Network


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

Hotspot


Dive into the research topics where Marie-Christophe Boissier is active.

Publication


Featured researches published by Marie-Christophe Boissier.


Joint Bone Spine | 2010

Vitamin D and inflammation.

Xavier Guillot; Luca Semerano; Nathalie Saidenberg-Kermanac’h; Géraldine Falgarone; Marie-Christophe Boissier

Calcitriol, or 1,25-dihydroxyvitamin D3 (1,25(OH)(2)D3) is a well-known endocrine regulator of calcium homeostasis. More recently, local calcitriol production by immune cells was shown to exert autocrine or paracrine immunomodulating effects. Immune cells that produce calcitriol also express the vitamin D receptor (VDR) and the enzymes needed to metabolize vitamin D3 (1α-, 25-, and 24-hydroxylases). Studies of animal models and cell cultures showed both direct and indirect immunomodulating effects involving the T cells, B cells, and antigen-presenting cells (dendritic cells and macrophages) and affecting both innate and adaptive immune responses. The overall effect is a switch from the Th1/Th17 response to the Th2/Treg profile. The immunomodulating effects of vitamin D may explain the reported epidemiological associations between vitamin D status and a large number of autoimmune and inflammatory diseases. Such associations have been suggested by observational studies not only in rheumatoid arthritis, lupus, inflammatory bowel disease, and type 1 diabetes; but also in infections, malignancies, transplant rejection, and cardiovascular disease. In animal models for these diseases, vitamin D supplementation has been found to produce therapeutic effects. Thus, vitamin D is a key focus for public health efforts and may hold promise for the treatment of dysimmune diseases.


Journal of Autoimmunity | 2012

Rheumatoid arthritis: from autoimmunity to synovitis and joint destruction.

Marie-Christophe Boissier; Luca Semerano; Salima Challal; Nathalie Saidenberg-Kermanach; Géraldine Falgarone

Rheumatoid arthritis is an autoimmune disease characterized by the production of two known antibodies - rheumatoid factor and anti-citrullinated peptide antibody (ACPA) - against common autoantigens that are widely expressed within and outside the joints. The interactions between genes and environment are crucial in all stages of the disease, involving namely genes from major histocompatibility complex locus, and antigens such as tobacco or microbes (e.g. Porphyromonas gingivalis). T and B cells are activated as soon as the earliest phases of the disease, rheumatoid arthritis appearing as a Th1 and Th17 disease. Inflammatory cytokines have a considerable importance in the hierarchy of the processes involved in RA. The joint destruction seen in RA is caused not only by cytokine imbalances, but also by specific effects of the Wnt system and osteoprotegerin on osteoclasts and by matrix production dysregulation responsible for cartilage damage. Both innate and adaptative immunity demonstrated their respective cornerstone position in rheumatoid arthritis, since targeted treatments has been efficiently developed against TNF-α, IL-6 receptor, IL-1β, CD20 B cells and T-cell/Dendritic cell interactions.


Annals of Medicine | 2011

Interleukin-23: A key cytokine in inflammatory diseases

Emilie Duvallet; Luca Semerano; Eric Assier; Géraldine Falgarone; Marie-Christophe Boissier

Abstract Interleukin-23 (IL-23) is a pro-inflammatory cytokine composed of two subunits, p19 and p40. The p40 subunit is shared with IL-12. IL-23 and IL-12 have different receptors and different effects. Whereas IL-12 induces development of Th1 cells, which produce interferon-γ, IL-23 is involved in differentiation of Th17 cells in a pro-inflammatory context and especially in the presence of TGF-β and IL-6. Activated Th17 cells produce IL-17A, IL-17F, IL-6, IL-22, TNF-α, and GM-CSF. Inflammatory macrophages express IL-23R and are activated by IL-23 to produce IL-1, TNF-α, and IL-23 itself. These effects identify IL-23 as a central cytokine in autoimmunity and a highly promising treatment target for inflammatory diseases. IL-23 is found in the skin of patients with psoriasis, in the bowel wall of patients with chronic inflammatory bowel disease, and in synovial membrane of patients with rheumatoid arthritis. IL-23 is involved in osteoclastogenesis, independently from IL-17, via induction of RANKL expression. Debate continues to surround the role for IL-23 in the pathophysiology of inflammatory joint diseases (rheumatoid arthritis and spondyloarthritis). Ustekinumab, which inhibits IL-12 and IL-23 by blocking p40, has been found effective in cutaneous psoriasis and psoriatic arthritis, as well as in Crohns disease. Treatments that specifically target IL-23 (antibodies to p19) are being developed.


Annals of the Rheumatic Diseases | 1987

Experimental autoimmune arthritis in mice. I. Homologous type II collagen is responsible for self-perpetuating chronic polyarthritis.

