Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Scott E. Guimond is active.

Publication


Featured researches published by Scott E. Guimond.


Journal of Endocrinology | 2011

Extracellular matrix and cell signalling: the dynamic cooperation of integrin, proteoglycan and growth factor receptor

Soo-Hyun Kim; Jeremy E. Turnbull; Scott E. Guimond

Extracellular matrices (ECM) are secreted molecules that constitute the cell microenvironment, composed of a dynamic and complex array of glycoproteins, collagens, glycosaminoglycans and proteoglycans. ECM provides the bulk, shape and strength of many tissues in vivo, such as basement membrane, bone and cartilage. In vitro, most animal cells can only grow when they are attached to surfaces through ECM. ECM is also the substrate for cell migration. However, ECM provides much more than just mechanical and structural support, with implications in developmental patterning, stem cell niches and cancer. ECM imparts spatial context for signalling events by various cell surface growth factor receptors and adhesion molecules such as integrins. The external physical properties of ECM may also have a role in the signalling process. ECM molecules can be flexible and extendable, and mechanical tension can expose cryptic sites, which could further interact with growth factors or their receptors. ECM proteins and structures can determine the cell behaviour, polarity, migration, differentiation, proliferation and survival by communicating with the intracellular cytoskeleton and transmission of growth factor signals. Integrins and proteoglycans are the major ECM adhesion receptors which cooperate in signalling events, determining the signalling outcomes, and thus the cell fate. This review focuses on the emerging concept of spatial cell biology of ECM, especially the current understanding of integrins and heparan sulphate proteoglycans as the essential cellular machineries that sense, integrate and respond to the physical and chemical environmental information either by directly connecting with the local adhesion sites or by regulating global cellular processes through growth factor receptor signalling pathways, leading to the integration of both external and internal signals in space and time.


Current Biology | 1999

Fibroblast growth factor receptor signalling is dictated by specific heparan sulphate saccharides

Scott E. Guimond; Jeremy E. Turnbull

Signalling by fibroblast growth factors (FGFs) through FGF receptors (FGFRs) depends on the cell-surface polysaccharide heparan sulphate (HS) [1] [2]. HS has an ordered domain structure of highly diverse saccharide motifs that present unique displays of sulphate, carboxyl and hydroxyl groups [3]. These motifs interact with many proteins, particularly growth factors. HS binds both to FGFs [4] [5] [6] and FGFRs [7], and probably activates signalling by facilitating ligand-induced receptor dimerisation [8] [9]. Nevertheless, the extent to which specific HS saccharide sequences play a regulatory role has not been established. By screening a library of structurally diverse HS decasaccharides in bioassays of FGF signalling mediated by three different FGFR isoforms, we found that saccharides showed specificity for both ligands and receptors; some saccharides selectively activated FGF signalling through different FGFR isoforms, others acted as negative regulators. We conclude that HS saccharides play critical roles in dictating the specificity of ligand-receptor interactions in FGFR signalling. Controlled alterations in HS structures [10] would provide a mechanism for regulation of cellular responsiveness to growth factors that bind HS.


The Journal of Neuroscience | 2004

Anosmin-1 Modulates Fibroblast Growth Factor Receptor 1 Signaling in Human Gonadotropin-Releasing Hormone Olfactory Neuroblasts through a Heparan Sulfate-Dependent Mechanism

David González-Martínez; Soo-Hyun Kim; Youli Hu; Scott E. Guimond; Jonathan Schofield; Paul Winyard; Gabriella Barbara Vannelli; Jeremy E. Turnbull; Pierre-Marc Bouloux

