Network


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

Hotspot


Dive into the research topics where Antoine Marçais is active.

Publication


Featured researches published by Antoine Marçais.


Nature Immunology | 2014

The metabolic checkpoint kinase mTOR is essential for IL-15 signaling during the development and activation of NK cells

Antoine Marçais; Julien Cherfils-Vicini; Charlotte Viant; Sophie Degouve; Sébastien Viel; Jessica Rabilloud; Katia Mayol; Armelle Tavares; Jacques Bienvenu; Yann-Gaël Gangloff; Eric Gilson; Eric Vivier; Thierry Walzer

Interleukin 15 (IL-15) controls both the homeostasis and the peripheral activation of natural killer (NK) cells. The molecular basis for this duality of action remains unknown. Here we found that the metabolic checkpoint kinase mTOR was activated and boosted bioenergetic metabolism after exposure of NK cells to high concentrations of IL-15, whereas low doses of IL-15 triggered only phosphorylation of the transcription factor STAT5. mTOR stimulated the growth and nutrient uptake of NK cells and positively fed back on the receptor for IL-15. This process was essential for sustaining NK cell proliferation during development and the acquisition of cytolytic potential during inflammation or viral infection. The mTORC1 inhibitor rapamycin inhibited NK cell cytotoxicity both in mice and humans; this probably contributes to the immunosuppressive activity of this drug in different clinical settings.


European Journal of Immunology | 2006

TLR2 engagement on CD8 T cells lowers the thresholdfor optimal antigen‐induced T cell activation

Anne Cottalorda; Claire Verschelde; Antoine Marçais; Martine Tomkowiak; Philippe Musette; Satoshi Uematsu; Shizuo Akira; Jacqueline Marvel; Nathalie Bonnefoy-Berard

TLR have a crucial role in the detection of microbial infection in mammals. Until recently, most investigations on TLR have focused on cells of the innate immune system and on the role of TLR in the initiation of antigen‐specific responses following recognition of microbial products by APC. Here, we report that murine T cells express TLR1, TLR2, TLR6, TLR7 and TLR9 mRNA. Using CD8 T cells from F5 TCR‐transgenic mice, we demonstrate that the lipopeptide Pam3CysSK4 (Pam), a synthetic analog of bacterial and mycoplasmal lipoproteins that recognizes TLR1/2 complex, costimulates antigen‐activated T cells. Costimulation with Pam permits an increased cell proliferation and survival associated with a sustained CD25 expression and an enhanced expression of Bcl‐xL anti‐apoptotic protein. In addition, we show that costimulation with Pam up‐regulates IFN‐γ production but also granzyme B secretion and cytotoxic activity of antigen‐activated T cells, indicating that TLR2 engagement enhances the major effector functions of CD8 T cells. Finally, we demonstrate that TLR2 engagement on T cells lowers the activation threshold for costimulatory signals delivered by APC.


Science Signaling | 2016

TGF-β inhibits the activation and functions of NK cells by repressing the mTOR pathway

Sébastien Viel; Antoine Marçais; Fernando Souza-Fonseca Guimaraes; Róisín M. Loftus; Jessica Rabilloud; Morgan Grau; Sophie Degouve; Sophia Djebali; Amélien Sanlaville; Emily Charrier; Jacques Bienvenu; Julien C. Marie; Christophe Caux; Jacqueline Marvel; Liam Town; Nicholas D. Huntington; Laurent Bartholin; David K. Finlay; Mark J. Smyth; Thierry Walzer

Blocking TGF-β signaling in natural killer cells enhances their metabolism and ability to kill tumor cells. Relieving NK cell suppression The immunosuppressive cytokine transforming growth factor–β (TGF-β) has beneficial effects when it resolves inflammation and prevents autoimmunity, but not when it inhibits antitumor immune responses. Viel et al. found that TGF-β signaling in mouse and human natural killer (NK) cells, cytotoxic cells that target tumor cells, inhibited the activation of the kinase mTOR, a central regulator of cellular metabolism and cytotoxic function. NK cells deficient in a TGF-β receptor subunit showed enhanced antitumor activity in mouse models of metastasis, suggesting that enhancing metabolism in NK cells may provide a therapeutic strategy to kill cancer cells. Transforming growth factor–β (TGF-β) is a major immunosuppressive cytokine that maintains immune homeostasis and prevents autoimmunity through its antiproliferative and anti-inflammatory properties in various immune cell types. We provide genetic, pharmacologic, and biochemical evidence that a critical target of TGF-β signaling in mouse and human natural killer (NK) cells is the serine and threonine kinase mTOR (mammalian target of rapamycin). Treatment of mouse or human NK cells with TGF-β in vitro blocked interleukin-15 (IL-15)–induced activation of mTOR. TGF-β and the mTOR inhibitor rapamycin both reduced the metabolic activity and proliferation of NK cells and reduced the abundances of various NK cell receptors and the cytotoxic activity of NK cells. In vivo, constitutive TGF-β signaling or depletion of mTOR arrested NK cell development, whereas deletion of the TGF-β receptor subunit TGF-βRII enhanced mTOR activity and the cytotoxic activity of the NK cells in response to IL-15. Suppression of TGF-β signaling in NK cells did not affect either NK cell development or homeostasis; however, it enhanced the ability of NK cells to limit metastases in two different tumor models in mice. Together, these results suggest that the kinase mTOR is a crucial signaling integrator of pro- and anti-inflammatory cytokines in NK cells. Moreover, we propose that boosting the metabolic activity of antitumor lymphocytes could be an effective strategy to promote immune-mediated tumor suppression.


