maltotriose 8,<br />

branched divalent glucose 9 and tetravalent glucose 10. Monovalent conjugates were obtained using appropriate<br />

carbohydrate phosphoramidite (1, 11 or 12) and branched glycoclusters were obtained using a branching<br />

phosphoramidite 13 and glucose phosphoramidite 1. All these conjugates were labeled with Alexa 488 at their 3’<br />

end in order to perform flow cytometry analysis with two types of cells (HeLa and U87) both in presence and in the<br />

absence of glucose in the culture media.


Oligonucleotide-Carbohydrate Conjugates Synthesis and Biological Applications of Glycoconjugates 147<br />

Figure 2: Monovalent, multivalent 5’-glycoconjugates and phosphoramidites for their preparation.<br />

Adsorption experiments highlighted slight difference of behavior of glycoconjugates in each cell type. For instance,<br />

the binding of tetravalent conjugate 10 was found to be less efficient with Hela cells than that with U87. The best<br />

cellular uptake was found for divalent 9 and maltose 7 conjugates. Conjugates linked through longer spacer (7 and<br />

9) showed better incorporation than these linked through shorter spacers (6 and 8). Surprisingly, tetravalent glucose<br />

conjugate 10 showed a bad incorporation presumably due to cell uptake hindering.<br />

Synthesis of heteroglycoconjugates [32], i.e. oligonucleotides bearing three different glycosyl groups (Fig. 3, 14) has<br />

also been achieved using appropriate orthogonally protected branching phosphoramidite 15 and carbohydratephosphoramidites<br />

(16-18). A derivative of bis(hydroxymethyl)-N,N’-bis(3-hydroxypropyl)malondiamide 15 was<br />

introduced during solid-phase synthesis. It contained a DMT protected hydroxyl and a phosphoramidite group<br />

allowing oligonucleotide elongation, and a levulinyl protected hydroxyl and a t-butyldiphenylsilyl protected<br />

hydroxyl for connecting glycosides residues. Conjugate syntheses were performed as follows: building block 15 was<br />

incorporated at the 5’ end of the oligonucleotide, DMT group was removed under acidic conditions and nucleotides<br />

were incorporated. At the end of the elongation, a carbohydrate-phosphoramidite was incorporated at the 5’ end.<br />

Levulinyl group was removed by hydrazinolysis allowing the incorporation of another carbohydrate<br />

phosphoramidite and finally TBDPS was removed prior to incorporation of the last glycosyl-phosphoramidite.<br />

This method allows the synthesis of heteroglycoclusters such as 14 in a reasonable yield. Melting temperature<br />

measurements showed that the presence of the bulky glycocluster at the 5’ terminus destabilises the duplex.<br />

In Sequence-Conjugation<br />

In the last example, glycosides were incorporated at the 5’ terminus, but building block 15 should allow in-sequence<br />

incorporation. This can also be performed using modified nucleoside phosphoramidite. This strategy has been used


164 Synthesis and Biological Applications of Glycoconjugates, 2011, 164-202<br />

Glycoliposomes and Metallic Glyconanoparticles in Glycoscience<br />

Marco Marradi a,b,* , Fabrizio Chiodo a , Isabel García a,b and Soledad Penadés a,b<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 10<br />

a Laboratory of GlycoNanotechnology, Biofunctional Nanomaterials Unit, CIC biomaGUNE, Parque Tecnológico de<br />

San Sebastián, Pº de Miramón 182, 20009 San Sebastián, Spain and b Biomedical Research Networking Center in<br />

Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Parque Tecnológico de San Sebastián, Pº de<br />

Miramón 182, 20009 San Sebastián, Spain<br />

Abstract: Multivalent sugar-based materials have attracted attention since the functional role of carbohydrates in<br />

biology has been disclosed. The design of artificial systems that mimics the polyvalent carbohydrate organization<br />

at cell surface has been envisaged as a strategy to study and intervene in carbohydrate-mediated interactions. One<br />

of the first synthetic glycomaterials which appeared in the literature were glycoliposomes, dynamic systems that<br />

resemble the glycocalix in the phospholipidic bilayer of cell membranes. Glycoliposomes are non-covalent<br />

systems which have been used since the seventies as multivalent tools in carbohydrate-based interactions against<br />

pathogens, for enhancing immunity and as molecular carriers in drug delivery. In former years, the advent of<br />

nanotechnology has allowed the design and construction of new materials similar in size to biologically relevant<br />

molecules (proteins, nucleic acids, etc) and displaying unique physical properties. The bio-functionalization of<br />

metallic nanomaterials with carbohydrates generated a new class of glycomaterials, named glyconanoparticles,<br />

which present carbohydrates in a highly multivalent way and in high local concentrations. At the same time, the<br />

quantum size properties of metallic nanoclusters can be used for biosensing, diagnostics, and (in perspective)<br />

therapy. This review focuses on glycoliposomes and covalently-functionalized glyconanoparticles which make<br />

use of the “glyco-code” to address specifically pathogens or pathological-related problems.<br />

Keywords: Glycoliposomes, glyconanoparticles, glycocalix, multivalency, neoglycolipids.<br />

INTRODUCTION<br />

The Functional Role of Carbohydrates<br />

Once thought to be only energetic sources for living systems or structural molecules protecting the cell surface,<br />

carbohydrates are nowadays recognized as essential tools for cell adhesion and recognition processes [1]. The surface of<br />

mammalian cells is covered by a dense coating of carbohydrates named glycocalyx. In the glycocalyx, carbohydrates<br />

appear mainly conjugated to proteins and lipids (glycoproteins, glycolipids and proteoglycans) and it is as clustered<br />

glycoconjugates that they develop their biological function. The high variety of carbohydrates, which have a much<br />

higher information store-capacity than amino acids and nucleotides, gives rise to a complex “glycocode” which is used<br />

by living systems to stock and transmit biological information. The (oligo)saccharides of the glycocalix are involved in<br />

carbohydrate-protein [2] and carbohydrate-carbohydrate [3] interactions that mediate many physiological and<br />

pathological processes [4, 5]. Carbohydrate-mediated interactions are generally very weak and the low affinity has to be<br />

compensated by multivalent presentation of the ligands. Polyvalent ligand–receptor interactions are characterized by the<br />

simultaneous binding of multiple ligands on one biological entity to multiple receptors on another biological entity and<br />

can be cooperatively much stronger than corresponding monovalent interactions [6]. These interactions are ubiquitous in<br />

biology and include both cellular signalling and matching contacts of viral and bacterial surfaces to host cells. For<br />

example, the weak association constant of carbohydrate-protein interactions in living systems is overcome both by<br />

clustering of sugar binding sites in the protein partner and presenting the carbohydrate at cell surface in a multivalent and<br />

orientated way [2, 7]. The structural diversity, the variability of sugar–units sequence and linkage points, the anomeric<br />

configuration, the chemical modification and/or substitution at different positions and with different groups (phosphate,<br />

sulphate, N-acetyl, etc), and the interconversion of conformers are some characteristics that create a unique level of<br />

diversity in the “glycocode”. Another key feature of the “glycocode” relies on the potential of glycoconjugates to adopt<br />

various defined shapes, each with its own particular ligand properties (differential conformer selection) [8].<br />

*Address correspondence to Marco Marradi: Laboratory of GlycoNanotechnology, Biofunctional Nanomaterials Unit, CIC biomaGUNE,<br />

Parque Tecnológico de San Sebastián, Pº de Miramón 182, 20009 San Sebastián, Spain: E-mail: mmarradi.ciber-bbn@cicbiomagune.es


Glycoliposomes and Metallic Glyconanoparticles in Glycoscience Synthesis and Biological Applications of Glycoconjugates 165<br />

