1
|
Chittum JE, Thompson A, Desai UR. Glycosaminoglycan microarrays for studying glycosaminoglycan-protein systems. Carbohydr Polym 2024; 335:122106. [PMID: 38616080 PMCID: PMC11032185 DOI: 10.1016/j.carbpol.2024.122106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/16/2024]
Abstract
More than 3000 proteins are now known to bind to glycosaminoglycans (GAGs). Yet, GAG-protein systems are rather poorly understood in terms of selectivity of recognition, molecular mechanism of action, and translational promise. High-throughput screening (HTS) technologies are critically needed for studying GAG biology and developing GAG-based therapeutics. Microarrays, developed within the past two decades, have now improved to the point of being the preferred tool in the HTS of biomolecules. GAG microarrays, in which GAG sequences are immobilized on slides, while similar to other microarrays, have their own sets of challenges and considerations. GAG microarrays are rapidly becoming the first choice in studying GAG-protein systems. Here, we review different modalities and applications of GAG microarrays presented to date. We discuss advantages and disadvantages of this technology, explain covalent and non-covalent immobilization strategies using different chemically reactive groups, and present various assay formats for qualitative and quantitative interpretations, including selectivity screening, binding affinity studies, competitive binding studies etc. We also highlight recent advances in implementing this technology, cataloging of data, and project its future promise. Overall, the technology of GAG microarray exhibits enormous potential of evolving into more than a mere screening tool for studying GAG - protein systems.
Collapse
Affiliation(s)
- John E Chittum
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States of America; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, United States of America
| | - Ally Thompson
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States of America; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, United States of America
| | - Umesh R Desai
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, United States of America; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, United States of America.
| |
Collapse
|
2
|
Zhang B, Chi L. Chondroitin Sulfate/Dermatan Sulfate-Protein Interactions and Their Biological Functions in Human Diseases: Implications and Analytical Tools. Front Cell Dev Biol 2021; 9:693563. [PMID: 34422817 PMCID: PMC8377502 DOI: 10.3389/fcell.2021.693563] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/20/2021] [Indexed: 01/12/2023] Open
Abstract
Chondroitin sulfate (CS) and dermatan sulfate (DS) are linear anionic polysaccharides that are widely present on the cell surface and in the cell matrix and connective tissue. CS and DS chains are usually attached to core proteins and are present in the form of proteoglycans (PGs). They not only are important structural substances but also bind to a variety of cytokines, growth factors, cell surface receptors, adhesion molecules, enzymes and fibrillary glycoproteins to execute series of important biological functions. CS and DS exhibit variable sulfation patterns and different sequence arrangements, and their molecular weights also vary within a large range, increasing the structural complexity and diversity of CS/DS. The structure-function relationship of CS/DS PGs directly and indirectly involves them in a variety of physiological and pathological processes. Accumulating evidence suggests that CS/DS serves as an important cofactor for many cell behaviors. Understanding the molecular basis of these interactions helps to elucidate the occurrence and development of various diseases and the development of new therapeutic approaches. The present article reviews the physiological and pathological processes in which CS and DS participate through their interactions with different proteins. Moreover, classic and emerging glycosaminoglycan (GAG)-protein interaction analysis tools and their applications in CS/DS-protein characterization are also discussed.
Collapse
Affiliation(s)
- Bin Zhang
- National Glycoengineering Research Center, Shandong University, Qingdao, China
| | - Lianli Chi
- National Glycoengineering Research Center, Shandong University, Qingdao, China
| |
Collapse
|
3
|
Strømme O, Psonka-Antonczyk KM, Stokke BT, Sundan A, Arum CJ, Brede G. Myeloma-derived extracellular vesicles mediate HGF/c-Met signaling in osteoblast-like cells. Exp Cell Res 2019; 383:111490. [PMID: 31283912 DOI: 10.1016/j.yexcr.2019.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 01/11/2023]
Abstract
Multiple myeloma is an incurable cancer of antibody-producing plasma cells. Hepatocyte growth factor (HGF), a cytokine aberrantly expressed in half of myeloma patients, is involved in myeloma pathogenesis by enhancing myeloma growth and invasiveness, and may play a role in myeloma bone disease by inhibiting osteoblastogenesis. In this study, we investigated whether extracellular vesicles (EVs) may play a role in HGF signaling between myeloma cells and osteoblast-like target cells. EVs from the HGF-positive cell line JJN-3 and the HGF-negative cell line INA-6, and from bone marrow plasma and primary human myeloma cells, were isolated using sequential centrifugation techniques and the presence of HGF on the EV-surface was investigated with ELISA. EVs from both cell lines were added to an established bioassay where HGF is known to induce interleukin-11 secretion in osteoblast-like cells. Our results show that HGF was bound to the surface of JJN-3-derived EVs, while INA-6-derived EVs were negative for HGF. Only JJN-3-derived EVs induced IL-11 secretion in osteoblast-like recipient cells. When osteoblast-like cells were preincubated with a specific HGF-receptor (c-Met) inhibitor, no induction of interleukin-11 was observed. Downstream c-Met phosphorylation was demonstrated by immunoblotting. EVs isolated from bone marrow plasma and primary myeloma cells were HGF-positive for a subset of myeloma patients. Taken together, this work shows for the first time that HGF bound on the surface of myeloma-derived EVs can effectuate HGF/c-Met signaling in osteoblast-like cells. Myeloma-derived EVs may play a role in myeloma bone disease by induction of the osteoclast-activating cytokine interleukin-11 in osteoblasts.
Collapse
Affiliation(s)
- Olaf Strømme
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Katarzyna M Psonka-Antonczyk
- Department of Physics, Faculty of Natural Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Bjørn Torger Stokke
- Department of Physics, Faculty of Natural Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Anders Sundan
- Centre of Molecular Inflammation Research and Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Carl-Jørgen Arum
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway and Department of Urology, St. Olavs University Hospital, Trondheim, Norway.
| | - Gaute Brede
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
4
|
Sakr HI, Coleman DT, Cardelli JA, Mathis JM. Characterization of an Oncolytic Adenovirus Vector Constructed to Target the cMet Receptor. Oncolytic Virother 2015; 4:119-132. [PMID: 26866014 PMCID: PMC4746000 DOI: 10.2147/ov.s87369] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The cMet receptor is a homodimer with tyrosine kinase activity. Upon stimulation with its ligand, hepatocyte growth factor (HGF), the receptor mediates wide physiologic actions. The HGF-cMet signaling pathway is dysregulated in many cancers, which makes cMet an important target for novel therapeutic interventions. Oncolytic adenoviruses (Ads) have been used for the past three decades as a promising therapeutic approach for a wide array of neoplastic diseases. To date, achieving cancer-specific replication of oncolytic Ads has been accomplished by either viral genome deletions or by incorporating tumor selective promoters. To achieve novel specificity of oncolytic Ad infection of cancer cells that overexpress cMet, we inserted the HGF NK2 sequence, corresponding to a competitive antagonist of HGF binding to the cMet receptor, into the Ad serotype 5 (Ad5) fiber gene. The resulting vector, Ad5-pIX-RFP-FF/NK2, was rescued, amplified in HEK293 cells, and characterized. Binding specificity and viral infectivity were tested in various cancer cell lines that express varying levels of cMet and hCAR (the Ad5 receptor). We found that Ad5-pIX-RFP-FF/NK2 demonstrated binding specificity to the cMet receptor. In addition, there was enhanced viral infectivity and virus replication compared with a non-targeted Ad vector. Although NK2 weakly induces cMet receptor activation, our results showed no receptor phosphorylation in the context of an oncolytic Ad virus. In summary, these results suggest that an oncolytic Ad retargeted to the cMet receptor is a promising vector for developing a novel cancer therapeutic agent.
Collapse
Affiliation(s)
- Hany I Sakr
- Department of Cellular Biology and Anatomy, LSU Health Shreveport, Shreveport, LA, USA; Gene Therapy Program, LSU Health Shreveport, Shreveport, LA, USA; Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA, USA
| | - David T Coleman
- Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA, USA; Department of Microbiology and Immunology, LSU Health Shreveport, Shreveport, LA, USA
| | - James A Cardelli
- Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA, USA; Department of Microbiology and Immunology, LSU Health Shreveport, Shreveport, LA, USA
| | - J Michael Mathis
- Gene Therapy Program, LSU Health Shreveport, Shreveport, LA, USA; Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA, USA; Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
5
|
Feng W, Wang X. Structure of decorin binding protein B from Borrelia burgdorferi and its interactions with glycosaminoglycans. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1823-1832. [PMID: 26275806 DOI: 10.1016/j.bbapap.2015.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/28/2015] [Accepted: 08/09/2015] [Indexed: 11/26/2022]
Abstract
Decorin-binding proteins (DBPs), DBPA and DBPB, are surface lipoproteins on Borrelia burgdorferi, the causative agent of Lyme disease. DBPs bind to the connective tissue proteoglycan decorin and facilitate tissue colonization by the bacterium. Although structural and biochemical properties of DBPA are well understood, little is known about DBPB. In current work, we determined the solution structure of DBPB from strain B31 of B. burgdorferi and characterized its interactions with glycosaminoglycans (GAGs). Our structure shows that DBPB adopts the same topology as DBPA, but possesses a much shorter terminal helix, resulting in a longer unstructured C-terminal tail, which is also rich in basic amino acids. Characterization of DBPB-GAG interactions reveals that, despite similar GAG affinities of DBPA and DBPB, the primary GAG-binding sites in DBPB are different from DBPA. In particular, our results indicate that lysines in the C-terminus of DBPB are vital to DBPB's ability to bind GAGs whereas C-terminal tail for DBPA from strain B31 only plays a minor role in facilitating GAG bindings. Furthermore, the traditional GAG-binding pocket important to DBPA-GAG interactions is only secondary to DBPB's GAG-binding ability.