Marie-Christophe Boissier; X Z Feng; A Carlioz; R Roudier; C Fournier

Immunisation with heterologous type II collagen (CII) induces arthritis in mice of the DBA/1 strain, which is genetically susceptible to this disease. To develop an experimental model of autoimmunity more adequate for the study of human rheumatoid arthritis (RA), DBA/1 mice were injected with 100 micrograms of native CII that had been purified from mouse xiphoid cartilage. About six weeks later the animals developed a chronic progressive polyarthritis involving the four paws but mainly confined to interphalangeal and metatarsophalangeal joints. The evolution of the disease fluctuated between remissions and exacerbations. The initial lesions assessed by clinical observations were more severe when the disease occurred early than in the case of late onset. Interestingly, the incidence of arthritis was clearly preponderant in males, and, moreover, the few female mice which developed arthritis had mild disease states with lower arthritic scores than the males. Varying levels of autoantibodies against mouse CII were found in the sera of immunised animals, regardless of the development of arthritis. These data indicate that the injection of homologous CII into mice caused a polyarthritis that is clinically closer to the human RA than the disease induced with heterologous CII and therefore will represent a useful tool for the study of the self-perpetuating mechanisms that characterise RA.


Joint Bone Spine | 2009

Regulatory T cells (Treg) in rheumatoid arthritis

Marie-Christophe Boissier; Eric Assier; Jérôme Biton; Anne Denys; Géraldine Falgarone; Natacha Bessis

Modulation of the T-cell response depends chiefly on regulatory T cells (Treg), which express CD4 and CD25. Some Treg cells are present naturally, whereas others are induced in response to antigens. The immunomodulating effects of Treg cells are mediated by membrane molecules (e.g., CTLA4, GITR, and OX40) and cytokines. IL-35 seems to be a crucial mediator, although IL-10 and TGFbeta are also important. The role for Treg cells in rheumatoid arthritis (RA) has been established in both patients and animal models. Treg function is deficient in RA, whereas Treg counts vary. Treg counts increase in patients who are responding to TNFalpha antagonist therapy. Among current hypotheses, Treg expansion or transfer may hold promise for the treatment of RA.


European Journal of Immunology | 2000

The type II decoy receptor of IL‐1 inhibits murine collagen‐induced arthritis

Natacha Bessis; Laurence Guéry; Alberto Mantovani; Annunciata Vecchi; John E. Sims; Didier Fradelizi; Marie-Christophe Boissier

IL‐1 is a key cytokine involved in the inflammatory response. The type II receptor of IL‐1 (IL‐1RII) acts as a decoy receptor, binding and inhibiting the effect of IL‐1. This study was undertaken to establish whether IL‐1RII can ameliorate collagen‐induced arthritis, a model of inflammatory arthritis in mice. We used human keratinocytes transfected with the human (h)IL‐1RII gene as a source of hIL‐1RII protein. We showed that these cells expressed both the membrane and soluble form of receptor. In vitro, IL‐1‐stimulated murine macrophage cells showed a decreased expression of TNF‐α in the presence of hIL‐1RII. We engrafted the hIL‐1RII‐transfected cells in the back of mice developing collagen‐induced arthritis. We found that clinical and histological parameters of arthritis were significantly decreased in mice treated with cells producing hIL‐1RII. In addition, hIL‐1RII administration was able to reduce the expression of mRNA for IL‐6 and myeloperoxidase in the joints of treated animals. These data show that hIL‐1RII anti‐inflammatory properties in the model of collagen‐induced arthritis in mice and could have a regulatory role in rheumatoid arthritis.


Proceedings of the National Academy of Sciences of the United States of America | 2006

TNFalpha kinoid vaccination-induced neutralizing antibodies to TNFalpha protect mice from autologous TNFalpha-driven chronic and acute inflammation.

Hélène Le Buanec; Laure Delavallée; Natacha Bessis; Sébastien Paturance; Bernard Bizzini; Robert C. Gallo; Daniel Zagury; Marie-Christophe Boissier

The proinflammatory cytokine TNFα is a potent mediator of septic shock and a therapeutic target for chronic inflammatory pathologies including rheumatoid arthritis and Crohns disease. As an alternative to anti-human TNFα (hTNFα) mAbs and other hTNFα blocker approved drugs, we developed an active anti-hTNFα immunotherapy, based on a vaccine comprised of a keyhole limpet hemocyanin-hTNFα heterocomplex immunogen (hTNFα kinoid) adjuvanted in incomplete Freunds adjuvant. In mice transgenic for hTNFα (TTg mice), hTNFα kinoid vaccination elicited high titers of Abs that neutralized hTNFα bioactivities but did not result in a cellular response to hTNFα. The vaccine was safe and effective in two experimental models. Kinoid-immunized but not control TTg mice resisted hTNFα-driven shock in one model and were prevented from spontaneous arthritis, inflammatory synovitis, and articular destruction in a second model. These data demonstrate an anti-cytokine induction of autoimmune protection against both acute and chronic hTNFα exposure. They show that active vaccination against a human cytokine can be achieved, and that the immune response can be effective and safe.