Synthetic mimics of the glycocalix, as well as small molecules which intervene in carbohydrate biosynthetic and<br />

processing pathways can be used to interfere with cellular interactions [9]. The design and development of<br />

therapeutic glycomimetics is a great challenge of current glycoscience and the concept of carbohydrate-based drugs<br />

has thus spread out in the last years [10, 11]. The key role of polyvalency in many biological interactions has<br />

suggested new strategies for the design of artificial systems which mimic nature organization and help<br />

understanding cellular events at molecular scale. In particular, different types of scaffolds such as glycopeptides and<br />

glycoproteins [2, 12-14], glycopolymers [15-17], and glycodendrimers [18] have been developed to multimerise<br />

biologically relevant carbohydrates and address biological problems. A big challenge in this direction relies in<br />

combining the effects of multivalency and high local concentrations of the carbohydrates in such systems, with the<br />

possibility of tailoring other types of molecules (vectors, drugs, etc.) on the selected scaffold (multifunctionality)<br />

and/or using different imaging techniques (fluorescence, magnetic resonance imaging [MRI], etc.) to track them in<br />

vitro and in vivo (multimodality). Glycocalixarenes [19] and cyclopeptide-based glycoclusters [20] are presented in<br />

different <strong>chapter</strong>s of this E-book. This <strong>chapter</strong> is focussed on glycoliposomes and glyconanoparticles (GNPs) as<br />

polyvalent systems for addressing biological problems.<br />

Glycoconjugation and Self-Assembly of Glycoconjugates<br />

The biosynthetic pathways of glycans are lead by enzymatic processes through the action of glycosidases and<br />

glycosyltransferases [21]. Although the isolation from natural sources can give access to biological relevant<br />

oligosaccharides, this method usually affords small amounts of the desired product and rarely provides high degree<br />

of purity. Chemical or chemoenzymatic syntheses still are the method of choice to obtain pure and well-defined<br />

oligosaccharides. The progress in glycoscience is undoubtedly related to the efficiency of routine synthetic methods<br />

of complex glycoconjugates. The developments in the set-up of general methods for the preparation of biologically<br />

relevant oligosaccharides, including multistep protecting-group manipulations and highly stereo- and<br />

chemoselective glycosylations, have recently been reviewed [13, 22, 23]. In addition to traditional solution phase<br />

synthesis, it is nowadays possible to use solid phase synthesis, even in automated fashion [24]. The preparation of<br />

neoglycoconjugates that mimic the functions of bioactive carbohydrates by using chemoselective approaches is of<br />

great importance for the development of new therapeutics and it is revised in two <strong>chapter</strong>s of this E-book [25, 26].<br />

The synthesis of suitable neoglycoconjugates is also essential for the construction of self-assembled glycosystems.<br />

There are two main strategies to construct multivalent carbohydrate-based systems. The first one is to create<br />

membrane-like systems by using carbohydrate-conjugates in the presence of self-assembly molecules, as it is the<br />

case of glycoliposomes (Fig. 1, left), in which a glycolipid is mixed with self-assembly phospholipids and/or<br />

cholesterol(Chol)-like molecules. The other way is to use a suitable scaffold to anchor the carbohydrates, as it is the<br />

case of GNPs (Fig. 1, right), in which a metallic nanocluster is used to covalently attach the glycoconjugates. Due to<br />

the advances in carbohydrates synthesis during the last 30 years, suitable glycoconjugates obtained by conventional<br />

coupling chemistry (amines with carboxy groups or isothiocyanates, thiols with maleimides, azides with alkynes,<br />

etc) have allowed the preparation of a great variety of glycoliposomes and glyconanoparticles. Carbohydrates have<br />

to be derivatised prior to the self-assembly process, although some approaches using unmodified sugars have been<br />

proposed. The main chemical methods used for immobilization and anchoring of oligosaccharides onto solid<br />

matrixes are also valid for surface-functionalization of liposomes and nanoparticles [27].<br />

The self-assembly process of glycolipids in the presence of phospholipids or the self-assembled monolayers (SAMs)<br />

of thiol-armed glycoconjugates are some examples of construction of complex systems in a straightforward way.<br />

Glycoliposomes are non-covalent systems, while glyconanoparticles are based on covalent chemistry. In this<br />

<strong>chapter</strong>, we review these two types of multivalent systems bio-functionalized with conjugates of biologically<br />

interesting carbohydrates and their application in glycoscience.<br />

GLYCOLIPOSOMES<br />

Amphiphilic phospholipids are able to self-assembly and form closed bilayered structures in water. The resulting<br />

vesicles (liposomes) enclose an aqueous compartment and their structure resembles the phospholipid membranes of<br />

living cells [28]. Liposomes can easily incorporate hydrophilic molecules in their aqueous compartment and


166 Synthesis and Biological Applications of Glycoconjugates Marradi et al.<br />

hydrophobic compounds in their lipophilic bilayer, they can interact with cells by lipid bilayers fusion phenomena,<br />

and they are biocompatible. In general, liposomes are formed when a thin film of lipids containing phospholipids<br />

and Chol is hydrated. The hydrated lipid sheets detach during agitation and self-close to form large, multilamellar<br />

vesicles which prevent interactions of water with the hydrocarbon core of the bilayer. Reducing the size of the soformed<br />

particles requires energy inputs in the form of sonic energy (sonication) or mechanical energy (extrusion),<br />

among others [29].<br />

Figure 1: Schematic representation of glycoliposomes (left) and calculated structure of a gold glyconanoparticle (right). The<br />

GNP is a model of a mannose-coated gold nanoparticle (the ligand being α-Man(CH 2) 5S) which was obtained using SYBYL suite<br />

program. Pictures are not in scale.<br />

It is nowadays widely accepted that liposomes belong to the first-generation of nanovectors, i.e. delivery systems<br />

which are governed by passive mechanisms of drug delivery [30]. Liposomal formulations of drugs have been<br />

approved and are currently in clinical use due to their relative safety and enhanced efficacy [31]. The enhanced<br />

permeation and retention effect of liposomes in vivo is the key factor of their success as drug carriers. In order to<br />

achieve selective targeting, strong efforts are being made to insert specific functionalities at liposome surface.<br />

Remote activation would be also desirable to trigger payloads release in a controlled way. New generations of<br />

liposomes have thus been designed to improve their potential in biomedical applications. From a simple<br />

phospholipids bilayer used for biophysical research, liposomes and analogous vesicular carriers which incorporate<br />

specific molecules for targeting purpose, fluorescent or magnetic dyes for diagnostic use, cell-penetrating peptides,<br />

pH-sensitive components or stimuli-responsive molecules for controlled release have been developed [32, 33]. A<br />

class of biomolecules which had an important role in liposome evolution are carbohydrates. Liposomes coated with<br />

multiple copies of carbohydrates (glycoliposomes) have been used since the early seventies to target protein<br />

receptors of pathogens, as we will account in the next section. Glycoliposomes are composed of phospho- and<br />

glycolipids and thus are good “dynamic scaffold” to mimic the glycocluster of cellular membranes. To understand<br />

the role that carbohydrates play in many biological processes is important to have in hand a dynamic cellular model<br />

in which multiple copies of sugar epitopes can diffuse in a membrane-like patch changing organization in front of<br />

external stimuli. Glycoliposomes have been proposed as potential systems for drug and gene targeting/delivery by<br />

exploiting the specificity of carbohydrates [34, 35].<br />

In here, we describe the historical excursus of glycoliposomes as multivalent systems for carbohydrates presentation<br />

and their current biological applications. We will especially focus on glycoliposomes containing well-defined<br />

glycolipids. The presence of well-defined carbohydrates has been exploited first to understand their interactions with<br />

lectins in simple cellular models and then to selectively target lectins involved in physiological or pathological<br />

processes like inflammation and/or cellular immune response. Polysaccharide-anchored liposomes as synthetic<br />

models to study cellular adhesion and as potential therapeutics have been reviewed elsewhere [36].<br />

The Seminal Works and Methods for Preparing Glycoliposomes<br />

Seminal works dealing with the construction of “model membranes” containing gangliosides, i.e. glycolipids<br />

containing N-acetyl neuraminic acid [5-(acetylamino)-3,5-dideoxy-D-glycero-α-D-galacto-non-2-ulopyranosonic<br />

acid; Neu5Ac], appeared in the early seventies [37-39]. Different gangliosides were inserted in bimolecular lipid


Synthesis and Biological Applications of Glycoconjugates, 2011, 203-225 203<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 11<br />

Chemoselective Glycosylation Techniques for the Synthesis of Bioactive<br />

Neoglycoconjugates, Glyconanoparticles and Glycoarrays<br />

Francesco Peri *<br />

Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2; 20126 Milano, Italy<br />