Journal of Immunology | 2009

Dicer-Dependent MicroRNA Pathway Controls Invariant NKT Cell Development

Maya Fedeli; Anna Napolitano; Molly Pui Man Wong; Antoine Marçais; Claudia de Lalla; Francesco Colucci; Matthias Merkenschlager; Paolo Dellabona; Giulia Casorati

Invariant NK T (iNKT) cells are a separate lineage of T lymphocytes with innate effector functions. They express an invariant TCR specific for lipids presented by CD1d and their development and effector differentiation rely on a unique gene expression program. We asked whether this program includes microRNAs, small noncoding RNAs that regulate gene expression posttranscriptionally and play a key role in the control of cellular differentiation programs. To this aim, we investigated iNKT cell development in mice in which Dicer, the RNase III enzyme that generates functional microRNAs, is deleted in cortical thymocytes. We find that Dicer deletion results in a substantial reduction of iNKT cells in thymus and their disappearance from the periphery, unlike mainstream T cells. Without Dicer, iNKT cells do not complete their innate effector differentiation and display a defective homeostasis due to increased cell death. Differentiation and homeostasis of iNKT cells require Dicer in a cell-autonomous fashion. Furthermore, we identify a miRNA profile specific for iNKT cells, which exhibits features of activated/effector T lymphocytes, consistent with the idea that iNKT cells undergo agonist thymic selection. Together, these results define a critical role of the Dicer-dependent miRNA pathway in the physiology of iNKT cells.


Frontiers in Immunology | 2013

Regulation of mouse NK cell development and function by cytokines

Antoine Marçais; Sébastien Viel; Morgan Grau; Thomas Henry; Jacqueline Marvel; Thierry Walzer

Natural Killer (NK) cells are innate lymphocytes with an important role in the early defense against intracellular pathogens and against tumors. Like other immune cells, almost every aspects of their biology are regulated by cytokines. Interleukin (IL)-15 is pivotal for their development, homeostasis, and activation. Moreover, numerous other activating or inhibitory cytokines such as IL-2, IL-4, IL-7, IL-10, IL-12, IL-18, IL-21, Transforming growth factor-β (TGFβ) and type I interferons regulate their activation and their effector functions at different stages of the immune response. In this review we summarize the current understanding on the effect of these different cytokines on NK cell development, homeostasis, and functions during steady-state or upon infection by different pathogens. We try to delineate the cellular sources of these cytokines, the intracellular pathways they trigger and the transcription factors they regulate. We describe the known synergies or antagonisms between different cytokines and highlight outstanding questions in this field of investigation. Finally, we discuss how a better knowledge of cytokine action on NK cells could help improve strategies to manipulate NK cells in different clinical situations.


Journal of Experimental Medicine | 2015

Terminal NK cell maturation is controlled by concerted actions of T-bet and Zeb2 and is essential for melanoma rejection.

Mary J. van Helden; Steven Goossens; Cécile Daussy; Anne-Laure Mathieu; Fabrice Faure; Antoine Marçais; Niels Vandamme; Natalie Farla; Katia Mayol; Sébastien Viel; Sophie Degouve; Emilie Debien; Eve Seuntjens; Andrea Conidi; Julie Chaix; Philippe Mangeot; Simon de Bernard; Laurent Buffat; Jody J. Haigh; Danny Huylebroeck; Bart M. Lambrecht; Geert Berx; Thierry Walzer

The transcription factor Zeb2 cooperates with T-bet to control NK cell maturation, viability, and exit from the bone marrow and is essential for rejection of melanoma lung metastasis.


Journal of Immunology | 2005

Flt3 Ligand-Generated Murine Plasmacytoid and Conventional Dendritic Cells Differ in Their Capacity to Prime Naive CD8 T Cells and to Generate Memory Cells In Vivo

Georgi S. Angelov; Martine Tomkowiak; Antoine Marçais; Yann Leverrier; Jacqueline Marvel

Mature dendritic cells (DCs) have the capacity to induce efficient primary T cell response and effector cell differentiation. Thus, these cells are a major tool in the design of various immunotherapeutic protocols. We have tested the capacity of different subsets of matured DCs pulsed with a peptide to induce the differentiation of naive CD8 T cells into memory cells in vivo. Flt3 ligand (FL) induces the differentiation of conventional DCs (cDCs) and plasmacytoid DCs (PDCs) from murine bone marrow precursors in vitro. After maturation, both subsets become strong stimulators of Ag-specific T cell responses in vitro. However, the in vivo T cell stimulatory capacity of these DC subsets has not been studied in detail. In the present study, we demonstrate that mature FL-generated DCs induce efficient peptide-specific CD8 T cell response and memory cell differentiation in vivo. This is mainly due to the cDC subset because the PDC subset induced only a negligible primary CD8 response without detectable levels of memory CD8 T cell differentiation. Thus, in vitro FL-generated mature cDCs, but not PDCs, are potent stimulators of peptide-specific CD8 T cell responses and memory generation in vivo.


Journal of Immunology | 2009

Characterization of a CD44/CD122int Memory CD8 T Cell Subset Generated under Sterile Inflammatory Conditions

Florentin-Martial Mbitikon-Kobo; Marc Vocanson; Marie-Cécile Michallet; Martine Tomkowiak; Anne Cottalorda; Georgi S. Angelov; Charles-Antoine Coupet; Sophia Djebali; Antoine Marçais; Bertrand Dubois; Nathalie Bonnefoy-Berard; Jean-François Nicolas; Christophe Arpin; Jacqueline Marvel

Most memory CD8 T cell subsets that have been hitherto defined are generated in response to infectious pathogens. In this study, we have characterized the CD8 T cells that survive priming conditions, devoid of pathogen-derived danger signals. In both a TCR-transgenic model and a model of contact hypersensitivity, we show that the priming of naive CD8 T cells under sterile inflammatory conditions generates memory. The corresponding memory CD8 T cells can be identified by their intermediate expression levels of CD44 and CD122. We also show that CD44/122int memory CD8 T cells spontaneously develop in wild type mice and that they display intermediate levels of several other memory traits including functional (IFN-γ secretion capacity, CCL5 messenger stores), phenotypic, and molecular (T-bet and eomesodermin expression levels) features. We finally show that they correspond to an early differentiation stage and can further differentiate in CD44/122high memory T cells. Altogether, our results identify a new memory CD8 T cell subset that is generated under sterile inflammatory conditions and involved in the recall contact hypersensitivity reactions that are responsible for allergic contact dermatitis.


Journal of Immunology | 2012

Negative Regulation of NKG2D Expression by IL-4 in Memory CD8 T Cells

Erwan Ventre; Lilia Brinza; Stéphane Schicklin; Julien Mafille; Charles-Antoine Coupet; Antoine Marçais; Sophia Djebali; Virginie Jubin; Thierry Walzer; Jacqueline Marvel

IL-4 is one of the main cytokines produced during Th2-inducing pathologies. This cytokine has been shown to affect a number of immune processes such as Th differentiation and innate immune responses. However, the impact of IL-4 on CD8 T cell responses remains unclear. In this study, we analyzed the effects of IL-4 on global gene expression profiles of Ag-induced memory CD8 T cells in the mouse. Gene ontology analysis of this signature revealed that IL-4 regulated most importantly genes associated with immune responses. Moreover, this IL-4 signature overlapped with the set of genes preferentially expressed by memory CD8 T cells over naive CD8 T cells. In particular, IL-4 downregulated in vitro and in vivo in a STAT6-dependent manner the memory-specific expression of NKG2D, thereby increasing the activation threshold of memory CD8 T cells. Furthermore, IL-4 impaired activation of memory cells as well as their differentiation into effector cells. This phenomenon could have an important clinical relevance as patients affected by Th2 pathologies such as parasitic infections or atopic dermatitis often suffer from viral-induced complications possibly linked to inefficient CD8 T cell responses.


OncoImmunology | 2013

Monitoring NK cell activity in patients with hematological malignancies

Sébastien Viel; Emily Charrier; Antoine Marçais; Paul Rouzaire; Jacques Bienvenu; Lionel Karlin; Gilles Salles; Thierry Walzer

Natural killer (NK) cells are lymphocytes of the innate immune system that can recognize and kill various types of malignant cells. Monitoring the activity of peripheral NK cells in patients affected by hematological malignancies may provide prognostic information or unveil ongoing tumor-specific immune responses. Moreover, further insights into the biology of NK cells might also promote the development of novel strategies for stimulating their anticancer activity. Here, we review the main methods to monitor phenotypic and functional NK cell properties in cancer patients, focusing on individuals affected by multiple myeloma, a hematological malignancy currently treated with immunomodulatory drugs.

Collaboration


Dive into the Antoine Marçais's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sébastien Viel

École normale supérieure de Lyon

View shared research outputs
Top Co-Authors

Avatar

Jacqueline Marvel

Claude Bernard University Lyon 1

View shared research outputs
Top Co-Authors

Avatar

Anne-Laure Mathieu

École normale supérieure de Lyon

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marie Marotel

École normale supérieure de Lyon

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric Vivier

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar

Sophia Djebali

École normale supérieure de Lyon

View shared research outputs
Researchain Logo
Decentralizing Knowledge