Network


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

Hotspot


Dive into the research topics where Bernard Martinet is active.

Publication


Featured researches published by Bernard Martinet.


Journal of Immunology | 2008

Myeloid-Derived Suppressor Cells Accumulate in Kidney Allograft Tolerance and Specifically Suppress Effector T Cell Expansion

Anne-Sophie Dugast; Thomas Haudebourg; Flora Coulon; Michèle Heslan; Fabienne Haspot; Nicolas Poirier; Romain Vuillefroy de Silly; Claire Usal; Helga Smit; Bernard Martinet; Pamela Thebault; Karine Renaudin; Bernard Vanhove

The immune tolerance to rat kidney allografts induced by a perioperative treatment with anti-CD28 Abs is associated with a severe unresponsiveness of peripheral blood cells to donor Ags. In this model, we identified an accumulation in the blood of CD3−class II−CD11b+CD80/86+ plastic-adherent cells that additionally expressed CD172a as well as other myeloid markers. These cells were able to inhibit proliferation, but not activation, of effector T cells and to induce apoptosis in a contact-dependent manner. Their suppressive action was found to be under the control of inducible NO synthase, an enzyme also up-regulated in tolerated allografts. Based on these features, these cells can be defined as myeloid-derived suppressor cells (MDSC). Interestingly, CD4+CD25highFoxP3+ regulatory T cells were insensitive in vitro to MDSC-mediated suppression. Although the adoptive transfer of MDSC failed to induce kidney allograft tolerance in recently transplanted recipients, the maintenance of tolerance after administration of anti-CD28 Abs was found to be dependent on the action of inducible NO synthase. These results suggest that increased numbers of MDSC can inhibit alloreactive T cell proliferation in vivo and that these cells may participate in the NO-dependent maintenance phase of tolerance.


Science Translational Medicine | 2010

Inducing CTLA-4–Dependent Immune Regulation by Selective CD28 Blockade Promotes Regulatory T Cells in Organ Transplantation

Nicolas Poirier; Agnes M. Azimzadeh; T. Zhang; Nahzli Dilek; Caroline Mary; B.N. Nguyen; Xavier Tillou; Guosheng Wu; Karine Reneaudin; Jeremy Hervouet; Bernard Martinet; Flora Coulon; Emma Allain-Launay; Georges Karam; Jean-Paul Soulillou; Richard N. Pierson; Gilles Blancho; Bernard Vanhove

An improved method of immunosuppression allows better immune function and prolongs the survival of transplanted organs in nonhuman primates. Increasing Tolerance with Less Toxicity According to Chinese legend, 2500 years ago the physician Pien Ch’iao exchanged the hearts of two warriors, one of strong spirit and weak will and the other of weak spirit and strong will, with the aim of achieving balance in their characters. Of course, without modern surgical techniques and the ability to suppress organ rejection by the recipient’s immune system, such transplants would have been impossible. It was not until the discovery of the powerful immunosuppressant drug cyclosporin that organ transplants between unrelated individuals became routinely successful, beginning in the 1980s. Cyclosporin and some newer similar drugs, however, have toxic side effects, and none of them can stop eventual rejection of the organ, so researchers are seeking other options. Now, Vanhove and colleagues test an alternative immunosuppressive strategy. T cells are a major culprit in the immune response directed against transplanted organs, and therefore a number of immunosuppressive drugs target T cells. Cyclosporin, for example, inhibits calcineurin, a protein phosphatase that under normal conditions increases the expression of interleukin 2, which—among other functions—promotes the proliferation of activated T cells. Certain newer approaches to immunosuppression target the process by which T cells become activated. In general, this process is induced by antigen binding to the T cell receptor and reinforced by costimulatory molecules—CD80 and CD86—binding to the T cell CD28 receptor. Additionally, CD80/86 can bind to the CTLA-4 receptor, resulting in T cell inhibition. CTLA-4 is also required for the function of regulatory T cells (Tregs), which suppress aberrant immune responses and are important for inducing tolerance toward transplanted tissue. Several drugs that block CD80/86 have been developed as a nontoxic alternative to calcineurin inhibitors, but in addition to affecting the CD28-mediated pathway of T cell stimulation, these reagents also inhibit the CTLA-4 signals required for Treg function. To avoid inhibiting CTLA-4–dependent regulation of the immune system while suppressing T cell activation, the Vanhove, Blancho, and Pierson labs aimed to block CD28 function directly in experiments on kidney and heart transplantation in baboons and monkeys. Previous work showed that down-regulation of CD28 activity with an anti-CD28 monoclonal antibody inhibited rejection of organ transplants in rodents. For the new nonhuman primate studies, the researchers used a fragment of a human CD28-specific monoclonal antibody fused to a carrier molecule to increase its half-life in vivo. This fusion antibody, sc28AT, recognizes CD28 from monkeys and baboons as well as humans, and it competes with CD80/86 for binding to CD28. sc28AT increased the number of functional Tregs and reduced chronic rejection of the transplanted tissue in the primates when used together with a calcineurin inhibitor. This is good news because it suggests that the dose of such inhibitors (and their toxic side effects) could be reduced in patients when given with a molecule targeting CD28. It remains to be determined whether this approach offers practical advantages over straight CD80/86 blockade when tested in the clinic. Transplantation is the treatment of choice for patients with end-stage organ failure. Its success is limited by side effects of immunosuppressive drugs, such as inhibitors of the calcineurin pathway that prevent rejection by reducing synthesis of interleukin-2 by T cells. Moreover, none of the existing drugs efficiently prevent the eventual rejection of the organ. Blocking the CD28-mediated T cell costimulation pathway is a nontoxic alternative immunosuppression strategy that is now achieved by blockade of CD80/86, the receptor for CD28 on antigen-presenting cells. However, interaction of CD80/86 with cytotoxic T lymphocyte–associated antigen 4 (CTLA-4) is required for immune regulation. Therefore, CD28 blockade, instead of CD80/86 blockade, might preserve regulatory signals mediated by CTLA-4 and preserve immune regulation. By using monovalent antibodies, we identified true CD28 antagonists that induced CTLA-4–dependent decreased T cell function compatible with regulatory T (Treg) cell suppression. In transplantation experiments in primates, blocking CD28 augmented intragraft and peripheral blood Treg cells, induced molecular signatures of immune regulation, and prevented graft rejection and vasculopathy in synergy with calcineurin inhibition. These findings suggest that targeting costimulation blockade at CD28 preserves CTLA-4–dependent immune regulation and promotes allograft survival.


American Journal of Transplantation | 2005

Prolongation of heart allograft survival by immature dendritic cells generated from recipient type bone marrow progenitors.

Hélène Pêche; Benjamin Trinité; Bernard Martinet; Maria Cristina Cuturi

Recent studies suggest that particular dendritic cells (DC) subpopulations may be tolerogenic. To test the capacity of different DC subpopulations to modulate allograft rejection, we generated two distinct populations of rat bone marrow‐derived DCs (BMDC) with low doses of GM‐CSF and IL‐4. The non‐adherent population (nBMDC), which are the ‘classical’ DCs was able to stimulate naïve allogeneic T cells and could be induced to completely mature using various stimuli. In contrast, the adherent population (aBMDC), which displayed an immature phenotype, was unable to stimulate T cells and was more resistant to maturation. We found that syngeneic aBMDCs, injected one day before transplantation, induced significant prolongation of heart allograft survival and decreased anti‐donor humoral and cellular responses. Similarly, syngeneic aBMDCs inhibited T‐cell responses to KLH in the spleen but not in lymph node in a KLH immunization model without graft. This effect was not antigen specific and could be reversed using an inhibitor of inducible nitric oxide synthase. This compartmentalized inhibition could be in part explained by the fact that the majority of syngeneic adherent cells administered intravenously were found in the spleen with some of them reaching the T‐cell areas. These data suggest that syngeneic aBMDCs can modulate immune responses.


Journal of Immunology | 2012

Control of Transplant Tolerance and Intragraft Regulatory T Cell Localization by Myeloid-Derived Suppressor Cells and CCL5

Nahzli Dilek; Nicolas Poirier; Claire Usal; Bernard Martinet; Gilles Blancho; Bernard Vanhove

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature cells that are believed to inhibit immune responses in the contexts of cancer and organ transplantation, in association with regulatory T cells (Treg). However, the way in which MDSC cooperate with Treg remains elusive. In this study, we used DNA microarrays to analyze gene expression in blood-derived MDSC from rat recipients of kidney allografts. We found CCL5 (Rantes), a chemotactic C-C motif 5 chemokine, to be strongly downregulated after treatment with a tolerizing regimen. The amount of CCL5 protein was also lower in the plasma of tolerant recipients, whereas intragraft CCL5 was unchanged. Because CCL5 is chemotactic for Treg, we hypothesized that a gradient of CCL5 between the graft and peripheral blood might contribute to the intragraft localization of Treg in tolerant animals. To test this hypothesis, we treated tolerant rat recipients of kidney allografts with recombinant rat CCL5 to restore normal plasma concentrations. This led to a strong reduction in intragraft Treg monitored by immunohistofluorescence and by quantitative real-time PCR measurement of Foxp3 mRNA. Ultimately, this treatment led to an increase in serum creatinine concentrations and to kidney graft rejection after about a month. The kidney function of syngeneic grafts was not affected by a similar administration of CCL5. These data highlight the contribution of MDSC to the establishment of a graft-to-periphery CCL5 gradient in tolerant kidney allograft recipients, which controls recruitment of Treg to the graft where they likely contribute to maintaining tolerance.


Journal of Clinical Investigation | 2014

IL-7 receptor blockade following T cell depletion promotes long-term allograft survival

Hoa-Le Mai; Françoise Boeffard; Julie Longis; Richard Danger; Bernard Martinet; Fabienne Haspot; Bernard Vanhove; Sophie Brouard; Jean-Paul Soulillou

T cell depletion is commonly used in organ transplantation for immunosuppression; however, a restoration of T cell homeostasis following depletion leads to increased memory T cells, which may promote transplant rejection. The cytokine IL-7 is important for controlling lymphopoiesis under both normal and lymphopenic conditions. Here, we investigated whether blocking IL-7 signaling with a mAb that targets IL-7 receptor α (IL-7Rα) alone or following T cell depletion confers an advantage for allograft survival in murine transplant models. We found that IL-7R blockade alone induced indefinite pancreatic islet allograft survival if anti-IL-7R treatment was started 3 weeks before graft. IL-7R blockade following anti-CD4- and anti-CD8-mediated T cell depletion markedly prolonged skin allograft survival. Furthermore, IL-7 inhibition in combination with T cell depletion synergized with either CTLA-4Ig administration or suboptimal doses of tacrolimus to induce long-term skin graft acceptance in this stringent transplant model. Together, these therapies inhibited T cell reconstitution, decreased memory T cell numbers, increased the relative frequency of Tregs, and abrogated both cellular and humoral alloimmune responses. Our data suggest that IL-7R blockade following T cell depletion has potential as a robust, immunosuppressive therapy in transplantation.


mAbs | 2013

Antagonist properties of monoclonal antibodies targeting human CD28: Role of valency and the heavy-chain constant domain

Caroline Mary; Flora Coulon; Nicolas Poirier; Nahzli Dilek; Bernard Martinet; Gilles Blancho; Bernard Vanhove

Antagonist antibodies targeting CD28 have been proposed as an alternative to the use of CD80/86 antagonists to modulate T cell responses in autoimmunity and transplantation. Advantages would be the blockade of CD28-mediated co-stimulatory signals without impeding the co-inhibitory signals dependent on CD80 interactions with CTLA-4 and PD-L1 that are important for the control of immune responses and for the function of regulatory T cells. Anti-CD28 antibodies are candidate antagonists only if they prevent access to the CD80/86 ligands without simultaneously stimulating CD28 itself, a process that is believed to depend on receptor multimerization. In this study, we evaluated the impact of different formats of a potentially antagonist anti-human CD28 antibody on T cell activation. In particular, we examined the role of valency and of the presence of an Fc domain, two components that might affect receptor multimerization either directly or in the presence of accessory cells expressing Fc receptors. Among monovalent (Fab’, scFv), divalent (Fab’2), monovalent-Fc (Fv-Fc) and divalent-Fc (IgG) formats, only the monovalent formats showed consistent absence of induced CD28 multimerization and absence of associated activation of phosphoinositol-3-kinase, and clear antagonist properties in T cell stimulation assays. In contrast, divalent antibodies showed agonist properties that resulted in cell proliferation and cytokine release in an Fc-independent manner. Conjugation of monovalent antibodies with polyethylene glycol, α-1-antitrypsin or an Fc domain significantly extended their in vivo half-life without modifying their antagonist properties. In conclusion, these data indicate that monovalency is mandatory for maintaining the antagonistic activity of anti-CD28 monoclonal antibodies.


JCI insight | 2017

Transient antibody targeting of CD45RC induces transplant tolerance and potent antigen-specific regulatory T cells

Elodie Picarda; Séverine Bézie; Laetitia Boucault; Elodie Autrusseau; Stéphanie Kilens; Dimitri Meistermann; Bernard Martinet; Véronique Daguin; Audrey Donnart; Eric Charpentier; Laurent David; Ignacio Anegon; Carole Guillonneau

Rat and human CD4+ and CD8+ Tregs expressing low levels of CD45RC have strong immunoregulatory properties. We describe here that human CD45 isoforms are nonredundant and identify distinct subsets of cells. We show that CD45RC is not expressed by CD4+ and CD8+ Foxp3+ Tregs, while CD45RA/RB/RO are. Transient administration of a monoclonal antibody (mAb) targeting CD45RC in a rat cardiac allotransplantation model induced transplant tolerance associated with inhibition of allogeneic humoral responses but maintained primary and memory responses against cognate antigens. Anti-CD45RC mAb induced rapid death of CD45RChigh T cells through intrinsic cell signaling but preserved and potentiated CD4+ and CD8+ CD45RClow/- Tregs, which are able to adoptively transfer donor-specific tolerance to grafted recipients. Anti-CD45RC treatment results in distinct transcriptional signature of CD4+ and CD8+ CD45RClow/- Tregs. Finally, we demonstrate that anti-human CD45RC treatment inhibited graft-versus-host disease (GVHD) in immune-humanized NSG mice. Thus, short-term anti-CD45RC is a potent therapeutic candidate to induce transplantation tolerance in human.


Journal of Immunology | 2015

Compensatory Regulatory Networks between CD8 T, B, and Myeloid Cells in Organ Transplantation Tolerance

Séverine Bézie; Elodie Picarda; Jason Ossart; Bernard Martinet; Ignacio Anegon; Carole Guillonneau

In transplantation tolerance, numerous regulatory populations have the capacity to inhibit allograft rejection; however, their compensatory capacities have never been clearly evidenced. We have previously demonstrated that the tolerogenic effect mediated by CD8+CD45RClow regulatory T cells (Tregs) in a model of organ transplantation with CD40Ig could be abrogated by permanent depletion of CD8+ cells that resulted in allograft rejection in half of the recipients. This result demonstrated that CD8+ Tregs were essential, but also that half of the recipients still survived indefinitely. We also demonstrated that no other regulatory populations, besides CD8+ Tregs, could induce and maintain allograft tolerance in CD40Ig-treated tolerant animals. In the current study, we analyzed the mechanisms that arose following CD8+ Treg depletion and allowed establishment of networks of new regulatory cells to maintain allograft survival. We identified regulatory B cells (Bregs) and regulatory myeloid cells (RegMCs) as being responsible of the maintenance of the long-term allograft survival. We demonstrated that both regulatory cell subsets efficiently inhibited antidonor immune responses in adoptively transferred recipients. Although Bregs were induced, they were not essential for the maintenance of the graft as demonstrated in IgM-deficient recipients. In addition, we showed that RegMCs were the most suppressive and acted alone, whereas Bregs activity was associated with increased suppressive activity of other subsets in adoptively transferred recipients. Altogether, to our knowledge, we demonstrated in this study for the first time the emergence of both Bregs and RegMCs following Tregs depletion and highlighted the importance of regulatory cell networks and their synergistic potential in transplantation.


PLOS ONE | 2016

Anti-EBOV GP IgGs Lacking α1-3-Galactose and Neu5Gc Prolong Survival and Decrease Blood Viral Load in EBOV-Infected Guinea Pigs.

Olivier Reynard; Frédéric Jacquot; Gwénaëlle Evanno; Hoa Le Mai; Apolline Salama; Bernard Martinet; Odile Duvaux; Jean-Marie Bach; Sophie Conchon; Jean-Paul Judor; Andrea Perota; Irina Lagutina; Roberto Duchi; Giovanna Lazzari; Ludmilla Le Berre; Hélène Perreault; Elsa Lheriteau; Hervé Raoul; Viktor E. Volchkov; Cesare Galli; Jean-Paul Soulillou

Polyclonal xenogenic IgGs, although having been used in the prevention and cure of severe infectious diseases, are highly immunogenic, which may restrict their usage in new applications such as Ebola hemorrhagic fever. IgG glycans display powerful xenogeneic antigens in humans, for example α1–3 Galactose and the glycolyl form of neuraminic acid Neu5Gc, and IgGs deprived of these key sugar epitopes may represent an advantage for passive immunotherapy. In this paper, we explored whether low immunogenicity IgGs had a protective effect on a guinea pig model of Ebola virus (EBOV) infection. For this purpose, a double knock-out pig lacking α1–3 Galactose and Neu5Gc was immunized against virus-like particles displaying surface EBOV glycoprotein GP. Following purification from serum, hyper-immune polyclonal IgGs were obtained, exhibiting an anti-EBOV GP titer of 1:100,000 and a virus neutralizing titer of 1:100. Guinea pigs were injected intramuscularly with purified IgGs on day 0 and day 3 post-EBOV infection. Compared to control animals treated with IgGs from non-immunized double KO pigs, the anti-EBOV IgGs-treated animals exhibited a significantly prolonged survival and a decreased virus load in blood on day 3. The data obtained indicated that IgGs lacking α1–3 Galactose and Neu5Gc, two highly immunogenic epitopes in humans, have a protective effect upon EBOV infection.


Journal of Translational Medicine | 2012

Antagonist properties of monoclonal antibodies to human CD28: role of valency and heavy-chain constant domain

Caroline Mary; Flora Coulon; Nicolas Poirier; Nahzli Dilek; Bernard Martinet; Gilles Blancho; Bernard Vanhove

Background Antagonist antibodies targeting CD28 have been proposed as an alternative to the use of CD80/86 antagonists to modulate T cell responses in autoimmunity and transplantation. Advantages would be the blockade of CD28mediated co-stimulatory signals without impeding co-inhibitory signals depending on CD80 interactions with CTLA-4 and PD-L1 that are important for the control of immune responses and for the function of regulatory T cells. Anti-CD28 antibodies are candidate antagonists only if they prevent access to the CD80/86 ligands without simultaneously stimulating CD28 itself, a process that is believed to depend on receptor multimerization.

Collaboration


Dive into the Bernard Martinet'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