Abstract: Chemoselective glycosylation (CG) methods have rapidly developed and diffused in glycochemist and<br />

glycobiologist’s community in the last fifteen years, and an increasing number of applications confirms the enormous<br />

potential of these techniques. CG has been successfully used for the synthesis of glycoproteins, glycopeptides,<br />

glycosylated natural compounds, carbohydrate-functionalized surfaces and nanoparticles. CG techniques can be<br />

considered as subset of click reactions that allow the efficient, high-yield conjugation of sugars with unprotected<br />

aglycons in aqueous media. In this review the CG methods have been divided into two main groups: “direct” CG that<br />

uses unmodified glycans, and “indirect” CG, in which the sugar has to be chemically modified before the glycosylation<br />

reaction. Recent applications of both CG techniques are reviewed, with special focus on the chemistry of CG reactions:<br />

mechanisms, stereochemistry issues, structure and stability of the glycoconjugates.<br />

Keywords: Chemoselective ligation, neoglycoconjugate, glyconanoparticle, microarray.<br />

DECONVOLUTING GLYCOME BY CHEMICAL TOOLS<br />

Like famous French and Italian sugar beads-covered cakes [1], cells from bacteria to mammals are covered by a<br />

sugar coating that presents the first information about the cell to the outside world. Increasing evidence indicates<br />

that carbohydrates have roles in a plethora of biological phenomena [2]. Carbohydrate-carbohydrate, carbohydrateprotein,<br />

and carbohydrate-nucleic acid interactions are the molecular basis of viral entry, signal transduction,<br />

inflammation, cell-cell interactions, bacteria-host interactions, fertility, and development, among other processes [3-<br />

5]. The discovery that a large part of the biological information is encoded in carbohydrate structures (or glycocode)<br />

rapidly became a fundamental concept in glycobiology, promoting the foundation of new biological disciplines first<br />

of all the specific branch of “omics” termed glycomics [6, 7]. In analogy with proteomics and genomics, glycomics<br />

explores the role of carbohydrates in biological processes. Carbohydrates, in the form of glycopeptides, glycolipids,<br />

glycosaminoglycans, proteoglycans, or other glycoconjugates have long been known to participate in all biological<br />

processes described above. Deconvoluting the importance of sugars in these biological events is challenging owing<br />

to many factors, including the complexity of the glycans (see below), the complex biosynthesis of the sugar part of<br />

glycoproteins (which is not under direct gene control), the multivalent nature of biological glycan recognition and<br />

the subtle phenotypes of glycan manipulation that often require multicellular environments to manifest.<br />

One of the principal obstacles to our study of carbohydrates comes from the complexity represented in the glycome<br />

itself, monosaccharides assemble into both linear and branched polymer structures, which are made more complex<br />

by the two possible stereochemical linkages between the units ( and ), resulting in a high degree of potential<br />

complexity. The complexity is further compounded by the different modes of attachment for cell-surface glycans,<br />

including glycolipids, and asparagine (-) and serine/threonine (O-) linkages to glycoproteins, and by further<br />

modifications of the monosaccharide units, such as methylation, sulfation, acetylation and phosphorylation.<br />

Organic chemists are therefore engaged in trying to reproduce synthetically the complexity of glycans. Synthesis<br />

provides access to relatively high quantities (mg) of glycans and glycoconjugates in a pure, chemically defined<br />

form. On the other and, milligram or sub-milligram amount of complex glycans (sugar epitopes, surface glycans,<br />

structural oligosaccharides) can be obtained by biological materials, and can be used as precious building blocks for<br />

the construction of chemically defined glycoconjugates. Despite how these complexes multivalent glycoconjugates<br />

have been obtained (by total synthesis or by chemical assembly of natural glycans) they are unique tools for basic<br />

structural glycobiology studies, and for use as therapeutics (Fig. 1).<br />

*Address correspondence to Francesco Peri: Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della<br />

Scienza, 2; 20126 Milano, Italy; E-mail: francesco.peri@unimib.it


204 Synthesis and Biological Applications of Glycoconjugates Francesco Peri<br />

Figure 1: Complex glycoconjugates as valuable tools for glycobiology and medicinal chemistry: A) modified surfaces for<br />

microarrays and SPR, B) neoglycoproteins, neoglycopeptides, carbohydrate vaccines, C) glycosylated natural products and drugs,<br />

D) multivalent carbohydrate gold or magnetic nanoparticles.<br />

This review presents how the development of chemoselective glycosylation (CG) reactions, as well as the use of<br />

click conjugation techniques, has in the last decade provided access to highly complex, multivalent,<br />

polyglycosylated structures. This can be done by a full chemical approach or by assembling by efficient conjugation<br />

reactions natural sugar building blocks deriving from mammalian or bacterial cells.<br />

The first part of the review the CG techniques will be described in detail, in particular that based on the reactivity of<br />

reducing sugars with hydroxylamine-containing scaffolds. These techniques have enormous potential and wide<br />

applicability, because are based on the conjugation of unmodified sugars in water. CG presents several advantage on<br />

more classical glycosylation methods. Their use not limited to organic chemistry laboratories specially equipped to<br />

work in strictly anhydrous conditions, but can be extended to biochemistry or biology labs. The most recent<br />

applications of CG will be presented and discussed in the second part of this review, in particular, the<br />

neoglycosylation of pharmacologically active natural compounds, of surfaces (glycoarrays), and of nanoparticles.<br />

CHEMOSELECTIVE LIGATION OF UNMODIFIED AND MODIFIED GLYCANS<br />

The fundamental reaction to covalently connect two monosaccharide units or to link a sugar to other biomolecules<br />

and biopolymers (proteins, lipids, or other aglycons), is the glycosylation reaction [8-10]. Nature executes this<br />

reaction with exquisite stereo- and regioselectivity and high efficiency to yield linear and branched poly- and<br />

oligosaccharides, glycoproteins, glycolipids, and glycosylated secondary metabolites. However, the installation of<br />

the glycosidic linkage by chemical means remains cumbersome, even with the aid of modern technologies [11].<br />

Owing to many recent breakthroughs in the field, the formation of most glycosidic bonds can be readily achieved. In<br />

the past three decades, much scientific effort has been dedicated to refining the glycosylation reaction through the<br />

development of new leaving groups, new promoters (activators), and through the optimization of the general<br />

reaction conditions. The “classical” glycosylation approaches for oligosaccharides, O- and N-linked glycopeptides<br />

and proteins and glycoconjugates (Scheme 1), require, regardless to the method used, protected sugar units, strictly<br />

anhydrous conditions, and the presence of glycosylation promoters or metal-based catalysts.


Chemoselective Glycosylation Techniques for the Synthesis Synthesis and Biological Applications of Glycoconjugates 205<br />

These techniques are often incompatible with aqueous conditions that are required when sugars should be linked to<br />

water-soluble molecules (such as proteins, peptides, lipids, polyphenols and other biological aglycones). On the<br />

contrary, strictly anhydrous conditions are required because water-labile glycosylation promoters are used. These<br />

glycosylation reactions therefore require specific equipment of organic chemistry lab and are not easily carried out<br />

in biochemistry or biology labs. Moreover, the extensive use of protecting groups and toxic and pollutant<br />

glycosylation promoters makes the classical glycosylation methods environmental unfriendly.<br />

O<br />

PO LG<br />

PO<br />

glycosyl donor oxacarbenium ion<br />

Scheme 1: Classical glycosylation reaction for the formation of O-glycosides.<br />

O<br />

O<br />

HO<br />

glycosyl<br />

PO<br />

O<br />

O<br />

O<br />

acceptor glycoside (stereoselectivity?)<br />

In a typical glycosylation reaction, after detachment of the leaving group in anomeric position, the glycosyl donor is<br />

converted into an oxycarbenium ion (Scheme 1) whose stability and conformational preferences strongly influence<br />

the yield and the stereoselectivity of the reaction. In the case of the most common O-glycosylations, the nucleophilic<br />

attack of the oxycarbenium ion by the oxygen nucleophile of the glycosyl acceptor (in the case of oligosaccharides)<br />

is always in competition with the attack of water. For this reason strictly anhydrous conditions are needed to avoid,<br />

or at least minimize, the unproductive hydrolysis of glycosyl donor.<br />

In sharp contrast, the so-called chemoselective glycosylation (CG) and click methods (Scheme 2), allow the<br />

formation of a glycoconjugate in the presence of water (or even directly in aqueous buffers). Chemoselective<br />

ligation methods, first developed in peptide and protein chemistry for the assembly of large unprotected peptide<br />

fragment to have access of totally synthetic proteins [12], have been extended to sugar chemistry for the preparation<br />