Collapse
Affiliation(s)
- Wei Feng
- Department of Chemistry & Biochemistry, Arizona State University, Tempe, AZ 85287, USA
| | - Xu Wang
- Department of Chemistry & Biochemistry, Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
6
|
Morgan A, Sepuru KM, Feng W, Rajarathnam K, Wang X. Flexible Linker Modulates Glycosaminoglycan Affinity of Decorin Binding Protein A. Biochemistry 2015. [PMID: 26223367 PMCID: PMC4873102 DOI: 10.1021/acs.biochem.5b00253] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Decorin
binding protein A (DBPA) is a glycosaminoglycan (GAG)-binding
adhesin found on the surface of the bacterium Borrelia
burgdorferi (B. burgdorferi), the causative agent of Lyme disease. DBPA facilitates bacterial
adherence to extracellular matrices of human tissues and is crucial
during the early stage of the infection process. Interestingly, DBPA
from different strains (B31, N40, and PBr) show significant differences
in GAG affinities, but the structural basis for the differences is
not clear. In this study, we show that GAG affinity of N40 DBPA is
modulated in part by flexible segments that control access to the
GAG binding site, such that shortening of the linker leads to higher
GAG affinity when analyzed using ELISA, gel mobility shift assay,
solution NMR, and isothermal titration calorimetry. Our observation
that GAG affinity differences among different B. burgdorferi strains can be attributed to a flexible linker domain regulating
access to the GAG-binding domain is novel. It also provides a rare
example of how neutral amino acids and dynamic segments in GAG binding
proteins can have a large influence on GAG affinity and provides insights
into why the number of basic amino acids in the GAG-binding site may
not be the only factor determining GAG affinity of proteins.
Collapse
Affiliation(s)
| | - Krishna Mohan Sepuru
- ‡Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | | | - Krishna Rajarathnam
- ‡Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | | |
Collapse
|
7
|
Gallagher J. Fell-Muir Lecture: Heparan sulphate and the art of cell regulation: a polymer chain conducts the protein orchestra. Int J Exp Pathol 2015; 96:203-31. [PMID: 26173450 PMCID: PMC4561558 DOI: 10.1111/iep.12135] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/22/2015] [Indexed: 12/12/2022] Open
Abstract
Heparan sulphate (HS) sits at the interface of the cell and the extracellular matrix. It is a member of the glycosaminoglycan family of anionic polysaccharides with unique structural features designed for protein interaction and regulation. Its client proteins include soluble effectors (e.g. growth factors, morphogens, chemokines), membrane receptors and cell adhesion proteins such as fibronectin, fibrillin and various types of collagen. The protein-binding properties of HS, together with its strategic positioning in the pericellular domain, are indicative of key roles in mediating the flow of regulatory signals between cells and their microenvironment. The control of transmembrane signalling is a fundamental element in the complex biology of HS. It seems likely that, in some way, HS orchestrates diverse signalling pathways to facilitate information processing inside the cell. A dictionary definition of an orchestra is 'a large group of musicians who play together on various instruments …' to paraphrase, the HS orchestra is 'a large group of proteins that play together on various receptors'. HS conducts this orchestra to ensure that proteins hit the right notes on their receptors but, in the manner of a true conductor, does it also set 'the musical pulse' and create rhythm and harmony attractive to the cell? This is too big a question to answer but fun to think about as you read this review.
Collapse
Affiliation(s)
- John Gallagher
- Cancer Research UK Manchester Institute, Institute of Cancer Sciences, Paterson Building, University of Manchester, Manchester, UK
| |
Collapse
|
8
|
Blaszczyk M, Harmer NJ, Chirgadze DY, Ascher DB, Blundell TL. Achieving high signal-to-noise in cell regulatory systems: Spatial organization of multiprotein transmembrane assemblies of FGFR and MET receptors. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2015; 118:103-11. [PMID: 25957048 PMCID: PMC4832006 DOI: 10.1016/j.pbiomolbio.2015.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/23/2015] [Accepted: 04/24/2015] [Indexed: 01/12/2023]
Abstract
How is information communicated both within and between cells of living systems with high signal to noise? We discuss transmembrane signaling models involving two receptor tyrosine kinases: the fibroblast growth factor receptor (FGFR) and the MET receptor. We suggest that simple dimerization models might occur opportunistically giving rise to noise but cooperative clustering of the receptor tyrosine kinases observed in these systems is likely to be important for signal transduction. We propose that this may be a more general prerequisite for high signal to noise in transmembrane receptor signaling.
Collapse
Affiliation(s)
- Michal Blaszczyk
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Nicholas J Harmer
- The Henry Wellcome Building for Biocatalysis, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | - Dimitri Y Chirgadze
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, UK
| | - David B Ascher
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, UK.
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, UK.
| |
Collapse
|
9
|
Structural mechanisms underlying sequence-dependent variations in GAG affinities of decorin binding protein A, a Borrelia burgdorferi adhesin. Biochem J 2015; 467:439-51. [DOI: 10.1042/bj20141201] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Decorin-binding protein A (DBPA) is an important surface adhesin of the bacterium Borrelia burgdorferi, the causative agent of Lyme disease. DBPA facilitates the bacteria's colonization of human tissue by adhering to glycosaminoglycan (GAG), a sulfated polysaccharide. Interestingly, DBPA sequence variation among different strains of Borrelia spirochetes is high, resulting in significant differences in their GAG affinities. However, the structural mechanisms contributing to these differences are unknown. We determined the solution structures of DBPAs from strain N40 of B. burgdorferi and strain PBr of Borrelia garinii, two DBPA variants whose GAG affinities deviate significantly from strain B31, the best characterized version of DBPA. Our structures revealed that significant differences exist between PBr DBPA and B31/N40 DBPAs. In particular, the C-terminus of PBr DBPA, unlike C-termini from B31 and N40 DBPAs, is positioned away from the GAG-binding pocket and the linker between helices one and two of PBr DBPA is highly structured and retracted from the GAG-binding pocket. The repositioning of the C-terminus allowed the formation of an extra GAG-binding epitope in PBr DBPA and the retracted linker gave GAG ligands more access to the GAG-binding epitopes than other DBPAs. Characterization of GAG ligands' interactions with wild-type (WT) PBr and mutants confirmed the importance of the second major GAG-binding epitope and established the fact that the two epitopes are independent of one another and the new epitope is as important to GAG binding as the traditional epitope.
Collapse
|
10
|
Simonneau C, Bérénice Leclercq, Mougel A, Adriaenssens E, Paquet C, Raibaut L, Ollivier N, Drobecq H, Marcoux J, Cianférani S, Tulasne D, de Jonge H, Melnyk O, Vicogne J. Semi-synthesis of a HGF/SF kringle one (K1) domain scaffold generates a potent in vivo MET receptor agonist. Chem Sci 2015; 6:2110-2121. [PMID: 28717459 PMCID: PMC5496502 DOI: 10.1039/c4sc03856h] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/19/2015] [Indexed: 12/21/2022] Open
Abstract
The development of MET receptor agonists is an important goal in regenerative medicine, but is limited by the complexity and incomplete understanding of its interaction with HGF/SF (Hepatocyte Growth Factor/Scatter Factor). NK1 is a natural occurring agonist comprising the N-terminal (N) and the first kringle (K1) domains of HGF/SF. In the presence of heparin, NK1 can self-associate into a "head to tail" dimer which is considered as the minimal structural module able to trigger MET dimerization and activation whereas isolated K1 and N domains showed a weak or a complete lack of agonistic activity respectively. Starting from these structural and biological observations, we investigated whether it was possible to recapitulate the biological properties of NK1 using a new molecular architecture of isolated N or K1 domains. Therefore, we engineered multivalent N or K1 scaffolds by combining synthetic and homogeneous site-specifically biotinylated N and K1 domains (NB and K1B) and streptavidin (S). NB alone or in complex failed to activate MET signaling and to trigger cellular phenotypes. Importantly and to the contrary of K1B alone, the semi-synthetic K1B/S complex mimicked NK1 MET agonist activity in cell scattering, morphogenesis and survival phenotypic assays. Impressively, K1B/S complex stimulated in vivo angiogenesis and, when injected in mice, protected the liver against fulminant hepatitis in a MET dependent manner whereas NK1 and HGF were substantially less potent. These data reveal that without N domain, proper multimerization of K1 domain is a promising strategy for the rational design of powerful MET agonists.
Collapse
Affiliation(s)
- Claire Simonneau
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Bérénice Leclercq
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Alexandra Mougel
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Eric Adriaenssens
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Charlotte Paquet
- SIRIC ONCOLille , Maison Régionale de la Recherche Clinique , 6 rue du Pr. Laguesse , 59037 Lille Cedex , France
| | - Laurent Raibaut
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Nathalie Ollivier
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Hervé Drobecq
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Julien Marcoux
- UMR 7178 CNRS , Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO) , IPHC-DSA , Université de Strasbourg , 25 rue Becquerel , 67087 Strasbourg , France
| | - Sarah Cianférani
- UMR 7178 CNRS , Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO) , IPHC-DSA , Université de Strasbourg , 25 rue Becquerel , 67087 Strasbourg , France
| | - David Tulasne
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Hugo de Jonge
- Division of Immunology and General Pathology , Department of Molecular Medicine , University of Pavia , 9 via A Ferrata , 27100 Pavia , Italy
| | - Oleg Melnyk
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| | - Jérôme Vicogne
- UMR CNRS 8161 CNRS , Université de Lille , Institut Pasteur de Lille , 1 rue du Pr Calmette , 59021 Lille Cedex , France . ;
| |
Collapse
|
11
|
Wright JW, Kawas LH, Harding JW. The development of small molecule angiotensin IV analogs to treat Alzheimer's and Parkinson's diseases. Prog Neurobiol 2014; 125:26-46. [PMID: 25455861 DOI: 10.1016/j.pneurobio.2014.11.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 11/17/2014] [Accepted: 11/19/2014] [Indexed: 02/07/2023]
Abstract
Alzheimer's (AD) and Parkinson's (PD) diseases are neurodegenerative diseases presently without effective drug treatments. AD is characterized by general cognitive impairment, difficulties with memory consolidation and retrieval, and with advanced stages episodes of agitation and anger. AD is increasing in frequency as life expectancy increases. Present FDA approved medications do little to slow disease progression and none address the underlying progressive loss of synaptic connections and neurons. New drug design approaches are needed beyond cholinesterase inhibitors and N-methyl-d-aspartate receptor antagonists. Patients with PD experience the symptomatic triad of bradykinesis, tremor-at-rest, and rigidity with the possibility of additional non-motor symptoms including sleep disturbances, depression, dementia, and autonomic nervous system failure. This review summarizes available information regarding the role of the brain renin-angiotensin system (RAS) in learning and memory and motor functions, with particular emphasis on research results suggesting a link between angiotensin IV (AngIV) interacting with the AT4 receptor subtype. Currently there is controversy over the identity of this AT4 receptor protein. Albiston and colleagues have offered convincing evidence that it is the insulin-regulated aminopeptidase (IRAP). Recently members of our laboratory have presented evidence that the brain AngIV/AT4 receptor system coincides with the brain hepatocyte growth factor/c-Met receptor system. In an effort to resolve this issue we have synthesized a number of small molecule AngIV-based compounds that are metabolically stable, penetrate the blood-brain barrier, and facilitate compromised memory and motor systems. These research efforts are described along with details concerning a recently synthesized molecule, Dihexa that shows promise in overcoming memory and motor dysfunctions by augmenting synaptic connectivity via the formation of new functional synapses.
Collapse
Affiliation(s)
- John W Wright
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA.
| | - Leen H Kawas
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA
| | - Joseph W Harding
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA
| |
Collapse
|
12
|
Morgan A, Wang X. The novel heparin-binding motif in decorin-binding protein A from strain B31 of Borrelia burgdorferi explains the higher binding affinity. Biochemistry 2013; 52:8237-45. [PMID: 24148022 DOI: 10.1021/bi401376u] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Decorin-binding protein A (DBPA), a glycosaminoglycan (GAG)-binding lipoprotein found in Borrelia burgdorferi, is crucial to the transmission of Lyme disease in its earliest stages. Because of its role in the initial transmission of the disease, DBPA is an ideal target for vaccine development. DBPA sequences from different strains also contain considerable heterogeneity, leading to differing affinities for GAGs and proteoglycans among different DBPA sequences. Through biophysical and structural analysis of DBPA from strain B31, we have discovered a novel and important GAG-binding epitope in B31 DBPA. Removal of the epitope greatly attenuated its affinity for DBPA and may explain the differential GAG affinities seen in DBPAs from other strains of B. burgdorferi. Paramagnetic perturbation of the protein with TEMPO-labeled heparin fragments showed bound GAGs are located close to the linker region containing the BXBB motif that plays a significant role in determining the specific affinity and orientation of binding of GAG to DBPA. Thermodynamic contributions of the new motif to GAG binding were also characterized by both nuclear magnetic resonance and isothermal titration calorimetry and compared with those of other DBPA residues previously known to be involved in GAG interactions. These analyses showed the motif is as important as other known binding epitopes. The discovery of the motif offers a possible structural explanation for the previously observed differences in GAG affinities of DBPA variants from different Borrelia strains and improves our understanding of DBPA-GAG interactions.
Collapse
Affiliation(s)
- Ashli Morgan
- Department of Chemistry & Biochemistry, Arizona State University , Tempe, Arizona 85287, United States
| | | |
Collapse
|
13
|
Targeting tumor micro-environment for design and development of novel anti-angiogenic agents arresting tumor growth. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2013; 113:333-54. [PMID: 24139944 DOI: 10.1016/j.pbiomolbio.2013.10.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 10/05/2013] [Accepted: 10/08/2013] [Indexed: 12/12/2022]
Abstract
Angiogenesis: a process of generation of new blood vessels has been proved to be necessary for sustained tumor growth and cancer progression. Inhibiting angiogenesis pathway has long been remained a significant hope for the development of novel, effective and target orientated antitumor agents arresting the tumor proliferation and metastasis. The process of neoangiogenesis as a biological process is regulated by several pro- and anti-angiogenic factors, especially vascular endothelial growth factor, fibroblast growth factor, epidermal growth factor, hypoxia inducible factor 1 and transforming growth factor. Every endothelial cell destined for vessel formation is equipped with receptors for these angiogenic peptides. Moreover, numerous other angiogenic cytokines such as platelet derived growth factor (PGDF), placenta growth factor (PGF), nerve growth factor (NGF), stem-cell factor (SCF), and interleukins-2, 4, 6 etc. These molecular players performs critical role in regulating the angiogenic switch. Couple of decade's research in molecular aspects of tumor biology has unraveled numerous structural and functional mysteries of these angiogenic peptides. In present article, a detailed update on the functional and structural peculiarities of the various angiogenic peptides is described focusing on structural opportunities made available that has potential to be used to modulate function of these angiogenic peptides in developing therapeutic agents targeting neoplastic angiogenesis. The data may be useful in the mainstream of developing novel anticancer agents targeting tumor angiogenesis. We also discuss major therapeutic agents that are currently used in angiogenesis associated therapies as well as those are subject of active research or are in clinical trials.
Collapse
|
14
|
Graveel CR, Tolbert D, Vande Woude GF. MET: a critical player in tumorigenesis and therapeutic target. Cold Spring Harb Perspect Biol 2013; 5:a009209. [PMID: 23818496 PMCID: PMC3685898 DOI: 10.1101/cshperspect.a009209] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Since its discovery more than 25 years ago, numerous studies have established that the MET receptor is unique among tyrosine kinases. Signaling through MET is necessary for normal development and for the progression of a wide range of human cancers. MET activation has been shown to drive numerous signaling pathways; however, it is not clear how MET signaling mediates diverse cellular responses such as motility, invasion, growth, and angiogenesis. Great strides have been made in understanding the pleotropic aspects of MET signaling using three-dimensional molecular structures, cell culture systems, human tumors, and animal models. These combined approaches have driven the development of MET-targeted therapeutics that have shown promising results in the clinic. Here we examine the unique features of MET and hepatocyte growth factor/scatter factor (HGF/SF) structure and signaling, mutational activation, genetic mouse models of MET and HGF/SF, and MET-targeted therapeutics.
Collapse
Affiliation(s)
- Carrie R Graveel
- Molecular Oncology, Van Andel Research Institute, Grand Rapids, Michigan 49503, USA
| | | | | |
Collapse
|
15
|
Minsky BB, Atmuri A, Kaltashov IA, Dubin PL. Counterion Condensation on Heparin Oligomers. Biomacromolecules 2013; 14:1113-21. [DOI: 10.1021/bm400006g] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Burcu Baykal Minsky
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street,
Amherst, Massachusetts, 01003, United States
| | - Anand Atmuri
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts,
01003, United States
| | - Igor A. Kaltashov
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street,
Amherst, Massachusetts, 01003, United States
| | - Paul L. Dubin
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street,
Amherst, Massachusetts, 01003, United States
| |
Collapse
|
16
|
Despras G, Bernard C, Perrot A, Cattiaux L, Prochiantz A, Lortat-Jacob H, Mallet JM. Toward libraries of biotinylated chondroitin sulfate analogues: from synthesis to in vivo studies. Chemistry 2012; 19:531-40. [PMID: 23154924 DOI: 10.1002/chem.201202173] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 08/28/2012] [Indexed: 12/14/2022]
Abstract
Chondroitin sulfate-E (CS-E) oligosaccharidic analogues (di to hexa) were prepared from lactose. In these compounds, the 2-acetamido group was replaced by a hydroxyl group. This modification speeded up the synthesis, and large oligosaccharides were constructed in a few steps from a lactose-originated block. The protecting groups used were as follows; Fmoc for hydroxyl groups to be glycosylated, allyl group for anomeric position protection, and trichoroacetimidate leaving groups were used to prepare up to octasaccharides. We took advantage of the presence of allyl group to develop a click biotinylation, through its transformation into a 3-azido-2-hydroxyl propyl group in two steps (epoxidation and sodium azide epoxide opening). The biotinylating agent was a water-soluble propargylated and biotinylated triethylene glycol (PEG). By using surface plasmon resonance (SPR), it was shown that the di-, tetra-, and hexasaccharides display a binding affinity and selectivity toward HSF/GSF and CXCL12 similar to that of CS-E. A parallel study confirmed their mimicry of natural compounds, based on the hexasaccharide interaction with Otx2, a homeodomain protein involved in brain maturation, thus validating our simplification approach to synthesize bioactive GAG.
Collapse
Affiliation(s)
- Guillaume Despras
- UPMC Paris 06, UMR 7203, Laboratoire des BioMolécules, Université P. et M. Curie, 4 Place Jussieu, 75005 Paris, France
| | | | | | | | | | | | | |
Collapse
|
17
|
Wang X. Solution structure of decorin-binding protein A from Borrelia burgdorferi. Biochemistry 2012; 51:8353-62. [PMID: 22985470 DOI: 10.1021/bi3007093] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Decorin-binding protein A (DBPA) is an important lipoprotein from the bacterium Borrelia burgdorferi, the causative agent of Lyme disease. The absence of DBPA drastically reduces the pathogenic potential of the bacterium, and biochemical evidence indicates DBPA's interactions with the glycosaminoglycan (GAG) portion of decorin are crucial to its function. We have determined the solution structure of DBPA and studied DBPA's interactions with various forms of GAGs. DBPA is determined to be a helical bundle protein consisting of five helices held together by a strong hydrophobic core. The structure also possesses a basic patch formed by portions of two helices and two flexible linkers. Low-molecular mass heparin-induced chemical shift perturbations for residues in the region as well as increases in signal intensities of select residues in their presence confirm residues in the pocket are perturbed by heparin binding. Dermatan sulfate fragments, the dominant GAG type found on decorin, were shown to have lower affinity than heparin but are still capable of binding DBPA.
Collapse
Affiliation(s)
- Xu Wang
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
18
|
Cecchi F, Pajalunga D, Fowler CA, Uren A, Rabe DC, Peruzzi B, MacDonald NJ, Blackman DK, Stahl SJ, Byrd RA, Bottaro DP. Targeted disruption of heparan sulfate interaction with hepatocyte and vascular endothelial growth factors blocks normal and oncogenic signaling. Cancer Cell 2012; 22:250-62. [PMID: 22897854 PMCID: PMC3422512 DOI: 10.1016/j.ccr.2012.06.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 04/04/2012] [Accepted: 06/26/2012] [Indexed: 12/16/2022]
Abstract
Hepatocyte growth factor (HGF) and vascular endothelial cell growth factor (VEGF) regulate normal development and homeostasis and drive disease progression in many forms of cancer. Both proteins signal by binding to receptor tyrosine kinases and heparan sulfate (HS) proteoglycans on target cell surfaces. Basic residues comprising the primary HS binding sites on HGF and VEGF provide similar surface charge distributions without underlying structural similarity. Combining three acidic amino acid substitutions in these sites in the HGF isoform NK1 or the VEGF isoform VEGF165 transformed each into potent, selective competitive antagonists of their respective normal and oncogenic signaling pathways. Our findings illustrate the importance of HS in growth factor driven cancer progression and reveal an efficient strategy for therapeutic antagonist development.
Collapse
Affiliation(s)
| | - Deborah Pajalunga
- Macromolecular NMR Section, Structural Biophysics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702-1201 USA
- EntreMed, Inc., Rockville, MD 20850 USA
| | - C. Andrew Fowler
- Macromolecular NMR Section, Structural Biophysics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702-1201 USA
| | - Aykut Uren
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C. 20057-1469
| | | | | | | | | | - Stephen J. Stahl
- Protein Expression Laboratory, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892-2775 USA
| | - R. Andrew Byrd
- Macromolecular NMR Section, Structural Biophysics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702-1201 USA
| | - Donald P. Bottaro
- Correspondence: Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bldg 10 CRC Rm 2-3952, 10 Center Drive MSC 1107, Bethesda, MD 20892-1107 USA Tel: 301-402-6499, Fax: 301-402-0922,
| |
Collapse
|
19
|
Preparation of heparin/heparan sulfate oligosaccharides with internal N-unsubstituted glucosamine residues for functional studies. Glycoconj J 2011; 28:525-35. [DOI: 10.1007/s10719-011-9352-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 09/03/2011] [Accepted: 09/07/2011] [Indexed: 10/17/2022]
|
20
|
Intracellular signaling cascades triggered by the NK1 fragment of hepatocyte growth factor in human prostate epithelial cell line PNT1A. Cell Signal 2011; 23:1961-71. [PMID: 21777671 DOI: 10.1016/j.cellsig.2011.07.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 06/21/2011] [Accepted: 07/04/2011] [Indexed: 01/09/2023]
Abstract
Hepatocyte Growth Factor (HGF)/c-MET signaling has an emerging role in promoting cell proliferation, survival, migration, wound repair and branching in a variety of cell types. HGF plays a crucial role as a mediator of stromal-epithelial interactions in the normal prostate but the precise biological function of HGF/c-Met interaction in the normal prostate and in prostate cancer is not clear. HGF has two naturally occurring splice variants and NK1, the smallest of these HGF variants, consists of the HGF amino terminus through the first kringle domain. We evaluated the intracellular signaling cascades and the morphological changes triggered by NK1 in human prostate epithelial cell line PNT1A which shows molecular and biochemical properties close to the normal prostate epithelium. We demonstrated that these cells express a functional c-MET, and cell exposure to NK1 induces the phosphorylation of tyrosines 1313/1349/1356 residues of c-MET which provide docking sites for signaling molecules. We observed an increased phosphorylation of ERK1/2, Akt, c-Src, p125FAK, SMAD2/3, and STAT3, down-regulation of the expression of epithelial cell-cell adhesion marker E-cadherin, and enhanced expression levels of mesenchymal markers vimentin, fibronectin, vinculin, α-actinin, and α-smooth muscle actin. This results in cell proliferation, in the appearance of a mesenchymal phenotype, in morphological changes resembling cell scattering and in wound healing. Our findings highlight the function of NK1 in non-tumorigenic human prostatic epithelial cells and provide a picture of the signaling pathways triggered by NK1 in a unique cell line.
Collapse
|
21
|
Seo ES, Blaum BS, Vargues T, De Cecco M, Deakin JA, Lyon M, Barran PE, Campopiano DJ, Uhrín D. Interaction of human β-defensin 2 (HBD2) with glycosaminoglycans. Biochemistry 2010; 49:10486-95. [PMID: 21062008 DOI: 10.1021/bi1011749] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human β-defensin 2 (HBD2) is a member of the defensin family of antimicrobial peptides that plays important roles in the innate and adaptive immune system of both vertebrates and invertebrates. In addition to their direct bactericidal action, defensins are also involved in chemotaxis and Toll-like receptor activation. In analogy to chemokine/glycosaminoglycan (GAG) interactions, GAG-defensin complexes are likely to play an important role in chemotaxis and in presenting defensins to their receptors. Using a gel mobility shift assay, we found that HBD2 bound to a range of GAGs including heparin/heparan sulfate (HS), dermatan sulfate (DS), and chondroitin sulfate. We used NMR spectroscopy of (15)N-labeled HBD2 to map the binding sites for two GAG model compounds, a heparin/HS pentasaccharide (fondaparinux sodium; FX) and enzymatically prepared DS hexasaccharide (DSdp6). We identified a number of basic amino acids that form a common ligand binding site, which indicated that these interactions are predominantly electrostatic. The dissociation constant of the [DSdp6-HBD2] complex was determined by NMR spectroscopy to be 5 ± 5 μM. Binding of FX could not be quantified because of slow exchange on the NMR chemical shift time scale. FX was found to induce HBD2 dimerization as evidenced by the analysis of diffusion coefficients, (15)N relaxation, and nESI-MS measurements. The formation of FX-bridged HBD2 dimers exhibited features of a cooperative binding mechanism. In contrast, the complex with DSdp6 was found to be mostly monomeric.
Collapse
Affiliation(s)
- Emily S Seo
- EastChem, School of Chemistry, The University of Edinburgh, King's Buildings, West Mains Road, Edinburgh EH9 3JJ, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Structural basis for agonism and antagonism of hepatocyte growth factor. Proc Natl Acad Sci U S A 2010; 107:13264-9. [PMID: 20624990 DOI: 10.1073/pnas.1005183107] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hepatocyte growth factor (HGF) is an activating ligand of the Met receptor tyrosine kinase, whose activity is essential for normal tissue development and organ regeneration but abnormal activation of Met has been implicated in growth, invasion, and metastasis of many types of solid tumors. HGF has two natural splice variants, NK1 and NK2, which contain the N-terminal domain (N) and the first kringle (K1) or the first two kringle domains of HGF. NK1, which is a Met agonist, forms a head-to-tail dimer complex in crystal structures and mutations in the NK1 dimer interface convert NK1 to a Met antagonist. In contrast, NK2 is a Met antagonist, capable of inhibiting HGF's activity in cell proliferation without clear mechanism. Here we report the crystal structure of NK2, which forms a "closed" monomeric conformation through interdomain interactions between the N- domain and the second kringle domain (K2). Mutations that were designed to open up the NK2 closed conformation by disrupting the N/K2 interface convert NK2 from a Met antagonist to an agonist. Remarkably, this mutated NK2 agonist can be converted back to an antagonist by a mutation that disrupts the NK1/NK1 dimer interface. These results reveal the molecular determinants that regulate the agonist/antagonist properties of HGF NK2 and provide critical insights into the dimerization mechanism that regulates the Met receptor activation by HGF.
Collapse
|
23
|
Sarrazin S, Lyon M, Deakin JA, Guerrini M, Lassalle P, Delehedde M, Lortat-Jacob H. Characterization and binding activity of the chondroitin/dermatan sulfate chain from Endocan, a soluble endothelial proteoglycan. Glycobiology 2010; 20:1380-8. [DOI: 10.1093/glycob/cwq100] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
24
|
Pan J, Qian Y, Zhou X, Lu H, Ramacciotti E, Zhang L. Chemically oversulfated glycosaminoglycans are potent modulators of contact system activation and different cell signaling pathways. J Biol Chem 2010; 285:22966-75. [PMID: 20418371 DOI: 10.1074/jbc.m109.063735] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Contaminated heparin was associated with adverse reactions by activating the contact system. Chemically oversulfated/modified glycosaminoglycans (GAGs) consisting of heparan sulfate, dermatan sulfate, and chondroitin sulfate have been identified as heparin contaminants. Current studies demonstrated that each component of oversulfated GAGs was comparable with oversulfated chondroitin sulfate in activating the contact system. By testing a series of unrelated negatively charged compounds, we found that the contact system recognized negative charges rather than specific chemical structures. We further tested how oversulfated GAGs and contaminated heparins affect different cell signaling pathways. Our data showed that chemically oversulfated GAGs and contaminated heparin had higher activity than the parent compounds and authentic heparin, indicative of sulfation-dominant and GAG sequence-dependent activities in BaF cell-based models of fibroblast growth factor/fibroblast growth factor receptor, glial cell line-derived neurotrophic factor/c-Ret, and hepatocyte growth factor/c-Met signaling. In summary, these data indicate that contaminated heparins intended for blood anticoagulation not only activated the contact system but also modified different GAG-dependent cell signaling pathways.
Collapse
Affiliation(s)
- Jing Pan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Targeting the HGF/Met signalling pathway in cancer. Eur J Cancer 2010; 46:1260-70. [PMID: 20303741 DOI: 10.1016/j.ejca.2010.02.028] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 02/16/2010] [Indexed: 12/13/2022]
Abstract
Under normal conditions, hepatocyte growth factor (HGF)-induced Met tyrosine kinase (TK) activation is tightly regulated by paracrine ligand delivery, ligand activation at the target cell surface, and ligand activated receptor internalisation and degradation. Despite these controls, HGF/Met signalling contributes to oncogenesis and tumour progression in several cancers and promotes aggressive cellular invasiveness that is strongly linked to tumour metastasis. The prevalence of HGF/Met pathway activation in human malignancies has driven rapid growth in cancer drug development programmes. Pathway inhibitors can be divided broadly into biologicals and low molecular weight synthetic TK inhibitors; of these, the latter now outnumber all other inhibitor types. We review here the basic properties of HGF/Met pathway antagonists now in preclinical and clinical development as well as the latest clinical trial results. The main challenges facing the effective use of HGF/Met-targeted antagonists for cancer treatment include optimal patient selection, diagnostic and pharmacodynamic biomarker development, and the identification and testing of optimal therapy combinations. The wealth of basic information, analytical reagents and model systems available concerning HGF/Met oncogenic signalling will continue to be invaluable in meeting these challenges and moving expeditiously toward more effective disease control.
Collapse
|
26
|
Zhang L. Glycosaminoglycan (GAG) biosynthesis and GAG-binding proteins. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 93:1-17. [PMID: 20807638 DOI: 10.1016/s1877-1173(10)93001-9] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Two major types of glycosaminoglycan (GAG) polysaccharides, heparan sulfate and chondroitin sulfate, are polymerized and modified by enzymes that are encoded by more than 40 genes in animal cells. Because of the expression repertoire of the GAG assembly and modification enzymes, each heparan sulfate and chondroitin sulfate chain has a sulfation pattern, chain length, and fine structure that is potentially unique to each animal cell. GAGs interact with hundreds of proteins. Such interactions protect growth factors, chemokines, and cytokines against proteolysis. GAGs catalyze protease (such as thrombin) inhibition by serpins. GAGs regulate multiple signaling pathways including, but not limited to, fibroblast growth factor (FGF)/FGFR, hepatocyte growth factor (HGF)/c-Met, glial cell line-derived neurotrophic factor (GDNF)/c-Ret/GFRalpha1, vascular endothelial growth factor (VEGF)/VEGFR, platelet derived growth factor (PDGF)/PDGFR, BAFF/TACI, Indian hedgehog, Wnt, and BMP signaling pathways,where genetic studies have revealed an absolute requirement for GAGs in these pathways. Most importantly, protein/GAG aggregates induce thrombin generation and immune system upregulation by activating the contact system. Abnormal protein/GAG aggregates are associated with a variety of devastating human diseases including, but not limited to, Alzheimer's, diabetes, prion or transmissible spongiform encephalopathies, Lupus, heparin-induced thrombocytopenia/thrombosis, and different kinds of cancers. Therefore, GAGs are essential components of modern molecular biology and human physiology. Understanding GAG structure and function at molecular level with regard to development and health represents a unique opportunity in combating different kinds of human diseases.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Pathology and Immunology, Washington University Medical School, St. Louis, MO, USA
| |
Collapse
|
27
|
Babu P, Kuberan B. Fluorescent-tagged heparan sulfate precursor oligosaccharides to probe the enzymatic action of heparitinase I. Anal Biochem 2009; 396:124-32. [PMID: 19732739 DOI: 10.1016/j.ab.2009.08.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 08/21/2009] [Accepted: 08/25/2009] [Indexed: 11/30/2022]
Abstract
Heparitinase I, a key lyase enzyme essential for structural analysis of heparan sulfate (HS), degrades HS domains that are undersulfated at glucuronyl residues through an elimination mechanism. Earlier studies employed viscosimetric measurements and electrophoresis to deduce the mechanism of action of heparitinase I and two other related lyases, heparitinase II and heparitinase III. However, these findings lack molecular evidence for the intermediates formed and could not distinguish whether the cleavage occurred from the reducing end or the nonreducing end. In the current study, 2-aminoacridone (2-AMAC)-labeled HS precursor oligosaccharides of various sizes were prepared to investigate the mechanism of heparitinase I-mediated depolymerization using sensitive and quantitative methodologies. Furthermore, fluorescent (2-AMAC) tagging of HS precursor oligosaccharides allowed us to distinguish fragments that result from cleavage of the substrates at various time intervals and sites farther away from the reducing and nonreducing ends of oligosaccharide substrates. This study provides the first direct molecular evidence for a predominantly random endolytic mechanism of cleavage of HS precursor oligosaccharides by heparitinase I. This robust strategy can be adapted to deduce the mechanism of action of other heparitinases and also to deduce structural information of complex HS oligosaccharides of biological importance.
Collapse
Affiliation(s)
- Ponnusamy Babu
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | | |
Collapse
|
28
|
Schultz JM, Khan SN, Ahmed ZM, Riazuddin S, Waryah AM, Chhatre D, Starost MF, Ploplis B, Buckley S, Velásquez D, Kabra M, Lee K, Hassan MJ, Ali G, Ansar M, Ghosh M, Wilcox ER, Ahmad W, Merlino G, Leal SM, Riazuddin S, Friedman TB, Morell RJ. Noncoding mutations of HGF are associated with nonsyndromic hearing loss, DFNB39. Am J Hum Genet 2009; 85:25-39. [PMID: 19576567 DOI: 10.1016/j.ajhg.2009.06.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 05/22/2009] [Accepted: 06/01/2009] [Indexed: 11/16/2022] Open
Abstract
A gene causing autosomal-recessive, nonsyndromic hearing loss, DFNB39, was previously mapped to an 18 Mb interval on chromosome 7q11.22-q21.12. We mapped an additional 40 consanguineous families segregating nonsyndromic hearing loss to the DFNB39 locus and refined the obligate interval to 1.2 Mb. The coding regions of all genes in this interval were sequenced, and no missense, nonsense, or frameshift mutations were found. We sequenced the noncoding sequences of genes, as well as noncoding genes, and found three mutations clustered in intron 4 and exon 5 in the hepatocyte growth factor gene (HGF). Two intron 4 deletions occur in a highly conserved sequence that is part of the 3' untranslated region of a previously undescribed short isoform of HGF. The third mutation is a silent substitution, and we demonstrate that it affects splicing in vitro. HGF is involved in a wide variety of signaling pathways in many different tissues, yet these putative regulatory mutations cause a surprisingly specific phenotype, which is nonsydromic hearing loss. Two mouse models of Hgf dysregulation, one in which an Hgf transgene is ubiquitously overexpressed and the other a conditional knockout that deletes Hgf from a limited number of tissues, including the cochlea, result in deafness. Overexpression of HGF is associated with progressive degeneration of outer hair cells in the cochlea, whereas cochlear deletion of Hgf is associated with more general dysplasia.
Collapse
|
29
|
Ferreira VP, Herbert AP, Cortés C, McKee KA, Blaum BS, Esswein ST, Uhrín D, Barlow PN, Pangburn MK, Kavanagh D. The binding of factor H to a complex of physiological polyanions and C3b on cells is impaired in atypical hemolytic uremic syndrome. THE JOURNAL OF IMMUNOLOGY 2009; 182:7009-18. [PMID: 19454698 DOI: 10.4049/jimmunol.0804031] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Factor H (fH) is essential for complement homeostasis in fluid-phase and on surfaces. Its two C-terminal domains (CCP 19-20) anchor fH to self-surfaces where it prevents C3b amplification in a process requiring its N-terminal four domains. In atypical hemolytic uremic syndrome (aHUS), mutations clustering toward the C terminus of fH may disrupt interactions with surface-associated C3b or polyanions and thereby diminish the ability of fH to regulate complement. To test this, we compared a recombinant protein encompassing CCP 19-20 with 16 mutants. The mutations had only very limited and localized effects on protein structure. Although we found four aHUS-linked fH mutations that decreased binding to C3b and/or to heparin (a model compound for cell surface polyanionic carbohydrates), we identified five aHUS-associated mutants with increased affinity for either or both ligands. Strikingly, these variable affinities for the individual ligands did not correlate with the extent to which all the aHUS-associated mutants were found to be impaired in a more physiological assay that measured their ability to inhibit cell surface complement functions of full-length fH. Taken together, our data suggest that disruption of a complex fH-self-surface recognition process, involving a balance of affinities for protein and physiological carbohydrate ligands, predisposes to aHUS.
Collapse
Affiliation(s)
- Viviana P Ferreira
- Department of Biochemistry, Center for Biomedical Research, University of Texas Health Science Center, Tyler, TX 75708, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Deakin JA, Blaum BS, Gallagher JT, Uhrín D, Lyon M. The binding properties of minimal oligosaccharides reveal a common heparan sulfate/dermatan sulfate-binding site in hepatocyte growth factor/scatter factor that can accommodate a wide variety of sulfation patterns. J Biol Chem 2008; 284:6311-21. [PMID: 19114710 DOI: 10.1074/jbc.m807671200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heparan sulfate (HS)/heparin and dermatan sulfate (DS) both bind with high affinity to hepatocyte growth factor/scatter factor (HGF/SF) and function as necessary co-factors in vitro. How both these two structurally distinct glycosaminoglycans (GAGs) are recognized has remained unclear. We have now reconciled this issue using a panel of minimal tri- and tetrasaccharide sequences of variable but well defined sulfation patterns in combination with further development of the gel mobility shift assay to allow simultaneous comparisons of relative protein affinities/selectivities for different oligosaccharides. From this approach it would seem that a minimum binding sequence is a disulfated trisaccharide comprised of an internal iduronate flanked by monosulfated hexosamine residues and that additional sulfation further enhances affinity. However, the similarity in recognition of HS/heparin and DS seems to arise primarily from a lack of any apparent positional requirement for sulfation. Thus, isomers of HS/heparin tetrasaccharides containing only two sulfates irrespective of whether they are purely N-, 2-O-, or 6-O-sulfates bind with equivalent apparent affinity as a disulfated DS tetrasaccharide. In addition, the NMR chemical shifts induced in NK1 (the truncated variant of HGF/SF comprised of the N-terminal and first Kringle domains) by titration with either heparin or DS oligosaccharides strongly indicate that both bind to essentially the same site. Together, these observations reveal an unexpected degree of flexibility in the GAG-HGF/SF interface, allowing a single binding site in the protein to accommodate iduronate-containing sequences of variable sulfation pattern and/or density from different GAGs.
Collapse
Affiliation(s)
- Jon A Deakin
- Cancer Research UK Glyco-Oncology Group, School of Cancer and Imaging Sciences, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Rd., Manchester M20 4BX, United Kingdom
| | | | | | | | | |
Collapse
|
31
|
Schmidt CQ, Herbert AP, Kavanagh D, Gandy C, Fenton CJ, Blaum BS, Lyon M, Uhrín D, Barlow PN. A new map of glycosaminoglycan and C3b binding sites on factor H. THE JOURNAL OF IMMUNOLOGY 2008; 181:2610-9. [PMID: 18684951 DOI: 10.4049/jimmunol.181.4.2610] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human complement factor H, consisting of 20 complement control protein (CCP) modules, is an abundant plasma glycoprotein. It prevents C3b amplification on self surfaces bearing certain polyanionic carbohydrates, while complement activation progresses on most other, mainly foreign, surfaces. Herein, locations of binding sites for polyanions and C3b are reexamined rigorously by overexpressing factor H segments, structural validation, and binding assays. As anticipated, constructs corresponding to CCPs 7-8 and 19-20 bind well in heparin-affinity chromatography. However, CCPs 8-9, previously reported to bind glycosaminoglycans, bind neither to heparin resin nor to heparin fragments in gel-mobility shift assays. Introduction of nonnative residues N-terminal to a construct containing CCPs 8-9, identical to those in proteins used in the previous report, converted this module pair to an artificially heparin-binding one. The module pair CCPs 12-13 does not bind heparin appreciably, notwithstanding previous suggestions to the contrary. We further checked CCPs 10-12, 11-14, 13-15, 10-15, and 8-15 for ability to bind heparin but found very low affinity or none. As expected, constructs corresponding to CCPs 1-4 and 19-20 bind C3b amine coupled to a CM5 chip (K(d)s of 14 and 3.5 microM, respectively) or a C1 chip (K(d)s of 10 and 4.5 microM, respectively). Constructs CCPs 7-8 and 6-8 exhibit measurable affinities for C3b according to surface plasmon resonance, although they are weak compared with CCPs 19-20. Contrary to expectations, none of several constructs encompassing modules from CCP 9 to 15 exhibited significant C3b binding in this assay. Thus, we propose a new functional map of factor H.
Collapse
Affiliation(s)
- Christoph Q Schmidt
- Schools of Biological Sciences and Chemistry, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Dons L, Jin Y, Kristensson K, Rottenberg ME. Axonal transport of Listeria monocytogenes and nerve-cell-induced bacterial killing. J Neurosci Res 2008; 85:2529-37. [PMID: 17387705 DOI: 10.1002/jnr.21256] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Listeria monocytogenes (L. monocytogenes) can cause fatal brainstem encephalitis in both sheep and humans. Here we review evidence that the bacteria can be incorporated into axons following a primary cycle of replication in macrophages/dendritic cells after subcutaneous injection in projection areas of peripheral neurons. The molecular mechanisms for the rocketing of L. monocytogenes in the cytosol by asymmetric cometic tails and the utility of this phenomenon for bacterial migration intraaxonally both in retro- and in anterograde directions to reach the central nervous system are described. The role of the immune response in the control of L. monocytogenes spread through peripheral neurons is highlighted, and a mechanism by which bacteria may be killed inside infected neurons through a nitric oxide-dependent pathway is pointed out.
Collapse
Affiliation(s)
- Lone Dons
- Department of Veterinary Pathobiology, Faculty of Life Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | | | | |
Collapse
|
33
|
Gemma E, Meyer O, Uhrín D, Hulme AN. Enabling methodology for the end functionalisation of glycosaminoglycan oligosaccharides. MOLECULAR BIOSYSTEMS 2008; 4:481-95. [DOI: 10.1039/b801666f] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
34
|
Catlow KR, Deakin JA, Wei Z, Delehedde M, Fernig DG, Gherardi E, Gallagher JT, Pavão MSG, Lyon M. Interactions of hepatocyte growth factor/scatter factor with various glycosaminoglycans reveal an important interplay between the presence of iduronate and sulfate density. J Biol Chem 2007; 283:5235-48. [PMID: 18156180 DOI: 10.1074/jbc.m706589200] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hepatocyte growth factor/scatter factor (HGF/SF) has a cofactor requirement for heparan sulfate (HS) and dermatan sulfate (DS) in the optimal activation of its signaling receptor MET. However, these two glycosaminoglycans (GAGs) have different sugar backbones and sulfation patterns, with only the presence of iduronate in common. The structural basis for GAG recognition and activation is thus very unclear. We have clarified this by testing a wide array of natural and modified GAGs for both protein binding and activation. Comparisons between Ascidia nigra (2,6-O-sulfated) and mammalian (mainly 4-O-sulfated) DS species, as well as between a panel of specifically desulfated heparins, revealed that no specific sulfate isomer, in either GAG, is vital for interaction and activity. Moreover, different GAGs of similar sulfate density had comparable properties, although affinity and potency notably increase with increasing sulfate density. The weaker interaction with CS-E, compared with DS, shows that GlcA-containing polymers can bind, if highly sulfated, but emphasizes the importance of the flexible IdoA ring. Our data indicate that the preferred binding sites in DS in vivo will be comprised of disulfated, IdoA(2S)-containing motifs. In HS, clustering of N-/2-O-/6-O-sulfation in S-domains will lead to strong reactivity, although binding can also be mediated by the transition zones where sulfates are mainly at the N- and 6-O- positions. GAG recognition of HGF/SF thus appears to be primarily driven by electrostatic interactions and exhibits an interesting interplay between requirements for iduronate and sulfate density that may reflect in part a preference for particular sugar chain conformations.
Collapse
Affiliation(s)
- Krista R Catlow
- Cancer Research UK Glyco-Oncology Group, School of Cancer and Imaging Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester M20 4BX, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Tolbert WD, Daugherty J, Gao C, Xie Q, Miranti C, Gherardi E, Vande Woude G, Xu HE. A mechanistic basis for converting a receptor tyrosine kinase agonist to an antagonist. Proc Natl Acad Sci U S A 2007; 104:14592-7. [PMID: 17804794 PMCID: PMC1965485 DOI: 10.1073/pnas.0704290104] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hepatocyte growth factor (HGF) activates the Met receptor tyrosine kinase by binding and promoting receptor dimerization. Here we describe a mechanistic basis for designing Met antagonists based on NK1, a natural variant of HGF containing the N-terminal and the first kringle domain. Through detailed biochemical and structural analyses, we demonstrate that both mouse and human NK1 induce Met dimerization via a conserved NK1 dimer interface. Mutations designed to alter the NK1 dimer interface abolish its ability to promote Met dimerization but retain full Met-binding activity. Importantly, these NK1 mutants act as Met antagonists by inhibiting HGF-mediated cell scattering, proliferation, branching, and invasion. The ability to separate the Met-binding activity of NK1 from its Met dimerization activity thus provides a rational basis for designing Met antagonists. This strategy of antagonist design may be applicable for other growth factor receptors by selectively abolishing the receptor activation ability but not the receptor binding of the growth factors.
Collapse
Affiliation(s)
| | | | | | - Qian Xie
- Laboratory of Molecular Oncology, and
| | - Cindy Miranti
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, 333 Bostwick Avenue, Grand Rapids, MI 49503; and
| | - Ermanno Gherardi
- Medical Research Council Centre, Laboratory of Molecular Biology, Hills Road, Cambridge CB2 2QH, United Kingdom
- To whom correspondence may be addressed. E-mail: or
| | | | - H. Eric Xu
- *Laboratory of Structural Sciences
- To whom correspondence may be addressed. E-mail: or
| |
Collapse
|
36
|
Raiber EA, Wilkinson JA, Manetti F, Botta M, Deakin J, Gallagher J, Lyon M, Ducki SW. Novel heparin/heparan sulfate mimics as inhibitors of HGF/SF-induced MET activation. Bioorg Med Chem Lett 2007; 17:6321-5. [PMID: 17870532 DOI: 10.1016/j.bmcl.2007.08.074] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Revised: 08/30/2007] [Accepted: 08/31/2007] [Indexed: 11/22/2022]
Abstract
The synthesis of simple, non-sugar glycosaminoglycan (GAG) mimics has been achieved and the analogues evaluated for their ability to inhibit the activation of the MET receptor by hepatocyte growth factor/scatter factor (HGF/SF).
Collapse
Affiliation(s)
- Eun-Ang Raiber
- Centre for Molecular Drug Design, Cockcroft Building, University of Salford, Salford, UK
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Herbert AP, Deakin JA, Schmidt CQ, Blaum BS, Egan C, Ferreira VP, Pangburn MK, Lyon M, Uhrín D, Barlow PN. Structure Shows That a Glycosaminoglycan and Protein Recognition Site in Factor H Is Perturbed by Age-related Macular Degeneration-linked Single Nucleotide Polymorphism. J Biol Chem 2007; 282:18960-8. [PMID: 17360715 DOI: 10.1074/jbc.m609636200] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A common single nucleotide polymorphism in the factor H gene predisposes to age-related macular degeneration. Factor H blocks the alternative pathway of complement on self-surfaces bearing specific polyanions, including the glycosaminoglycan chains of proteoglycans. Factor H also binds C-reactive protein, potentially contributing to noninflammatory apoptotic processes. The at risk sequence contains His (rather than Tyr) at position 402 (384 in the mature protein), in the seventh of the 20 complement control protein (CCP) modules (CCP7) of factor H. We expressed both His(402) and Tyr(402) variants of CCP7, CCP7,8, and CCP6-8. We determined structures of His(402) and Tyr(402) CCP7 and showed them to be nearly identical. The side chains of His/Tyr(402) have similar, solvent-exposed orientations far from interfaces with CCP6 and -8. Tyr(402) CCP7 bound significantly more tightly than His(402) CCP7 to a heparin affinity column as well as to defined-length sulfated heparin oligosaccharides employed in gel mobility shift assays. This observation is consistent with the position of the 402 side chain on the edge of one of two glycosaminoglycan-binding surface patches on CCP7 that we inferred on the basis of chemical shift perturbation studies with a sulfated heparin tetrasaccharide. According to surface plasmon resonance measurements, Tyr(402) CCP6-8 binds significantly more tightly than His(402) CCP6-8 to immobilized C-reactive protein. The data support a causal link between H402Y and age-related macular degeneration in which variation at position 402 modulates the response of factor H to age-related changes in the glycosaminoglycan composition and apoptotic activity of the macula.