Defects of either anosmin-1 or fibroblast growth factor receptor 1 (FGFR1) are known to underlie hereditary Kallmanns syndrome (KS), a human disorder of olfactory and gonadotropin-releasing hormone (GnRH) neuronal ontogeny. Here, we report a functional interaction between anosmin-1 and the FGFR1-FGF2-heparan sulfate complex, leading to amplified responses in the FGFR1 signaling pathway. In human embryonic GnRH olfactory neuroblasts, wild-type anosmin-1, but not proteins with loss-of-function KS mutations, induces neurite outgrowth and cytoskeletal rearrangements through FGFR1-dependent mechanisms involving p42/44 and p38 mitogen-activated protein kinases and Cdc42/Rac1 activation. Furthermore, anosmin-1 enhances FGF2 signaling specifically through FGFR1 IIIc in heterologous BaF3 lymphoid cells in a heparan sulfate-dependent manner. Our study provides compelling evidence for anosmin-1 as an isoform-specific co-ligand modulator of FGFR signaling that amplifies and specifies FGFR1 signaling responses during human nervous system development and defines a mechanism underlying the link between autosomal and X-linked KS.


Methods in Enzymology | 1994

[11] Regulation by heparan sulfate in fibroblast growth factor signaling

Alan C. Rapraeger; Scott E. Guimond; Alison Krufka; Bradley B. Olwin

Abstract The integral role of heparan sulfate proteoglycans in FGF signaling provides a potential means of regulating FGF activity. This regulation may be used by the cell, where the modification of heparan sulfate glycosaminoglycans during their synthesis in the Golgi can produce cell type- and potentially ligand-specific sulfation sequences. The description of these sequences will not only provide information on how this regulation is achieved, perhaps lending insight into other heparan sulfate-ligand interactions, but may also discern sulfated mimetics that can be used to disrupt or alter FGF signaling. These mimetics may be useful in the treatment of disease, or in understanding how FGF signaling via discrete pathways within the cell leads to specific cellular responses, such as activation of mitogenic signaling pathways, calcium fluxes, and cellular differentiation.


Developmental Dynamics | 2003

Localisation of specific heparan sulfate proteoglycans during the proliferative phase of brain development

M. Ford-Perriss; Kirsty Turner; Scott E. Guimond; Anwyn Apedaile; Hans-Dieter Haubeck; Jeremy E. Turnbull; Mark Murphy

Early brain development is characterised by the proliferation of neural precursor cells. Several families of signalling molecules such as the fibroblast growth factors (FGFs) and Wnts are known to play important roles in this early phase of brain development. Accumulating evidence demonstrates that signalling of these molecules requires the presence of heparan sulfate chains attached to a proteoglycan core protein (HSPG). However, the specific identity of the HSPG components in the developing brain is unknown. To determine which HSPGs might be involved at this early phase, we analysed the expression of the major cell surface HSPG families in the developing brain at a time of most active proliferation. Syndecan‐1 and glypican‐4 were the most highly expressed in the developing brain during the time of peak proliferation and localise to ventricular regions of the brain, where the precursor cells are proliferating. Syndecan‐4, although less abundant, also localises to cells in the ventricular zone. We have also examined HSPG involvement in brain development using cultures of embryonic neural precursor cells. We find that FGF2 stimulation of proliferation is inhibited in the presence of sodium chlorate, an inhibitor of heparan sulfate synthesis, and is rescued by addition of exogenous heparan sulfate. These data support a requirement for heparan sulfate in FGF signalling for proliferation of brain precursor cells. The expression of these specific HSPGs within the proliferative zone of the brain suggests that they may be involved in regulation of early brain development, such as FGF‐stimulated proliferation. Developmental Dynamics 227:170–184, 2003.


Journal of Biological Chemistry | 2009

Novel mechanisms of fibroblast growth factor receptor 1 regulation by extracellular matrix protein anosmin-1.

Youli Hu; Scott E. Guimond; Paul J. Travers; Steven Mark Cadman; Erhard Hohenester; Jeremy E. Turnbull; Soo-Hyun Kim; Pierre-Marc Bouloux