of different types of glycoconjugates [13].<br />

A<br />

B<br />

C<br />

HO<br />

O<br />

HO<br />

HO<br />

O OH<br />

Y aglycon<br />

O<br />

X + Y aglycon<br />

linker/peptide<br />

+<br />

X<br />

+<br />

Y aglycon<br />

direct<br />

chemoslective<br />

glycosylation<br />

indirect<br />

chemoslective<br />

glycosylation<br />

HO<br />

chemoselective HO<br />

ligation of<br />

glycosylated fragments<br />

HO<br />

O<br />

O<br />

Y aglycon<br />

O X Y aglycon<br />

linker/peptide<br />

X<br />

Y aglycon<br />

Scheme 2: Chemoselective glycosylation methods: A) Direct chemoselective glycosylation of unmodified glycans; B) indirect<br />

chemoselective glycosylation of glycans chemically modified in anomeric position; C) chemoselective ligation of glycosylated<br />

fragments.<br />

These methods have been designed so that a natural reducing sugar (mono or oligosaccharide) is reacted<br />

chemoselectively with a molecule bearing a nucleophile X group (Scheme 2A). Several copies of the reactive<br />

nucleophile X can be linked to the same scaffold when a multivalent glycoconjugate should be obtained. These<br />

methods, mainly based on the use of hydroxylamine and hydrazide nucleophiles, rely on the use of unmodified<br />

sugars, and will be treated here as “direct” CG methods because the glycosidic bond is formed in the ligation step. A<br />

second type of techniques has been developed based on the previous sugar modification, with introduction of a<br />

group X at the sugar’s anomeric position. Group X reacts selectively with a Y group on the aglycone moiety<br />

(protein, peptide, etc...) (Scheme 2B). These methods are somehow “indirect” because the real glycosidic bond<br />

(involving the anomeric C-1 carbon of the sugar) should be formed before the ligation step.


226 Synthesis and Biological Applications of Glycoconjugates, 2011, 226-239<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 12<br />

Glycosidases in Synthesis of Glycomimetics and Unnatural Carbohydrates<br />

Vladimír Křen *<br />

Institute of Microbiology, Centre of Biocatalysis and Biotransformation, Academy of <strong>Science</strong>s of the Czech<br />

Republic, Vídeňská 1083, CZ 142 20 Prague 4, Czech Republic<br />

Abstract: In recent years, carbohydrate-processing enzymes have become the enzymes of choice in many<br />

applications thanks to their stereoselectivity and efficiency. This <strong>chapter</strong> presents recent development in<br />

glycosidase-catalyzed synthesis of unnatural semisynthetic carbohydrate structures via two complementary<br />

approaches: the use of wild-type enzymes with engineered substrates and mutant glycosidases. Genetic<br />

engineering has recently produced glucuronyl synthases, an inverting xylosynthase and the first mutant endo-β-Nacetylglucosaminidase.<br />

A careful selection of enzyme producing strains and aptly modified substrates has resulted<br />

in rare glycostructures, such as N-acetyl--galactosaminuronates, 1,4-linked mannosides and 1,4-linked<br />

galactosides. The efficient selection of mutant enzymes is facilitated by high-throughput screening assays<br />

involving the co-expression of coupled enzymes or chemical complementation. Selective glycosidase inhibitors<br />

and highly specific glycosidases are finding attractive applications in biomedicine, biology and proteomics.<br />

Keywords: Glycosidase, glycosynthase, thioglycoligase, glycosyl azide, transglycosylation.<br />

INTRODUCTION<br />

In the past decades, the need for new carbohydrate materials and the development of glycomics have provided a<br />

boost to carbohydrate chemistry. Oligosaccharides can be prepared by classical organic chemistry methods [1] with<br />

all necessary tedious protection, deprotection, and activation strategies leading often to low yields and increased<br />

costs. As an alternative to the synthetic approach, enzymatic synthesis, ideally in a stereo- and regiospecific manner,<br />

is a valued option (Fig.1). Elegant glycosidic bond formation can be accomplished using two main enzyme groups –<br />

glycosyltransferases (EC 2.4.1.-) and glycosidases (EC 3.2.1.-) [2-5]. In this <strong>chapter</strong>, we will focus on recent<br />

advances in carbohydrate synthesis by glycosidases based on two approaches: the use of naturally occurring<br />

glycosidases with specially engineered substrates, and, use of glycosynthases leading to unnatural and modified<br />

carbohydrate structures, eventually to the glycomimetics.<br />

ENZYME ENGINEERING – MUTANT GLYCOSIDASES<br />

Glycosynthases, created by site-directed mutagenesis, represent a large group of mutant glycosidases. Since their<br />

introduction in 1998 [6, 7] they have caused a revolution in the high-yield enzymatic synthesis of carbohydrates and<br />

have been the topic of numerous reviews [8, 9].<br />

The mutation of the active site nucleophile to a non-nucleophile, such as alanine, renders a retaining glycosidase<br />

hydrolytically inactive but the resulting mutant – glycosynthase – can transfer an activated sugar donor, such as a<br />

glycosyl fluoride, onto a suitable acceptor substrate (Fig. 2). All aspects of using glycosyl fluorides in enzymatic<br />

reactions were described in a classical review by Williams and Withers [10].<br />

A number of new glycosynthases have been generated in recent years; for example, the glycosynthase derived from<br />

Thermus thermophilus -glycosidase, which can selectively synthesize the 1,3 glycosidic linkages in yields of up<br />

to 90 % [12], and the xylosynthase derived from Agrobacterium sp. -glucosidase [13]. Other xylosynthases<br />

followed, which were based on endo-1,4--xylanase from Cellulomonas fimi [14] and -xylosidase from<br />

Geobacillus stearothermophilus [15]. A noteworthy case of substrate engineering with hyperthermophilic<br />

*Address correspondence to Vladimír Křen: Institute of Microbiology, Centre of Biocatalysis and Biotransformation, Academy of <strong>Science</strong>s of<br />

the Czech Republic, Vídeňská 1083, CZ 142 20 Prague 4, Czech Republic; E-mail: kren@biomed.cas.cz


Glycosidases in Synthesis of Glycomimetics Synthesis and Biological Applications of Glycoconjugates 227<br />

glycosynthase from Sulfolobus solfataricus using glycosyl acceptors of varying anomeric configuration and<br />

lipophilicity was shown by Moracci et al. [16]. Glycosynthases have proved their potential in a number of<br />

applications involving the preparation of difficult-to-obtain glycosylated structures; for example glycosphingolipids,<br />

which are potential therapeutics for a range of pathologies including Alzheimer´s, Parkinson´s diseases and stroke<br />

[17], and, recently, glycosylated flavonoids, acting for example as immunosuppressants [18].<br />

Figure 1: Hydrolysis mechanisms: A) of retaining and B) inverting glycosidase; C) of -N-acetylhexosaminidase, which uses a<br />

modified retaining mechanism.


228 Synthesis and Biological Applications of Glycoconjugates Vladimír Křen<br />

A<br />

B<br />

mutated acid/ base (Glu Ala)<br />

O<br />

O<br />

CH3<br />

O<br />

F<br />

O -<br />

- O O<br />

H<br />

HO<br />

O<br />

O<br />

nucleophile<br />

acid/ base<br />

CH3<br />

NO2<br />

HS<br />

HO<br />

OH<br />

O<br />

OH<br />

NO2<br />

OR<br />

mutated nucleophile (Glu Ala)<br />

C<br />

mutated acid/ base<br />

(Glu Ala)<br />

OH<br />

OR<br />

O<br />

OH<br />

O2N<br />

HO<br />

HF<br />

NO2<br />

O<br />

CH3<br />

O<br />

O<br />

O<br />

HO O<br />

-<br />

S<br />

HO<br />

HO<br />

O<br />

CH3<br />

CH3<br />

F<br />

O -<br />

S<br />

HO<br />

OH<br />

OR<br />

O<br />

O<br />

S<br />

HO<br />

OH<br />

OR<br />

O<br />

F<br />

H<br />

OH<br />

H<br />

OH<br />

-<br />

mutated nucleophile<br />

(Glu Gly)<br />

CH3<br />

OH<br />

O<br />

OH<br />

OH<br />

OR<br />

O<br />

OH<br />

OR<br />

O<br />

O<br />

CH3<br />

S<br />

HO<br />

O -<br />

OH<br />

OR<br />

O<br />

OH<br />

Figure 2: Reaction mechanisms of mutant glycosidases [11]: Transglycosylation mechanism: A) of a glycosynthase; B) of<br />

glycosyl thioglycoligase; C) of thioglycosynthase. A) Glycosynthase, in which the catalytic nucleophile Glu was mutated to Ala,<br />

uses fluoride of the opposite anomeric configuration as a donor. Its mechanism is common both for exo- and endoglycosynthases.<br />

R = aglycon, e. g., p-nitrophenyl; B) Thioglycoligase, in which the catalytic acid-base Glu was mutated to Ala,<br />

uses reactive 2,4-dinitrophenyl glycoside donors and nucleophilic thiosugar acceptors (R = aglycon); C) Double mutant<br />

thioglycosynthase, with the catalytic acid-base Glu mutated to Ala and the catalytic nucleophile Glu mutated to Gly, uses<br />

inverted glycosyl fluoride donors and thiosugar acceptors (R = aglycon).<br />

In 2006, the Withers group presented the first glycosynthase that was able to transfer glucuronyl residues [19].<br />

Interestingly, the alanine mutant (Glu383Ala) was the most efficient in the transfer of both glucuronyl and<br />

galacturonyl moieties, which is in contrast to previous observations with other glycosynthases, where generally<br />

serine [20] and glycine [21] mutants exhibited superior activity to those with alanine. Another glucuronyl synthase,<br />

Glu504Gly mutant of -glucuronidase from E. coli, was applied in the -glucuronylation of a range of alcohols [22].<br />

Important advances have also been accomplished in the development of thioglycoligases – retaining glycosidases in<br />

which the acid–base catalytic residue is substituted by another amino acid, mainly Ala and Gln (Fig. 2B) [23]. These<br />

mutant enzymes use activated donor glycosides, such as dinitrophenyl glycosides with thiol acceptors. Müllegger et<br />

al. [24] discovered an improved mutant thioglycoligase (Glu170Gln) from Agrobacterium sp. by saturation<br />

mutagenesis, which can use donor sugars with a relatively poor leaving group such as -D-glucopyranosyl azide.


240 Synthesis and Biological Applications of Glycoconjugates, 2011, 240-254<br />

Diels-Alder Based Synthesis of Glycomimetics<br />

Cristina Nativi a,* , Elisa Dragoni b , Barbara Richichi a and Stefano Roelens c<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 13<br />

a Dipartimento di Chimica, Universita’ di Firenze, via della Lastruccia, 3,13 I-50019 Sesto F.no (FI), Italy;<br />

b ProtEra, Polo Scientifico Universita’ di Firenze, viale delle Idee, 22 I-50019 Sesto F.no (FI), Italy and c CNR,<br />

Istituto Metodologie Chimiche (IMC), Dipartimento di Chimica, Universita’ di Firenze, via della Lastruccia, 3,13 I-<br />

50019 Sesto F.no (FI), Italy.<br />

Abstract: [4+2] Cycloadditions between “in situ” generated ,’-dioxothiones and unprotected or variously<br />

protected glycals provide a powerful synthetic way to obtain glycomimetics chemo-, regio- and stereoselectively.<br />

Examples of glycopeptidomimetics and mimetic of tumor antigens are reported.<br />

Keywords: Hetero Diels-Alder, mimetics, tumor antigens, glycopeptides, matrix metalloproteinases.<br />

INTRODUCTION<br />

Diels-Alder reactions are a powerful tool to obtain regio- and stereoselectively complex molecules containing an<br />

unsaturated six member ring under mild reaction conditions. Owing to the simplicity of operation and to mildness of<br />

reaction conditions, [4+2] cycloadditions are largely preferred to other reactions when a suitable diene and<br />

dienophile can be envisioned. In addition, the variant in which either the diene or the dienophile can contain a<br />

heteroatom makes them attractive substrates for the synthesis of six-member ring heterocycles.<br />

Complex chiral molecules can effectively be prepared relying on the stereochemical features of [4+2]<br />

cycloadditions. Indeed, although the formation of more than one regioisomeric/stereoisomeric cycloadduct is<br />

possible in Diels-Alder reactions, the propensity for one regio- and/or stereoisomer is predictable. A representative<br />

example is the recently reported synthesis of the oxadecalin core of phomactin A 1 (Fig. 1), a selective antagonist of<br />

platelet activating factor (PAF) [1], via a stereoselective Diels-Alder reaction between the appropriately<br />

functionalized diene 2 and maleic anhydride (Fig. 1, transition state 3).<br />

Figure 1: Structure of phomactin 1, diene 2 and transition state 3.<br />

Me<br />

Me<br />

O<br />

Me<br />

1<br />

OTBS OTBDPS<br />

O<br />

Si<br />

O<br />

O<br />

iPr<br />

iPr<br />

Me<br />

2<br />

OH OH<br />

O<br />

Other literature examples concern the total synthesis of the natural product vernolepin 4 [2] or the Diels-Alder<br />

reactions between pyran-based alkoxy-dienes 5 and various dienophiles [3-5]. In these reactions, the endo addition is<br />

highly favored with a suitable dienophile like maleic anhydride [6] (Scheme 1).<br />

*Address correspondence to Cristina Nativi: Dipartimento di Chimica, Universita’ di Firenze, via della Lastruccia, 3,13 I-50019 Sesto F.no<br />

(FI), Italy; E-mail: cristina.nativi@unifi.it<br />

Me<br />

O<br />

TBSO<br />

O<br />

O<br />

Si<br />

O<br />

O<br />

O<br />

TBDPSO Me<br />

3


Diels-Alder Based Synthesis of Glycomimetics Synthesis and Biological Applications of Glycoconjugates 241<br />

Scheme 1: Structure of vemolepin 4 and Diels-Alder reaction between 5 and maleic anhydride.<br />

More recently, the synthesis of an interesting glycodendrimer showing both water-solubility and lipophilicity was<br />

accomplished employing Schimdt glycosylation and Diels-Alder cycloaddition recursively [7] (Scheme 2).<br />

In the last decade we successfully developed hetero Diels-Alder reactions between unusual heterodynes, i.e. ,’dioxothiones<br />

and 1-,4- and 5-glycals [8-10]. Glycals are versatile unsaturated sugar derivatives, widely used in<br />

carbohydrate chemistry as enantiomerically pure precursors [11-13] of saccharidic and non-saccharidic compounds.<br />

On the other hand, ,’-dioxothiones were a class of heterodienes little known until Capozzi and co-workers<br />

published the first applications in hetero Diels-Alder reactions [14, 15].<br />

Scheme 2: Synthesis of aromatic glycodendrimers.<br />

O<br />

AcO<br />

O<br />

AcO<br />

AcO<br />

AcO<br />

AcO<br />

AcO<br />

AcO<br />

AcO<br />

AcO<br />

AcO<br />

AcO<br />

4<br />

O<br />

OAc<br />

5<br />

AcO<br />

AcO O<br />

O<br />

O<br />

OH<br />

O<br />

O<br />

NPht<br />

O<br />

PHtN<br />

Diels-Alder<br />

Reaction<br />

O<br />

PHtN<br />

O<br />

PHtN<br />

O<br />

O<br />

O<br />

O<br />

O<br />

PhH, reflux<br />

68%<br />

OMP<br />

NH<br />

CHl 3<br />

Schmidt<br />

Glycosylation<br />

AcO<br />

AcO<br />

AcO<br />

AcO<br />

AcO<br />

AcO<br />

O<br />

PHtN<br />

O<br />

PHtN<br />

AcO<br />

AcO<br />

AcO<br />

AcO<br />

O<br />

O<br />

OAc<br />

O<br />

O<br />

OAc<br />

O<br />

O<br />

O<br />

HO OH<br />

AcO<br />

AcO O<br />

O<br />

O<br />

O<br />

PHtN<br />

OR<br />

R=MP<br />

R = C(=NH)CCl3<br />

O<br />

O<br />

5:1<br />

O


242 Synthesis and Biological Applications of Glycoconjugates Nativi et al.<br />

’-Dioxothiones as Electron-Poor Dienes<br />

’-Dioxothiones are highly reactive conjugated thiocarbonyl derivatives formed in situ and trapped as dienes in<br />

[4+2] Diels-Alder reactions [16-18]. Some years ago we developed the efficient synthesis of ’-dioxothiones from<br />

sufenylphthalimides and their trapping as electron-poor heterodienes in highly selective Diels-Alder reactions.<br />

Phthalimidesulfenyl chloride 6, (PhthNSCl, Phth = phthaloyl), reacts smoothly with -diketones, -ketoester or ketotiolester<br />