Joint Bone Spine | 2003

Recent data on the role for angiogenesis in rheumatoid arthritis

Gaëlle Clavel; Natacha Bessis; Marie-Christophe Boissier

Angiogenesis is central to the development and perpetuation of rheumatoid synovitis. Vascular endothelial growth factor (VEGF), the main mediator of angiogenesis, is found in the synovial fluid and serum of patients with rheumatoid arthritis (RA), and its expression is correlated with disease severity. Compelling evidence that VEGF is involved in synovitis has been obtained from experimental models of RA. In particular, VEGF inhibition by synthetic compounds (e.g. TNP-470) or by naturally occurring factors (e.g., the soluble VEGF receptor) produce therapeutic effects. Angiopoietin-1, a recently discovered growth factor specific for neovascularization, is expressed within the rheumatoid synovium and may be stimulated by TNF-alpha. Other compounds, including integrins, fibroblast growth factor, and proinflammatory cytokines contribute to joint angiogenesis and, therefore, to the development of rheumatoid synovitis. Assessing vascularity may prove useful for evaluating or even predicting bone destruction. Furthermore, inhibition of angiogenesis may prove useful as an adjunct to current anti-inflammatory treatments.


Journal of Gene Medicine | 2002

Gene therapy for rheumatoid arthritis

Natacha Bessis; Christelle Doucet; Virginie Cottard; Anne‐Marie Douar; Hüseyin Firat; Christian Jorgensen; Mauro Mezzina; Marie-Christophe Boissier

Rheumatoid arthritis (RA) is a severe autoimmune systemic disease. Chronic synovial inflammation results in destruction of the joints. No conventional treatment is efficient in RA. Gene therapy of RA targets mainly the players of inflammation or articular destruction: TNF‐α or IL‐1 blocking agents (such as anti‐TNF‐α monoclonal antibodies, soluble TNF‐α receptor, type II soluble receptor of IL‐1, IL‐1 receptor antagonist), antiinflammatory cytokines (such as IL‐4, IL‐10, IL‐1), and growth factors. In this polyarticular disease, the vector expressing the therapeutic protein can be administered as a local (intra‐articular injection) or a systemic treatment (extra‐articular injection). All the main vectors have been used in experimental models, including the more recent lentivirus and adeno‐associated virus. Ex vivo gene transfer was performed with synovial cells, fibroblasts, T cells, dendritic cells, and different cells from xenogeneic origin. In vivo gene therapy is simpler, although a less controlled method. Clinical trials in human RA have started with ex vivo retrovirus‐expressing IL‐1 receptor antagonists and have demonstrated the feasibility of the strategy of gene therapy. The best target remains to be determined and extensive research has to be conducted in preclinical studies. Copyright


Human Gene Therapy | 2004

Gene Therapy of Collagen-Induced Arthritis by Electrotransfer of Human Tumor Necrosis Factor-α Soluble Receptor I Variants

Carole Bloquel; Natacha Bessis; Marie-Christophe Boissier; Daniel Scherman; Pascal Bigey

Electrotransfer is a simple and efficient strategy of nonviral gene delivery. We have used this method to deliver plasmids encoding three human tumor necrosis factor-alpha soluble receptor I variants (hTNFR-Is) a monomeric hTNFR-Is, a chimeric hTNFR-Is/mIgG1, and a dimeric (hTNFR-Is)(2) form. Electrotransfer parameters were studied and because anti-TNF strategies have proven efficient for the treatment of rheumatoid arthritis in clinics, we used a collagen-induced arthritis (CIA) mouse model to assess the efficacy of our constructs in the treatment of the disease. All proteins were proven bioactive, both in vitro and ex vivo. Plasmid intramuscular electrotransfer in mice resulted in a local expression of the three variants for at least 6 months; systemic expression lasted also more than 6 months for the hTNFR-Is/mIgG1 form, while it was shorter for the two other forms. This expression was plasmid dose-dependent. Electrotransfer of 50 microg of hTNFR-Is/mIgG1 at the onset of a CIA induced a clear-cut decrease in both clinical and histologic signs of the disease; the dimeric form also showed some efficacy. Moreover, the long-lasting protective effect was observed for more than 5 weeks. Comparison of this electrotransfer approach with repeated recombinant protein (etanercept) injections highlighted the potential practical interest of gene therapy approach for CIA, which leads to sustained therapeutic effect after single treatment. These results show that electrotransfer may be a useful method to deliver cytokine or anticytokine therapy in rheumatoid arthritis and also illustrate the potentiality of plasmid intramuscular electrotransfer for the rapid screening and assessment of different variant forms of secreted proteins.

Collaboration


Dive into the Marie-Christophe Boissier'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge