1
|
Su D, Li Y, Yates EA, Skidmore MA, Lima MA, Fernig DG. Analysis of protein-heparin interactions using a portable SPR instrument. PEERJ ANALYTICAL CHEMISTRY 2022. [DOI: 10.7717/peerj-achem.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Optical biosensors such as those based on surface plasmon resonance (SPR) are a key analytical tool for understanding biomolecular interactions and function as well as the quantitative analysis of analytes in a wide variety of settings. The advent of portable SPR instruments enables analyses in the field. A critical step in method development is the passivation and functionalisation of the sensor surface. We describe the assembly of a surface of thiolated oleyl ethylene glycol/biotin oleyl ethylene glycol and its functionalisation with streptavidin and reducing end biotinylated heparin for a portable SPR instrument. Such surfaces can be batch prepared and stored. Two examples of the analysis of heparin-binding proteins are presented. The binding of fibroblast growth factor 2 and competition for the binding of a heparan sulfate sulfotransferase by a library of selectively modified heparins and suramin, which identify the selectivity of the enzyme for sulfated structures in the polysaccharide and demonstrate suramin as a competitor for the enzyme’s sugar acceptor site. Heparin functionalised surfaces should have a wide applicability, since this polysaccharide is a close structural analogue of the host cell surface polysaccharide, heparan sulfate, a receptor for many endogenous proteins and viruses.
Collapse
Affiliation(s)
- Dunhao Su
- Biochemistry, University of Liverpool, Liverpool, United Kingdom
| | - Yong Li
- Biochemistry, University of Liverpool, Liverpool, United Kingdom
| | - Edwin A. Yates
- Biochemistry, University of Liverpool, Liverpool, United Kingdom
| | - Mark A. Skidmore
- Molecular & Structural Biosciences, School of Life Sciences, University of Keele, Newcastle-Under-Lyme, United Kingdom
| | - Marcelo A. Lima
- Molecular & Structural Biosciences, School of Life Sciences, University of Keele, Newcastle-Under-Lyme, United Kingdom
| | - David G. Fernig
- Biochemistry, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
2
|
Sun C, Liu M, Sun P, Yang M, Yates EA, Guo Z, Fernig DG. Sulfated polysaccharides interact with fibroblast growth factors and protect from denaturation. FEBS Open Bio 2019; 9:1477-1487. [PMID: 31271519 PMCID: PMC6668377 DOI: 10.1002/2211-5463.12696] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/26/2019] [Accepted: 07/03/2019] [Indexed: 01/08/2023] Open
Abstract
Fibroblast growth factors (FGFs) regulate embryonic development and homeostasis, including tissue and organ repair and specific aspects of metabolism. The basic FGF and acidic FGF, now known as FGF2 and FGF1, are widely used protein drugs for tissue repair. However, they are susceptible to denaturation at ambient temperatures and during long-time storage, which will reduce their biological activity. The interaction of FGFs with the sulfated domains of heparan sulfate and heparin is essential for their cellular signaling and stability. Therefore, we analyzed the interactions of FGF1 and FGF2 with four sulfated polysaccharides: heparin, dextran sulfate (DXS), λ-carrageenan, and chondroitin sulfate. The results of thermal stability and cell proliferation assays demonstrate that heparin, DXS, and λ-carrageenan bound to both FGFs and protected them from denaturation. Our results suggest heparin, DXS, and λ-carrageenan are potential formulation materials that bind and stabilize FGFs, and which may also potentiate their activity and control their delivery.
Collapse
Affiliation(s)
- Changye Sun
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityChina
| | - Mengxin Liu
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityChina
| | - Panwen Sun
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityChina
| | - Mingming Yang
- Department of CardiologySchool of MedicineAffiliated Zhongda HospitalSoutheast UniversityNanjingChina
| | - Edwin A. Yates
- Department of BiochemistryInstitute of Integrative BiologyUniversity of LiverpoolUK
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityChina
| | - David G. Fernig
- Department of BiochemistryInstitute of Integrative BiologyUniversity of LiverpoolUK
| |
Collapse
|
3
|
Collins LE, Troeberg L. Heparan sulfate as a regulator of inflammation and immunity. J Leukoc Biol 2018; 105:81-92. [PMID: 30376187 DOI: 10.1002/jlb.3ru0618-246r] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/30/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022] Open
Abstract
Heparan sulfate is found on the surface of most cell types, as well as in basement membranes and extracellular matrices. Its strong anionic properties and highly variable structure enable this glycosaminoglycan to provide binding sites for numerous protein ligands, including many soluble mediators of the immune system, and may promote or inhibit their activity. The formation of ligand binding sites on heparan sulfate (HS) occurs in a tissue- and context-specific fashion through the action of several families of enzymes, most of which have multiple isoforms with subtly different specificities. Changes in the expression levels of these biosynthetic enzymes occur in response to inflammatory stimuli, resulting in structurally different HS and acquisition or loss of binding sites for immune mediators. In this review, we discuss the multiple roles for HS in regulating immune responses, and the evidence for inflammation-associated changes to HS structure.
Collapse
Affiliation(s)
- Laura E Collins
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Linda Troeberg
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
4
|
Abstract
Simple and complex carbohydrates (glycans) have long been known to play major metabolic, structural and physical roles in biological systems. Targeted microbial binding to host glycans has also been studied for decades. But such biological roles can only explain some of the remarkable complexity and organismal diversity of glycans in nature. Reviewing the subject about two decades ago, one could find very few clear-cut instances of glycan-recognition-specific biological roles of glycans that were of intrinsic value to the organism expressing them. In striking contrast there is now a profusion of examples, such that this updated review cannot be comprehensive. Instead, a historical overview is presented, broad principles outlined and a few examples cited, representing diverse types of roles, mediated by various glycan classes, in different evolutionary lineages. What remains unchanged is the fact that while all theories regarding biological roles of glycans are supported by compelling evidence, exceptions to each can be found. In retrospect, this is not surprising. Complex and diverse glycans appear to be ubiquitous to all cells in nature, and essential to all life forms. Thus, >3 billion years of evolution consistently generated organisms that use these molecules for many key biological roles, even while sometimes coopting them for minor functions. In this respect, glycans are no different from other major macromolecular building blocks of life (nucleic acids, proteins and lipids), simply more rapidly evolving and complex. It is time for the diverse functional roles of glycans to be fully incorporated into the mainstream of biological sciences.
Collapse
Affiliation(s)
- Ajit Varki
- Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center, University of California at San Diego, La Jolla, CA 92093-0687, USA
| |
Collapse
|
5
|
Li Y, Sun C, Yates EA, Jiang C, Wilkinson MC, Fernig DG. Heparin binding preference and structures in the fibroblast growth factor family parallel their evolutionary diversification. Open Biol 2016; 6:rsob.150275. [PMID: 27030175 PMCID: PMC4821243 DOI: 10.1098/rsob.150275] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The interaction of a large number of extracellular proteins with heparan sulfate (HS) regulates their transport and effector functions, but the degree of molecular specificity underlying protein–polysaccharide binding is still debated. The 15 paracrine fibroblast growth factors (FGFs) are one of the paradigms for this interaction. Here, we measure the binding preferences of six FGFs (FGF3, FGF4, FGF6, FGF10, FGF17, FGF20) for a library of modified heparins, representing structures in HS, and model glycosaminoglycans, using differential scanning fluorimetry. This is complemented by the identification of the lysine residues in the primary and secondary binding sites of the FGFs by a selective labelling approach. Pooling these data with previous sets provides good coverage of the FGF phylogenetic tree, deduced from amino acid sequence alignment. This demonstrates that the selectivity of the FGFs for binding structures in sulfated polysaccharides and the pattern of secondary binding sites on the surface of FGFs follow the phylogenetic relationship of the FGFs, and so are likely to be the result of the natural selection pressures that led to the expansion of the FGF family in the course of the evolution of more complex animal body plans.
Collapse
Affiliation(s)
- Yong Li
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool L69 7ZB, UK
| | - Changye Sun
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool L69 7ZB, UK
| | - Edwin A Yates
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool L69 7ZB, UK
| | - Chao Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Mark C Wilkinson
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool L69 7ZB, UK
| | - David G Fernig
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool L69 7ZB, UK
| |
Collapse
|
6
|
Meneghetti MCZ, Hughes AJ, Rudd TR, Nader HB, Powell AK, Yates EA, Lima MA. Heparan sulfate and heparin interactions with proteins. J R Soc Interface 2016; 12:0589. [PMID: 26289657 DOI: 10.1098/rsif.2015.0589] [Citation(s) in RCA: 213] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Heparan sulfate (HS) polysaccharides are ubiquitous components of the cell surface and extracellular matrix of all multicellular animals, whereas heparin is present within mast cells and can be viewed as a more sulfated, tissue-specific, HS variant. HS and heparin regulate biological processes through interactions with a large repertoire of proteins. Owing to these interactions and diverse effects observed during in vitro, ex vivo and in vivo experiments, manifold biological/pharmacological activities have been attributed to them. The properties that have been thought to bestow protein binding and biological activity upon HS and heparin vary from high levels of sequence specificity to a dependence on charge. In contrast to these opposing opinions, we will argue that the evidence supports both a level of redundancy and a degree of selectivity in the structure-activity relationship. The relationship between this apparent redundancy, the multi-dentate nature of heparin and HS polysaccharide chains, their involvement in protein networks and the multiple binding sites on proteins, each possessing different properties, will also be considered. Finally, the role of cations in modulating HS/heparin activity will be reviewed and some of the implications for structure-activity relationships and regulation will be discussed.
Collapse
Affiliation(s)
- Maria C Z Meneghetti
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), Rua Três de Maio, São Paulo 40440-020, Brazil
| | - Ashley J Hughes
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg 40530, Sweden Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Timothy R Rudd
- The National Institute for Biological Standards and Control (NIBSC), South Mimms, Potters Bar, Hertfordshire EN6 3QC, UK Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Helena B Nader
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), Rua Três de Maio, São Paulo 40440-020, Brazil
| | - Andrew K Powell
- School of Pharmacy and Biomolecular Science, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, UK Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Edwin A Yates
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), Rua Três de Maio, São Paulo 40440-020, Brazil
| | - Marcelo A Lima
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), Rua Três de Maio, São Paulo 40440-020, Brazil Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| |
Collapse
|
7
|
Sun C, Marcello M, Li Y, Mason D, Lévy R, Fernig DG. Selectivity in glycosaminoglycan binding dictates the distribution and diffusion of fibroblast growth factors in the pericellular matrix. Open Biol 2016; 6:150277. [PMID: 27009190 PMCID: PMC4821244 DOI: 10.1098/rsob.150277] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/02/2016] [Indexed: 12/13/2022] Open
Abstract
The range of biological outcomes generated by many signalling proteins in development and homeostasis is increased by their interactions with glycosaminoglycans, particularly heparan sulfate (HS). This interaction controls the localization and movement of these signalling proteins, but whether such control depends on the specificity of the interactions is not known. We used five fibroblast growth factors with an N-terminal HaloTag (Halo-FGFs) for fluorescent labelling, with well-characterized and distinct HS-binding properties, and measured their binding and diffusion in pericellular matrix of fixed rat mammary 27 fibroblasts. Halo-FGF1, Halo-FGF2 and Halo-FGF6 bound to HS, whereas Halo-FGF10 also interacted with chondroitin sulfate/dermatan sulfate, and FGF20 did not bind detectably. The distribution of bound FGFs in the pericellular matrix was not homogeneous, and for FGF10 exhibited striking clusters. Fluorescence recovery after photobleaching showed that FGF2 and FGF6 diffused faster, whereas FGF1 diffused more slowly, and FGF10 was immobile. The results demonstrate that the specificity of the interactions of proteins with glycosaminoglycans controls their binding and diffusion. Moreover, cells regulate the spatial distribution of different protein-binding sites in glycosaminoglycans independently of each other, implying that the extracellular matrix has long-range structure.
Collapse
Affiliation(s)
- Changye Sun
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Marco Marcello
- Centre for Cell Imaging, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Yong Li
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - David Mason
- Centre for Cell Imaging, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Raphaël Lévy
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - David G Fernig
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| |
Collapse
|
8
|
Advances in the molecular functions of syndecan-1 (SDC1/CD138) in the pathogenesis of malignancies. Crit Rev Oncol Hematol 2014; 94:1-17. [PMID: 25563413 DOI: 10.1016/j.critrevonc.2014.12.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 11/28/2014] [Accepted: 12/10/2014] [Indexed: 01/08/2023] Open
Abstract
Syndecan-1 (SDC1, synd, CD138) is the most widely studied member of four structurally related cell surface heparan sulfate proteoglycans (HSPG). Although SDC1 has been implicated in a wide range of biological functions, its altered expression often produces malignant phenotypes, which arise from increased cell proliferation and cell growth, cell survival, cell invasion and metastasis, and angiogenesis. Recent studies revealed much about the underlying molecular roles of SDC1 in these processes. The changes in SDC1 expression also have a direct impact on the clinical course of cancers, as evident by its prognostic significance. Accumulating evidence suggest that SDC1 is involved in stimulation of cancer stem cells (CSC) or tumor initiating cells (TIC) and this may affect disease relapse, and resistance to therapy. This review discusses the progress on the pro-tumorigenic role(s) of SDC1 and how these roles may impact the clinical aspect of the disease. Also discussed, are the current strategies for targeting SDC1 or its related signaling.
Collapse
|
9
|
Angiogenic growth factors interactome and drug discovery: The contribution of surface plasmon resonance. Cytokine Growth Factor Rev 2014; 26:293-310. [PMID: 25465594 DOI: 10.1016/j.cytogfr.2014.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 11/21/2022]
Abstract
Angiogenesis is implicated in several pathological conditions, including cancer, and in regenerative processes, including the formation of collateral blood vessels after stroke. Physiological angiogenesis is the outcome of a fine balance between the action of angiogenic growth factors (AGFs) and anti-angiogenic molecules, while pathological angiogenesis occurs when this balance is pushed toward AGFs. AGFs interact with multiple endothelial cell (EC) surface receptors inducing cell proliferation, migration and proteases upregulation. On the contrary, free or extracellular matrix-associated molecules inhibit angiogenesis by sequestering AGFs (thus hampering EC stimulation) or by interacting with specific EC receptors inducing apoptosis or decreasing responsiveness to AGFs. Thus, angiogenesis results from an intricate network of interactions among pro- and anti-angiogenic molecules, EC receptors and various modulators. All these interactions represent targets for the development of pro- or anti-angiogenic therapies. These aims call for suitable technologies to study the countless interactions occurring during neovascularization. Surface plasmon resonance (SPR) is a label-free optical technique to study biomolecular interactions in real time. It has become the golden standard technology for interaction analysis in biomedical research, including angiogenesis. From a survey of the literature it emerges that SPR has already contributed substantially to the better understanding of the neovascularization process, laying the basis for the decoding of the angiogenesis "interactome" and the identification of "hub molecules" that may represent preferential targets for an efficacious modulation of angiogenesis. Here, the still unexploited full potential of SPR is enlightened, pointing to improvements in its use for a deeper understanding of the mechanisms of neovascularization and the identification of novel anti-angiogenic drugs.
Collapse
|
10
|
Dos Santos C, Blanc C, Elahouel R, Prescott M, Carpentier G, Ori A, Courty J, Hamma-Kourbali Y, Fernig DG, Delbé J. Proliferation and migration activities of fibroblast growth factor-2 in endothelial cells are modulated by its direct interaction with heparin affin regulatory peptide. Biochimie 2014; 107 Pt B:350-7. [PMID: 25315978 DOI: 10.1016/j.biochi.2014.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 10/02/2014] [Indexed: 11/15/2022]
Abstract
Angiogenesis is the physiological process involving the growth of new blood vessels from pre-existing vessels. In normal or pathological angiogenesis, angiogenic growth factors activate cognate receptors on endothelial cells. Fibroblast growth factor-2 (FGF-2) and heparin affin regulatory peptide (HARP) are two heparin-binding growth factors and were described for their pro-angiogenic properties on human umbilical endothelial cells (HUVEC). We now show that HARP acts as a mediator of FGF-2's stimulatory effects, since it is able to inhibit the proliferation and migration of HUVEC induced by FGF-2. We demonstrate by ELISA and optical biosensor binding assay that HARP and FGF-2 interact through direct binding. We have adapted a previously developed structural proteomics method for the identification of residues involved in protein-protein interactions. Application of this method showed that two sequences in HARP were involved in binding FGF-2. One was in the C-thrombospondin type 1 repeat (C-TSR-1) domain and the other in the C-terminal domain of HARP. The identification of these regions as mediating the binding of FGF-2 was confirmed by ELISA using synthetic peptides, which are as well mediators of FGF-2-induced proliferation, migration and tubes formation on HUVEC in vitro. These results imply that besides a regulation of the proliferation, migration and angiogenesis of HUVEC by direct interaction of FGF-2 with its receptors, an alternative pathway exists involving its binding to growth factors such as HARP.
Collapse
Affiliation(s)
- Célia Dos Santos
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France.
| | - Charly Blanc
- IMRB INSERM, U955, Equipe 07, Faculté de Médecine, 8 rue du Général Sarrail, 94010 Créteil, France
| | - Rania Elahouel
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | - Mark Prescott
- Department of Structural and Chemical Biology, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool L69 7ZB, UK
| | - Gilles Carpentier
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | - Alessandro Ori
- Department of Structural and Chemical Biology, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool L69 7ZB, UK
| | - José Courty
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | - Yamina Hamma-Kourbali
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | - David G Fernig
- Department of Structural and Chemical Biology, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool L69 7ZB, UK
| | - Jean Delbé
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| |
Collapse
|
11
|
Uniewicz KA, Ori A, Ahmed YA, Yates EA, Fernig DG. Characterisation of the interaction of neuropilin-1 with heparin and a heparan sulfate mimetic library of heparin-derived sugars. PeerJ 2014; 2:e461. [PMID: 25024924 PMCID: PMC4089425 DOI: 10.7717/peerj.461] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/09/2014] [Indexed: 12/22/2022] Open
Abstract
Background. Neuropilin-1 (NRP-1) is a multidomain membrane protein with soluble isoforms interacting with a complex network of other membrane receptors, their respective ligands and heparan sulfate (HS). It is involved in the development of vasculature, neural patterning, immunological responses and pathological angiogenesis. Methods. We have characterised the binding of a Fc fusion of rat NRP-1 (Fc rNRP-1) and of a soluble isoform, corresponding to the first four extracellular domains of human NRP-1, shNRP-1, using optical biosensor-based binding assays with a library of heparin derivatives. Selective labelling of lysines protected upon heparin binding allowed their identification by mass spectrometry. Results. Fc rNRP-1 bound to heparin with high affinity (2.5 nM) and fast ka (9.8 × 10(6) M(-1)s(-1)). Unusually, NRP-1 bound both highly sulfated and completely desulfated stretches of heparin and exhibited a complex pattern of preferences for chemically modified heparins possessing one or two sulfate groups, e.g., it bound heparin with just a 6-O sulfate group better than heparin with any two of N-sulfate, 6-O sulfate and 2-O sulfate. Mass-spectrometry based mapping identified that, in addition to the expected the b1 domain, the a1, and c domains and the L2 linker were also involved in the interaction. In contrast, shNRP-1 bound heparin far more weakly. This could only be shown by affinity chromatography and by differential scanning fluorimetry. Discussion. The results suggest that the interaction of NRP-1 with HS is more complex than anticipated and involving a far greater extent of the protein than just the b1-b2 domains. NRP-1's preference for binding long saccharide structures suggests it has the potential to bind large segments of HS chains and so organise their local structure. In contrast, the four domain soluble isoform, shNRP-1 binds heparin weakly and so would be expected to diffuse away rapidly from the source cell.
Collapse
Affiliation(s)
- Katarzyna A Uniewicz
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool , Liverpool , United Kingdom
| | - Alessandro Ori
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool , Liverpool , United Kingdom
| | - Yassir A Ahmed
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool , Liverpool , United Kingdom
| | - Edwin A Yates
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool , Liverpool , United Kingdom
| | - David G Fernig
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool , Liverpool , United Kingdom
| |
Collapse
|
12
|
Nishida M, Kozakai T, Nagami K, Kanamaru Y, Yabe T. Structural alteration of cell surface heparan sulfate through the stimulation of the signaling pathway for heparan sulfate 6-O-sulfotransferase-1 in mouse fibroblast cells. Biosci Biotechnol Biochem 2014; 78:770-9. [PMID: 25035978 DOI: 10.1080/09168451.2014.905178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Heparan sulfate (HS) is a randomly sulfated polysaccharide that is present on the cell surface and in the extracellular matrix. The sulfated structures of HS were synthesized by multiple HS sulfotransferases, thereby regulating various activities such as growth factor signaling, cell differentiation, and tumor metastasis. Therefore, if the sulfated structures of HS could be artificially controlled, those manipulations would help to understand the various functions depending on HS. However, little knowledge is currently available to realize the mechanisms controlling the expression of such enzymes. In this study, we found that the ratio of 6-O-sulfated disaccharides increased at 3 h after adrenaline stimulation in mouse fibroblast cells. Furthermore, adrenaline-induced up-regulation of HS 6-O-sulfotransferase-1 (6-OST-1) was controlled by Src-ERK1/2 signaling pathway. Finally, inhibiting the signaling pathways for 6-OST-1 intentionally suppressed the adrenaline-induced structural alteration of HS. These observations provide fundamental insights into the understanding of structural alterations in HS by extracellular cues.
Collapse
Affiliation(s)
- Mitsutaka Nishida
- a United Graduate School of Agricultural Science, Gifu University , Gifu , Japan
| | | | | | | | | |
Collapse
|
13
|
Favretto ME, Wallbrecher R, Schmidt S, van de Putte R, Brock R. Glycosaminoglycans in the cellular uptake of drug delivery vectors – Bystanders or active players? J Control Release 2014; 180:81-90. [DOI: 10.1016/j.jconrel.2014.02.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 02/07/2014] [Accepted: 02/09/2014] [Indexed: 12/30/2022]
|
14
|
Szatmári T, Dobra K. The role of syndecan-1 in cellular signaling and its effects on heparan sulfate biosynthesis in mesenchymal tumors. Front Oncol 2013; 3:310. [PMID: 24392351 PMCID: PMC3867677 DOI: 10.3389/fonc.2013.00310] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/04/2013] [Indexed: 12/23/2022] Open
Abstract
Proteoglycans (PGs) and in particular the syndecans are involved in the differentiation process across the epithelial-mesenchymal axis, principally through their ability to bind growth factors and modulate their downstream signaling. Malignant tumors have individual proteoglycan profiles, which are closely associated with their differentiation and biological behavior, mesenchymal tumors showing a different profile from that of epithelial tumors. Syndecan-1 is the main syndecan of epithelial malignancies, whereas in sarcomas its expression level is generally low, in accordance with their mesenchymal phenotype and highly malignant behavior. This proteoglycan is often overexpressed in adenocarcinoma cells, whereas mesothelioma and fibrosarcoma cells express syndecan-2 and syndecan-4 more abundantly. Increased expression of syndecan-1 in mesenchymal tumors changes the tumor cell morphology to an epithelioid direction whereas downregulation results in a change in shape from polygonal to spindle-like morphology. Although syndecan-1 plays major roles on the cell-surface, there are also intracellular functions, which are not very well studied. On the functional level, syndecan-1 affects mesenchymal tumor cell proliferation, adhesion, migration and motility, and the effect varies with the different domains of the core protein. Syndecan-1 may exert stimulatory or inhibitory effects, depending on the concentration of various mitogens, enzymes, and signaling molecules, the ratio between the shed and membrane-associated syndecan-1 and histological grade of the tumour. Growth factor signaling seems to be delicately controlled by regulatory loops involving the syndecan expression levels and their sulfation patterns. Overexpression of syndecan-1 modulates the biosynthesis and sulfation of heparan sulfate and it also affects the expression of other PGs. On transcriptomic level, syndecan-1 modulation results in profound effects on genes involved in regulation of cell growth.
Collapse
Affiliation(s)
- Tünde Szatmári
- Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital , Stockholm , Sweden
| | - Katalin Dobra
- Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital , Stockholm , Sweden
| |
Collapse
|
15
|
Abstract
Syndecan-4, a ubiquitous cell surface proteoglycan, mediates numerous cellular processes through signaling pathways that affect cellular proliferation, migration, mechanotransduction and endocytosis. These effects are achieved through syndecan-4 functioning as both a co-receptor for the fibroblast growth factor receptors (FGFR1-FGFR4) and its ability to independently activate signaling pathways upon ligand binding. As an FGFR co-receptor, syndecan-4 strengthens the duration and intensity of downstream signaling upon ligand binding; this is particularly evident with regard to mitogen-activated protein kinase (MAPK) signaling. In contrast, syndecan-4 also functions as an independent receptor for heparin-binding growth factors, such as fibroblast growth factors (FGFs), vascular endothelial growth factors (VEGFs) and platelet-derived growth factors (PDGFs). These signaling cascades affect canonical signaling components, such as the mammalian target of rapamycin (mTOR), AKT1 and the Rho family of GTPases. In combination with the integrin family of proteins, syndecan-4 is also able to form physical connections between the extracellular matrix (ECM) and cytoskeletal signaling proteins, and it has a key role in regulation of integrin turnover. This unique versatility of the interactions of syndecan-4 is characterized in this Cell Science at a Glance article and illustrated in the accompanying poster.
Collapse
Affiliation(s)
- Arye Elfenbein
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06520, USA
| | | |
Collapse
|
16
|
Adage T, Piccinini AM, Falsone A, Trinker M, Robinson J, Gesslbauer B, Kungl AJ. Structure-based design of decoy chemokines as a way to explore the pharmacological potential of glycosaminoglycans. Br J Pharmacol 2013; 167:1195-205. [PMID: 22747966 DOI: 10.1111/j.1476-5381.2012.02089.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Glycosaminoglycans (GAGs) are a class of highly negatively charged, unbranched, O-linked polysaccharides that are involved in many diseases. Their role as a protein-binding matrix on cell surfaces has long been recognized, but therapeutic approaches to interfere with protein-GAG interactions have been limited due to the complex chemistry of GAGs, on one hand, and due to the lack of specific antibodies against GAGs, on the other hand. We have developed a protein engineering platform (the so-called CellJammer(®) technology), which enables us to introduce higher GAG-binding affinity into wild-type GAG-binding proteins and to combine this with impaired biological, receptor-binding function. Chemokines are among the prototypic GAG-binding proteins and here we present selected results of our CellJammer technology applied to several of these proinflammatory proteins. An overview is given of our lead decoy protein, PA401, which is a CXCL8-based mutant protein with increased GAG-binding affinity and decreased CXCR1/2 binding and activation. Major results from our CCL2 and CCL5 programmes are also summarized and the potential for clinical application of these decoy proteins is presented.
Collapse
|
17
|
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: 1.2] [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
|
18
|
Cattaruzza S, Ozerdem U, Denzel M, Ranscht B, Bulian P, Cavallaro U, Zanocco D, Colombatti A, Stallcup WB, Perris R. Multivalent proteoglycan modulation of FGF mitogenic responses in perivascular cells. Angiogenesis 2012; 16:309-27. [PMID: 23124902 DOI: 10.1007/s10456-012-9316-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Accepted: 10/12/2012] [Indexed: 01/10/2023]
Abstract
Sprouting of angiogenic perivascular cells is thought to be highly dependent upon autocrine and paracrine growth factor stimulation. Accordingly, we report that corneal angiogenesis induced by ectopic FGF implantation is strongly impaired in NG2/CSPG4 proteoglycan (PG) null mice known to harbour a putative deficit in pericyte proliferation/mobilization. Conversely, no significant differences were seen between wild type and knockout corneas when VEGF was used as an angiocrine factor. Perturbed responsiveness of NG2-deficient pericytes to paracrine and autocrine stimulation by several FGFs could be confirmed in cells isolated from NG2 null mice, while proliferation induced by other growth factors was equivalent in wild type and knockout cells. Identical results were obtained after siRNA-mediated knock-down of NG2 in human smooth muscle-like cell lines, as also demonstrated by the decreased levels of FGF receptor phosphorylation detected in these NG2 deprived cells. Binding assays with recombinant proteins and molecular interactions examined on live cells asserted that FGF-2 bound to NG2 in a glycosaminoglycan-independent, core protein-mediated manner and that the PG was alone capable of retaining FGF-2 on the cell membrane for subsequent receptor presentation. The use of dominant-negative mutant cells, engineered by combined transduction of NG2 deletion constructs and siRNA knock-down of the endogenous PG, allowed us to establish that the FGF co-receptor activity of NG2 is entirely mediated by its extracellular portion. In fact, forced overexpression of the NG2 ectodomain in human smooth muscle-like cells increased their FGF-2-induced mitosis and compensated for low levels of FGF receptor surface expression, in a manner equivalent to that produced by overexpression of the full-length NG2. Upon FGF binding, the cytoplasmic domain of NG2 is phosphorylated, but there is no evidence that this event elicits signal transductions that could bypass the FGFR-mediated ones. Pull-down experiments, protein-protein binding assays and flow cytometry FRET coherently revealed an elective ligand-independent association of NG2 with FGFR1 and FGFR3. The NG2 cooperation with these receptors was also corroborated functionally by the outcome of FGF-2 treatments of cells engineered to express diverse NG2/FGFR combinations. Comprehensively, the findings suggest that perivascular NG2 may serve as a dual modulator of the availability/accessibility of FGF at the cell membrane, as well as the resulting FGFR transducing activity.
Collapse
Affiliation(s)
- Sabrina Cattaruzza
- S.O.C. for Experimental Oncology 2, The National Cancer Institute Aviano, CRO-IRCCS, Via Pedemontana Occidentale 12, 33081, Aviano, PN, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Xu R, Ori A, Rudd TR, Uniewicz KA, Ahmed YA, Guimond SE, Skidmore MA, Siligardi G, Yates EA, Fernig DG. Diversification of the structural determinants of fibroblast growth factor-heparin interactions: implications for binding specificity. J Biol Chem 2012; 287:40061-73. [PMID: 23019343 DOI: 10.1074/jbc.m112.398826] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The functions of a large number (>435) of extracellular regulatory proteins are controlled by their interactions with heparan sulfate (HS). In the case of fibroblast growth factors (FGFs), HS binding determines their transport between cells and is required for the assembly of high affinity signaling complexes with their cognate FGF receptor. However, the specificity of the interaction of FGFs with HS is still debated. Here, we use a panel of FGFs (FGF-1, FGF-2, FGF-7, FGF-9, FGF-18, and FGF-21) spanning five FGF subfamilies to probe their specificities for HS at different levels as follows: binding parameters, identification of heparin-binding sites (HBSs) in the FGFs, changes in their secondary structure caused by heparin binding and structures in the sugar required for binding. For interaction with heparin, the FGFs exhibit K(D) values varying between 38 nM (FGF-18) and 620 nM (FGF-9) and association rate constants spanning over 20-fold (FGF-1, 2,900,000 M(-1) s(-1) and FGF-9, 130,000 M(-1) s(-1)). The canonical HBS in FGF-1, FGF-2, FGF-7, FGF-9, and FGF-18 differs in its size, and these FGFs have a different complement of secondary HBS, ranging from none (FGF-9) to two (FGF-1). Differential scanning fluorimetry identified clear preferences in these FGFs for distinct structural features in the polysaccharide. These data suggest that the differences in heparin-binding sites in both the protein and the sugar are greatest between subfamilies and may be more restricted within a FGF subfamily in accord with the known conservation of function within FGF subfamilies.
Collapse
Affiliation(s)
- Ruoyan Xu
- Institute of Integrative Biology, Department of Chemical and Structural Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Duchesne L, Octeau V, Bearon RN, Beckett A, Prior IA, Lounis B, Fernig DG. Transport of fibroblast growth factor 2 in the pericellular matrix is controlled by the spatial distribution of its binding sites in heparan sulfate. PLoS Biol 2012; 10:e1001361. [PMID: 22815649 PMCID: PMC3398970 DOI: 10.1371/journal.pbio.1001361] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 06/06/2012] [Indexed: 12/31/2022] Open
Abstract
The heparan sulfate (HS) chains of proteoglycans are a key regulatory component of the extracellular matrices of animal cells, including the pericellular matrix around the plasma membrane. In these matrices they regulate transport, gradient formation, and effector functions of over 400 proteins central to cell communication. HS from different matrices differs in its selectivity for its protein partners. However, there has been no direct test of how HS in the matrix regulates the transport of its partner proteins. We address this issue by single molecule imaging and tracking in fibroblast pericellular matrix of fibroblast growth factor 2 (FGF2), stoichiometrically labelled with small gold nanoparticles. Transmission electron microscopy and photothermal heterodyne imaging (PHI) show that the spatial distribution of the HS-binding sites for FGF2 in the pericellular matrix is heterogeneous over length scales ranging from 22 nm to several µm. Tracking of individual FGF2 by PHI in the pericellular matrix of living cells demonstrates that they undergo five distinct types of motion. They spend much of their time in confined motion (∼110 nm diameter), but they are not trapped and can escape by simple diffusion, which may be slow, fast, or directed. These substantial translocations (µm) cover distances far greater than the length of a single HS chain. Similar molecular motion persists in fixed cells, where the movement of membrane PGs is impeded. We conclude that FGF2 moves within the pericellular matrix by translocating from one HS-binding site to another. The binding sites on HS chains form non-random, heterogeneous networks. These promote FGF2 confinement or substantial translocation depending on their spatial organisation. We propose that this spatial organisation, coupled to the relative selectivity and the availability of HS-binding sites, determines the transport of FGF2 in matrices. Similar mechanisms are likely to underpin the movement of many other HS-binding effectors.
Collapse
Affiliation(s)
- Laurence Duchesne
- Department of Structural and Chemical Biology, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Institut du Fer à Moulin, UMR-S 839 INSERM, University Pierre and Marie Curie, Paris, France
- UMR 6290 CNRS, Institut de Génétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France
- * E-mail: (LD); (DGF)
| | - Vivien Octeau
- Laboratoire Photonique Numérique et Nanosciences, Université de Bordeaux, UMR 5298 CNRS and Institut d'Optique Graduate School, Talence, France
| | - Rachel N. Bearon
- Department of Mathematical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Alison Beckett
- Physiological Laboratory, University of Liverpool, Liverpool, United Kingdom
| | - Ian A. Prior
- Physiological Laboratory, University of Liverpool, Liverpool, United Kingdom
| | - Brahim Lounis
- Laboratoire Photonique Numérique et Nanosciences, Université de Bordeaux, UMR 5298 CNRS and Institut d'Optique Graduate School, Talence, France
| | - David G. Fernig
- Department of Structural and Chemical Biology, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- * E-mail: (LD); (DGF)
| |
Collapse
|
21
|
Thompson SM, Connell MG, van Kuppevelt TH, Xu R, Turnbull JE, Losty PD, Fernig DG, Jesudason EC. Structure and epitope distribution of heparan sulfate is disrupted in experimental lung hypoplasia: a glycobiological epigenetic cause for malformation? BMC DEVELOPMENTAL BIOLOGY 2011; 11:38. [PMID: 21672206 PMCID: PMC3127989 DOI: 10.1186/1471-213x-11-38] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Accepted: 06/14/2011] [Indexed: 12/14/2022]
Abstract
BACKGROUND Heparan sulfate (HS) is present on the surface of virtually all mammalian cells and is a major component of the extracellular matrix (ECM), where it plays a pivotal role in cell-cell and cell-matrix cross-talk through its large interactome. Disruption of HS biosynthesis in mice results in neonatal death as a consequence of malformed lungs, indicating that HS is crucial for airway morphogenesis. Neonatal mortality (~50%) in newborns with congenital diaphragmatic hernia (CDH) is principally associated with lung hypoplasia and pulmonary hypertension. Given the importance of HS for lung morphogenesis, we investigated developmental changes in HS structure in normal and hypoplastic lungs using the nitrofen rat model of CDH and semi-synthetic bacteriophage ('phage) display antibodies, which identify distinct HS structures. RESULTS The pulmonary pattern of elaborated HS structures is developmentally regulated. For example, the HS4E4V epitope is highly expressed in sub-epithelial mesenchyme of E15.5 - E17.5 lungs and at a lower level in more distal mesenchyme. However, by E19.5, this epitope is expressed similarly throughout the lung mesenchyme.We also reveal abnormalities in HS fine structure and spatiotemporal distribution of HS epitopes in hypoplastic CDH lungs. These changes involve structures recognised by key growth factors, FGF2 and FGF9. For example, the EV3C3V epitope, which was abnormally distributed in the mesenchyme of hypoplastic lungs, is recognised by FGF2. CONCLUSIONS The observed spatiotemporal changes in HS structure during normal lung development will likely reflect altered activities of many HS-binding proteins regulating lung morphogenesis. Abnormalities in HS structure and distribution in hypoplastic lungs can be expected to perturb HS:protein interactions, ECM microenvironments and crucial epithelial-mesenchyme communication, which may contribute to lung dysmorphogenesis. Indeed, a number of epitopes correlate with structures recognised by FGFs, suggesting a functional consequence of the observed changes in HS in these lungs. These results identify a novel, significant molecular defect in hypoplastic lungs and reveals HS as a potential contributor to hypoplastic lung development in CDH. Finally, these results afford the prospect that HS-mimetic therapeutics could repair defective signalling in hypoplastic lungs, improve lung growth, and reduce CDH mortality.
Collapse
Affiliation(s)
- Sophie M Thompson
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, UK
- Division of Child Health, School of Reproductive and Developmental Medicine, Royal Liverpool Children's Hospital, Alder Hey, Liverpool, UK
| | - Marilyn G Connell
- Division of Child Health, School of Reproductive and Developmental Medicine, Royal Liverpool Children's Hospital, Alder Hey, Liverpool, UK
| | - Toin H van Kuppevelt
- Department of Biochemistry 280, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Ruoyan Xu
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, UK
| | - Jeremy E Turnbull
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, UK
| | - Paul D Losty
- Division of Child Health, School of Reproductive and Developmental Medicine, Royal Liverpool Children's Hospital, Alder Hey, Liverpool, UK
| | - David G Fernig
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, UK
| | - Edwin C Jesudason
- Division of Child Health, School of Reproductive and Developmental Medicine, Royal Liverpool Children's Hospital, Alder Hey, Liverpool, UK
| |
Collapse
|
22
|
Interaction of ATP with fibroblast growth factor 2: biochemical characterization and consequence for growth factor stability. BMC BIOCHEMISTRY 2011; 12:14. [PMID: 21447157 PMCID: PMC3072325 DOI: 10.1186/1471-2091-12-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 03/29/2011] [Indexed: 11/30/2022]
Abstract
Background Fibroblast growth factor 2, a well-characterized heparin-binding growth factor, is involved in many biological processes like embryogenesis, cell proliferation and angiogenesis. However, this growth factor is very unstable and shows rapid degradation in aqueous solution. Beside the well-known stabilization of FGF2 by heparin or heparan sulphate, the recently discovered binding to ATP also shows a stabilizing and protective effect on this growth factor. Results Here we determined the dissociation constant of ATP on FGF2 by equilibrium microdialysis (KD: 59.8 μM) and analyzed the impact of this binding on secondary structure by CD-spectroscopy. ATP-binding to FGF2 significantly changed the secondary structure of this growth factor with a shift to random coil structure elements. We also analyzed the influence of this binding on the stability of FGF2 in aqueous solution over a period of 2 h. While the amount of untreated FGF2 is reduced drastically over this period of time, ATP-binding reduces the degradation considerably. Conclusions Taken together, our data suggest an important role of ATP in FGF2-stabilization beside the well known-role of heparin and heparan sulphate.
Collapse
|
23
|
Becher J, Möller S, Weiss D, Schiller J, Schnabelrauch M. Synthesis of New Regioselectively Sulfated Hyaluronans for Biomedical Application. ACTA ACUST UNITED AC 2010. [DOI: 10.1002/masy.201051060] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
24
|
Thompson SM, Jesudason EC, Turnbull JE, Fernig DG. Heparan sulfate in lung morphogenesis: The elephant in the room. ACTA ACUST UNITED AC 2010; 90:32-44. [PMID: 20301217 DOI: 10.1002/bdrc.20169] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Heparan sulfate (HS) is a structurally complex polysaccharide located on the cell surface and in the extracellular matrix, where it participates in numerous biological processes through interactions with a vast number of regulatory proteins such as growth factors and morphogens. HS is crucial for lung development; disruption of HS synthesis in flies and mice results in a major aberration of airway branching, and in mice, it results in neonatal death as a consequence of malformed lungs and respiratory distress. Epithelial-mesenchymal interactions governing lung morphogenesis are directed by various diffusible proteins, many of which bind to, and are regulated by HS, including fibroblast growth factors, sonic hedgehog, and bone morphogenetic proteins. The majority of research into the molecular mechanisms underlying defective lung morphogenesis and pulmonary pathologies, such as bronchopulmonary dysplasia and pulmonary hypoplasia associated with congenital diaphragmatic hernia (CDH), has focused on abnormal protein expression. The potential contribution of HS to abnormalities of lung development has yet to be explored to any significant extent, which is somewhat surprising given the abnormal lung phenotype exhibited by mutant mice synthesizing abnormal HS. This review summarizes our current understanding of the role of HS and HS-binding proteins in lung morphogenesis and will present in vitro and in vivo evidence for the fundamental importance of HS in airway development. Finally, we will discuss the future possibility of HS-based therapeutics for ameliorating insufficient lung growth associated with lung diseases such as CDH.
Collapse
Affiliation(s)
- Sophie M Thompson
- School of Biological Sciences, University of Liverpool, Liverpool L69 7ZB, United Kingdom.
| | | | | | | |
Collapse
|
25
|
Zhu H, Duchesne L, Rudland PS, Fernig DG. The heparan sulfate co-receptor and the concentration of fibroblast growth factor-2 independently elicit different signalling patterns from the fibroblast growth factor receptor. Cell Commun Signal 2010; 8:14. [PMID: 20576134 PMCID: PMC2912315 DOI: 10.1186/1478-811x-8-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 06/24/2010] [Indexed: 01/12/2023] Open
Abstract
Background The fibroblast growth factor receptor (FGFR) interprets concentration gradients of FGF ligands and structural changes in the heparan sulfate (HS) co-receptor to generate different cellular responses. However, whether the FGFR generates different signals is not known. Results We have previously shown in rat mammary fibroblasts that in cells deficient in sulfation, and so in HS co-receptor, FGF-2 can only stimulate a transient phosphorylation of p42/44 MAPK and so cannot stimulate DNA synthesis. Here we demonstrate that this is because in the absence of HS, FGF-2 fails to stimulate the phosphorylation of the adaptor FGFR substrate 2 (FRS2). In cells possessing the HS co-receptor, FGF-2 elicits a bell-shaped dose response: optimal concentrations stimulate DNA synthesis, but supramaximal concentrations (≥ 100 ng/mL) have little effect. At optimal concentrations (300 pg/mL) FGF-2 stimulates a sustained dual phosphorylation of p42/44 MAPK and tyrosine phosphorylation of FRS2. In contrast, 100 ng/mL FGF-2 only stimulates a transient early peak of p42/44 MAPK phosphorylation and fails to stimulate appreciably the phosphorylation of FRS2 on tyrosine. Conclusions These results suggest that the nature of the FGFR signal produced is determined by a combination of the HS co-receptor and the concentration of FGF ligand. Both the phosphorylation of the adaptor FRS2, the kinetics (sustained or transient) of phosphorylation of p42/44(MAPK) are varied, and so differing cellular responses are produced.
Collapse
Affiliation(s)
- Hongyan Zhu
- School of Biological Sciences, Biosciences Building, Crown Street, University of Liverpool, Liverpool, L69 7ZB, UK.
| | | | | | | |
Collapse
|
26
|
Tong M, Tuk B, Hekking IM, Vermeij M, Barritault D, van Neck JW. Stimulated neovascularization, inflammation resolution and collagen maturation in healing rat cutaneous wounds by a heparan sulfate glycosaminoglycan mimetic, OTR4120. Wound Repair Regen 2010; 17:840-52. [PMID: 19903305 DOI: 10.1111/j.1524-475x.2009.00548.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Heparan sulfate glycosaminoglycans (HS-GAGs) are not only the structural elements of tissue architecture but also regulate the bioavailability and transduction pathways of heparan sulfate-bound polypeptides released by cells or the extracellular matrix. Heparan sulfate-bound polypeptides include inflammatory mediators, chemokines, angiogenic factors, morphogens, and growth-promoting factors that induce cell migration, proliferation, and differentiation in wound healing. OTR4120, a polymer engineered to mimic the properties of HS-GAGs, is used to replace the natural HS-GAGs that are degraded during wound repair, and enhance the tissue regeneration by preserving the cellular microenvironment and the endogenous signals needed for tissue regeneration. We previously demonstrated that OTR4120 treatment had a long-term effect on increasing breaking strength and vasodilation in healing rat full-thickness excisional wounds. The present study investigates the underlying mechanisms of the effects of OTR4120 treatment in improving the quality of cutaneous wound repair. We found that OTR4120 treatment stimulated inflammation resolution and increased neovascularization. OTR4120 treatment also promoted epidermal migration and proliferation during reepithelialization. Moreover, the granulation tissue formation and collagen maturation were improved in OTR4120-treated wounds. Three months after wounding, the effects of OTR4120 treatment on vascularization and inflammation resolution were normalized, except for an improved neodermis. We conclude that OTR4120 is a potential matrix therapeutic agent that ensures the quality of normal cutaneous wound repair and may restore impaired wound healing characterized by deficient angiogenesis and prolonged inflammation.
Collapse
Affiliation(s)
- Miao Tong
- Department of Plastic and Reconstructive Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
27
|
Thompson SM, Fernig DG, Jesudason EC, Losty PD, van de Westerlo EMA, van Kuppevelt TH, Turnbull JE. Heparan sulfate phage display antibodies identify distinct epitopes with complex binding characteristics: insights into protein binding specificities. J Biol Chem 2009; 284:35621-31. [PMID: 19837661 PMCID: PMC2790993 DOI: 10.1074/jbc.m109.009712] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 10/02/2009] [Indexed: 01/03/2023] Open
Abstract
Heparan sulfate (HS) binds and modulates the transport and activity of a large repertoire of regulatory proteins. The HS phage display antibodies are powerful tools for the analysis of native HS structure in situ; however, their epitopes are not well defined. Analysis of the binding specificities of a set of HS antibodies by competitive binding assays with well defined chemically modified heparins demonstrates that O-sulfates are essential for binding; however, increasing sulfation does not necessarily correlate with increased antibody reactivity. IC50 values for competition with double modified heparins were not predictable from IC50 values with corresponding singly modified heparins. Binding assays and immunohistochemistry revealed that individual antibodies recognize distinct epitopes and that these are not single linear sequences but families of structurally similar motifs in which subtle variations in sulfation and conformation modify the affinity of interaction. Modeling of the antibodies demonstrates that they possess highly basic CDR3 and surrounding surfaces, presenting a number of possible orientations for HS binding. Unexpectedly, there are significant differences between the existence of epitopes in tissue sections and observed in vitro in dot blotted tissue extracts, demonstrating that in vitro specificity does not necessarily correlate with specificity in situ/vivo. The epitopes are therefore more complex than previously considered. Overall, these data have significance for structure-activity relationships of HS, because the model of one antibody recognizing multiple HS structures and the influence of other in situ HS-binding proteins on epitope availability are likely to reflect the selectivity of many HS-protein interactions in vivo.
Collapse
Affiliation(s)
- Sophie M. Thompson
- From the School of Biological Sciences, University of Liverpool, Liverpool L69 7ZB, United Kingdom
| | - David G. Fernig
- From the School of Biological Sciences, University of Liverpool, Liverpool L69 7ZB, United Kingdom
| | - Edwin C. Jesudason
- the Division of Child Health, School of Reproductive and Developmental Medicine, Royal Liverpool Children's Hospital, Alder Hey, Liverpool L12 2AP, United Kingdom, and
| | - Paul D. Losty
- the Division of Child Health, School of Reproductive and Developmental Medicine, Royal Liverpool Children's Hospital, Alder Hey, Liverpool L12 2AP, United Kingdom, and
| | - Els M. A. van de Westerlo
- the Department of Matrix Biochemistry, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Toin H. van Kuppevelt
- the Department of Matrix Biochemistry, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Jeremy E. Turnbull
- From the School of Biological Sciences, University of Liverpool, Liverpool L69 7ZB, United Kingdom
| |
Collapse
|
28
|
Ismail TM, Zhang S, Fernig DG, Gross S, Martin-Fernandez ML, See V, Tozawa K, Tynan CJ, Wang G, Wilkinson MC, Rudland PS, Barraclough R. Self-association of calcium-binding protein S100A4 and metastasis. J Biol Chem 2009; 285:914-22. [PMID: 19917604 DOI: 10.1074/jbc.m109.010892] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Elevated levels of the calcium-binding protein S100A4 promote metastasis and in carcinoma cells are associated with reduced survival of cancer patients. S100A4 interacts with target proteins that affect a number of activities associated with metastatic cells. However, it is not known how many of these interactions are required for S100A4-promoted metastasis, thus hampering the design of specific inhibitors of S100A4-induced metastasis. Intracellular S100A4 exists as a homodimer through previously identified, well conserved, predominantly hydrophobic key contacts between the subunits. Here it is shown that mutating just one key residue, phenylalanine 72, to alanine is sufficient to reduce the metastasis-promoting activity of S100A4 to 50% that of the wild type protein, and just 2 or 3 specific mutations reduces the metastasis-promoting activity of S100A4 to less than 20% that of the wild type protein. These mutations inhibit the self-association of S100A4 in vivo and reduce markedly the affinity of S100A4 for at least two of its protein targets, a recombinant fragment of non-muscle myosin heavy chain isoform A, and p53. Inhibition of the self-association of S100 proteins might be a novel means of inhibiting their metastasis-promoting activities.
Collapse
Affiliation(s)
- Thamir M Ismail
- School of Biological Sciences, University of Liverpool, Liverpool L69 7ZB, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Hintze V, Moeller S, Schnabelrauch M, Bierbaum S, Viola M, Worch H, Scharnweber D. Modifications of Hyaluronan Influence the Interaction with Human Bone Morphogenetic Protein-4 (hBMP-4). Biomacromolecules 2009; 10:3290-7. [DOI: 10.1021/bm9008827] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Vera Hintze
- Institute of Material Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, 01069 Dresden, Germany, Biomaterials Department, INNOVENT e.V., 07745 Jena, Germany, and Department of Experimental and Clinical Biomedical Sciences, Università dell’Insubria, 21100 Varese, Italy
| | - Stephanie Moeller
- Institute of Material Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, 01069 Dresden, Germany, Biomaterials Department, INNOVENT e.V., 07745 Jena, Germany, and Department of Experimental and Clinical Biomedical Sciences, Università dell’Insubria, 21100 Varese, Italy
| | - Matthias Schnabelrauch
- Institute of Material Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, 01069 Dresden, Germany, Biomaterials Department, INNOVENT e.V., 07745 Jena, Germany, and Department of Experimental and Clinical Biomedical Sciences, Università dell’Insubria, 21100 Varese, Italy
| | - Susanne Bierbaum
- Institute of Material Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, 01069 Dresden, Germany, Biomaterials Department, INNOVENT e.V., 07745 Jena, Germany, and Department of Experimental and Clinical Biomedical Sciences, Università dell’Insubria, 21100 Varese, Italy
| | - Manuela Viola
- Institute of Material Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, 01069 Dresden, Germany, Biomaterials Department, INNOVENT e.V., 07745 Jena, Germany, and Department of Experimental and Clinical Biomedical Sciences, Università dell’Insubria, 21100 Varese, Italy
| | - Hartmut Worch
- Institute of Material Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, 01069 Dresden, Germany, Biomaterials Department, INNOVENT e.V., 07745 Jena, Germany, and Department of Experimental and Clinical Biomedical Sciences, Università dell’Insubria, 21100 Varese, Italy
| | - Dieter Scharnweber
- Institute of Material Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, 01069 Dresden, Germany, Biomaterials Department, INNOVENT e.V., 07745 Jena, Germany, and Department of Experimental and Clinical Biomedical Sciences, Università dell’Insubria, 21100 Varese, Italy
| |
Collapse
|
30
|
Zong F, Fthenou E, Wolmer N, Hollósi P, Kovalszky I, Szilák L, Mogler C, Nilsonne G, Tzanakakis G, Dobra K. Syndecan-1 and FGF-2, but not FGF receptor-1, share a common transport route and co-localize with heparanase in the nuclei of mesenchymal tumor cells. PLoS One 2009; 4:e7346. [PMID: 19802384 PMCID: PMC2750749 DOI: 10.1371/journal.pone.0007346] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Accepted: 09/07/2009] [Indexed: 11/18/2022] Open
Abstract
Syndecan-1 forms complexes with growth factors and their cognate receptors in the cell membrane. We have previously reported a tubulin-mediated translocation of syndecan-1 to the nucleus. The transport route and functional significance of nuclear syndecan-1 is still incompletely understood. Here we investigate the sub-cellular distribution of syndecan-1, FGF-2, FGFR-1 and heparanase in malignant mesenchymal tumor cells, and explore the possibility of their coordinated translocation to the nucleus. To elucidate a structural requirement for this nuclear transport, we have transfected cells with a syndecan-1/EGFP construct or with a short truncated version containing only the tubulin binding RMKKK sequence. The sub-cellular distribution of the EGFP fusion proteins was monitored by fluorescence microscopy. Our data indicate that syndecan-1, FGF-2 and heparanase co-localize in the nucleus, whereas FGFR-1 is enriched mainly in the perinuclear area. Overexpression of syndecan-1 results in increased nuclear accumulation of FGF-2, demonstrating the functional importance of syndecan-1 for this nuclear transport. Interestingly, exogenously added FGF-2 does not follow the route taken by endogenous FGF-2. Furthermore, we prove that the RMKKK sequence of syndecan-1 is necessary and sufficient for nuclear translocation, acting as a nuclear localization signal, and the Arginine residue is vital for this localization. We conclude that syndecan-1 and FGF-2, but not FGFR-1 share a common transport route and co-localize with heparanase in the nucleus, and this transport is mediated by the RMKKK motif in syndecan-1. Our study opens a new perspective in the proteoglycan field and provides more evidence of nuclear interactions of syndecan-1.
Collapse
Affiliation(s)
- Fang Zong
- Department of Laboratory Medicine, Division of Pathology, Huddinge University Hospital, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Eleni Fthenou
- Department of Histology, Division of Morphology, School of Medicine, University of Crete, Heraklion, Greece
| | - Nina Wolmer
- Department of Laboratory Medicine, Division of Pathology, Huddinge University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Péter Hollósi
- 1st Institute of Pathology and Experimental Cancer Research, Semmelweis University Budapest, Hungary
| | - Ilona Kovalszky
- 1st Institute of Pathology and Experimental Cancer Research, Semmelweis University Budapest, Hungary
| | - László Szilák
- 1st Institute of Pathology and Experimental Cancer Research, Semmelweis University Budapest, Hungary
| | - Carolin Mogler
- Department of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Gustav Nilsonne
- Department of Laboratory Medicine, Division of Pathology, Huddinge University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Georgios Tzanakakis
- Department of Histology, Division of Morphology, School of Medicine, University of Crete, Heraklion, Greece
| | - Katalin Dobra
- Department of Laboratory Medicine, Division of Pathology, Huddinge University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Polanska UM, Fernig DG, Kinnunen T. Extracellular interactome of the FGF receptor-ligand system: complexities and the relative simplicity of the worm. Dev Dyn 2009; 238:277-93. [PMID: 18985724 DOI: 10.1002/dvdy.21757] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) regulate a multitude of biological functions in embryonic development and in adult. A major question is how does one family of growth factors and their receptors control such a variety of functions? Classically, specificity was thought to be imparted by alternative splicing of the FGFRs, resulting in isoforms that bind specifically to a subset of the FGFs, and by different saccharide sequences in the heparan sulfate proteoglycan (HSPG) co-receptor. A growing number of noncanonical co-receptors such as integrins and neural cell adhesion molecule (NCAM) are now recognized as imparting additional complexity to classic FGFR signaling. This review will discuss the noncanonical FGFR ligands and speculate on the possibility that they provide additional and alternative means to determining the functional specificity of FGFR signaling. We will also discuss how invertebrate models such as C. elegans may advance our understanding of noncanonical FGFR signaling.
Collapse
Affiliation(s)
- Urszula M Polanska
- School of Biological Sciences, University of Liverpool, Liverpool, United Kingdom
| | | | | |
Collapse
|
32
|
Kumarasuriyar A, Murali S, Nurcombe V, Cool SM. Glycosaminoglycan composition changes with MG-63 osteosarcoma osteogenesis in vitro and induces human mesenchymal stem cell aggregation. J Cell Physiol 2009; 218:501-11. [PMID: 18988189 DOI: 10.1002/jcp.21620] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Osteogenic differentiation is coordinated by the exposure of cells to temporal changes in a combination of growth factors and elements within the extracellular matrix (ECM). Many of the key proteins that drive these changes share the property of being dependent on ECM glycosaminoglycans (GAGs) for their activity. Here, we examined whether GAGs isolated from proliferating, differentiating and mineralizing MG-63 osteosarcoma cells differed in their physical properties, and thus in their capacities to coordinate the osteogenic cascade both in human MG-63 osteosarcoma cells and primary human mesenchymal stem cells (hMSCs). Our results show that the size distribution of GAGs, the expression of GAG-carrying proteoglycan cores and the expression of enzymes involved in their modification systematically change as MG-63 cells mature in culture. When dosed back onto cells exogenously in soluble form, GAGs regulated MG-63 survival and growth in a dose-dependent manner, but not differentiation in either cell type. In contrast, hMSCs aggregated into distinct colonies when grown on GAG-coated substrates, while MG-63 cells did not. Heparin-coated substrates improved hMSC viability without inducing aggregation. These results suggest a complex role for GAGs in coordinating the emergence of the osteoblast phenotype, and provide further evidence for the use of heparans in bone tissue repair applications.
Collapse
Affiliation(s)
- A Kumarasuriyar
- Institute of Molecular and Cell Biology, Proteos, Singapore, Singapore
| | | | | | | |
Collapse
|
33
|
Ismail TM, Fernig DG, Rudland PS, Terry CJ, Wang G, Barraclough R. The basic C-terminal amino acids of calcium-binding protein S100A4 promote metastasis. Carcinogenesis 2008; 29:2259-66. [PMID: 18784356 DOI: 10.1093/carcin/bgn217] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The calcium-binding protein S100A4 can induce a metastatic phenotype in animal model systems and its expression in various human cancers has been shown to be associated with metastasis and reduced patient survival. Using a series of nested deletion mutants, it is now shown that the two C-terminal lysine residues are required for the enhanced metastasis, invasion and migration abilities that S100A4 confers on cells in a model system of metastasis. Basic C-terminal residues enhance the affinity between S100A4 and its best characterized target, a recombinant C-terminal fragment of non-muscle myosin II heavy chain isoform A (NMMHC-IIA). In wild-type S100A4 protein, the presence of the C-terminal lysine, residue 101, enhances the rate of association between S100A4 and NMMHC-IIA. These results identify the amino acids of S100A4 that are involved in metastasis induction and show that the C-terminal region of S100A4 is a possible target for inhibitors of its metastatic action.
Collapse
Affiliation(s)
- Thamir M Ismail
- Cancer and Polio Research Fund Laboratories, Biosciences Building, University of Liverpool, Liverpool, UK
| | | | | | | | | | | |
Collapse
|
34
|
Mapili G, Lu Y, Chen S, Roy K. Laser-layered microfabrication of spatially patterned functionalized tissue-engineering scaffolds. J Biomed Mater Res B Appl Biomater 2008; 75:414-24. [PMID: 16025464 DOI: 10.1002/jbm.b.30325] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Understanding cell behavior inside complex, three-dimensional (3D) microenvironments with controlled spatiotemporal patterning of physical and biochemical factors would provide significant insights into the basic biology of organ development and tissue functions. One of the fundamental limitations in studying such behavior has been the inability to create patterned microenvironments within 3D scaffold structures. Here a simple, layer-by-layer stereolithography (SL) method that can precisely pattern ligands, extracellular-matrix (ECM) components, and growth factors, as well as controlled release particles inside a single scaffold, has been developed. The process also allows fabrication of predesigned internal architectures and porosities. Photocrosslinkable poly(ethylene glycol) dimethacrylate (PEGDMA) was used as the basic structural component of these microfabricated scaffolds. PEG acrylates, covalently modified with the cell adhesive peptide arginine-glycine-aspartic acid (RGD) or the ECM component heparan sulfate, was incorporated within the scaffolds to facilitate cell attachment and to allow spatial sequestration of heparan-binding growth factors. Fluorescently labeled polymer microparticles and basic fibroblast growth factor (FGF-2) were chosen to illustrate the capability of SL to spatiotemporally pattern scaffolds. The results demonstrate that a precise, predesigned distribution of single or multiple factors within a single 3D structure can be created, and specific internal architectures can be fabricated. Functionalization of these scaffolds with RGD is demonstrated, and heparan sulfate allows efficient cell attachment and spatial localization of growth factors. Such patterned scaffolds might provide effective systems to study cell behavior in complex microenvironments and could eventually lead to engineering of complex, hybrid tissue structures through predesigned, multilineage differentiation of a single stem-cell population.
Collapse
Affiliation(s)
- Gazell Mapili
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78712, USA
| | | | | | | |
Collapse
|
35
|
Gemma E, Meyer O, Uhrín D, Hulme AN. Enabling methodology for the end functionalization of glycosaminoglycan oligosaccharides. MOLECULAR BIOSYSTEMS 2008; 4:481-95. [PMID: 18493641 DOI: 10.1039/b801666f] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2025]
Abstract
The chemical functionalization of glycosaminoglycans is very challenging due to their structural heterogeneity and polyanionic character; but as an enabling technology it promises rich rewards in terms of the structural and biological data it will afford. This review surveys the known methods for the preparation of glycosaminoglycan oligosaccharides and conditions for the selective functionalization of both the reducing and non-reducing ends. The synthetic merits of each approach are discussed, together with the structural modification of the glycosaminoglycan oligosaccharide which they confer. Recent applications of this methodology are highlighted, including introduction of functional labels for gel mobility shift assays and NMR studies of glycosaminoglycan-protein complexes, and synthesis of immobilised glycosaminoglycan arrays.
Collapse
Affiliation(s)
- Emiliano Gemma
- School of Chemistry, The University of Edinburgh, Kings Buildings, West Mains Road, Edinburgh, UK
| | | | | | | |
Collapse
|
36
|
Bermek O, Diamantopoulou Z, Polykratis A, Dos Santos C, Hamma-Kourbali Y, Burlina F, Delbé J, Chassaing G, Fernig DG, Katsoris P, Courty J. A basic peptide derived from the HARP C-terminus inhibits anchorage-independent growth of DU145 prostate cancer cells. Exp Cell Res 2007; 313:4041-50. [PMID: 17727841 DOI: 10.1016/j.yexcr.2007.07.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Revised: 07/31/2007] [Accepted: 07/31/2007] [Indexed: 12/27/2022]
Abstract
Heparin affin regulatory peptide (HARP) is an 18 kDa heparin-binding protein that plays a key role in tumor growth. We showed previously that the synthetic peptide P(111-136) composed of the last 26 HARP amino acids inhibited HARP-induced mitogenesis. Here, to identify the exact molecular domain involved in HARP inhibition, we investigated the effect of the shorter basic peptide P(122-131) on DU145 cells, which express HARP and its receptor protein tyrosine phosphatase beta/zeta (RPTPbeta/zeta). P(122-131) was not cytotoxic; it dose-dependently inhibited anchorage-independent growth of DU145 cells. Binding studies using biotinylated P(122-131) indicated that this peptide interfered with HARP binding to DU145 cells. Investigation of the mechanisms involved suggested interference, under anchorage-independent conditions, of P(122-131) with a HARP autocrine loop in an RPTPbeta/zeta-dependent fashion. Thus, P(122-131) may hold potential for the treatment of disorders involving RPTPbeta/zeta.
Collapse
Affiliation(s)
- Oya Bermek
- Laboratoire de recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires (CRRET), CNRS UMR 7149, Université Paris 12, 61 Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Vanpouille C, Deligny A, Delehedde M, Denys A, Melchior A, Liénard X, Lyon M, Mazurier J, Fernig DG, Allain F. The heparin/heparan sulfate sequence that interacts with cyclophilin B contains a 3-O-sulfated N-unsubstituted glucosamine residue. J Biol Chem 2007; 282:24416-29. [PMID: 17588944 DOI: 10.1074/jbc.m701835200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Many of the biological functions of heparan sulfate (HS) proteoglycans can be attributed to specialized structures within HS moieties, which are thought to modulate binding and function of various effector proteins. Cyclophilin B (CyPB), which was initially identified as a cyclosporin A-binding protein, triggers migration and integrin-mediated adhesion of peripheral blood T lymphocytes by a mechanism dependent on interaction with cell surface HS. Here we determined the structural features of HS that are responsible for the specific binding of CyPB. In addition to the involvement of 2-O,6-O, and N-sulfate groups, we also demonstrated that binding of CyPB was dependent on the presence of N-unsubstituted glucosamine residues (GlcNH2), which have been reported to be precursors for sulfation by 3-O-sulfotransferases-3 (3-OST-3). Interestingly, 3-OST-3B isoform was found to be the main 3-OST isoenzyme expressed in peripheral blood T lymphocytes and Jurkat T cells. Moreover, down-regulation of the expression of 3-OST-3 by RNA interference potently reduced CyPB binding and consequent activation of p44/42 mitogen-activated protein kinases. Altogether, our results strongly support the hypothesis that 3-O-sulfation of GlcNH2 residues could be a key modification that provides specialized HS structures for CyPB binding to responsive cells. Given that 3-O-sulfation of GlcNH2-containing HS by 3-OST-3 also provides binding sites for glycoprotein gD of herpes simplex virus type I, these findings suggest an intriguing structural linkage between the HS sequences involved in CyPB binding and viral infection.
Collapse
Affiliation(s)
- Christophe Vanpouille
- Unité de Glycobiologie Structurale et Fonctionnelle, Unité Mixte de Recherche Number 8576 du CNRS, Institut de Recherche Fédératif No. 147, Université des Sciences et Technologies de Lille, 59655 Villeneuve d'Ascq, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kobayashi T, Habuchi H, Tamura K, Ide H, Kimata K. Essential role of heparan sulfate 2-O-sulfotransferase in chick limb bud patterning and development. J Biol Chem 2007; 282:19589-97. [PMID: 17493930 DOI: 10.1074/jbc.m610707200] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The interactions of heparan sulfate (HS) with heparin-binding growth factors, such as fibroblast growth factors (FGFs), depend greatly on the chain structures. O-Sulfations at various positions on the chain are major factors determining HS structure; therefore, O-sulfation patterns may play a crucial role in controlling the developmental and morphogenetic processes of various tissues and organs by spatiotemporally regulating the activities of heparin-binding growth factors. In a previous study, we found that HS-2-O-sulfotransferase is strongly expressed throughout the mesoderm of chick limb buds during the early stages of development. Here we show that inhibition of HS-2-O-sulfotransferase in the prospective limb region by small inhibitory RNA resulted in the truncation of limb buds and reduced Fgf-8 expression in the apical ectodermal ridge. The treatment also reduced Fgf-10 expression in the mesenchyme. Moreover 2-O-sulfated HS, normally abundant in the basement membranes and mesoderm under ectoderm in limb buds, was significantly reduced in the treated buds. Phosphorylation levels of ERK and Akt were up-regulated in such truncated buds. Thus, we have shown for the first time that 2-O-sulfation of HS is essential for the FGF signaling required for limb bud development and outgrowth.
Collapse
Affiliation(s)
- Takashi Kobayashi
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | | | | | | | | |
Collapse
|
39
|
Campbell EJ, Owen CA. The sulfate groups of chondroitin sulfate- and heparan sulfate-containing proteoglycans in neutrophil plasma membranes are novel binding sites for human leukocyte elastase and cathepsin G. J Biol Chem 2007; 282:14645-54. [PMID: 17384412 DOI: 10.1074/jbc.m608346200] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human leukocyte elastase (HLE) and cathepsin G (CG) are expressed at high levels on the surface of activated human neutrophils (PMN) in catalytically active but inhibitor-resistant forms having the potential to contribute to tissue injury. Herein we have investigated the mechanisms by which HLE and CG bind to PMN plasma membranes. (125)I-Labeled HLE and CG bind to PMN at 0 degrees C in a saturable and reversible manner (K(D) = 5.38 and 4.36 x 10(-7) m and 11.5 and 8.1 x 10(6) binding sites/cell, respectively). Incubation of PMN with radiolabeled HLE and CG in the presence of a 200-fold molar excess of unlabeled HLE, CG, myeloperoxidase, lactoferrin, proteinase 3, phenylmethylsulfonyl fluoride (PMSF)-inactivated HLE, or PMSF-inactivated CG inhibited binding of radiolabeled ligands. This indicates that these PMN granule proteins share binding sites on PMN and that functional active sites of HLE and CG are not required for their binding to PMN. The sulfate groups of heparan sulfate- and chondroitin sulfate-containing proteoglycans are the PMN binding sites for HLE and CG since binding of HLE and CG to PMN was inhibited by incubating PMN with 1) trypsin, chondroitinase ABC, and heparitinases, but not other glycanases, and 2) purified chondroitin sulfates, heparan sulfate, and other sulfated molecules, but not with non-sulfated glycans. Thus, heparan sulfate- and chondroitin sulfate-containing proteoglycans are low affinity, high volume PMN surface binding sites for HLE and CG, which are well suited to bind high concentrations of active serine proteinases released from degranulating PMN.
Collapse
Affiliation(s)
- Edward J Campbell
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
40
|
Hamma-Kourbali Y, Bernard-Pierrot I, Heroult M, Dalle S, Caruelle D, Milhiet PE, Fernig DG, Delbé J, Courty J. Inhibition of the mitogenic, angiogenic and tumorigenic activities of pleiotrophin by a synthetic peptide corresponding to its C-thrombospondin repeat-I domain. J Cell Physiol 2007; 214:250-9. [PMID: 17607711 DOI: 10.1002/jcp.21191] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pleiotrophin (PTN), is a heparin-dependent growth factor involved in angiogenesis and tumor growth. PTN contains a thrombospondin repeat-I (TSR-I) motif in its two beta-sheet domains that are involved in its binding to heparin and its neurite outgrowth activity. Based on the importance of the binding of PTN to heparin in its dimerization and biological activities, we have designed two synthetic peptides, P(13-39) and P(65-97) corresponding to a part of the N-terminal and C-terminal TSR-I motif of PTN, respectively. P(65-97) inhibited the mitogenic, tumorigenic and angiogenic activities of PTN, as well as the mitogenic and an angiogenic activity of fibroblast growth factor-2 (FGF-2). However, P(65-97) had no effect on the mitogenic activity of epidermal growth factor, which does not bind heparin. P(65-97) but not P(13-39) inhibited the binding of PTN and to a lesser extent of FGF-2 to heparin using an immunoassay and an optical biosensor assay and bound directly to heparin with a K(d) of 120 nM. These findings suggest that P(65-97), containing amino acids 65-97 of the TSR-I motif of the C-terminal domain of PTN, inhibits the activities of PTN and FGF-2 by virtue of its ability to bind heparin very effectively and so compete with the growth factors for their polysaccharide co-receptor.
Collapse
Affiliation(s)
- Yamina Hamma-Kourbali
- Laboratoire de Recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires (CRRET), CNRS UMR 7149, Université Paris XII, Créteil Cedex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ricard-Blum S, Beraud M, Raynal N, Farndale RW, Ruggiero F. Structural requirements for heparin/heparan sulfate binding to type V collagen. J Biol Chem 2006; 281:25195-204. [PMID: 16815843 DOI: 10.1074/jbc.m603096200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Collagen-proteoglycan interactions participate in the regulation of matrix assembly and in cell-matrix interactions. We reported previously that a fragment (Ile824-Pro950) of the collagen alpha1(V) chain, HepV, binds to heparin via a cluster of three major basic residues, Arg912, Arg918, and Arg921, and two additional residues, Lys905 and Arg909 (Delacoux, F., Fichard, A., Cogne, S., Garrone, R., and Ruggiero, F. (2000) J. Biol. Chem. 275, 29377-29382). Here, we further characterized the binding of HepV and collagen V to heparin and heparan sulfate by surface plasmon resonance assays. HepV bound to heparin and heparan sulfate with a similar affinity (KD approximately 18 and 36 nM, respectively) in a cation-dependent manner, and 2-O-sulfation of heparin was shown to be crucial for the binding. An octasaccharide of heparin and a decasaccharide of heparan sulfate were required for HepV binding. Studies with HepV mutants showed that the same basic residues were involved in the binding to heparin, to heparan sulfate, and to the cell surface. The contribution of Lys905 and Arg909 was found to be significant. The triple-helical peptide GPC(GPP)5G904-R918(GPP)5GPC-NH2 and native collagen V molecules formed much more stable complexes with heparin than HepV, and collagen V bound to heparin/heparan sulfate with a higher affinity (in the nanomolar range) than HepV. Heat and chemical denaturation strongly decreased the binding, indicating that the triple helix plays a major role in stabilizing the interaction with heparin. Collagen V and HepV may play different roles in cell-matrix interactions and in matrix assembly or remodeling mediated by their specific interactions with heparan sulfate.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- Institut de Biologie et Chimie des Protéines, UMR CNRS 5086, Université Claude Bernard Lyon 1, Institut Féderatif de Recherche 128 BioSciences Gerland, 7 Passage du Vercors, 69367 Lyon Cedex 07, Lyon, France
| | | | | | | | | |
Collapse
|
42
|
Reiland J, Kempf D, Roy M, Denkins Y, Marchetti D. FGF2 binding, signaling, and angiogenesis are modulated by heparanase in metastatic melanoma cells. Neoplasia 2006; 8:596-606. [PMID: 16867222 PMCID: PMC1601937 DOI: 10.1593/neo.06244] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Heparanase (HPSE) and fibroblast growth factor-2 (FGF2) are critical regulators of melanoma angiogenesis and metastasis. Elevated HPSE expression contributes to melanoma progression; however, further augmentation of HPSE presence can inhibit tumorigenicity. HPSE enzymatically cleaves heparan sulfate glycosaminoglycan chains (HS) from proteoglycans. HS act as both low-affinity FGF2 receptors and coreceptors in the formation of high-affinity FGF2 receptors. We have investigated HPSE's ability to modulate FGF2 activity through HS remodeling. Extensive HPSE degradation of human metastatic melanoma cells (70W) inhibited FGF2 binding. Unexpectedly, treatment of 70W cells with low HPSE concentrations enhanced FGF2 binding. In addition, HPSE-unexposed cells did not phosphorylate extracellular signal-related kinase (ERK) or focal adhesion kinase (FAK) in response to FGF2. Conversely, in cells treated with HPSE, FGF2 stimulated ERK and FAK phosphorylation. Secondly, the presence of soluble HPSE-degraded HS enhanced FGF2 binding and ERK phosphorylation at low HS concentrations. Higher concentrations of soluble HS inhibited FGF2 binding, but FGF2 signaling through ERK remained enhanced. Soluble HS were unable to support FGF2-stimulated FAK phosphorylation irrespective of HPSE treatment. Finally, cell exposure to HPSE or to HPSE-degraded HS modulated FGF2-induced angiogenesis in melanoma. In conclusion, these effects suggest relevant mechanisms for the HPSE modulation of melanoma growth factor responsiveness and tumorigenicity.
Collapse
Affiliation(s)
- Jane Reiland
- Department of Comparative Biomedical Sciences-SVM, Louisiana State University-Baton Rouge, Baton Rouge, LA 70803, USA
| | | | | | | | | |
Collapse
|
43
|
Abstract
[Image: see text] Heparin, the well-known anticoagulant polysaccharide, is also active in many other biological systems owing to its structural similarity to HS, but usually lacks selectivity because it is more highly sulfated. A series of straightforward chemical reactions (de-O-sulfation, de-N-sulfation and re-N-acetylation), carried out to partial or complete extent, were combined, resulting in a number of modified heparin polysaccharide derivatives with altered properties. These exhibited a range of abilities to promote cell signalling through the FGF/FGFR tyrosine kinase signalling system, in an in vitro cell assay with combinations of FGF-1, -2, -3 and FGFR 1 and 3. One polysaccharide (N-acetylated, 6-O- and 2-O-sulfated heparin), with only a fraction (<10(-3)) of the anticoagulant activity of heparin (200 U . mg(-1)), promoted FGF-2-mediated angiogenesis (10-fold) and therefore had an improved ratio of pro-angiogenic activity to anticoagulant activity in excess of 10(4) compared to heparin. These results demonstrate that heparin-derived polysaccharides can be engineered for selected activities and have potential in a wide range of medical, biotechnological and tissue-engineering applications. Effect of selected engineered heparin polysaccharides on angiogenesis.
Collapse
Affiliation(s)
- Scott E Guimond
- School of Biological Sciences, University of Liverpool, L69 7ZB, UK
| | | | | |
Collapse
|
44
|
Duchesne L, Tissot B, Rudd TR, Dell A, Fernig DG. N-glycosylation of fibroblast growth factor receptor 1 regulates ligand and heparan sulfate co-receptor binding. J Biol Chem 2006; 281:27178-89. [PMID: 16829530 DOI: 10.1074/jbc.m601248200] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The regulation of cell function by fibroblast growth factors (FGF) occurs through a dual receptor system consisting of a receptor-tyrosine kinase, FGFR and the glycosaminoglycan heparan sulfate (HS). Mutations of some potential N-glycosylation sites in human fgfr lead to phenotypes characteristic of receptor overactivation. To establish how N-glycosylation may affect FGFR function, soluble- and membrane-bound recombinant receptors corresponding to the extracellular ligand binding domain of FGFR1-IIIc were produced in Chinese Hamster Ovary cells. Both forms of FGFR1-IIIc were observed to be heavily N-glycosylated and migrated on SDS-PAGE as a series of multiple bands between 50 and 75 kDa, whereas the deglycosylated receptors migrated at 32 kDa, corresponding to the expected molecular weight of the polypeptides. Optical biosensor and quartz crystal microbalance-dissipation binding assays show that the removal of the N-glycans from FGFR1-IIIc caused an increase in the binding of the receptor to FGF-2 and to heparin-derived oligosaccharides, a proxy for cellular HS. This effect is mediated by N-glycosylation reducing the association rate constant of the receptor for FGF-2 and heparin oligosaccharides. N-Glycans were analyzed by mass spectrometry, which demonstrates a predominance of bi- and tri-antennary core-fucosylated complex type structures carrying one, two, and/or three sialic acids. Modeling of such glycan structures on the receptor protein suggests that at least some may be strategically positioned to interfere with interactions of the receptor with FGF ligand and/or the HS co-receptor. Thus, the N-glycans of the receptor represent an additional pathway for the regulation of the activity of FGFs.
Collapse
Affiliation(s)
- Laurence Duchesne
- School of Biological Sciences, University of Liverpool, Liverpool L69 7ZB, United Kingdom.
| | | | | | | | | |
Collapse
|
45
|
Manton KJ, Sadasivam M, Cool SM, Nurcombe V. Bone-specific heparan sulfates induce osteoblast growth arrest and downregulation of retinoblastoma protein. J Cell Physiol 2006; 209:219-29. [PMID: 16826571 DOI: 10.1002/jcp.20727] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The heparan sulfate (HSs) sugars of the extracellular matrix (ECM) play a key role during both development and wound repair in regulating the flow of growth and adhesive factors across their cell surface receptors. The aim of this study was to assess the structural and functional differences of HS chains extracted from the conditioned media (soluble), cell surface, and ECM of primary human osteoblast cultures, and to analyze their effects on osteoblast cell growth. HS chains from these compartments were characterized through a combination of enzymatic degradation, anion exchange chromatography, and molecular sieving. Although the chains were all approximately the same size, they varied systematically in their sulfate content, suggesting differences in their protein-binding domains. When added to pre-confluent hFOB1.19 osteoblast cultures, HS doses exceeding 500 ng/ml inhibited proliferation, without affecting viability, irrespective of their origin. Furthermore, HS doses of 500 ng/ml also downregulated retinoblastoma, Cyclin A and CDK1 protein expression, indicating that high doses of osteoblast HS negatively regulate cell cycle, resulting in growth arrest; when high doses of HS were withdrawn after a prolonged period, linear cell growth was reestablished. Thus, despite differences in sulfation, HS from either the soluble, cell surface, or matrix compartments of primary human osteoblast cultures are functionally similar with respect to their effects on growth. Binding assays revealed that the HS chains bound TGFbeta1, a known inhibitor of osteoprogenitor growth, at higher affinity than a suite of other bone-related, heparin-binding growth factors. Overcoming such sugar-mediated inhibition may prove important for wound repair.
Collapse
Affiliation(s)
- Kerry J Manton
- Institute of Molecular and Cell Biology, Proteos, Singapore
| | | | | | | |
Collapse
|
46
|
Vandermoere F, El Yazidi-Belkoura I, Adriaenssens E, Lemoine J, Hondermarck H. The antiapoptotic effect of fibroblast growth factor-2 is mediated through nuclear factor-kappaB activation induced via interaction between Akt and IkappaB kinase-beta in breast cancer cells. Oncogene 2005; 24:5482-91. [PMID: 15856005 DOI: 10.1038/sj.onc.1208713] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Fibroblast growth factor-2 (FGF-2) is known for its mitogenic and motogenic effects on breast cancer cells. Here, we demonstrate that FGF-2 is also a potent stimulator of breast cancer cell survival, as it counteracts the apoptotic activity of the C2 ceramide analogue and various chemotherapeutic agents (5-fluorouracil, camptothecin, etoposide) in MCF-7, T47-D and BT-20 cells. The use of pharmacological inhibitors (PD98059, wortmannin, LY294002, SN50) and transfection with negative dominants (IkappaBm, p110(PI3K (phosphoinositide 3-kinase))*DeltaK, AktND) or small interfering RNA targeted against Akt indicated that PI3K/Akt and nuclear factor-kappaB (NF-kappaB), but not p42/p44 MAP-kinases, were required to stimulate FGF-2 antiapoptotic activity. The activation of NF-kappaB was dependent on PI3K/Akt, and using a combination of approaches based on immunoprecipitation, Western blotting and proteomics (two-dimensional electrophoresis and mass spectrometry), we identified the beta form of IkappaB kinase (IKKbeta) as a target of Akt signaling. The selective disruption of IKKbeta using small interfering RNA induced a potent inhibition of Akt-mediated activation of NF-kappaB and cell survival, indicating the functional involvement of IKKbeta in FGF-2 antiapoptotic signaling. Together, these results demonstrate Akt/IKKbeta interaction in NF-kappaB pathways, thereby emphasizing the potential of these proteins as therapeutic targets in breast cancer.
Collapse
Affiliation(s)
- Franck Vandermoere
- ERI-8 INSERM, UPRES-EA-1033, batiment SN3, 59655 Villeneuve d'Ascq, France
| | | | | | | | | |
Collapse
|
47
|
Zhang S, Wang G, Liu D, Bao Z, Fernig DG, Rudland PS, Barraclough R. The C-terminal region of S100A4 is important for its metastasis-inducing properties. Oncogene 2005; 24:4401-11. [PMID: 15856021 DOI: 10.1038/sj.onc.1208663] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The EF-hand protein, S100A4, binds calcium ions and interacts specifically in vitro with protein targets. Elevated levels of S100A4 have been shown to produce a metastatic phenotype in independent models of breast cancer. The presence of S100A4 in the carcinoma cells of patients with different carcinomas is associated with reduced patient survival. In order to identify the region of the S100A4 molecule that is responsible for its metastasis-inducing properties, specific mutant S100A4 genes and proteins have been produced which contain targeted mutations to the two calcium-binding sites and a deletion of the last 15 amino-acid residues of the protein. The ability of the mutant proteins to bind to a potential specific target in vitro, nonmuscle myosin heavy chain, is correlated with their ability to cause motile, invasive and metastatic phenotypes. Mutation of the C-EF hand of S100A4 virtually abolished calcium binding, and motility/invasion in vitro, abolished interaction with a molecular target, and reduced metastasis induction by 2.5-3-fold. However, deletion of the last 15 amino acids of S100A4 reduced motility/invasion, target binding and metastasis-induction to similar extents as the C-EF-hand mutant, but reduced calcium binding by only 26%. The results suggest that the ability to interact with protein target(s) is important in S100A4-induced metastasis.
Collapse
Affiliation(s)
- Shu Zhang
- Cancer and Polio Research Fund Laboratories, Biosciences Building, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | | | | | | | | | | | | |
Collapse
|
48
|
Vanpouille C, Denys A, Carpentier M, Pakula R, Mazurier J, Allain F. Octasaccharide is the minimal length unit required for efficient binding of cyclophilin B to heparin and cell surface heparan sulphate. Biochem J 2005; 382:733-40. [PMID: 15109301 PMCID: PMC1133832 DOI: 10.1042/bj20031453] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2003] [Revised: 04/19/2004] [Accepted: 04/26/2004] [Indexed: 12/11/2022]
Abstract
Cyclophilin B (CyPB) is a heparin-binding protein first identified as a receptor for cyclosporin A. In previous studies, we reported that CyPB triggers chemotaxis and integrin-mediated adhesion of T-lymphocytes by way of interaction with two types of binding sites. The first site corresponds to a signalling receptor; the second site has been identified as heparan sulphate (HS) and appears crucial to induce cell adhesion. Characterization of the HS-binding unit is critical to understand the requirement of HS in pro-adhesive activity of CyPB. By using a strategy based on gel mobility shift assays with fluorophore-labelled oligosaccharides, we demonstrated that the minimal heparin unit required for efficient binding of CyPB is an octasaccharide. The mutants CyPB(KKK-) [where KKK- refers to the substitutions K3A(Lys3-->Ala)/K4A/K5A] and CyPB(DeltaYFD) (where Tyr14-Phe-Asp16 has been deleted) failed to interact with octasaccharides, confirming that the Y14FD16 and K3KK5 clusters are required for CyPB binding. Molecular modelling revealed that both clusters are spatially arranged so that they may act synergistically to form a binding site for the octasaccharide. We then demonstrated that heparin-derived octasaccharides and higher degree of polymerization oligosaccharides inhibited the interaction between CyPB and fluorophore-labelled HS chains purified from T-lymphocytes, and strongly reduced the HS-dependent pro-adhesive activity of CyPB. However, oligosaccharides or heparin were unable to restore adhesion of heparinase-treated T-lymphocytes, indicating that HS has to be present on the cell membrane to support the pro-adhesive activity of CyPB. Altogether, these results demonstrate that the octasaccharide is likely to be the minimal length unit required for efficient binding of CyPB to cell surface HS and consequent HS-dependent cell responses.
Collapse
Affiliation(s)
- Christophe Vanpouille
- Unité de Glycobiologie Structurale et Fonctionnelle, Unité Mixte de Recherche No. 8576 du CNRS, Institut de Recherche Fédératif No. 118, Université des Sciences et Technologies de Lille, 59655 Villeneuve d'Ascq Cedex, France
| | - Agnès Denys
- Unité de Glycobiologie Structurale et Fonctionnelle, Unité Mixte de Recherche No. 8576 du CNRS, Institut de Recherche Fédératif No. 118, Université des Sciences et Technologies de Lille, 59655 Villeneuve d'Ascq Cedex, France
| | - Mathieu Carpentier
- Unité de Glycobiologie Structurale et Fonctionnelle, Unité Mixte de Recherche No. 8576 du CNRS, Institut de Recherche Fédératif No. 118, Université des Sciences et Technologies de Lille, 59655 Villeneuve d'Ascq Cedex, France
| | - Rachel Pakula
- Unité de Glycobiologie Structurale et Fonctionnelle, Unité Mixte de Recherche No. 8576 du CNRS, Institut de Recherche Fédératif No. 118, Université des Sciences et Technologies de Lille, 59655 Villeneuve d'Ascq Cedex, France
| | - Joël Mazurier
- Unité de Glycobiologie Structurale et Fonctionnelle, Unité Mixte de Recherche No. 8576 du CNRS, Institut de Recherche Fédératif No. 118, Université des Sciences et Technologies de Lille, 59655 Villeneuve d'Ascq Cedex, France
| | - Fabrice Allain
- Unité de Glycobiologie Structurale et Fonctionnelle, Unité Mixte de Recherche No. 8576 du CNRS, Institut de Recherche Fédératif No. 118, Université des Sciences et Technologies de Lille, 59655 Villeneuve d'Ascq Cedex, France
- To whom correspondence should be addressed (email )
| |
Collapse
|
49
|
West DC, Rees CG, Duchesne L, Patey SJ, Terry CJ, Turnbull JE, Delehedde M, Heegaard CW, Allain F, Vanpouille C, Ron D, Fernig DG. Interactions of multiple heparin binding growth factors with neuropilin-1 and potentiation of the activity of fibroblast growth factor-2. J Biol Chem 2005; 280:13457-64. [PMID: 15695515 DOI: 10.1074/jbc.m410924200] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The hypothesis that neuropilin-1 (Npn-1) may interact with heparin-binding proteins other than vascular endothelial growth factor has been tested using an optical biosensor-based binding assay. The results show that fibroblast growth factor (FGF) 1, 2, 4, and 7, FGF receptor 1, hepatocyte growth factor/scatter factor (HGF/SF), FGF-binding protein, normal protease sensitive form of prion protein, antithrombin III, and Npn-1 itself are all able to interact with Npn-1 immobilized on the sensor surface. FGF-2, FGF-4, and HGF/SF are also shown to interact with Npn-1 in a solution assay. Moreover, these protein-protein interactions are dependent on the ionic strength of the medium and are inhibited by heparin, and the kinetics of binding of FGF-2, FGF-4 and HGF/SF to Npn-1 are characterized by fast association rate constants (270,000-1,600,000 m(-1) s(-1)). These results suggest that Npn-1 possesses a "heparin" mimetic site that is able to interact at least in part through ionic bonding with the heparin binding site on many of the proteins studied. Npn-1 was also found to potentiate the growth stimulatory activity of FGF-2 on human umbilical vein endothelial cells, indicating that Npn-1 may not just bind but also regulate the activity of heparin-binding proteins.
Collapse
Affiliation(s)
- David C West
- School of Biological Sciences, Biosciences Building, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Barbosa I, Morin C, Garcia S, Duchesnay A, Oudghir M, Jenniskens G, Miao HQ, Guimond S, Carpentier G, Cebrian J, Caruelle JP, van Kuppevelt T, Turnbull J, Martelly I, Papy-Garcia D. A synthetic glycosaminoglycan mimetic (RGTA) modifies natural glycosaminoglycan species during myogenesis. J Cell Sci 2005; 118:253-64. [PMID: 15615789 DOI: 10.1242/jcs.01607] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Crucial events in myogenesis rely on the highly regulated spatiotemporal distribution of cell surface heparan sulfate proteoglycans to which are associated growth factors, thus creating a specific microenvironment around muscle cells. Most growth factors involved in control of myoblast growth and differentiation are stored in the extracellular matrix through interaction with specific sequences of glycosaminoglycan oligosaccharides, mainly heparan sulfate (HS). Different HS subspecies revealed by specific antibodies, have been shown to provide spatiotemporal regulation during muscle development. We have previously shown that glycosaminoglycan (GAG) mimetics called RGTA (ReGeneraTing Agent), stimulate muscle precursor cell growth and differentiation. These data suggest an important role of GAGs during myogenesis; however, little is yet known about the different species of GAGs synthesized during myogenesis and their metabolic regulation. We therefore quantified GAGs during myogenesis of C2.7 cells and show that the composition of GAG species was modified during myogenic differentiation. In particular, HS levels were increased during this process. In addition, the GAG mimetic RGTA, which stimulated both growth and differentiation of C2.7 cells, increased the total amount of GAG produced by these cells without significantly altering their rate of sulfation. RGTA treatment further enhanced HS levels and changed its sub-species composition. Although mRNA levels of the enzymes involved in HS biosynthesis were almost unchanged during myogenic differentiation, heparanase mRNA levels decreased. RGTA did not markedly alter these levels. Here we show that the effects of RGTA on myoblast growth and differentiation are in part mediated through an alteration of GAG species and provide an important insight into the role of these molecules in normal or pathologic myogenic processes.
Collapse
Affiliation(s)
- Isabelle Barbosa
- Laboratoire CRRET, CNRS UMR 7149, Université Paris 12-Val de Marne, 61 Avenue du Général de Gaulle, 94010 Créteil CEDEX, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|