7 or with silylderivatives 8 to afford -acyl-N-thiophthalimide derivatives 9, which easily and<br />

quantitatively generate the reactive thiones 10 in the presence of mild bases. The transient thiones 10 can be trapped<br />

in Diels-Alder reactions with electron rich alkenes thus forming 5,6-dihydro-1,4-oxathiin derivatives 11 [14, 19, 20]<br />

(Scheme 3). Phthalimidesulfenyl chloride as well as phthalimide derivatives, 9 are stable compounds which can be<br />

prepared in grams scale and obtained pure by crystallization.<br />

In these inverse electron-demand Diels-Alder reactions, the molecular orbitals involved in the cycloadditions are the<br />

HOMO of the dienophile and the LUMO of the diene [15], therefore the 1,4-oxathiins formed through this synthetic<br />

strategy are always single chemo- and regioisomers. Indeed, the shape of the molecular orbitals obtained by<br />

molecular calculations ¥ showed that the symmetry of the HOMO of dienophiles and of the LUMO of dienes (’dioxothiones)<br />

is optimal for superimposition and that the orbitals with the largest coefficients are found on the sulfur<br />

atom of the thione and on the vinyl methylene of the alkene (Scheme 3).<br />

6<br />

Scheme 3: Synthesis of phthalimide derivative 9 and oxathiin 11.<br />

O<br />

O<br />

N<br />

O O<br />

S +<br />

Cl<br />

R R'<br />

R=R'=Alk,Ph<br />

R = Alk, Ph; R' = OR, SR, CN, COR<br />

9<br />

6 +<br />

PhthN =<br />

Me 3SiO O<br />

R<br />

Py, CHCl3<br />

- PhthNH<br />

O<br />

N<br />

O<br />

R<br />

R'<br />

O<br />

7<br />

S<br />

10<br />

O<br />

CHCl 3<br />

22, CH 2Cl 2, -18 °C<br />

-Me3SiCl<br />

8<br />

R=R'=Alk,Ph<br />

R=Alk,Ph;R'=OR,SR,CN,COR<br />

R' OR''<br />

H<br />

O O<br />

R R'<br />

SNPhth<br />

9<br />

Cycloaddition reactions of ’-dioxothiones with electron-rich alkenes are also characterized by high<br />

stereoselectivity. Retention of the dienophile Z/E geometry was always observed with cyclic or linear alkenes.<br />

Moreover, the use of chiral non-racemic ’-dioxothiones, obtained from -ketoesters bearing a chiral auxiliary in<br />

the ester and/or in the ketone residue, gave an appreciable discrimination between the enantiotopic faces of several<br />

dienophiles [21] (Scheme 4).<br />

¥<br />

Molecular orbitals were obtained by geometry optimization through ab initio Hartree-Fock calculation, using a 3-21G* basis set implemented in<br />

the Spartan program for PC<br />

R'<br />

R<br />

O<br />

9<br />

O<br />

S<br />

11<br />

O R''


Synthesis and Biological Applications of Glycoconjugates, 2011, 255-266 255<br />

Analytical Tools for Protein-Carbohydrate Interaction Studies<br />

Cédric Goyer and Pierre Labbé *<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers<br />

CHAPTER 14<br />

Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph Fourier, 38041<br />

Grenoble Cedex 9, France<br />

Abstract: A wide range of assays has been used to characterize protein-carbohydrates interactions allowing access to<br />

various quantitative information including thermodynamic ones such as stoichiometry of binding, binding constant,<br />

enthalpy of binding as well as kinetics and mechanistic ones. An understanding of each technique is essential in order<br />

to propose and validate appropriate interaction models that rationalize experimental data. Due to the complexity of<br />

carbohydrate-lectins interactions as an inherent consequence of multivalent interactions, a careful control of<br />

experimental conditions is necessary in order to avoid misinterpretation. This article will briefly present the various<br />

techniques that are commonly used to characterize sugar-lectin interaction: inhibition of hemagglutination assay<br />

(HIA), enzyme-linked lectin assays (ELLA), isothermal titration calorimetry (ITC) and surface plasmon resonance<br />

(SPR). Recently introduced, force spectroscopy by atomic force microscopy (AFM) is a new approach for measuring<br />

carbohydrate-protein interaction at the level of a single protein.<br />

Keywords: Biomolecular interaction, biosensors, surface plasmon resonance, isothermal calorimetric titration,<br />

atomic force spectroscopy.<br />

INTRODUCTION<br />

Lectins are carbohydrate binding proteins other than enzymes and antibodies [1] and exist in most living organisms<br />

ranging from viruses and bacteria to plants and animals [2]. They constitute cellular receptors that play a fundamental<br />

role in a wide range of biological processes including physiological and pathological events [3] such as adhesion of<br />

infectious agents to host cells, inflammatory processes, cell interactions in the immune system, growth and metastasis<br />

of tumour cells. Interactions between lectins and carbohydrates occur in the highly hydrated biological environment<br />

where hydration and solvation effects play a determinant role [4]. Carbohydrates primarily bind to polar groups of the<br />

lectin active site through hydrogen-bonding of their hydroxyl groups. Complementary hydrophobic interactions also<br />

occur between hydrophobic patches of the carbohydrate structure and the aromatic moieties that are present in the lectin<br />

constituents. It has been early recognized that an individual carbohydrate ligand binds to a lectin site weakly, with<br />

affinity constant generally in the range 1µm-1mM. The reason for this weakness lies in the solvent-exposed nature of<br />

the lectin binding-site, which are shallow pockets making few direct contacts with the ligands [5]. Considering the high<br />

specificity of biorecognition processes, such a weak binding is unlikely to be of any biological significance. However it<br />

is now well established that the weak binding affinities are efficiently overcome through so-called multivalent<br />

interactions, also known as “cluster glycoside effect”, that allow increased binding affinities and higher specificity<br />

required for a biologically relevant recognition process. Polyvalent associations that occur throughout biology present a<br />

number of characteristics that monovalent interactions do not exhibit. In most cases, polyvalent interactions rather than<br />

a strong single one are used in biological systems. Thus, the ability of lectins to distinguish between subtle variations of<br />

sugar structure makes them particularly efficient for decoding such carbohydrate-encoded information known as<br />

“glycocode”. Since lectins and carbohydrates are both commonly present at the cell surface and because sugars posses<br />

tremendous coding capacity, they constitute excellent cell recognition markers [6] that can be consequently exploited<br />

for therapeutic and diagnostic applications.<br />

A complete comprehension of the mechanism of protein-carbohydrate interaction at the molecular scale requires the<br />

elucidation of both structural and energetic aspects of the process. A large amount of structural data is now<br />

available, essentially from X-ray crystallography and NMR spectroscopy that have been used to elucidate the<br />

*Address correspondence to Pierre Labbé: Département de Chimie Moléculaire, UMR-CNRS 5250 & ICMG FR 2607, Université Joseph<br />

Fourier, 38041 Grenoble Cedex 9, France; E-mail: pierre.labbé@ujf-grenoble.fr


256 Synthesis and Biological Applications of Glycoconjugates Goyer and Labbé<br />

structures of lectins, sugars and their complexes. In addition an increasing number of experimental methods have<br />

been employed to determine the thermodynamics and kinetics of the binding process. Whereas the initial research’s<br />

were focusing on large glycoproteins and neoglycoconjugates, the recent progress in engineering new polymeric or<br />

small molecular weight sugar ligands provide new molecular tools for unravelling these complex ligand-receptor<br />

interactions. The main goal of this <strong>chapter</strong> is to give an overview of the various analytical tools that are classically<br />

used for studying sugar-lectin interactions. Since this topic was discussed in several reviews [4, 6-8] we will briefly<br />

present them and discuss their specificity and limitations. A special focus will be devoted to the new expanding tools<br />

and techniques that allow improving characterization at molecular level.<br />

A wide range of assays has been used to characterize protein-carbohydrates interactions allowing access to various<br />

quantitative information including thermodynamic ones such as stoichiometry of binding, binding constant, enthalpy<br />

of binding as well as kinetics and mechanistic ones. An understanding of each technique is essential in order to<br />

propose and validate appropriate interaction models that rationalize experimental data. Due to the complexity of<br />

carbohydrate-lectins interactions as an inherent consequence of multivalent interactions, a careful control of<br />

experimental conditions is necessary in order to avoid misinterpretation. Historically, the strength of sugar-lectin<br />

interaction has been quantified by inhibition of hemagglutination assay (HIA [9]), enzyme-linked lectin assays<br />

(ELLA [10]) although several spectroscopic techniques (NMR [11, 12], UV spectroscopy [13], fluorescence<br />

polarization [14-16]), have also been utilized. Finally among the more advanced methods, isothermal titration<br />

calorimetry (ITC) and surface plasmon resonance (SPR) constitutes two complementary and powerful techniques.<br />

Recently introduced, force spectroscopy by atomic force microscopy (AFM) has been applied for measuring<br />

carbohydrate-protein interaction at the level of a single protein.<br />

INHIBITION OF HEMAGGLUTINATION ASSAY (HIA)<br />

Hemagglutination is a specific form of agglutination that involves red blood cells. These erythrocytes present on<br />

their surface N-Acetylneuraminic acid, the predominant sialic acid found in mammalian cells. This negatively<br />

charged residue is found in complex glycan on mucins and glycoproteins found at the cell membrane. Chicken or<br />

porcine erythrocytes are most commonly used for this assay [9]. Starting from an anticoagulated erythrocytes<br />

suspension, addition of a soluble multimeric lectin cross-links erythrocytes and produces a gel-phase that does not<br />

settle down. Addition of soluble lectin in the presence of soluble carbohydrate ligands inhibits this hemagglutination<br />

process as a consequence of a competitive binding of the carbohydrate ligand of interest to the lectin active site. At<br />

high ligand concentration the hemagglutination reaction is completely inhibited and the anticoagulated erythrocytes<br />

settle down. The assay reports the minimum ligands concentration (IC50) that inhibits the hemagglutination process<br />

[7]. This historical assay suffers quite a lack of reproducibility and was used as a simple and qualitative method that<br />

allowed ordering soluble ligands in a rough order of binding affinity.<br />

ENZYME-LINKED LECTIN ASSAY (ELLA)<br />

Enzyme-linked lectin assays (ELLA) [17] assays are conceptually similar to the common ELISA (enzyme-linked<br />

immunosorbent assay) but are unique in that they rely upon non-immunologic reagents. ELLA assays constitutes a<br />

rapid method for evaluating lectin binding properties of soluble natural or synthetic lectin ligands in solution [18-<br />

21]. Microtiter plates wells are coated with a reference ligand that is often a (neo)glycoprotein or a polysaccharide.<br />

Lectin-enzyme conjugate and the soluble ligand are added to the wells and after an incubation period, the wells are<br />

evacuated, rinsed and refilled with a prodye substrate for the enzyme conjugate. The amount of color formed,<br />

monitored spectrophotometrically, is proportional to the concentration of lectin-enzyme conjugate immobilized on<br />

the wells and inversely proportional to the affinity of the soluble ligand. IC50 values for inhibition of the binding of<br />

an enzyme-labeled lectin to the immobilized reference ligand (the matrix) by the soluble ligands to be tested are thus<br />

determined. Generally the relative inhibitory powers are similar to those deduced from HIA assays although large<br />

differences in IC50 are often observed. One advantage of ELLA compared to solid-phase binding assays such as<br />

surface plasmon resonance (SPR) is that both, lectin and ligand are in solution. However, the use of glycoproteins<br />

from natural sources as reference ligands is limited because their varying complex carbohydrate composition due to<br />

microheterogeneity leads to reduced reproducibility of IC50 values. Furthermore, incomplete inhibition of lectin<br />

binding to the plate surface is often observed, that is, maximal inhibition occurs at considerably less than 100%.<br />

Finally ELLA provides inhibition constant and not a concentration binding constant.


Analytical Tools for Protein-Carbohydrate Interaction Studies Synthesis and Biological Applications of Glycoconjugates 257<br />

ISOTHERMAL TITRATION MICROCALORIMETRY (ITC)<br />

Isothermal Titration Calorimetry (ITC) is a well-established and powerful methodology for studying biomolecular<br />

interactions. Dam and Brewer [22] have published a very comprehensive review on the use of ITC to study lectincarbohydrates<br />

interactions whereas Lundquist and Toone [7] discussed the advantages and limitations of this technique.<br />

Titration calorimetry measures the heat evolved during ligand binding as a function of titrant concentration. Usually the<br />

lectin is placed in the calorimetric cell, the carbohydrate ligand is added in a series of 20 to 50 injections and the heat<br />

absorbed or produced during the binding is recorded. The shape of the resulting titration curve is a function of the<br />

number of lectin binding sites in the cell and of the binding constant. Analysis of the experimental data needs the use of<br />

a binding model. This model is then used to fit the experimental binding curve, which hopefully gives access to binding<br />

constant (Ka), free energy (G°), enthalpy (H°), entropy (S°) of binding and stoichiometry (n) of interaction for each<br />

class of binding sites that are assumed in the model. From temperature dependence of enthalpy and entropy changes,<br />

the change in heat capacity (Cp°) can be evaluated. Importantly, ITC is the only technique that allows the<br />

simultaneous determination of the thermodynamic binding parameters (Ka, G°, H°, S°) and stoichiometry (n) in a<br />

single experiment. Due to material availability, solubility limitations and sensitivity issues, ITC is practically effective<br />

for studying systems exhibiting binding constants ranging roughly from 10 3 to 10 7 M -1 .<br />

As previously discussed [7], ITC presents many advantages as compared to other methods. First ITC gives a<br />

concentration binding constant rather than an IC50, which can be related directly to free energy change of the<br />

interaction. It is also the only methods able to provide directly binding enthalpies in contrast to other methods that are<br />

based on the temperature dependence of binding constant and use of Gibbs-Helmholtz relationship. Thus ITC is unique<br />

in allowing direct measurement of the enthalpy of binding as a function of temperature. This in turn provides a direct<br />

measure of the change in molar heat capacity accompanying binding. This molar heat capacity change is of primary<br />

importance when studying the effect of solvent reorganization on the thermodynamic of ligand binding [23].<br />

Main drawbacks concerns the large amount of needed material (typically milligram quantities of both lectin and<br />

ligand), which is often a strong practical limitation. As a matter of fact, an adequate model describing the binding<br />

process has to be elaborated and validated before using it for fitting experimental data in order to extract quantitative<br />

thermodynamic information.<br />

SURFACE PLASMON RESONANCE (SPR)<br />

Surface plasmon resonance (SPR) biosensor instruments are sensitive to the common property of refractive index or<br />

mass change that is induced by complex formation at the surface of a biospecific sensor [24-28]. This optical<br />

method can provide both equilibrium and dynamic characterization of the interaction process [29, 30]. The method<br />

is label-free since it is based on refractive index changes, and monitors the interactions in real time. Although SPR<br />

was first observed in 1902 [31], its use for biosensors was only developed in the mid 1980s. Today the technique is<br />

widely used for studying bio-interactions, in particular because of the commercial availability of several<br />

instruments. Assessment of carbohydrate-lectin interactions by SPR has been reviewed in several articles [4, 7, 32].<br />

A very complete and detailed article by Wilson et al. [33] focusing on the use of SPR for quantitative analysis of<br />

small molecule- nucleic acid interactions provides fundamental and practical information of general interest.<br />

The physical basis of SPR has been presented in several reviews [34-35]. SPR phenomenon is observed under<br />

condition of total internal reflexion (incidence angle higher than critical angle, see Fig. 1) at an interface between a<br />

dielectric medium of high refractive index (usually a prism) that is covered by a thin film (2.5 to few tens of nm) of an<br />

adequate metal (aluminium, silver, gold), in contact with a dielectric medium of low refractive index (usually the liquid<br />

buffer of interest).<br />

Gold is usually preferred due to its inertness under biological conditions and to its easy and versatile<br />

functionalization using Au-S bond formation by reaction with thiolated derivatives. In absence of a metallic film, the<br />

fully reflected beam leaks an electrical field intensity (i.e. the evanescent field) into the medium of low refractive<br />

index. No photons exit the reflecting surface but the intensity of the corresponding evanescent electrical field<br />

decreases exponentially with distance from the interface, decaying over a distance of about ¼ wavelength beyond<br />

the surface in the low refractive index medium (Fig. 2a).


