Network


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

Hotspot


Dive into the research topics where Christiane Mousson is active.

Publication


Featured researches published by Christiane Mousson.


Transplantation | 2003

Detection of donor-specific anti-HLA antibodies with flow cytometry in eluates and sera from renal transplant recipients with chronic allograft nephropathy.

Laurent Martin; Fredy Guignier; Christiane Mousson; David Rageot; Eve Justrabo; Gérard Rifle

Background. Chronic allograft nephropathy (CAN), which remains the main cause of graft loss after kidney transplantation, is still poorly understood. Because anti-HLA antibodies may be involved in the pathogenesis of CAN, this study was performed to look for donor-specific antibodies (DSA) fixed onto renal transplants with CAN. Methods. DSA were identified after elution with flow cytometric assay and/or flow cytometric crossmatches in 20 transplants removed after irreversible graft failure caused by CAN and in control samples from 2 transplants with relapsing glomerulopathy, 2 transplants lost after vascular thrombosis, and 4 normal kidneys. The results were compared with those obtained in the serum samples 1 year after grafting, at the time of transplantectomy, and within 2 months after transplantectomy. Results. IgG anti-class I, anti-class II, or both DSA were identified in 70.6% of eluates versus 73.6% of posttransplantectomy serum samples (NS), 42.1% of 1-year postgrafting serum samples (P <0.05), and 31.6% of serum samples at the time of transplantectomy (P <0.05). Our data show a good correlation between the target of anti-HLA antibodies found in both eluates and posttransplantectomy serum samples, but the precise specificity of anti-HLA antibodies is more often assigned in posttransplantectomy serum samples than in eluates. This problem needs further evaluation. Conclusion. This study shows that testing for anti-HLA DSA in eluates from removed kidney transplants using flow cytometry can be achieved and is highly efficient. It already suggests that both anti-class I and anti-class II HLA antibodies can be involved in CAN. Further studies are now needed to evaluate the possibility of identifying such antibodies in the eluates of transplant biopsy specimens from recipients experiencing CAN.


American Journal of Transplantation | 2011

Autoantibodies Specific for the Phospholipase A2 Receptor in Recurrent and De Novo Membranous Nephropathy

H. Debiec; L. Martin; Chantal Jouanneau; G. Dautin; Laurent Mesnard; Eric Rondeau; Christiane Mousson; Pierre Ronco

Recent findings in idiopathic membranous nephropathy (MN) suggest that in most patients, the disease is because of anti‐phospholipase A2 receptor (PLA2R1) autoantibodies. Our aim was to analyze the prevalence and significance of anti‐PLA2R1 antibodies in recurrent and de novo MN after transplantation. We assessed circulating PLA2R1 autoantibodies by a direct immunofluorescence assay based on human embryonic kidney cells transfected with a PLA2R1 cDNA, and the presence of PLA2R1 antigen in immune deposits. We showed that PLA2R1 was involved in 5 of 10 patients with recurrent MN, but in none of the 9 patients with de novo MN. We also showed a marked heterogeneity in the kinetics and titers of anti‐PLA2R1, which may relate to different pathogenic potential. We provide evidence that some patients with PLA2R1‐related idiopathic MN and anti‐PLA2R1 antibodies at the time of transplantation will not develop recurrence. Because PLA2R1 autoantibody was not always associated with recurrence, its predictive value should be carefully analyzed in prospective studies.


Transplantation | 2005

Donor-specific antibodies in allograft rejection: clinical and experimental data.

Gérard Rifle; Christiane Mousson; Laurent Martin; Fredy Guignier; Kais Hajji

The ability of donor-specific major histocompatibility complex alloantibodies to destroy a transplanted organ within minutes, the so-called hyperacute rejection phenomenon, has been known for a long time. It is a clear demonstration of the possible cytotoxic effect of antibodies. Apart from this particular situation, the role of antibodies in inducing acute or chronic allograft rejection remains controversial. Many clinical data have shown that transplant recipients capable of developing class I or class II anti-HLA antibodies experienced shorter survival periods than those who were not. This fact, in accordance with experimental data, only demonstrates that high antibody responders reject a transplant more easily than low responders. More interestingly, there is now increasing evidence that posttransplant appearance of donor-specific alloantibodies, and probably of alloreactive-induced autoantibodies, is strongly correlated with reduced graft survival rate, especially from chronic rejection. We demonstrated that donor-specific HLA antibodies can be found in more than 70% of transplanted kidneys with chronic allograft nephropathy, and that the intragraft presence of such antibodies is significantly correlated with high numbers of plasma cells on early biopsies and C4d deposits, a recognized marker of humoral rejection. It is likely that non-HLA antibodies also play a deleterious role in organ transplant outcome, particularly the heterogeneous group of anti-endothelial cells antibodies, anti-MIC antibodies, autoantibodies and some others with no recognized target. Convincing experimental data, especially using B cell and T cell deficient mice, strongly suggest that B cells and donor-specific antibodies are required for fully developed chronic allograft rejection. The role of antibodies in inducing the cascade of cytokines and growth factors leading to tissue lesions is of increasing interest since it is now possible to control B cell proliferation and antibody production.


Journal of Clinical Oncology | 2013

Post-Transplantation Lymphoproliferative Disorder After Kidney Transplantation: Report of a Nationwide French Registry and the Development of a New Prognostic Score

Sophie Caillard; Raphael Porcher; François Provôt; Jacques Dantal; Sylvain Choquet; Antoine Durrbach; Emmanuel Morelon; Valérie Moal; Bénédicte Janbon; Eric Alamartine; Claire Pouteil Noble; Delphine Morel; Nassim Kamar; M. Buchler; Marie Noelle Peraldi; Christian Hiesse; Edith Renoult; Olivier Toupance; Jean Philippe Rerolle; Sylvie Delmas; Philippe Lang; Yvon Lebranchu; Anne Elisabeth Heng; Jean Michel Rebibou; Christiane Mousson; Joseph Rivalan; Antoine Thierry; Isabelle Etienne; Marie Christine Moal; Laetitia Albano

PURPOSE Post-transplantation lymphoproliferative disorder (PTLD) is associated with significant mortality in kidney transplant recipients. We conducted a prospective survey of the occurrence of PTLD in a French nationwide population of adult kidney recipients over 10 years. PATIENTS AND METHODS A French registry was established to cover a nationwide population of transplant recipients and prospectively enroll all adult kidney recipients who developed PTLD between January 1, 1998, and December 31, 2007. Five hundred patient cases of PTLD were referred to the French registry. The prognostic factors for PTLD were investigated using Kaplan-Meier and Cox analyses. RESULTS Patients with PTLD had a 5-year survival rate of 53% and 10-year survival rate of 45%. Multivariable analyses revealed that age > 55 years, serum creatinine level > 133 μmol/L, elevated lactate dehydrogenase levels, disseminated lymphoma, brain localization, invasion of serous membranes, monomorphic PTLD, and T-cell PTLD were independent prognostic indicators of poor survival. Considering five variables at diagnosis (age, serum creatinine, lactate dehydrogenase, PTLD localization, and histology), we constructed a prognostic score that classified patients with PTLD as being at low, moderate, high, or very high risk for death. The 10-year survival rate was 85% for low-, 80% for moderate-, 56% for high-, and 0% for very high-risk recipients. CONCLUSION This nationwide study highlights the prognostic factors for PTLD and enables the development of a new prognostic score. After validation in an independent cohort, the use of this score should allow treatment strategies to be better tailored to individual patients in the future.


Transplantation | 2004

Ultraviolet light-induced regulatory (suppressor) T cells: An approach for promoting induction of operational allograft tolerance?

F. Aubin; Christiane Mousson

Ultraviolet (UV) light is known to induce skin cancers by causing DNA gene mutations and inducing immunosuppression. Taking advantage of these immunosuppressive capacities, UV light has been used, with different modalities, as an immunosuppressive therapy in a variety of diseases including allograft rejection and graft-versus-host disease. Phototherapy includes UVB irradiation, UVA irradiation, oral psoralen+UVA irradiation (PUVA), photodynamic therapy, and extracorporeal photopheresis, which consists of infusion of UVA-irradiated autologous leukocytes collected by apheresis and incubated with 8-methoxypsoralen. According to numerous experimental models and human data, there is increasing evidence that UVB irradiation and extracorporeal photopheresis can induce regulatory T cells and anticlonotypic activity. These therapies induce apoptosis of activated T cells or of extracorporally treated mononuclear cells, and up-regulate the expression of costimulary molecules and adhesion molecules on antigen presenting cells. UVB- or UVA-induced apoptotic cells could secrete immune suppressive cytokines (interleukin (IL)-4, IL-10). The processing and presentation of apoptotic T cell antigens from clones of pathogenic T cells by activated antigen presenting cells might explain the induction of systemic anticlonotypic activity by photopheresis. This induction of cell-mediated suppressive activity opens up future prospects with the aim of expanding regulatory T cells and/or anticlonotypic activity, especially by photopheresis in organ and cell transplantation.


Journal of The American Society of Nephrology | 2017

Rituximab for Severe Membranous Nephropathy: A 6-Month Trial with Extended Follow-Up

Karine Dahan; Hanna Debiec; Emmanuelle Plaisier; Marine Cachanado; Alexandra Rousseau; Laura Wakselman; P.A. Michel; Fabrice Mihout; Bertrand Dussol; Marie Matignon; Christiane Mousson; Tabassome Simon; Pierre Ronco

Randomized trials of rituximab in primary membranous nephropathy (PMN) have not been conducted. We undertook a multicenter, randomized, controlled trial at 31 French hospitals (NCT01508468). Patients with biopsy-proven PMN and nephrotic syndrome after 6 months of nonimmunosuppressive antiproteinuric treatment (NIAT) were randomly assigned to 6-month therapy with NIAT and 375 mg/m2 intravenous rituximab on days 1 and 8 (n=37) or NIAT alone (n=38). Median times to last follow-up were 17.0 (interquartile range, 12.5-24.0) months and 17.0 (interquartile range, 13.0-23.0) months in NIAT-rituximab and NIAT groups, respectively. Primary outcome was a combined end point of complete or partial remission of proteinuria at 6 months. At month 6, 13 (35.1%; 95% confidence interval [95% CI], 19.7 to 50.5) patients in the NIAT-rituximab group and eight (21.1%; 95% CI, 8.1 to 34.0) patients in the NIAT group achieved remission (P=0.21). Rates of antiphospholipase A2 receptor antibody (anti-PLA2R-Ab) depletion in NIAT-rituximab and NIAT groups were 14 of 25 (56%) and one of 23 (4.3%) patients at month 3 (P<0.001) and 13 of 26 (50%) and three of 25 (12%) patients at month 6 (P=0.004), respectively. Eight serious adverse events occurred in each group. During the observational phase, remission rates before change of assigned treatment were 24 of 37 (64.9%) and 13 of 38 (34.2%) patients in NIAT-rituximab and NIAT groups, respectively (P<0.01). Positive effect of rituximab on proteinuria remission occurred after 6 months. These data suggest that PLA2R-Ab levels are early markers of rituximab effect and that addition of rituximab to NIAT does not affect safety.


Transplantation | 2003

Donor-derived hematopoietic cells in organ transplantation: a major step toward allograft tolerance?

Gérard Rifle; Christiane Mousson

Infusion of donor-derived cells can improve organ allograft survival in animal models. Under certain conditions, it can even induce tolerance (i.e., unlimited organ survival without any maintenance immunosuppressive therapy). Use of nonmyeloablative regimens allows engraftment of donor-derived bone marrow cells, induction of mixed chimerism, and tolerance in rodents. High doses of bone marrow cells together with anti–T-cell antibodies can even result in mixed chimerism without cytoablative host conditioning. Cultured donor-derived CD34+ cells or donor-derived immature (or even mature) dendritic cells associated with monoclonal antibodies directed against co-stimulatory molecules might also induce tolerance. Among the numerous experimental protocols leading to tolerance of solid organs in animal models, how can we find our bearings in human transplantation? Numerous problems have yet to be solved: the type and amount of donor-derived cells (including stromal cells) to be used, the timing for infusion of donor cells in keeping with organ transplantation, the route of infusion (should it be intravenous, into the portal vein?), and the conditioning regimen. The first clinical trials would appear to indicate that tolerance induction in humans using donor-derived cells is a relatively safe solution that is both promising and realistic.


American Journal of Transplantation | 2006

Successful Hepatorenal Transplantation in Hereditary Amyloidosis Caused by a Frame‐Shift Mutation in Fibrinogen Aα‐Chain Gene

Christiane Mousson; Bruno Heyd; Eve Justrabo; Jean-Michel Rebibou; Yves Tanter; Jean-Philippe Miguet; Gérard Rifle

Hereditary systemic amyloidosis comprises several autosomal dominant diseases caused by mutations in a number of plasma proteins, including the fibrinogen Aα‐chain. Four mutations in the fibrinogen Aα‐chain that are able to induce amyloidosis have been identified so far, the most common being the Glu526Val mutation. We have observed a family in which the father and his son reached end‐stage renal failure because of renal amyloidosis induced by a frame‐shift mutation in the fibrinogen Aα‐chain gene producing a novel amyloid protein. Two kidney transplantations in the father and one in the son resulted in fast graft loss caused by recurrence of amyloid deposition. We then performed hepatorenal transplantation in the son. Three years later, liver and kidney functions are normal without recurrence of amyloid deposition. This case, together with three others with the Glu526Val mutation in the extensive literature, suggests that liver transplantation can cure hereditary fibrinogen amyloidosis, whatever the mutation may be.


Transplantation | 1999

Foscarnet-induced crystalline glomerulonephritis with nephrotic syndrome and acute renal failure after kidney transplantation.

Gilbert Zanetta; Laurence Maurice-Estepa; Christiane Mousson; Eve Justrabo; Michel Daudon; Gérard Rifle; Yves Tanter

Foscarnet nephrotoxicity has been reported to be associated with acute tubulointerstitial nephritis. Crystals in glomerular capillary lumens have also been observed in patients with acquired immunodeficiency syndrome who were treated with foscarnet for cytomegalovirus disease. We describe a kidney transplant recipient who developed a nephrotic syndrome with microscopic hematuria and nonoliguric acute renal failure within 15 days after starting foscarnet therapy for cytomegalovirus infection. A kidney biopsy specimen showed the presence of crystals in all glomeruli and in proximal tubules. Fourier transform infrared microscopy analysis demonstrated that crystals were made from several forms of foscarnet salts: mixed calcium and sodium salts, and unchanged trisodium foscarnet salts. Renal function and proteinuria spontaneously improved, and a second transplant biopsy performed 8 months after the first one revealed fibrotic organization of half of the glomeruli and of interstitial tissue, and crystal vanishing. We were thus able to provide proof of the possible precipitation of foscarnet in a transplanted kidney.


Transplantation | 2011

Early steroid withdrawal and optimization of mycophenolic acid exposure in kidney transplant recipients receiving mycophenolate mofetil.

Yannick Le Meur; Antoine Thierry; François Glowacki; Jean-Philippe Rerolle; Valérie Garrigue; Nacera Ouali; Anne-Elisabeth Heng; Michel Delahousse; Laeticia Albano; Philippe Lang; Lionel Couzi; Maïté Jauréguy; Yvon Lebranchu; Christiane Mousson; Michèle Kessler; François Vrtovsnik; Stephanie Rouanet; Nailya Tagieva; Nassim Kamar

Background. Early posttransplant steroid withdrawal may increase the risk of acute rejection and the occurrence of subclinical acute rejection (SCAR). We assessed the feasibility and safety of early steroid withdrawal in low-risk patients receiving cyclosporine A (CsA) and the impact of optimization of mycophenolic acid exposure on steroid withdrawal success. Methods. De novo, low-immunological risk kidney recipients received an anti-interleukin-2-receptor-&agr; antibody induction, a short course of 7 days of corticosteroids, and CsA with 2-hr postdose concentration monitoring. They were randomized to adjusted dose (AD) of mycophenolate mofetil (MMF) using therapeutic drug monitoring (TDM) or a fixed-dose (FD) regimen. MMF 3 g was initiated posttransplant and then adjusted starting at week 2 to a 0 to 12 hr area under the concentration time curve of 40 mg · h/L versus 2 g daily, respectively. The primary endpoint was a composite of the proportion of patients experiencing biopsy-proven acute rejection (BPAR) and those with SCAR identified on the 3-month protocol biopsy. Results. Among 247 analyzed patients, only 22 in the AD group and 17 in the FD group experienced BPAR or SCAR (P=0.46). The rate of SCAR was low: 4% (AD) and 2.5% (FD). No between-group difference in the incidence of BPAR was observed. TDM yielded MMF doses ranging from 1 to 4 g/d and significantly reduced interpatient variability at weeks 26 and 52 in the AD group. Conclusions. In low-immunological risk kidney recipients, MMF combined with CsA allows early corticosteroid discontinuation with good tolerability. In this group of patients, TDM of MMF does not improve clinical outcome.

Collaboration


Dive into the Christiane Mousson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claire Presne

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

Paul Coppo

Institut Gustave Roussy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

D. Krausé

University of Burgundy

View shared research outputs
Top Co-Authors

Avatar

Jean-Michel Halimi

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

M. Buchler

François Rabelais University

View shared research outputs
Researchain Logo
Decentralizing Knowledge