Network


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

Hotspot


Dive into the research topics where Jean Sévigny is active.

Publication


Featured researches published by Jean Sévigny.


Purinergic Signalling | 2006

The E-NTPDase family of ectonucleotidases: Structure function relationships and pathophysiological significance.

Simon C. Robson; Jean Sévigny; Herbert Zimmermann

Ectonucleotidases are ectoenzymes that hydrolyze extracellular nucleotides to the respective nucleosides. Within the past decade, ectonucleotidases belonging to several enzyme families have been discovered, cloned and characterized. In this article, we specifically address the cell surface-located members of the ecto-nucleoside triphosphate diphosphohydrolase (E-NTPDase/CD39) family (NTPDase1,2,3, and 8). The molecular identification of individual NTPDase subtypes, genetic engineering, mutational analyses, and the generation of subtype-specific antibodies have resulted in considerable insights into enzyme structure and function. These advances also allow definition of physiological and patho-physiological implications of NTPDases in a considerable variety of tissues. Biological actions of NTPDases are a consequence (at least in part) of the regulated phosphohydrolytic activity on extracellular nucleotides and consequent effects on P2-receptor signaling. It further appears that the spatial and temporal expression of NTPDases by various cell types within the vasculature, the nervous tissues and other tissues impacts on several patho-physiological processes. Examples include acute effects on cellular metabolism, adhesion, activation and migration with other protracted impacts upon developmental responses, inclusive of cellular proliferation, differentiation and apoptosis, as seen with atherosclerosis, degenerative neurological diseases and immune rejection of transplanted organs and cells. Future clinical applications are expected to involve the development of new therapeutic strategies for transplantation and various inflammatory cardiovascular, gastrointestinal and neurological diseases.


Hepatology | 2007

Transforming growth factor‐β and substrate stiffness regulate portal fibroblast activation in culture

Zhaodong Li; Jonathan A. Dranoff; Erick P. Chan; Masayuki Uemura; Jean Sévigny; Rebecca G. Wells

Myofibroblasts derived from portal fibroblasts are important fibrogenic cells in the early stages of biliary fibrosis. In contrast to hepatic stellate cells, portal fibroblasts have not been well studied in vitro, and little is known about their myofibroblastic differentiation. In this article we report the isolation and characterization of rat portal fibroblasts in culture. We demonstrate that primary portal fibroblasts undergo differentiation to α‐smooth muscle actin–expressing myofibroblasts over 10–14 days. Marker analysis comparing portal fibroblasts to hepatic stellate cells demonstrated that these are distinct populations and that staining with elastin and desmin can differentiate between them. Portal fibroblasts expressed elastin at all stages in culture but never expressed desmin, whereas hepatic stellate cells consistently expressed desmin but never elastin. Immunostaining of rat liver tissue confirmed these results in vivo. Characterization of portal fibroblast differentiation in culture demonstrated that these cells required transforming growth factor‐β (TGF‐β): cells remained quiescent in the presence of a TGF‐β receptor kinase inhibitor, whereas exogenous TGF‐β1 enhanced portal fibroblast α‐smooth muscle actin expression and stress fiber formation. In contrast, platelet‐derived growth factor inhibited myofibroblastic differentiation. Portal fibroblasts were also dependent on mechanical tension for myofibroblastic differentiation, and cells cultured on polyacrylamide supports of variable stiffness demonstrated an increasingly myofibroblastic phenotype as stiffness increased. Conclusion: Portal fibroblasts are morphologically and functionally distinct from hepatic stellate cells. Portal fibroblast myofibroblastic differentiation can be modeled in culture and requires both TGF‐β and mechanical tension. (HEPATOLOGY 2007.)


Purinergic Signalling | 2005

Comparative hydrolysis of P2 receptor agonists by NTPDases 1, 2, 3 and 8

Filip Kukulski; Sébastien A. Lévesque; Elise G. Lavoie; Joanna Lecka; François Bigonnesse; Aileen F. Knowles; Simon C. Robson; Terence L. Kirley; Jean Sévigny

Nucleoside triphosphate diphosphohydrolases 1, 2, 3 and 8 (NTPDases 1, 2, 3 and 8) are the dominant ectonucleotidases and thereby expected to play important roles in nucleotide signaling. Distinct biochemical characteristics of individual NTPDases should allow them to regulate P2 receptor activation differentially. Therefore, the biochemical and kinetic properties of these enzymes were compared. NTPDases 1, 2, 3 and 8 efficiently hydrolyzed ATP and UTP with Km values in the micromolar range, indicating that they should terminate the effects exerted by these nucleotide agonists at P2X1- and P2Y2,4,11 receptors. Since NTPDase1 does not allow accumulation of ADP, it should terminate the activation of P2Y1,12,13 receptors far more efficiently than the other NTPDases. In contrast, NTPDases 2, 3 and 8 are expected to promote the activation of ADP specific receptors, because in the presence of ATP they produce a sustained (NTPDase2) or transient (NTPDases 3 and 8) accumulation of ADP. Interestingly, all plasma membrane NTPDases dephosphorylate UTP with a significant accumulation of UDP, favoring P2Y6 receptor activation. NTPDases differ in divalent cation and pH dependence, although all are active in the pH range of 7.0-.5. Various NTPDases may also distinctly affect formation of extracellular adenosine and therefore adenosine receptor-mediated responses, since they generate different amounts of the substrate (AMP) and inhibitor (ADP) of ecto-5-nucleotidase, the rate limiting enzyme in the production of adenosine. Taken together, these data indicate that plasma membrane NTPDases hydrolyze nucleotides in a distinctive manner and may therefore differentially regulate P2 and adenosine receptor signaling.


Circulation Research | 2004

Targeted Disruption of cd73/Ecto-5′-Nucleotidase Alters Thromboregulation and Augments Vascular Inflammatory Response

Patrycja Koszalka; Burcin Özüyaman; Yuqing Huo; Alma Zernecke; Ulrich Flögel; Norbert Braun; Anja Buchheiser; Michael L. Smith; Jean Sévigny; Adrian R. L. Gear; Artur Aron Weber; Andrei Molojavyi; Zhaoping Ding; Christian Weber; Klaus Ley; Herbert Zimmermann; Axel Gödecke; Jürgen Schrader

To investigate the role of adenosine formed extracellularly in vascular homeostasis, mice with a targeted deletion of the cd73/ecto-5′-nucleotidase were generated. Southern blot, RT-PCR, and Western blot analysis confirmed the constitutive knockout. In vivo analysis of hemodynamic parameters revealed no significant differences in systolic blood pressure, ejection fraction, or cardiac output between strains. However, basal coronary flow measured in the isolated perfused heart was significantly lower (−14%; P<0.05) in the mutant. Immunohistochemistry revealed strong CD73 expression on the endothelium of conduit vessels in wild-type (WT) mice. Time to carotid artery occlusion after ferric chloride (FeCl3) was significantly reduced by 20% in cd73−/− mice (P<0.05). Bleeding time after tail tip resection tended to be shorter in cd73−/− mice (−35%). In vivo platelet cAMP levels were 0.96±0.46 in WT versus 0.68±0.27 pmol/106 cells in cd73−/− mice (P<0.05). Under in vitro conditions, platelet aggregation in response to ADP (0.05 to 10 &mgr;mol/L) was undistinguishable between the two strains. In the cremaster model of ischemia–reperfusion, the increase in leukocyte attachment to endothelium was significantly higher in cd73−/− compared with WT littermates (WT 98% versus cd73−/− 245%; P<0.005). The constitutive adhesion of monocytes in ex vivo–perfused carotid arteries of WT mice was negligible but significantly increased in arteries of cd73−/− mice (P<0.05). Thus, our data provide the first evidence that adenosine, extracellularly formed by CD73, can modulate coronary vascular tone, inhibit platelet activation, and play an important role in leukocyte adhesion to the vascular endothelium in vivo.


The Journal of Comparative Neurology | 2006

Nucleoside triphosphate diphosphohydrolase-2 is the ecto-ATPase of type I cells in taste buds.

Dianna L. Bartel; Susan L. Sullivan; Elise G. Lavoie; Jean Sévigny; Thomas E. Finger

The presence of one or more calcium‐dependent ecto‐ATPases (enzymes that hydrolyze extracellular 5′‐triphosphates) in mammalian taste buds was first shown histochemically. Recent studies have established that dominant ecto‐ATPases consist of enzymes now called nucleoside triphosphate diphosphohydrolases (NTPDases). Massively parallel signature sequencing (MPSS) from murine taste epithelium provided molecular evidence suggesting that NTPDase2 is the most likely member present in mouse taste papillae. Immunocytochemical and enzyme histochemical staining verified the presence of NTPDase2 associated with plasma membranes in a large number of cells within all mouse taste buds. To determine which of the three taste cell types expresses this enzyme, double‐label assays were performed with antisera directed against the glial glutamate/aspartate transporter (GLAST), the transduction pathway proteins phospholipase Cβ2 (PLCβ2) or the G‐protein subunit α‐gustducin, and serotonin (5HT) as markers of type I, II, and III taste cells, respectively. Analysis of the double‐labeled sections indicates that NTPDase2 immunoreactivity is found on cell processes that often envelop other taste cells, reminiscent of type I cells. In agreement with this observation, NTPDase2 was located to the same membrane as GLAST, indicating that this enzyme is present in type I cells. The presence of ecto‐ATPase in taste buds likely reflects the importance of ATP as an intercellular signaling molecule in this system. J. Comp. Neurol. 497:1–12, 2006.


Development | 2006

Extracellular nucleotide signaling in adult neural stem cells: synergism with growth factor-mediated cellular proliferation

Santosh K. Mishra; Norbert Braun; Varsha Shukla; Marc Füllgrabe; Christof Schomerus; Horst-Werner Korf; Christian Gachet; Yukio Ikehara; Jean Sévigny; Simon C. Robson; Herbert Zimmermann

We have previously shown that the extracellular nucleoside triphosphate-hydrolyzing enzyme NTPDase2 is highly expressed in situ by stem/progenitor cells of the two neurogenic regions of the adult murine brain: the subventricular zone (type B cells) and the dentate gyrus of the hippocampus (residual radial glia). We explored the possibility that adult multipotent neural stem cells express nucleotide receptors and investigated their functional properties in vitro. Neurospheres cultured from the adult mouse SVZ in the presence of epidermal growth factor and fibroblast growth factor 2 expressed the ecto-nucleotidases NTPDase2 and the tissue non-specific isoform of alkaline phosphatase, hydrolyzing extracellular ATP to adenosine. ATP, ADP and, to a lesser extent, UTP evoked rapid Ca2+ transients in neurospheres that were exclusively mediated by the metabotropic P2Y1 and P2Y2 nucleotide receptors. In addition, agonists of these receptors and low concentrations of adenosine augmented cell proliferation in the presence of growth factors. Neurosphere cell proliferation was attenuated after application of the P2Y1-receptor antagonist MRS2179 and in neurospheres from P2Y1-receptor knockout mice. In situ hybridization identified P2Y1-receptor mRNA in clusters of SVZ cells. Our results infer nucleotide receptor-mediated synergism that augments growth factor-mediated cell proliferation. Together with the in situ data, this supports the notion that extracellular nucleotides contribute to the control of adult neurogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2009

From the Cover: CD39 deletion exacerbates experimental murine colitis and human polymorphisms increase susceptibility to inflammatory bowel disease.

David J. Friedman; Beat M. Künzli; Yousif I. A-Rahim; Jean Sévigny; Pascal O. Berberat; Keiichi Enjyoji; Eva Csizmadia; Helmut Friess; Simon C. Robson

CD39/ENTPD1 hydrolyzes proinflammatory nucleotides to generate adenosine. As purinergic mediators have been implicated in intestinal inflammation, we hypothesized that CD39 might protect against inflammatory bowel disease. We studied these possibilities in a mouse model of colitis using mice with global CD39 deletion. We then tested whether human genetic polymorphisms in the CD39 gene might influence susceptibility to Crohns disease. We induced colitis in mice using Dextran Sodium Sulfate (DSS). Readouts included disease activity scores, histological evidence of injury, and markers of inflammatory activity. We used HapMap cell lines to find SNPs that tag for CD39 expression, and then compared the frequency of subjects with high vs. low CD39-expression genotypes in a case-control cohort for Crohns disease. Mice null for CD39 were highly susceptible to DSS injury, with heterozygote mice showing an intermediate phenotype compared to wild type (WT). We identified a common SNP that tags CD39 mRNA expression levels in man. The SNP tagging low levels of CD39 expression was associated with increased susceptibility to Crohns disease in a case-control cohort comprised of 1,748 Crohns patients and 2,936 controls (P = 0.005–0.0006). Our data indicate that CD39 deficiency exacerbates murine colitis and suggest that CD39 polymorphisms are associated with inflammatory bowel disease in humans.


European Journal of Neuroscience | 2003

Expression of the ecto-ATPase NTPDase2 in the germinal zones of the developing and adult rat brain

Norbert Braun; Jean Sévigny; Santosh K. Mishra; Simon C. Robson; Stephan W. Barth; Ruediger Gerstberger; Klaus Hammer; Herbert Zimmermann

In the adult nervous system, multipotential stem cells of the subventricular zone of the lateral ventricles generate neuron precursors (type‐A cells) that migrate via the rostral migratory stream to the olfactory bulb where they differentiate into neurons. The migrating neuroblasts are surrounded by a sheath of astrocytes (type‐B cells). Using immunostaining, in situ hybridization and enzyme histochemistry, we demonstrate that the ecto‐ATPase nucleoside triphosphate diphosphohydrolase 2 (NTPDase2) is expressed in the subventricular zone and the rostral migratory stream of the adult rat brain. This enzyme hydrolyses extracellular nucleoside triphosphates to the respective nucleoside diphosphates and is thought to directly modulate ATP receptor‐mediated cell communication. Double labelling for the astrocyte intermediate filament protein GFAP and the glial glutamate transporter GLAST identifies the NTPDase2‐positive cells as type‐B cells. During development the enzyme protein is first detected at E18, long before expression of the astrocyte marker GFAP. It gradually becomes expressed along the ventricular and subventricular zone of the brain, followed by complete retraction to the adult expression pattern at P21. NTPDase2 is transiently expressed in the outer molecular layer of the dentate gyrus and within the cerebellar white matter and is associated with select microvessels, tanycytes of the third ventricle, and subpial astrocytes of the adult brain. Our results suggest that NTPDase2 can serve as a novel marker for specifying subsets of cells during in vivo and in vitro studies of neural development and raise the possibility that ATP‐mediated signalling pathways play a role in neural development and differentiation.


British Journal of Pharmacology | 2007

Specificity of the ecto-ATPase inhibitor ARL 67156 on human and mouse ectonucleotidases

Sébastien A. Lévesque; Elise G. Lavoie; Joanna Lecka; François Bigonnesse; Jean Sévigny

ARL 67156, 6‐N,N‐Diethyl‐D‐β‐γ‐dibromomethylene adenosine triphosphate, originally named FPL 67156, is the only commercially available inhibitor of ecto‐ATPases. Since the first report on this molecule, various ectonucleotidases responsible for the hydrolysis of ATP at the cell surface have been cloned and characterized. In this work, we identified the ectonucleotidases inhibited by ARL 67156.


Neuroscience | 2006

Nucleoside triphosphate diphosphohydrolase-2 (NTPDase2/CD39L1) is the dominant ectonucleotidase expressed by rat astrocytes

Márcia R. Wink; Elizandra Braganhol; Alessandra Sayuri Kikuchi Tamajusuku; Guido Lenz; Luiz F. Zerbini; Towia A. Libermann; Jean Sévigny; Ana Maria Oliveira Battastini; Simon C. Robson

Inflammatory and degenerative pathophysiological processes within the CNS are important causes of human disease. Astrocytes appear to modulate these reactions and are a major source of inflammatory mediators, e.g. extracellular adenine nucleotides, in nervous tissues. Actions following extracellular nucleotides binding to type 2 purinergic receptors are regulated by ectonucleotidases, including members of the CD39/ecto-nucleoside triphosphate diphosphohydrolase family. The ectonucleotidases of astrocytes expressed by rat brain rapidly convert extracellular ATP to ADP, ultimately to AMP. RT-PCR, immunocytochemistry as well as Western blotting analysis demonstrated expression of multiple ecto-nucleoside triphosphate diphosphohydrolase family members at both the mRNA and protein level. By quantitative real-time PCR, we identified Entpd2 (CD39L1) as the dominant Entpd gene expressed by rat hippocampal, cortical and cerebellar astrocytes. These data in combination with the elevated ecto-ATPase activity observed in these brain regions, suggest that NTPDase2, an ecto-enzyme that preferentially hydrolyzes ATP, is the major ecto-nucleoside triphosphate diphosphohydrolase expressed by rat astrocytes. NTPDase2 may modulate inflammatory reactions within the CNS and could represent a useful therapeutic target in human disease.

Collaboration


Dive into the Jean Sévigny's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Simon C. Robson

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jamshed Iqbal

COMSATS Institute of Information Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michel Fausther

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Syeda Abida Ejaz

COMSATS Institute of Information Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elise G. Lavoie

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Herbert Zimmermann

Goethe University Frankfurt

View shared research outputs
Researchain Logo
Decentralizing Knowledge