A<br />

Synthesis and Biological Applications of Glycoconjugates, 2011, 267-276 267<br />

Index<br />

β-N-acetylglucosaminidase 226<br />

β-N-acetylhexosaminidase 227, 229-234-236<br />

N-acetyllactosamine 93, 99<br />

Activated donor 228-230<br />

Acylhydrazide 206-207, 209, 218<br />

Adhesin 3-5, 7, 9, 12-16, 21, 23-24, 29-31, 186<br />

Agglutination 13, 15, 23, 49-50, 58-59, 167, 170, 256<br />

Agglutinin<br />

- Wheat Germ Agglutinin (WGA) 51, 94, 133, 169, 186<br />

- Helix pomatia agglutinin (HPA) 140, 263<br />

- peanut (Arachis hypogaea) agglutinin (PNA) 58, 94, 133-134, 140<br />

- Vicia Villosa Agglutinin (VVA) 50-51<br />

- Viscum album L. agglutinin (VAA) 52, 95, 136, 186, 246-248<br />

Alkyl mannoside 20<br />

Allosteric control 30<br />

1,2-alternate 40-41<br />

1,3-alternate 40-41, 46, 52-54, 105<br />

Amide bond formation 43, 45, 69, 78, 220<br />

Aminooxy 131-132, 134, 141, 153, 158, 179, 183, 208,<br />

216, 220<br />

Aminooxyacetyl 208<br />

Analytical ultracentrifugation (AUC) 86<br />

Anionic guest 57<br />

Anti-adhesion 12, 28, 29, 55, 58, 186<br />

Anti-adhesive 29, 31, 187, 188<br />

Antibody (Ab) 19, 56, 57, 139, 159, 185, 187, 220<br />

Antigen Presenting Cells (APC) 171-172, 176<br />

Anti-tumor effect 101, 139, 172, 174, 212, 244<br />

-L-arabinopyranoside 233<br />

Aromatic amino acid 57<br />

Asialoglycophorin 51<br />

Asialoglycoprotein receptor (ASGP-R) 94, 190<br />

Aspergillus oryzae 230-231, 234-236<br />

Atomic force microscopy (AFM) 29, 49, 59, 68, 86, 178, 184, 255-256, 260-263<br />

Azide 26-27, 47, 68, 71-72, 75, 82, 100, 153, 157,<br />

168, 212, 220, 226, 228-229, 233, 235<br />

B<br />

Bacillus circulans 230-231<br />

B-cell 137, 168<br />

-bromoalanine 214<br />

Betulinic acid (BA) 217<br />

Bingel reaction 69-73<br />

Biochips 145, 155-156<br />

Bioisosterism 207<br />

Biomaterials 61<br />

Biosensor 70, 145, 185-187, 255, 257<br />

BK virus 51<br />

Blood brain barrier 189<br />

Olivier Renaudet and Nicolas Spinelli (Eds)<br />

All rights reserved - © 2011 <strong>Bentham</strong> <strong>Science</strong> Publishers


268 Synthesis and Biological Applications of Glycoconjugates Renaudet and Spinelli<br />

Bolaforms 58-59<br />

C<br />

(Glyco)Calixarene 36-61, 85, 105-106, 129, 165, 185<br />

Capillary tubes 141<br />

Carbohydrate mediated adhesion 60<br />

Carbohydrate phosphoramidite 146-147, 154, 160<br />

Carbohydrate recognition domain (CRD) 8-9, 15-17, 20-25, 27, 30, 64-66, 73, 92-93, 96,<br />

97<br />

Carbohydrate-carbohydrate interaction 169-170, 177-178, 184-185, 219<br />

Carbohydrate-processing enzyme 73, 226<br />

Carbohydrate-protein interaction 36, 129, 135, 140, 164, 169, 171, 184, 186,<br />

190, 220, 255-256, 264<br />

Carbopeptoid 150-151<br />

CD69 52, 138-139, 235<br />

C-glycoside 48, 82, 116, 212, 243<br />

Chaperone/usher pathway 13-14, 16<br />

Chemoselective 131-132, 151, 160, 165, 167-168, 177, 179,<br />

203-221<br />

Cholera toxin 6-7, 9, 44-45, 55, 65, 78-81, 87, 136, 167, 185,<br />

191, 200<br />

CH-π interaction 57, 146<br />

Cycloaddition 65-66, 73, 82, 242, 244<br />

- Azide-alkyne 46-47, 71, 86, 132, 153, 180<br />

- Cu(I)-catalyzed alkyne-azide cycloaddition (CuAAc) 46-47, 71-73, 75, 132, 139, 153, 154, 155- 158<br />

- Diels-Alder 67, 213, 240-252<br />

- 1,3-dipolar 46-47, 66, 72, 99, 103, 105<br />

- Huisgen 46, 71-73, 153, 157, 180<br />

- [3+2] 68, 214<br />

- [4+2] 240, 243, 248<br />

Click chemistry 46-47, 64, 71, 73- 75, 151, 168, 214<br />

Cluster glycoside effect 92, 94, 104, 255, 264<br />

Cluster mannoside 12, 20-21<br />

Combinatorial<br />

- chemistry 95, 116, 118, 129, 133, 141<br />

- libraries 95, 116-117, 119-120, 123, 133<br />

Computer modeling 24<br />

Cone 40-41, 43-44, 46-47, 51-54, 56, 71, 105<br />

Conformational flexibility 17, 24<br />

(glyco) Conjugation 22, 156-157, 160, 165, 168, 176-177, 179-181,<br />

183, 190, 204, 207-208, 212-215, 219-220<br />

Cross-linking 27-28, 49-50, 55, 58-59, 68, 75, 137, 168, 178<br />

Cyclodextrin 37, 65, 102-103, 105, 129<br />

Cyclopeptide-based glycocluster (CBG) 129-141<br />

D<br />

Delivery 58, 79, 118, 123, 145, 151-152, 166-167, 171-<br />

172, 190<br />

- drug 36, 57, 60, 61, 68, 92, 116, 123, 125, 164, 166,<br />

172-173, 213<br />

- gene 36, 61, 94, 145, 175, 192<br />

Deltorphin 209


Index Synthesis and Biological Applications of Glycoconjugates 269<br />

(glyco)Dendrimer 9, 20, 37, 64, 69, 75, 78, 83-86, 102, 108-109,<br />

116-125, 129, 165, 170, 177, 184, 187, 219, 241<br />

Dendri-RAFT 134-136<br />

4-deoxy sugar 234<br />

N,N´-diacetylchitobiose 231-232, 234-235<br />

Diazirine 25-27<br />

Digitoxin 216-217<br />

Dithiogalactoside 100-102<br />

Dithiopyridyl 215<br />

DNA binder 60<br />

DNA encoded library 159<br />

DNA-Directed Immobilization (DDI) 155<br />

Docking 12, 22-24, 55, 234-235<br />

Donor Strand Complementation (DSC) 15<br />

Donor Strand Exchange (DSE) 13-15<br />

DOSY NMR 71, 184<br />

D-ribose-phosphate 59<br />

Dynamic Light Scattering (DLS) 49, 59, 68, 84, 86-87, 169<br />

E<br />

Electrode 140, 185<br />

Electrophoretic mobility 49<br />

ELISA (Enzyme-Linked Immunosorbent Assay) 19, 20-21, 23-24, 27, 85, 86, 94, 171-172, 174-<br />

175, 185, 187, 256<br />

ELLA (Enzyme-Linked Lectin Assay) 51, 105, 121-122, 133, 255, 256<br />

Enantioselectivity 57<br />

Enhancement factor 37-38, 54, 56<br />

Epimerization 131, 234<br />

Epitope orientation 52<br />

Erythropoietin (EPO) 215<br />

Escherichia coli (E. coli) 4, 6-7, 9, 12-16, 18-19, 23, 27-28, 31, 75, 78,<br />

81, 87, 181-182, 186-187, 190-191, 228-229,<br />

233<br />

ESI-MS 57<br />

F<br />

Fibrillum 13-14<br />

fim gene cluster 13, 18<br />

Fimbriae 3-4, 9, 12-21, 23, 27, 29, 31<br />

FimC/FimH chaperone-adhesin complex 16<br />

Flow cytometry 94, 146<br />

Fluorescence 20, 49, 56, 58, 82, 85, 105, 165, 186, 189, 190,<br />

219, 256<br />

- activated cell sorter scans 53<br />

- anisotropy 132, 140<br />

Frontal affinity chromatography (FAC) 94, 97<br />

Fucose 7-8, 95, 116, 119-122, 131, 146, 172-173, 189<br />

-D-fucopyranoside 233<br />

-D-fucosidase 229<br />

(glycol)Fullerene 64-75, 94, 129<br />

Fullerenol 68<br />

Functionalisation 40-42

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!