Network


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

Hotspot


Dive into the research topics where Stéphane Brézillon is active.

Publication


Featured researches published by Stéphane Brézillon.


Experimental Cell Research | 2010

Lumican inhibits cell migration through α2β1 integrin.

Cédric Zeltz; Stéphane Brézillon; Jarmo Käpylä; Johannes A. Eble; Hélène Bobichon; Corinne Perreau; Clemens M. Franz; Jyrki Heino; François-Xavier Maquart; Yanusz Wegrowski

Lumican, an extracellular matrix protein of the small leucine-rich proteoglycan family, has been shown to impede melanoma progression by inhibiting cell migration. In the present study, we show that lumican targets α2β1 integrin thereby inhibiting cell migration. A375 melanoma cells were transfected with siRNA directed against the α2 integrin subunit. Compared to A375 control cells, the anti-migratory effect of lumican was abrogated on transfected A375 cells. Moreover, lumican inhibited the chemotactic migration of Chinese hamster ovary (CHO) cells stably transfected with α2 integrin subunit (CHO-A2) but not that of wild-type CHO cells (CHO-WT) lacking this subunit. In contrast to CHO-WT cells, we observed in time-lapse microscopy a decrease of CHO-A2 cell migration speed in presence of lumican. Focal adhesion kinase phosphorylated at tyrosine-397 (pFAK) and total FAK were analysed in CHO-WT and CHO-A2 cells. A significant decrease of the ratio pFAK/FAK was shown in presence of recombinant human lumican. Using solid phase assays, a direct binding between lumican and the α2β1 integrin was demonstrated. This interaction did not involve the glycan moiety of lumican and was cation independent. Lumican was also able to bind the activated I domain of the α2 integrin subunit with a K(d)≥200nM. In conclusion, we demonstrated for the first time that the inhibition of cell migration by lumican depends on a direct binding between the core protein of lumican and the α2β1 integrin.


Thrombosis Research | 2011

Lumican inhibits angiogenesis by interfering with α2β1 receptor activity and downregulating MMP-14 expression

Jolanta Niewiarowska; Stéphane Brézillon; Radoslaw Bednarek; François-Xavier Maquart; Mariusz Malinowski; Magdalena Wiktorska; Yanusz Wegrowski; Czeslaw S. Cierniewski

INTRODUCTION Previous studies showed that lumican, a small leucine-rich proteoglycan that binds to α2 integrin I domain, is an efficient inhibitor of cell adhesion and migration. In this report, we tested its effect on angiogenesis in vitro and in vivo. MATERIALS AND METHODS Effect of lumican on angiogenesis was evaluated by in vitro capillary tube formation test performed between Fibrin II Gels or in Matrigel™ and in vivo by Matrigel(™) plug assay in BALB/c mice. Changes in matrix metalloproteinases expression caused by lumican were analyzed in endothelial cells by real-time PCR, Western immunoblotting and gelatin zymography. RESULTS In unchallenged endothelial cells, Matrigel™ induced robust capillary morphogenesis. In contrast, tube formation was dramatically reduced by lumican, and by siRNA to β1 integrin subunit mRNA but not by control siRNA. Similarly, lumican effectively inhibited neovascularization in vivo in assays using Matrigel™ plugs formed in BALB/c mice. Interestingly, lumican significantly reduced expression of matrix metalloproteinases, particularly MMP-14 that is known to activate other MMPs in close vicinity of endothelial cell membranes. CONCLUSIONS Our results provide strong evidence that lumican affects angiogenesis both by interfering with α2β1 receptor activity and downregulating proteolytic activity associated with surface membranes of endothelial cells.


FEBS Journal | 2013

Lumican effects in the control of tumour progression and their links with metalloproteinases and integrins

Stéphane Brézillon; Katarzyna Pietraszek; François-Xavier Maquart; Yanusz Wegrowski