Collapse
Affiliation(s)
- Andrew P Herbert
- Edinburgh Biomolecular NMR Unit, School of Chemistry and School of Biological Sciences, University of Edinburgh, West Mains Road, Edinburgh EH9 3JJ, Scotland, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Gemma E, Hulme AN, Jahnke A, Jin L, Lyon M, Müller RM, Uhrín D. DMT-MM mediated functionalisation of the non-reducing end of glycosaminoglycans. Chem Commun (Camb) 2007:2686-8. [PMID: 17594020 DOI: 10.1039/b617038b] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Efficient functionalisation of the non-reducing end of uronic acid derivatives and glycosaminoglycan-derived disaccharides using peptide coupling has been achieved, mediated by the water-soluble agent DMT-MM.
Collapse
Affiliation(s)
- Emiliano Gemma
- School of Chemistry, University of Edinburgh, West Mains Road, Edinburgh, UK EH9 3JJ
| | | | | | | | | | | | | |
Collapse
|
39
|
Sakiyama R, Fukuta K, Matsumoto K, Furukawa M, Takahashi Y, Nakamura T. Stimulation of Hepatocyte Growth Factor Production by Heparin-derived Oligosaccharides. ACTA ACUST UNITED AC 2007; 141:653-60. [PMID: 17317686 DOI: 10.1093/jb/mvm067] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
We previously reported that heparin post-transcriptionally stimulates the production of hepatocyte growth factor (HGF). In this study, we addressed the size-dependency of heparin fragments on the HGF-inducing activity aiming to obtain fragments without antiblood coagulant activity. Heparin fragments, produced by digestion with heparinase, were size-fractionated and tested for HGF-inducing activity in cultured human fibroblasts. The HGF-inducing activity deceased with the reduction in oligosaccharide size. Decasaccharides exerted an activity comparable with undigested heparin, while smaller oligosaccharides showed lesser activities. The anticoagulant activity of heparin fragments also decreased with size and anticoagulant activity of decasaccharides was <13% that of undigested heparin. Further fractionation of decasaccharides by anion-exchange chromatography revealed that most of the decasaccharides had HGF-inducing activity and the extent of sulfation was roughly related to the activity. The lack of N-sulfation in heparin markedly reduced HGF-inducing activity, whereas 2-O-desulfation or 6-O-desulation had a lesser influence. Moreover, an N-sulfated disaccharide showed significant HGF-inducing activity, suggesting the involvement of N-sulfation in HGF-inducing activity. Because of the much reduced anticoagulant activity, potential applications of heparin-derived oligosaccharides such as decasaccharides is considerable as a therapeutic agent for many diseases.
Collapse
Affiliation(s)
- Ryoichi Sakiyama
- Division of Molecular Regenerative Medicine, Department of Biochemistry and Molecular Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Holmes O, Pillozzi S, Deakin JA, Carafoli F, Kemp L, Butler PJG, Lyon M, Gherardi E. Insights into the structure/function of hepatocyte growth factor/scatter factor from studies with individual domains. J Mol Biol 2007; 367:395-408. [PMID: 17258232 DOI: 10.1016/j.jmb.2006.12.061] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 12/14/2006] [Accepted: 12/19/2006] [Indexed: 11/20/2022]
Abstract
Hepatocyte growth factor/scatter factor (HGF/SF), the ligand for the receptor tyrosine kinase encoded by the c-Met proto-oncogene, is a multidomain protein structurally related to the pro-enzyme plasminogen and with major roles in development, tissue regeneration and cancer. We have expressed the N-terminal (N) domain, the four kringle domains (K1 to K4) and the serine proteinase homology domain (SP) of HGF/SF individually in yeast or mammalian cells and studied their ability to: (i) bind the Met receptor as well as heparan sulphate and dermatan sulphate co-receptors, (ii) activate Met in target cells and, (iii) map their binding sites onto the beta-propeller domain of Met. The N, K1 and SP domains bound Met directly with comparable affinities (K(d)=2.4, 3.3 and 1.4 microM). The same domains also bound heparin with decreasing affinities (N>K1>>SP) but only the N domain bound dermatan sulphate. Three kringle domains (K1, K2 and K4) displayed agonistic activity on target cells. In contrast, the N and SP domains, although capable of Met binding, displayed no or little activity. Further, cross-linking experiments demonstrated that both the N domain and kringles 1-2 bind the beta-chain moiety (amino acid residues 308-514) of the Met beta-propeller. In summary, the K1, K2 and K4 domains of HGF/SF are sufficient for Met activation, whereas the N and SP domains are not, although the latter domains contribute additional binding sites necessary for receptor activation by full length HGF/SF. The results provide new insights into the structure/function of HGF/SF and a basis for engineering the N and K1 domains as receptor antagonists for cancer therapy.
Collapse
Affiliation(s)
- O Holmes
- MRC Centre, Laboratory of Molecular Biology, Hills Road, Cambridge, CB2 2QH, UK
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Sasisekharan R, Raman R, Prabhakar V. GLYCOMICS APPROACH TO STRUCTURE-FUNCTION RELATIONSHIPS OF GLYCOSAMINOGLYCANS. Annu Rev Biomed Eng 2006; 8:181-231. [PMID: 16834555 DOI: 10.1146/annurev.bioeng.8.061505.095745] [Citation(s) in RCA: 230] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Extracellular modulation of phenotype is an emerging paradigm in this current postgenomics age of molecular and cell biology. Glycosaminoglycans (GAGs) are primary components of the cell surface and the cell-extracellular matrix (ECM) interface. Advances in the technology to analyze GAGs and in whole-organism genetics have led to a dramatic increase in the known important biological role of these complex polysaccharides. Owing to their ubiquitous distribution at the cell-ECM interface, GAGs interact with numerous proteins and modulate their activity, thus impinging on fundamental biological processes such as cell growth and development. Many recent reviews have captured important aspects of GAG structure and biosynthesis, GAG-protein interactions, and GAG biology. GAG research is currently at a stage where there is a need for an integrated systems or glycomics approach, which involves an integration of all of the above concepts to define their structure-function relationships. Focusing on heparin/heparan (HSGAGs) and chondroitin/dermatan sulfate (CSGAGs), this review highlights the important aspects of GAGs and summarizes these aspects in the context of taking a glycomics approach that integrates the different technologies to define structure-function relationships of GAGs.
Collapse
Affiliation(s)
- Ram Sasisekharan
- Biological Engineering Division, Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| | | | | |
Collapse
|
42
|
Kemp LE, Mulloy B, Gherardi E. Signalling by HGF/SF and Met: the role of heparan sulphate co-receptors. Biochem Soc Trans 2006; 34:414-7. [PMID: 16709175 DOI: 10.1042/bst0340414] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The receptor tyrosine kinase Met and its ligand HGF/SF (hepatocyte growth factor/scatter factor) are essential in the signalling pathways required for embryogenesis and tissue regeneration. Aberrant signalling of this complex is also a feature of many tumours and appears to contribute to the growth, invasiveness and metastasis of both carcinomas and sarcomas. HGF/SF, like many other angiogenic growth factors, employs heparan sulphate as co-receptor. The role of this interaction has not been completely defined but appears to be physiologically relevant. Thus the presence of heparin increases the potency of HGF/SF in experiments with cells in culture leading to elevated downstream signalling effects and, although not vital for the Met-HGF/SF interaction, heparin or heparan sulphate is essential for the activity of certain isoforms of HGF/SF, such as NK1 and NK2. Here, we summarize the progress made in understanding the interaction between heparin and heparan sulphate and NK1, NK2 and HGF/SF and we discuss their role in HGF/SF-Met signalling.
Collapse
Affiliation(s)
- L E Kemp
- MRC Centre, Hills Road, Cambridge, UK
| | | | | |
Collapse
|
43
|
Gallagher JT. Multiprotein signalling complexes: regional assembly on heparan sulphate. Biochem Soc Trans 2006; 34:438-41. [PMID: 16709181 DOI: 10.1042/bst0340438] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Heparan sulphate (HS) is an abundant component of cell surfaces and the extracellular matrix. It binds to a wide variety of peptide growth factors, morphogens, chemokines and extracellular matrix proteins (e.g. fibronectin) and many of these interactions are essential for these effector proteins to transduce signals across the plasma membrane. The unique molecular design and flexibility of HS are essential for its ability to exert control over the cellular response to proteinaceous ligands. The clustering of sulphated sugar residues in a series of complex domains with variable sulphation patterns generates considerable diversity in the molecular fine structure of HS. This diversity reflects a high degree of selectivity in protein recognition and in the assembly of functional multiprotein complexes on the HS polymer chain.
Collapse
Affiliation(s)
- J T Gallagher
- CRUK Department of Medical Oncology, University of Manchester, Paterson Institute for Cancer Research, UK.
| |
Collapse
|
44
|
Herbert AP, Uhrín D, Lyon M, Pangburn MK, Barlow PN. Disease-associated sequence variations congregate in a polyanion recognition patch on human factor H revealed in three-dimensional structure. J Biol Chem 2006; 281:16512-20. [PMID: 16533809 DOI: 10.1074/jbc.m513611200] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mutations and polymorphisms in the regulator of complement activation, factor H, have been linked to atypical hemolytic uremic syndrome (aHUS), membranoproliferative glomerulonephritis, and age-related macular degeneration. Many aHUS patients carry mutations in the two C-terminal modules of factor H, which normally confer upon this abundant 155-kDa plasma glycoprotein its ability to selectively bind self-surfaces and prevent them from inappropriately triggering the complement cascade via the alternative pathway. In the current study, the three-dimensional solution structure of the C-terminal module pair of factor H has been determined. A binding site for a fully sulfated heparin-derived tetrasaccharide has been delineated using chemical shift mapping and the C3d/C3b-binding site inferred from sequence comparisons and computational docking. The resultant information allows assessment of the likely consequences of aHUS-associated amino acid substitutions in this critical region of factor H. It is striking that, excepting those likely to perturb the three-dimensional structure, aHUS-associated missense mutations congregate in the polyanion-binding site delineated in this study, thus potentially disrupting a vital mechanism for control of complement on self-surfaces in the microvasculature of the kidney. It is intriguing that a single nucleotide polymorphism predisposing to age-related macular degeneration occupies another region of factor H that harbors a polyanion-binding site.
Collapse
Affiliation(s)
- Andrew P Herbert
- Edinburgh Biomolecular NMR Unit, University of Edinburgh, West mains Road, Edinburgh EH9 3JJ, United Kingdom
| | | | | | | | | |
Collapse
|
45
|
Robinson CJ, Harmer NJ, Goodger SJ, Blundell TL, Gallagher JT. Cooperative Dimerization of Fibroblast Growth Factor 1 (FGF1) upon a Single Heparin Saccharide May Drive the Formation of 2:2:1 FGF1·FGFR2c·Heparin Ternary Complexes. J Biol Chem 2005; 280:42274-82. [PMID: 16219767 DOI: 10.1074/jbc.m505720200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The related glycosaminoglycans heparin and heparan sulfate are essential for the activity of the fibroblast growth factor (FGF) family as they form an integral part of the signaling complex at the cell surface. Using size-exclusion chromatography we have studied the capacities of a variety of heparin oligosaccharides to bind FGF1 and FGFR2c both separately and together in ternary complexes. In the absence of heparin, FGF1 had no detectable affinity for FGFR2c. However, 2:2:1 complexes formed spontaneously in solution between FGF1, FGFR2c, and heparin octasaccharide (dp8). The dp8 sample was the shortest chain length that bound FGFR2c, that dimerized FGF1, and that promoted a strong mitogenic response to FGF1 through FGFR2c. Heparin hexasaccharide and various selectively desulfated heparin dp12s failed to bind FGFR2c and could only interact with FGF1 monomerically. These saccharides formed 1:1:1 complexes with FGF1 and FGFR2c, which had no tendency to self-associate, suggesting that binding of two FGF1 molecules to the same saccharide chain is a prerequisite for subsequent FGFR2c dimerization. We found that FGF1 dimerization upon heparin was favored over monomeric interactions even when a large excess of saccharide was present. A cooperative mechanism of FGF1 dimerization could explain how 2:2:1 signaling complexes form at the cell surface, an environment rich in heparan sulfate.
Collapse
Affiliation(s)
- Christopher J Robinson
- Cancer Research UK and Department of Medical Oncology, University of Manchester, Christie Hospital National Health Service Trust, Wilmslow Road, Manchester M20 4BX.
| | | | | | | | | |
Collapse
|
46
|
Sarrazin S, Adam E, Lyon M, Depontieu F, Motte V, Landolfi C, Lortat-Jacob H, Bechard D, Lassalle P, Delehedde M. Endocan or endothelial cell specific molecule-1 (ESM-1): a potential novel endothelial cell marker and a new target for cancer therapy. Biochim Biophys Acta Rev Cancer 2005; 1765:25-37. [PMID: 16168566 DOI: 10.1016/j.bbcan.2005.08.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Revised: 08/08/2005] [Accepted: 08/10/2005] [Indexed: 12/21/2022]
Abstract
Endocan, previously called endothelial cell specific molecule-1, is a soluble proteoglycan of 50 kDa, constituted of a mature polypeptide of 165 amino acids and a single dermatan sulphate chain covalently linked to the serine residue at position 137. This dermatan sulphate proteoglycan, which is expressed by the vascular endothelium, has been found freely circulating in the bloodstream of healthy subjects. Experimental evidence is accumulating that implicates endocan as a key player in the regulation of major processes such as cell adhesion, in inflammatory disorders and tumor progression. Inflammatory cytokines such as TNF-alpha, and pro-angiogenic growth factors such as VEGF, FGF-2 and HGF/SF, strongly increased the expression, synthesis or the secretion of endocan by human endothelial cells. Endocan is clearly overexpressed in human tumors, with elevated serum levels being observed in late-stage lung cancer patients, as measured by enzyme-linked immunoassay, and with its overexpression in experimental tumors being evident by immunohistochemistry. Recently, the mRNA levels of endocan have also been recognized as being one of the most significant molecular signatures of a bad prognosis in several types of cancer including lung cancer. Overexpression of this dermatan sulphate proteoglycan has also been shown to be directly involved in tumor progression as observed in mouse models of human tumor xenografts. Collectively, these results suggest that endocan could be a biomarker for both inflammatory disorders and tumor progression as well as a validated therapeutic target in cancer. On the basis of the recent successes of immunotherapeutic approaches in cancer, the preclinical data on endocan suggests that an antibody raised against the protein core of endocan could be a promising cancer therapy.
Collapse
Affiliation(s)
- S Sarrazin
- ENDOTIS PHARMA, Parc Eurasanté, 70 rue du Dr. Yersin, 59120 Loos, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Raman R, Sasisekharan V, Sasisekharan R. Structural insights into biological roles of protein-glycosaminoglycan interactions. ACTA ACUST UNITED AC 2005; 12:267-77. [PMID: 15797210 DOI: 10.1016/j.chembiol.2004.11.020] [Citation(s) in RCA: 301] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2004] [Revised: 10/27/2004] [Accepted: 11/15/2004] [Indexed: 02/02/2023]
Abstract
The extracellular environment is largely comprised of complex polysaccharides, which were historically considered inert materials that hydrated the cells and contributed to the structural scaffolds. Recent advances in development of sophisticated analytical techniques have brought about a dramatic transformation in understanding the numerous biological roles of these complex polysaccharides. Glycosaminoglycans (GAGs) are a class of these polysaccharides, which bind to a wide variety of proteins and signaling molecules in the cellular environment and modulate their activity, thus impinging on fundamental biological processes. Despite the importance of GAGs modulating biological functions, there are relatively few examples that demonstrate specificity of GAG-protein interactions, which in turn define the structure-function relationships of these polysaccharides. Focusing on heparin/heparan (HSGAGs) and chondroitin/dermatan sulfate (CSGAGs), this review provides structural insights into the oligosaccharide-protein interactions and discusses some key and challenging aspects of understanding GAG structure-function relationships.
Collapse
Affiliation(s)
- Rahul Raman
- Biological Engineering Division, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
48
|
Abstract
Listeria monocytogenes is a Gram-positive bacterium that is able to invade and multiply within eukaryotic cells. Its intracellular life-cycle includes pathogen-induced phagocytosis, lysis of the phagocytic vacuole, movement in the cytoplasmic environment, and a cell-to-cell spread mechanism. Many L. monocytogenes virulence factors have been studied in detail, certain of which subvert specific eukaryotic cell functions in order to favour infection. During entry, the invasion protein InlA takes advantage of the adhesion molecule E-cadherin and the adherens junction machinery to adhere to target and invade polarized epithelial cells. Another invasion protein of the internalin family, InlB, subverts the signalling pathway of the hepatocyte growth factor receptor Met to induce endocytosis of the receptor and also to favour internalization of the bacteria in non-polarized epithelial cells. Once inside the cell, the haemolysin of L. monocytogenes--the listeriolysin O or LLO--is secreted to lyse the phagocytic vacuole, and when the bacteria is freed in the cytoplasm, the activity of the LLO is in part regulated by the infected cell itself, taking advantage of the pH sensitivity of the LLO that leads to its inactivation in the neutral eukaryotic cell cytoplasm. Finally, to induce bacterial movement in the cytoplasm, the L. monocytogenes surface protein ActA mimics the activity of the eukaryotic WASP family of proteins to recruit to the bacteria the actin nucleation machinery required for actin polymerization and for the formation of the actin structures (called 'actin comet tails') that propel the parasite in the cytosol and help it to invade neighbouring cells.
Collapse
Affiliation(s)
- Javier Pizarro-Cerdá
- Unité des Interactions Bactéries-Cellules/INSERM U604/INRA USC2020, Institut Pasteur, Paris, France.
| | | |
Collapse
|
49
|
Seyfried NT, Blundell CD, Day AJ, Almond A. Preparation and application of biologically active fluorescent hyaluronan oligosaccharides. Glycobiology 2004; 15:303-12. [PMID: 15496500 DOI: 10.1093/glycob/cwi008] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We report the production of biologically active hyaluronan (HA) oligosaccharides labeled with the fluorophore 2-aminobenzoic acid (2AA). Oligosaccharides from 4 to 40 residues in length were purified to homogeneity by ion exchange chromatography using a logarithmic gradient. Molecular weight and purity characterization of HA oligosaccharides is facilitated by 2AA derivatization because it enhances signals in MALDI-TOF MS and improves FACE (fluorophore-assisted carbohydrate electrophoresis) analysis by avoiding the inverted parabolic migration characteristic of 2-aminoacridone (AMAC)-labeled sugars. The small size and shape of the fluorophore maintains the biological activity of the derivatized oligosaccharides, as demonstrated by their ability to compete for polymeric HA binding to the G1-domain of human recombinant versican (VG1). An electrophoretic mobility shift assay was used to study VG1 binding to labeled HA 8-, 10-, 20-, 30-, and 40-mers, and although no stable VG1 binding was observed to labeled 8-mers, the equilibrium dissociation constant (100 nM) for VG1 with HA(10) was estimated from densitometry analysis of the free oligosaccharide. Interactions involving HA 20-, 30-, and 40-mers (proposed to be multivalent) could also be studied using this protocol. Oligosaccharides labeled with 2AA therefore show excellent potential as probes in fluorescence-based assays that investigate protein-carbohydrate interactions.
Collapse
Affiliation(s)
- Nicholas T Seyfried
- MRC Immunochemistry Unit, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | | | | | | |
Collapse
|