Arnaud Foussat
French Institute of Health and Medical Research
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Arnaud Foussat.
Journal of Immunology | 2003
Arnaud Foussat; Valérie Brun; Nathalie Fournier; Jean-Philippe Breittmayer; Hervé Groux
There is now compelling evidence that CD4+CD25+ T cells play a major role in the maintenance of tolerance. Besides CD4+CD25+ T cells, different populations of regulatory CD4+ T cells secreting high amounts of IL-10 (T regulatory type 1 (Tr1)) or TGF-β (Th3) have also been described in in vivo models. In the lymphocyte transfer model of inflammatory bowel disease, we show here that the control of inflammation during the first weeks is not due to a complete inhibition of differentiation of aggressive proinflammatory T cells, but is the result of a balance between proinflammatory and Tr cells. We also show that in the first weeks continuous IL-10 secretion was required to actively control inflammation. Indeed, treatment with anti-IL-10R Abs 3 wk after the start of the experiment completely reversed the protective effect of Tr cells. IL-10 secretion and control of inflammation could be provided by late injection of Tr1 cells that efficiently cure ongoing inflammatory responses in two different models of inflammation. In contrast, inflammation was not controlled when high numbers of CD4+CD45RBlow or CD4+CD25+ T cells were injected as early as 1 wk after the start of the experiment. These results confirm in vitro studies showing that CD4+CD45RBlow do not contain high IL-10-producing cells and suggest that CD4+CD45RBlow Tr cells maintain tolerance in vivo, in part indirectly, through the differentiation of IL-10-secreting Tr1 cells.
European Journal of Immunology | 2000
Arnaud Foussat; Aurore Coulomb-L'Hermine; Jennifa Gosling; Roman Krzysiek; Ingrid Durand-Gasselin; Thomas J. Schall; Axel Balian; Yolande Richard; Pierre Galanaud; Dominique Emilie
Expression and function of the fractalkine receptor CX3CR1 by T lymphocyte subpopulations was evaluated in healthy individuals. In CD8+ T lymphocytes, CX3CR1 was expressed by and functional in both CD45RO– and CD45RO+ cells. In CD4+ T lymphocytes, CX3CR1 was expressed mainly by CD45RO+ cells, and almost exclusively by activated HLA‐DR+ T lymphocytes. This receptor was functional in CD45RO+ cells, but not in CD45RO– cells. Expression of fractalkine was detected by in situ hybridization and immunohistochemistry in endothelial cells of normal lung and thymus. In hyperplastic lymph nodes, fractalkine was expressed by endothelial cells of high endothelial venules and of subcapsular vessels, by follicular dendritic cells (FDC) and by some follicle lymphocytes. Fractalkine mRNA was constitutively present in the HK FDC‐like cell line, and it was induced in vitro in B lymphocytes stimulated by an anti‐μ or by a CD40 mAb. These findings indicate that fractalkine may contribute to the recruitment of effector T helper lymphocytes, either in peripheral tissues or in lymphoid organs. In these tissues, fractalkine and its receptor may favor contact within follicles between activated T helper lymphocytes, activated B lymphocytes and FDC, thus contributing to the maturation of the B lymphocyte response.
Journal of Immunology | 2007
Laurence Lamy; Arnaud Foussat; Eric J. Brown; Paul Bornstein; Michel Ticchioni; Alain Bernard
CD47 on the surface of T cells was shown in vitro to mediate either T cell activation or, in the presence of high amounts of thrombospondin (TSP), T cell apoptosis. We report here that CD47-deficient mice, as well as TSP-1 or TSP-2-deficient mice, sustain oxazolone-induced inflammation for more than four days, whereas wild-type mice reduce the inflammation within 48 h. We observe that prolonged inflammation in CD47-, TSP-1-, or TSP-2-deficient mice is accompanied by a local deficiency of T cell apoptosis. Finally, we show that upon activation normal T cells increase the expression of the proapoptotic Bcl-2 family member BNIP3 (Bcl-2/adenovirus E1B 19-kDa interacting protein) and undergo CD47-mediated apoptosis. This finding is consistent with our previous demonstration of a physical interaction between BNIP3 and CD47 that inhibits BNIP3 degradation by the proteasome, sensitizing T cells to CD47-induced apoptosis. Overall, these results reveal an important role in vivo for this new CD47/BNIP3 pathway in limiting inflammation by controlling the number of activated T cells.
Journal of Immunology | 2003
Karl Balabanian; Jacques Couderc; Laurence Bouchet-Delbos; Ali Amara; Dominique Berrebi; Arnaud Foussat; Françoise Baleux; Alain Portier; Ingrid Durand-Gasselin; Robert L. Coffman; Pierre Galanaud; Michel Peuchmaur; Dominique Emilie
In normal mice, stromal cell-derived factor 1 (SDF-1/CXCL12) promotes the migration, proliferation, and survival of peritoneal B1a (PerB1a) lymphocytes. Because these cells express a self-reactive repertoire and are expanded in New Zealand Black/New Zealand White (NZB/W) mice, we tested their response to SDF-1 in such mice. PerB1a lymphocytes from NZB/W mice were exceedingly sensitive to SDF-1. This greater sensitivity was due to the NZB genetic background, it was not observed for other B lymphocyte subpopulations, and it was modulated by IL-10. SDF-1 was produced constitutively in the peritoneal cavity and in the spleen. It was also produced by podocytes in the glomeruli of NZB/W mice with nephritis. The administration of antagonists of either SDF-1 or IL-10 early in life prevented the development of autoantibodies, nephritis, and death in NZB/W mice. Initiation of anti-SDF-1 mAb treatment later in life, in mice with established nephritis, inhibited autoantibody production, abolished proteinuria and Ig deposition, and reversed morphological changes in the kidneys. This treatment also counteracted B1a lymphocyte expansion and T lymphocyte activation. Therefore, PerB1a lymphocytes are abnormally sensitive to the combined action of SDF-1 and IL-10 in NZB/W mice, and SDF-1 is key in the development of autoimmunity in this murine model of lupus.
European Journal of Immunology | 2001
Arnaud Foussat; Karl Balabanian; Ali Amara; Laurence Bouchet-Delbos; Ingrid Durand-Gasselin; Françoise Baleux; Jacques Couderc; Pierre Galanaud; Dominique Emilie
B1a lymphocytes accumulate and proliferate in the peritoneal cavity. Stromal cell‐derived factor 1 (SDF‐1) is a chemotactic and growth promoting factor for B cell precursors. It is required for fetal liver B cell lymphopoiesis, which generates mostly B1a lymphocytes. Using immunohistochemistry with an anti‐SDF‐1 monoclonal antibody, we found that SDF‐1 was produced by peritoneal mesothelial cells in adult mice. Peritoneal B1a lymphocytes expressed a functional SDF‐1 receptor, as shown by actin polymerization experiments. In vitro, SDF‐1 stimulated migration, proliferation of a minority of peritoneal B1a lymphocytes, and prevented apoptosis in a large fraction of cells. B1a cells migrating in response to SDF‐1 were largely enriched in the CD5highCD43highB220–CD1d– subpopulation. In vivo, neutralization of SDF‐1 for 3 weeks significantly decreased the number of peritoneal B1 cells. SDF‐1 also acted on peritoneal B2 cells. These findings show that after the cessation of B cell lymphopoiesis in the liver, around birth, the persistence of B1a cells remains SDF‐1 dependent, and that SDF‐1 production by mesothelial cells plays a role in the peritoneal location of B1a cells. Thus, the role of mesothelial cells for B1a cells in adults may be similar to that of SDF‐1‐producing biliary ductal plate cells in the fetus, and to that of bone marrow stromal cells for B2 cell precursors.
AIDS | 1999
Weiping Zou; Arnaud Foussat; Sami Houhou; Ingrid Durand-Gasselin; Anne Dulioust; Laurence Bouchet; Pierre Galanaud; Yves Levy; Dominique Emilie
BACKGROUND The treatment of HIV-infected patients with interleukin (IL)-2 causes a sustained increase in CD4+ T-lymphocyte counts, involving both naive and memory cells. However, the short-term immunological effects of IL-2, which may shed light on the mechanism of immune reconstitution by this cytokine, are unknown. OBJECTIVE To evaluate the acute effect of IL-2 on circulating T-lymphocyte subpopulations and their expression of chemokine receptors. DESIGN AND METHODS Flow cytometry, reverse transcriptase polymerase chain reaction and chemokine receptor function experiments were performed before and after 5 days of IL-2 administration in 30 HIV-infected patients. RESULTS IL-2 induced an acute lymphopenia of both naive and memory T-helper (TH) lymphocytes. This was associated with a large increase in CC-chemokine receptor (CCR)-5 and CCR-2b expression by TH cells. Before IL-2 treatment, CCR-5 was mostly produced by CD62L- memory TH lymphocytes. After 5 days of IL-2 administration, the level of CCR-5 mRNA in circulating cells was 18.6 times higher than before treatment (P < 0.002). CCR-5 expression was upregulated in CD62L- memory TH lymphocytes, but also in CD62L+ memory and in naive (CD62L+ CD45RO-) TH lymphocytes. IL-2 treatment also increased the function of CCR-5 in TH cells. CONCLUSIONS Chemokine receptors are involved in trafficking of lymphocytes. The IL-2-induced upregulation of chemokine receptors in TH cells may thus play a role in the acute effects of this cytokine in TH lymphocyte redistribution.
International Immunopharmacology | 2009
Valérie Brun; Hervé Bastian; Virginie Neveu; Arnaud Foussat
IL-10 producing regulatory type 1 (Tr1) cells represents a subpopulation of CD4(+) regulatory cells able to prevent in vitro bystander T-cell proliferation and to cure ongoing chronic colitis in mice. In order to assess the efficacy and tolerance of Tr1 cell therapy in a Phase I/IIa clinical trial in patients displaying severe Crohns disease, we set up a reproducible manufacturing process for the GMP production of human ovalbumin specific Tr1 cells. Procedures used for Tr1-cell production include the use of Drosophila derived artificial Antigen Presenting Cells transfected with specific stimulatory molecules. Characterization of the human cell therapy product shows an in vitro suppressive activity on T-cell proliferation dependent on the production of both IL-10 and TGF-beta. Manufactured Tr1 cells display a regulatory phenotype including Foxp3, GITR and CTLA-4 surface expression. In vitro toxicity studies of human Tr1 cell product show a safety profile compatible with the use of these regulatory Tr1 lymphocytes for cell therapy.
Journal of Immunology | 2012
Cécile Chenivesse; Ying Chang; Imane Azzaoui; Saliha Ait Yahia; Olivier Morales; C. Ple; Arnaud Foussat; André-Bernard Tonnel; Nadira Delhem; Hans Yssel; Han Vorng; Benoit Wallaert; Anne Tsicopoulos
CCL18 is both a constitutively expressed and an inducible chemokine, whose role in the inflammatory reaction is poorly known. The aim of this study was to evaluate whether CCL18 has the capacity to attract human T cells with a regulatory function (regulatory T cells [Treg]). Results from chemotaxis assays performed on different types of Treg showed that CD4+CD25+CD127low cells, but neither T regulatory type 1 clones nor Treg differentiated in vitro with anti-CD3/CD46 mAbs, were recruited by CCL18 in a dose-dependent manner. CCL18-recruited memory CD4+ T cells were enriched in CD25high, CD25+CD127low, latency-associated peptide/TGF-β1, and CCR4-expressing T cells, whereas there was no enrichment in Foxp3+ cells as compared with controls. Stimulated CCL18-recruited memory T cells produced significantly increased amounts of the regulatory cytokines IL-10 and TGF-β1, as well as IL-4, but not IFN-γ and IL-17. Cell surface CCL18 binding was found predominantly on IL-10+ (26.3 ± 5.8%) and on a few latency-associated peptide/TGF-β1+ (18.1 ± 1.9%) and IL-4+ (14.5 ± 2.9%) memory T cells. In an in vivo model of SCID mice grafted with human skin and reconstituted with autologous PBMCs, the intradermal injection of CCL18 led to the cutaneous recruitment of CD4+, CD25+, and IL-10+ cells, but not Foxp3+ cells. Furthermore, CCL18-recruited memory T cells inhibited the proliferation of CD4+CD25− effector T cells through an IL-10–dependent mechanism. These data suggest that CCL18 may contribute to maintaining tolerance and/or suppressing deleterious inflammation by attracting memory Tregs into tissues, particularly in the lung, where it is highly and constitutively expressed.
International Archives of Allergy and Immunology | 2006
Massilva Rahmoun; Arnaud Foussat; Hervé Groux; Jérôme Pène; Hans Yssel; Pascal Chanez
Background: Results from a transcriptome analysis of human CD4+ T regulatory type 1 (Tr1) clones have indicated that transcripts for the integrins CD18 and CD49b are overexpressed in these cells. The aim of this study was to investigate whether the presence of T cells concomitantly expressing these molecules could be detected in asthmatic patients and represent Tr1 cells. Methods:Expression of CD18 and CD49b was analyzed by flow cytometry on peripheral blood mononuclear cells from asthmatic patients of various severity and healthy subjects. The cytokine production profile of purified CD4+ CD18high CD49b+ T cells was analyzed by ELISA. The effect of glucocorticoid treatment on the expression of CD18 and CD49b was determined. Results: The frequency of peripheral blood CD18high CD49b+ T cells was significantly elevated in severe asthmatic patients, as compared with both mild asthmatic and healthy donors, and was diminished in asthmatic patients with a controlled status of the disease. Neither short-course oral glucocorticoid treatment of asthmatic patients ex vivo, nor culture of their peripheral blood mononuclear cells with dexamethasone in vitro, increased the frequency of CD18high CD49b+ T cells, indicating that their presence seems to be independent from recent anti-inflammatory treatment. However, purified CD4+ CD18high CD49b+ T cells from these patients, in contrast to those from healthy donors, lacked the production of the immunosuppressive cytokine interleukin-10. Conclusion: In contrast to healthy donors, peripheral blood CD18high CD49b+ T cells of asthmatic patients do not fulfill the phenotypic criteria of Tr1 cells. Nevertheless, the presence of elevated numbers of peripheral blood CD18high CD49b+ T cells is characteristic for patients with severe and uncontrolled asthma.
Arthritis Research & Therapy | 2014
Hélène Asnagli; Delphine Martire; Nathalie Belmonte; Julie Quentin; Hervé Bastian; Mathilde Boucard-Jourdin; Papa Babacar Fall; Anne-Laure Mausset-Bonnefont; Amélie Mantello-Moreau; Sandrine Rouquier; Irène Marchetti; Christian Jorgensen; Arnaud Foussat; Pascale Louis-Plence
IntroductionRegulatory T (Treg) cells play a crucial role in preventing autoimmune diseases and are an ideal target for the development of therapies designed to suppress inflammation in an antigen-specific manner. Type 1 regulatory T (Tr1) cells are defined by their capacity to produce high levels of interleukin 10 (IL-10), which contributes to their ability to suppress pathological immune responses in several settings. The aim of this study was to evaluate the therapeutic potential of collagen type II–specific Tr1 (Col-Treg) cells in two models of rheumatoid arthritis (RA) in mice.MethodsCol-Treg clones were isolated and expanded from collagen-specific TCR transgenic mice. Their cytokine secretion profile and phenotype characterization were studied. The therapeutic potential of Col-Treg cells was evaluated after adoptive transfer in collagen-antibody– and collagen-induced arthritis models. The in vivo suppressive mechanism of Col-Treg clones on effector T-cell proliferation was also investigated.ResultsCol-Treg clones are characterized by their specific cytokine profile (IL-10highIL-4negIFN-γint) and mediate contact-independent immune suppression. They also share with natural Tregs high expression of GITR, CD39 and granzyme B. A single infusion of Col-Treg cells reduced the incidence and clinical symptoms of arthritis in both preventive and curative settings, with a significant impact on collagen type II antibodies. Importantly, injection of antigen-specific Tr1 cells decreased the proliferation of antigen-specific effector T cells in vivo significantly.ConclusionsOur results demonstrate the therapeutic potential of Col-Treg cells in two models of RA, providing evidence that Col-Treg could be an efficient cell-based therapy for RA patients whose disease is refractory to current treatments.