Lumican is a member of the small leucine‐rich proteoglycan family. It is present in numerous extracellular matrices of different tissues, such as muscle, cartilage, and cornea. In skin, lumican is present as a glycoprotein. It plays a critical role in collagen fibrillogenesis, as shown by knocking out of its gene in mice. A direct link between lumican expression and melanoma progression and metastasis has been demonstrated. Lumican was shown to impede tumour cell migration and invasion by directly interacting with the α2β1 integrin. In addition, an active sequence of the lumican core protein, called lumcorin, was identified as being responsible for inhibition of melanoma cell migration. Lumican was also shown to exert angiostatic properties by downregulating the proteolytic activity associated with endothelial cell membranes, particularly matrix metalloproteinase (MMP)‐14 and MMP‐9. Globally, lumican appears to be a potent agent for inhibiting tumour progression rather than tumorigenesis. However, progressive changes in proteoglycans occur in the tumour environment. The complexity and diversity of proteoglycan structure might be responsible for a variety of functions that regulate cell behaviour. Through their core protein and their glycosaminoglycan chains, proteoglycans can interact with growth factors and chemokines. These interactions affect cell signalling, motility, adhesion, growth, and apoptosis. This review summarizes recent data concerning lumican control of tumour progression in different cancers, with a particular focus on its interactions with MMPs and integrins. Its potential therapeutic implications are discussed.


PLOS ONE | 2012

Effect of lumican on the migration of human mesenchymal stem cells and endothelial progenitor cells: involvement of matrix metalloproteinase-14.

Mariusz Malinowski; Katarzyna Pietraszek; Corinne Perreau; Mateusz Boguslawski; Véronique Decot; Jean-François Stoltz; Laurent Vallar; Jolanta Niewiarowska; Czeslaw S. Cierniewski; François-Xavier Maquart; Yanusz Wegrowski; Stéphane Brézillon

Background Increasing number of evidence shows that soluble factors and extracellular matrix (ECM) components provide an optimal microenvironment controlling human bone marrow mesenchymal stem cell (MSC) functions. Successful in vivo administration of stem cells lies in their ability to migrate through ECM barriers and to differentiate along tissue-specific lineages, including endothelium. Lumican, a protein of the small leucine-rich proteoglycan (SLRP) family, was shown to impede cell migration and angiogenesis. The aim of the present study was to analyze the role of lumican in the control of MSC migration and transition to functional endothelial progenitor cell (EPC). Methodology/Principal Findings Lumican inhibited tube-like structures formation on Matrigel® by MSC, but not EPC. Since matrix metalloproteinases (MMPs), in particular MMP-14, play an important role in remodelling of ECM and enhancing cell migration, their expression and activity were investigated in the cells grown on different ECM substrata. Lumican down-regulated the MMP-14 expression and activity in MSC, but not in EPC. Lumican inhibited MSC, but not EPC migration and invasion. The inhibition of MSC migration and invasion by lumican was reversed by MMP-14 overexpression. Conclusion/Significance Altogether, our results suggest that lumican inhibits MSC tube-like structure formation and migration via mechanisms that involve a decrease of MMP-14 expression and activity.


Journal of Structural Biology | 2013

Quantitative analysis of type I collagen fibril regulation by lumican and decorin using AFM

Dimitar R. Stamov; Anna Müller; Yanusz Wegrowski; Stéphane Brézillon; Clemens M. Franz

Lumican and decorin, two members of the small leucine-rich repeat proteoglycan (SLRP) family, have been implicated as regulators of collagen I fibril structure in different tissues. Both proteoglycans consist of a core protein and a glycosaminoglycan (GAG) chain, but quantitative information regarding the precise role of the protein and GAG moieties in regulating collagen structure is still limited. In this study, we used AFM imaging and a model system of aligned collagen I nanofibrils to investigate the role of lumican and decorin on collagen I fibril structure with high resolution. When co-assembled with collagen I, recombinant lumican or decorin proteins lacking the GAG chains decreased collagen fibril width to values below <100nm and increased interfibrillar spacing in a dose-dependent manner. At lower concentrations, lumican appeared to have a stabilizing effect on newly-formed collagen fibrils, while at higher concentrations both lumican and decorin inhibited collagen fibrillogenesis. GAG-containing decorin also increased interfibrillar spacing, decreased fibril width and ultimately inhibited fibrillogenesis, but these effects required lower concentrations compared to recombinant decorin, indicating that the decorin core protein alone cannot compensate for the full regulatory and structural contribution of the GAG chain during collagen I fibrillogenesis. Using a 2D autocorrelation approach, we furthermore analyzed and compared the effects of recombinant and glycosylated decorin on collagen ultrastructure, providing a quantitative measure for the observed structural differences. AFM analysis of ordered fibrillar collagen arrays in combination with quantitative autocorrelation image analysis thus provides a useful tool for investigating SLRP-dependent nanoscale effects on collagen fibril structure.


Life Sciences | 2008

Lumican affects actin cytoskeletal organization in human melanoma A375 cells

Agata Radwańska; Dagmara Baczynska; Dorota Nowak; Stéphane Brézillon; Agnieszka Popow; François-Xavier Maquart; Yanusz Wegrowski; Maria Malicka-Błaszkiewicz

AIMS Lumican, a small leucine-rich proteoglycan (SLRP), has attracted attention as a molecule of the extracellular matrix possibly involved in signalling pathways affecting cancer cell behaviour. The remodelling of the actin cytoskeleton, induced in response to external stimuli, is crucial for cell motility and intracellular signal transduction. The main goal of this study was to examine the effects of recombinant lumican on actin organization, the state of actin polymerization, actin isoform expression, and their sub-cellular distribution in the A375 human melanoma cell line. MAIN METHODS Fluorescence and confocal microscopy were used to observe actin cytoskeletal organization and the sub-cellular distribution of cytoplasmic beta- and gamma-actins. The ability of actin to inhibit DNaseI activity was used to quantify actin. Western blotting and real-time PCR were used to determine the expression levels of the actin isoforms. KEY FINDINGS A375 cells grown on lumican coatings changed in morphology and presented rearranged actin filament organization: from filaments evenly spread throughout the whole cell body to their condensed sub-membrane localization. In the presence of lumican, both actin isoforms were concentrated under the cellular membrane. A statistically significant increase in the total, filamentous, and monomeric actin pools was observed in A375 cells grown on lumican. SIGNIFICANCE Novel biological effects of lumican, an extracellular matrix SLRP, on the actin pool and organization are identified, which may extend our understanding of the mechanism underlying the inhibitory effect of lumican on the migration of melanoma cells.


FEBS Letters | 2014

Lumican: A new inhibitor of matrix metalloproteinase‐14 activity

Katarzyna Pietraszek; Aurore Chatron-Colliet; Stéphane Brézillon; Corinne Perreau; Anna Jakubiak-Augustyn; Hubert Krotkiewski; François-Xavier Maquart; Yanusz Wegrowski

We previously showed that lumican regulates MMP‐14 expression. The aim of this study was to compare the effect of lumican and decorin on MMP‐14 activity. In contrast to decorin, the glycosylated form of lumican was able to significantly decrease MMP‐14 activity in B16F1 melanoma cells. Our results suggest that a direct interaction occurs between lumican and MMP‐14. Lumican behaves as a competitive inhibitor which leads to a complete blocking of the activity of MMP‐14. It binds to the catalytic domain of MMP‐14 with moderate affinity (K D ∼ 275 nM). Lumican may protect collagen against MMP‐14 proteolysis, thus influencing cell–matrix interaction in tumor progression.


PLOS ONE | 2012

Tetrastatin, the NC1 domain of the α4(IV) collagen chain: a novel potent anti-tumor matrikine.

Sylvie Brassart-Pasco; Karine Sénéchal; Jessica Thevenard; Laurent Ramont; Jérôme Devy; Ludivine Di Stefano; Aurélie Dupont-Deshorgue; Stéphane Brézillon; Jezabel Feru; Jean-François Jazeron; Marie-Danièle Diebold; Sylvie Ricard-Blum; François-Xavier Maquart; Jean Claude Monboisse

Background NC1 domains from α1, α2, α3 and α6(IV) collagen chains were shown to exert anti-tumor or anti-angiogenic activities, whereas the NC1 domain of the α4(IV) chain did not show such activities so far. Methodology/Principal Findings We demonstrate in the present paper that the NC1 α4(IV) domain exerts a potent anti-tumor activity both in vitro and in an experimental human melanoma model in vivo. The overexpression of NC1 α4(IV) in human UACC-903 melanoma cells strongly inhibited their in vitro proliferative (–38%) and invasive (–52%) properties. MT1-MMP activation was largely decreased and its cellular distribution was modified, resulting in a loss of expression at the migration front associated with a loss of migratory phenotype. In an in vivo xenograft model in athymic nude mice, the subcutaneous injection of NC1 α4(IV)-overexpressing melanoma cells induced significantly smaller tumors (–80% tumor volume) than the Mock cells, due to a strong inhibition of tumor growth. Exogenously added recombinant human NC1 α4(IV) reproduced the inhibitory effects of NC1 α4(IV) overexpression in UACC-903 cells but not in dermal fibroblasts. An anti-αvβ3 integrin blocking antibody inhibited cell adhesion on recombinant human NC1 α4(IV) substratum. The involvement of αvβ3 integrin in mediating NC1 α4(IV) effect was confirmed by surface plasmon resonance (SPR) binding assays showing that recombinant human NC1 α4(IV) binds to αvβ3 integrin (KD = 148±9.54 nM). Conclusion/Significance Collectively, our results demonstrate that the NC1 α4(IV) domain, named tetrastatin, is a new endogenous anti-tumor matrikine.


PLOS ONE | 2013

Lumican - derived peptides inhibit melanoma cell growth and migration.

Katarzyna Pietraszek; Stéphane Brézillon; Corinne Perreau; Maria Malicka-Błaszkiewicz; François-Xavier Maquart; Yanusz Wegrowski

Lumican, a small leucine-rich proteoglycan of the extracellular matrix, presents potent anti-tumor properties. Previous works from our group showed that lumican inhibited melanoma cell migration and tumor growth in vitro and in vivo. Melanoma cells adhered to lumican, resulting in a remodeling of their actin cytoskeleton and preventing their migration. In addition, we identified a sequence of 17 amino acids within the lumican core protein, named lumcorin, which was able to inhibit cell chemotaxis and reproduce anti-migratory effect of lumican in vitro. The aim of the present study was to characterize the anti-tumor mechanism of action of lumcorin. Lumcorin significantly decreased the growth in monolayer and in soft agar of two melanoma cell lines – mice B16F1 and human SK-MEL-28 cells – in comparison to controls. Addition of lumcorin to serum free medium significantly inhibited spontaneous motility of these two melanoma cell lines. To characterize the mechanisms involved in the inhibition of cell migration by lumcorin, the status of the phosphorylation/dephosphorylation of proteins was examined. Inhibition of focal adhesion kinase phosphorylation was observed in presence of lumcorin. Since cancer cells have been shown to migrate and to invade by mechanisms that involve matrix metalloproteinases (MMPs), the expression and activity of MMPs were analyzed. Lumcorin induced an accumulation of an intermediate form of MMP-14 (~59kDa), and inhibited MMP-14 activity. Additionally, we identified a short, 10 amino acids peptide within lumcorin sequence, which was able to reproduce its anti-tumor effect on melanoma cells. This peptide may have potential pharmacological applications.


PLOS ONE | 2016

Lumican Inhibits SNAIL-Induced Melanoma Cell Migration Specifically by Blocking MMP-14 Activity

Marta Stasiak; Joanna Boncela; Corinne Perreau; Konstantina Karamanou; Aurore Chatron-Colliet; Isabelle Proult; Patrycja Przygodzka; Shukti Chakravarti; François Xavier Maquart; M. Anna Kowalska; Yanusz Wegrowski; Stéphane Brézillon

Lumican, a small leucine rich proteoglycan, inhibits MMP-14 activity and melanoma cell migration in vitro and in vivo. Snail triggers epithelial-mesenchymal transitions endowing epithelial cells with migratory and invasive properties during tumor progression. The aim of this work was to investigate lumican effects on MMP-14 activity and migration of Snail overexpressing B16F1 (Snail-B16F1) melanoma cells and HT-29 colon adenocarcinoma cells. Lumican inhibits the Snail induced MMP-14 activity in B16F1 but not in HT-29 cells. In Snail-B16F1 cells, lumican inhibits migration, growth, and melanoma primary tumor development. A lumican-based strategy targeting Snail-induced MMP-14 activity might be useful for melanoma treatment.

Collaboration


Dive into the Stéphane Brézillon's collaboration.

Top Co-Authors

Avatar

François-Xavier Maquart

University of Reims Champagne-Ardenne

View shared research outputs
Top Co-Authors

Avatar

Yanusz Wegrowski

University of Reims Champagne-Ardenne

View shared research outputs
Top Co-Authors

Avatar

Corinne Perreau

University of Reims Champagne-Ardenne

View shared research outputs
Top Co-Authors

Avatar

Aurore Chatron-Colliet

University of Reims Champagne-Ardenne

View shared research outputs
Top Co-Authors

Avatar

Katarzyna Pietraszek

University of Reims Champagne-Ardenne

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cédric Zeltz

University of Reims Champagne-Ardenne

View shared research outputs
Top Co-Authors

Avatar

François Xavier Maquart

University of Reims Champagne-Ardenne

View shared research outputs
Top Co-Authors

Avatar

Hélène Bobichon

University of Reims Champagne-Ardenne

View shared research outputs
Top Co-Authors

Avatar

Jean Claude Monboisse

University of Reims Champagne-Ardenne

View shared research outputs
Researchain Logo
Decentralizing Knowledge