Activation of fibroblast growth factor (FGF) signaling is initiated by a multiprotein complex formation between FGF, FGF receptor (FGFR), and heparan sulfate proteoglycan on the cell membrane. Cross-talk with other factors could affect this complex assembly and modulate the biological response of cells to FGF. We have previously demonstrated that anosmin-1, a glycosylated extracellular matrix protein, interacts with the FGFR1 signaling complex and enhances its activity in an IIIc isoform-specific and HS-dependent manner. The molecular mechanism of anosmin-1 action on FGFR1 signaling, however, remains unknown. Here, we show that anosmin-1 directly binds to FGFR1 with high affinity. This interaction involves domains in the N terminus of anosmin-1 (cysteine-rich region, whey acidic protein-like domain and the first fibronectin type III domain) and the D2–D3 extracellular domains of FGFR1. In contrast, anosmin-1 binds to FGFR2IIIc with much lower affinity and displays negligible binding to FGFR3IIIc. We also show that FGFR1-bound anosmin-1, although capable of binding to FGF2 alone, cannot bind to a FGF2·heparin complex, thus preventing FGFR1·FGF2·heparin complex formation. By contrast, heparin-bound anosmin-1 binds to pre-formed FGF2·FGFR1 complex, generating an anosmin-1·FGFR1·FGF2·heparin complex. Furthermore, a functional interaction between anosmin-1 and the FGFR1 signaling complex is demonstrated by immunofluorescence co-localization and Transwell migration assays where anosmin-1 was shown to induce opposing effects during chemotaxis of human neuronal cells. Our study provides molecular and cellular evidence for a modulatory action of anosmin-1 on FGFR1 signaling, whereby binding of anosmin-1 to FGFR1 and heparin can play a dual role in assembly and activity of the ternary FGFR1·FGF2·heparin complex.


Biochemistry | 1996

Two hierarchies of FGF-2 signaling in heparin: mitogenic stimulation and high-affinity binding/receptor transphosphorylation.

Alison Krufka; Scott E. Guimond; Alan C. Rapraeger

FGF-2 activates multiple signaling pathways by a cell surface signaling complex assembled with FGF, its receptor tyrosine kinase, and heparan sulfate proteoglycan. Heparan sulfate binds to a site on the receptor and at least one site on the growth factor. Several models propose an important role for heparan sulfate not only in facilitating FGF-2 binding to its receptor tyrosine kinase but also in promoting signaling via formation of receptor dimers. Such dimers are capable of transphosphorylation of the cytoplasmic domain of the receptor, leading to the generation of phosphotyrosines that are important initiators of intracellular signaling pathways. To explore the participation of heparan sulfates in the formation of a signaling complex that activates these pathways, the binding and activity of FGF-2 on Swiss 3T3 fibroblasts and F32 lymphoid cells is examined with either native or modified forms of heparin. As shown previously, fibroblasts treated with chlorate, which inhibits the sulfation of heparan sulfate and its subsequent binding to FGF-2, display a dramatically reduced response to picomolar concentrations of FGF-2, but binding to receptors and a mitogenic response is restored by heparin. However, the restoration of high-affinity binding is seen only at an optimal concentration of heparin. Excess heparin competes for binding sites within the signaling complex such that high-affinity binding and receptor transphosphorylation are reduced. Despite this, mitogenic signaling is not diminished. A similar result is observed using heparin fragments that promote mitogenesis but not high-affinity binding. These results suggest that the high-affinity signaling complex that is necessary for stable receptor transphosphorylation differs from the signaling complex sufficient for triggering mitogenesis. We speculate that heparan sulfate in vivo participates in two hierarchies of receptor activation. In one, heparan sulfate participates in FGF-2 binding to its receptor tyrosine kinase and activation of mitogenic signaling, perhaps through monomeric receptors or the transient formation of receptor dimers. In the second hierarchy, heparan sulfate participates in the stabilization of a signaling complex that is likely to be comprised of receptor multimers that carry out effective receptor transphosphorylation. A further description of this mechanism may lead to an understanding of how heparan sulfate or its homologues can regulate specific signaling pathways within the cell.


Journal of Biological Chemistry | 2012

Diversification of the Structural Determinants of Fibroblast Growth Factor-Heparin Interactions IMPLICATIONS FOR BINDING SPECIFICITY

Ruoyan Xu; Alessandro Ori; Timothy R. Rudd; Katarzyna A. Uniewicz; Yassir Ahmed; Scott E. Guimond; Mark A. Skidmore; Giuliano Siligardi; Edwin A. Yates; David G. Fernig

Background: Heparan sulfate (HS) regulates the transport and signaling activities of fibroblast growth factors (FGF). Results: The molecular determinants of the interactions of FGFs and heparin were identified. Conclusion: There are clear molecular specificities determining the interactions of FGFs with the polysaccharide. Significance: The expansion of the FGFs in metazoan evolution parallels the diversification of the specificity of their interactions with heparin. 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 KD 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.


Glycobiology | 2009

Glycosaminoglycan origin and structure revealed by multivariate analysis of NMR and CD spectra

Timothy R. Rudd; Mark A. Skidmore; Scott E. Guimond; Cesare Cosentino; Giangiacomo Torri; David G. Fernig; Robert M. Lauder; Marco Guerrini; Edwin A. Yates

Principal component analysis (PCA) is a method of simplifying complex datasets to generate a lower number of parameters, while retaining the essential differences and allowing objective comparison of large numbers of datasets. Glycosaminoglycans (GAGs) are a class of linear sulfated carbohydrates with diverse sequences and consequent complex conformation and structure. Here, PCA is applied to three problems in GAG research: (i) distinguishing origins of heparin preparations, (ii) structural analysis of heparin derivatives, and (iii) classification of chondroitin sulfates (CS). The results revealed the following. (i) PCA of heparin (13)C NMR spectra allowed their origins to be distinguished and structural differences were identified. (ii) Analysis of the information-rich (1)H and (13)C NMR spectra of a series of systematically modified heparin derivatives uncovered underlying properties. These included the presence of interactions between residues, providing evidence that a degree of degeneracy exists in linkage geometry and that a different degree of variability exists for the two types of glycosidic linkage. The relative sensitivity of each position (C or H nucleus) in the disaccharide repeating unit to changes in O-, N-sulfation and N-acetylation was also revealed. (iii) Analysis of the (1)H NMR and CD spectra of a series of CS samples from different origins allowed their structural classification and highlighted the power of employing complementary spectroscopic methods in concert with PCA.


Journal of Biological Chemistry | 2009

Rapid Purification and High Sensitivity Analysis of Heparan Sulfate from Cells and Tissues TOWARD GLYCOMICS PROFILING

Scott E. Guimond; Tania M. Puvirajesinghe; Mark A. Skidmore; Ina Kalus; Thomas Dierks; Edwin A. Yates; Jeremy E. Turnbull

Studies on glycosaminoglycans and proteoglycans (PGs) have been hampered by difficulties in isolation and analysis by traditional methods that are laborious and lack sensitivity and throughput. Here we demonstrate a simple method for rapid isolation of proteoglycans (RIP) employing phenol/guanidine/chloroform reagent to purify heparan sulfate (HS) PGs quantitatively from various tissues and cells. We further show that this generic purification methodology, when applied in concert with a BODIPYTM fluorescent label, permits structural analyses on RIP-purified HS at ∼1,000-fold higher sensitivity than standard UV detection methods and ∼10–100-fold higher sensitivity than previous fluorescence detection methods. The utility of RIP-BODIPY methodology was demonstrated by rapid profiling of HS structural composition from small tissue samples, multiple mouse organs, and as little as a few thousand cultured cells. It was also used to generate novel insights into in vivo structural changes in HS from Sulf1 knock-out mice for the first time that differed significantly from previous observations limited to tissue culture experiments. RIP was also applied to purify HS for bioassay testing, exemplified by cell assays of fibroblast growth factor signaling activation; this generated data from 2-O-sulfotransferase knock-out mice and revealed an unexpected deficiency in fibroblast growth factor activation by HS from heterozygous mice. These data demonstrate that RIP will underpin emerging efforts to develop glycomics profiling strategies for HS and other glycosaminoglycans to explore their structure-function relationships in complex biological systems.

Collaboration


Dive into the Scott E. Guimond's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giangiacomo Torri

Federal University of Paraná

View shared research outputs
Top Co-Authors

Avatar

Marco Guerrini

Federal University of Paraná

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge