Network


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

Hotspot


Dive into the research topics where Julien C. Marie is active.

Publication


Featured researches published by Julien C. Marie.


Nature Immunology | 2002

Linking innate and acquired immunity: divergent role of CD46 cytoplasmic domains in T cell–induced inflammation

Julien C. Marie; Anne Astier; Pierre Rivailler; Chantal Rabourdin-Combe; T. Fabian Wild; Branka Horvat

CD46 is a widely expressed transmembrane protein that was initially identified as binding and inactivating C3b and C4b complement products. We used mice that were transgenic for one of two human CD46 isoforms that differ in their cytoplasmic domains (termed CD46-1 and CD46-2) to analyze the effect of CD46 stimulation on the immune response. We show here that CD46 can regulate inflammatory responses, either by inhibiting (CD46-1) or increasing (CD46-2) the contact hypersensitivity reaction. We found that engagement of CD46-1 or CD46-2 differentially affected CD8+ T cell cytotoxicity, CD4+ T cell proliferation, interleukin 2 (IL-2) and IL-10 production as well as tyrosine phosphorylation of Vav in T lymphocytes. These results indicate that CD46 plays a role in regulating the T cell–induced inflammatory reaction and in fine-tuning the cellular immune response by bridging innate and acquired immunity.


Immunity | 2001

Mechanism of Measles Virus–Induced Suppression of Inflammatory Immune Responses

Julien C. Marie; Jeanne Kehren; Marie-Claude Trescol-Biémont; Alexey Evlashev; Hélène Valentin; Thierry Walzer; R. Tedone; Bruce E. Loveland; Jean-François Nicolas; Chantal Rabourdin-Combe; Branka Horvat

Measles virus (MV) causes profound immunosuppression, resulting in high infant mortality. The mechanisms are poorly understood, largely due to the lack of a suitable animal model. Here, we report that particular MV proteins, in the absence of MV replication, could generate a systemic immunosuppression in mice through two pathways: (1) via MV-nucleoprotein and its receptor FcgammaR on dendritic cells; and (2) via virus envelope glycoproteins and the MV-hemagglutinin cellular receptor, CD46. The effects comprise reduced hypersensitivity responses associated with impaired function of dendritic cells, decreased production of IL-12, and the loss of antigen-specific T cell proliferation. These results introduce a novel model for testing the immunosuppressive potential of anti-measles vaccines and reveal a specific mechanism of MV-induced modulation of inflammatory reactions.


Journal of Virology | 2003

Measles Virus (MV) Nucleoprotein Binds to a Novel Cell Surface Receptor Distinct from FcγRII via Its C-Terminal Domain: Role in MV-Induced Immunosuppression

David Laine; Marie-Claude Trescol-Biémont; Sonia Longhi; Geneviève Libeau; Julien C. Marie; Pierre-Olivier Vidalain; Olga Azocar; Adama Diallo; Bruno Canard; Chantal Rabourdin-Combe; Hélène Valentin

ABSTRACT During acute measles virus (MV) infection, an efficient immune response occurs, followed by a transient but profound immunosuppression. MV nucleoprotein (MV-N) has been reported to induce both cellular and humoral immune responses and paradoxically to account for immunosuppression. Thus far, this latter activity has been attributed to MV-N binding to human and murine FcγRII. Here, we show that apoptosis of MV-infected human thymic epithelial cells (TEC) allows the release of MV-N in the extracellular compartment. This extracellular N is then able to bind either to MV-infected or uninfected TEC. We show that recombinant MV-N specifically binds to a membrane protein receptor, different from FcγRII, highly expressed on the cell surface of TEC. This new receptor is referred to as nucleoprotein receptor (NR). In addition, different Ns from other MV-related morbilliviruses can also bind to FcγRII and/or NR. We show that the region of MV-N responsible for binding to NR maps to the C-terminal fragment (NTAIL). Binding of MV-N to NR on TEC triggers sustained calcium influx and inhibits spontaneous cell proliferation by arresting cells in the G0 and G1 phases of the cell cycle. Finally, MV-N binds to both constitutively expressed NR on a large spectrum of cells from different species and to human activated T cells, leading to suppression of their proliferation. These results provide evidence that MV-N, after release in the extracellular compartment, binds to NR and thereby plays a role in MV-induced immunosuppression.


Journal of Experimental Medicine | 2009

iNKT cell development is orchestrated by different branches of TGF-beta signaling.

Jean-Marc Doisne; Laurent Bartholin; Kai-Ping Yan; Celine Garcia; Nadia Duarte; Jean-Benoît Le Luduec; David F. Vincent; Farhan S. Cyprian; Branka Horvat; Sylvie Martel; Ruth Rimokh; Régine Losson; Kamel Benlagha; Julien C. Marie

Invariant natural killer T (iNKT) cells constitute a distinct subset of T lymphocytes exhibiting important immune-regulatory functions. Although various steps of their differentiation have been well characterized, the factors controlling their development remain poorly documented. Here, we show that TGF-β controls the differentiation program of iNKT cells. We demonstrate that TGF-β signaling carefully and specifically orchestrates several steps of iNKT cell development. In vivo, this multifaceted role of TGF-β involves the concerted action of different pathways of TGF-β signaling. Whereas the Tif-1γ branch controls lineage expansion, the Smad4 branch maintains the maturation stage that is initially repressed by a Tif-1γ/Smad4-independent branch. Thus, these three different branches of TGF-β signaling function in concert as complementary effectors, allowing TGF-β to fine tune the iNKT cell differentiation program.


Immunity | 2015

Integrin αvβ8-Mediated TGF-β Activation by Effector Regulatory T Cells Is Essential for Suppression of T-Cell-Mediated Inflammation

John J. Worthington; Aoife Kelly; Catherine Smedley; David Bauché; Simon Campbell; Julien C. Marie; Mark A. Travis

Summary Regulatory T (Treg) cells play a pivotal role in suppressing self-harmful T cell responses, but how Treg cells mediate suppression to maintain immune homeostasis and limit responses during inflammation is unclear. Here we show that effector Treg cells express high amounts of the integrin αvβ8, which enables them to activate latent transforming growth factor-β (TGF-β). Treg-cell-specific deletion of integrin αvβ8 did not result in a spontaneous inflammatory phenotype, suggesting that this pathway is not important in Treg-cell-mediated maintenance of immune homeostasis. However, Treg cells lacking expression of integrin αvβ8 were unable to suppress pathogenic T cell responses during active inflammation. Thus, our results identify a mechanism by which Treg cells suppress exuberant immune responses, highlighting a key role for effector Treg-cell-mediated activation of latent TGF-β in suppression of self-harmful T cell responses during active inflammation.


Journal of Clinical Investigation | 2014

TGF-β prevents T follicular helper cell accumulation and B cell autoreactivity

Mark J. McCarron; Julien C. Marie

T follicular helper (Tfh) cells contribute to the establishment of humoral immunity by controlling the delivery of helper signals to activated B cells; however, Tfh development must be restrained, as aberrant accumulation of these cells is associated with positive selection of self-reactive germinal center B cells and autoimmunity in both humans and mice. Here, we show that TGF-β signaling in T cells prevented Tfh cell accumulation, self-reactive B cell activation, and autoantibody production. Using mice with either T cell-specific loss or constitutive activation of TGF-β signaling, we demonstrated that TGF-β signaling is required for the thymic maturation of CD44⁺CD122⁺Ly49⁺CD8⁺ regulatory T cells (Tregs), which induce Tfh apoptosis and thus regulate this cell population. Moreover, peripheral Tfh cells escaping TGF-β control were resistant to apoptosis, exhibited high levels of the antiapoptotic protein BCL2, and remained refractory to regulation by CD8+ Tregs. The unrestrained accumulation of Tfh cells in the absence of TGF-β was dependent on T cell receptor engagement and required B cells. Together, these data indicate that TGF-β signaling restrains Tfh cell accumulation and B cell-associated autoimmunity and thereby controls self-tolerance.


Journal of Virology | 2004

Cell Surface Delivery of the Measles Virus Nucleoprotein: a Viral Strategy To Induce Immunosuppression

Julien C. Marie; Frédéric Saltel; Jean-Michel Escola; Pierre Jurdic; T. Fabian Wild; Branka Horvat

ABSTRACT Although only a few blood cells are infected during measles, this infectious disease is followed by acute immunosuppression, associated with high infant mortality. Measles virus nucleoprotein has been suggested to contribute to virus-induced inhibition of the immune response. However, it has been difficult to understand how this cytosolic viral protein could leave an infected cell and then perturb the immune response. Here we demonstrate that intracellularly synthesized nucleoprotein enters the late endocytic compartment, where it recruits its cellular ligand, the Fcγ receptor. Nucleoprotein is then expressed at the surfaces of infected leukocytes associated with the Fcγ receptor and is secreted into the extracellular compartment, allowing its interaction with uninfected cells. Finally, cell-derived nucleoprotein inhibits the secretion of interleukin-12 and the generation of the inflammatory reaction, both shown to be impaired during measles. These results reveal nucleoprotein egress from infected cells as a novel strategy in measles-induced immunosuppression.


Journal of Virology | 2000

Productive Measles Virus Brain Infection and Apoptosis in CD46 Transgenic Mice

Alexey Evlashev; Emmanuel Moyse; Hélène Valentin; Olga Azocar; Marie-Claude Trescol-Biémont; Julien C. Marie; Chantal Rabourdin-Combe; Branka Horvat

ABSTRACT Measles virus (MV) infection causes acute childhood disease, associated in certain cases with infection of the central nervous system (CNS) and development of neurological disease. To develop a murine model of MV-induced pathology, we generated several lines of transgenic mice ubiquitously expressing as the MV receptor a human CD46 molecule with either a Cyt1 or Cyt2 cytoplasmic tail. All transgenic lines expressed CD46 protein in the brain. Newborn transgenic mice, in contrast to nontransgenic controls, were highly sensitive to intracerebral infection by the MV Edmonston strain. Signs of clinical illness (lack of mobility, tremors, and weight loss) appeared within 5 to 7 days after infection, followed by seizures, paralysis, and death of the infected animals. Virus replication was detected in neurons from infected mice, and virus was reproducibly isolated from transgenic brain tissue. MV-induced apoptosis observed in different brain regions preceded the death of infected animals. Similar results were obtained with mice expressing either a Cyt1 or Cyt2 cytoplasmic tail, demonstrating the ability of different isoforms of CD46 to function as MV receptors in vivo. In addition, maternally transferred immunity delayed death of offspring given a lethal dose of MV. These results document a novel CD46 transgenic murine model where MV neuronal infection is associated with the production of infectious virus, similarly to progressive infectious measles encephalitis seen in immunocompromised patients, and provide a new means to study pathogenesis of MV infection in the CNS.


PLOS ONE | 2009

Interplay between Virus-Specific Effector Response and Foxp3 + Regulatory T Cells in Measles Virus Immunopathogenesis

Caroline I. Sellin; Jean-François Jégou; Joëlle Renneson; Johan Druelle; T. Fabian Wild; Julien C. Marie; Branka Horvat

Measles is a highly contagious childhood disease associated with an immunological paradox: although a strong virus-specific immune response results in virus clearance and the establishment of a life-long immunity, measles infection is followed by an acute and profound immunosuppression leading to an increased susceptibility to secondary infections and high infant mortality. In certain cases, measles is followed by fatal neurological complications. To elucidate measles immunopathology, we have analyzed the immune response to measles virus in mice transgenic for the measles virus receptor, human CD150. These animals are highly susceptible to intranasal infection with wild-type measles strains. Similarly to what has been observed in children with measles, infection of suckling transgenic mice leads to a robust activation of both T and B lymphocytes, generation of virus-specific cytotoxic T cells and antibody responses. Interestingly, Foxp3+CD25+CD4+ regulatory T cells are highly enriched following infection, both in the periphery and in the brain, where the virus intensively replicates. Although specific anti-viral responses develop in spite of increased frequency of regulatory T cells, the capability of T lymphocytes to respond to virus-unrelated antigens was strongly suppressed. Infected adult CD150 transgenic mice crossed in an interferon receptor type I-deficient background develop generalized immunosuppression with an increased frequency of CD4+CD25+Foxp3+ T cells and strong reduction of the hypersensitivity response. These results show that measles virus affects regulatory T-cell homeostasis and suggest that an interplay between virus-specific effector responses and regulatory T cells plays an important role in measles immunopathogenesis. A better understanding of the balance between measles-induced effector and regulatory T cells, both in the periphery and in the brain, may be of critical importance in the design of novel approaches for the prevention and treatment of measles pathology.


Genesis | 2008

Generation of mice with conditionally activated transforming growth factor beta signaling through the TβRI/ALK5 receptor

Laurent Bartholin; Farhan S. Cyprian; David F. Vincent; Celine Garcia; Sylvie Martel; Branka Horvat; Cyril Berthet; Sophie Goddard-Léon; Isabelle Treilleux; Ruth Rimokh; Julien C. Marie

We generated a transgenic mouse strain (LSL‐TβRICA) containing a latent constitutively active TGFβ type I receptor (TβRI/ALK5) by using a knock‐in strategy into the X chromosome‐linked hypoxanthine phosphoribosyl‐transferase (Hprt) locus. Transgene expression, under the control of the ubiquitous CAG (human cytomegalovirus enhancer and chicken β‐actin) promoter, is repressed by a floxed transcriptional “Stop” (LSL, Lox‐Stop‐Lox). In the presence of cre‐recombinase, the “Stop” is excised to allow TβRICA transgene expression. We showed that restricted expression of TβRICA in T lymphocytes efficiently activates TGFβ signaling and rescues the T‐cell autoimmune disorders of TGFβRII conditional knockouts. Unexpectedly, our study reveals that TGFβ signaling upregulation controls T‐cell activation but does not impair their development or their peripheral homeostasis. In addition to the information provided on TGFβ effects on T‐cell biology, LSL‐TβRICA mouse constitutes an attractive tool to address the effect of TGFβ signaling upregulation in any cell type expressing the cre‐recombinase. genesis 46:724–731, 2008. Published 2008 Wiley‐Liss, Inc.

Collaboration


Dive into the Julien C. Marie's collaboration.

Top Co-Authors

Avatar

Branka Horvat

École normale supérieure de Lyon

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Isabelle Treilleux

École normale supérieure de Lyon

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marie-Claude Trescol-Biémont

École normale supérieure de Lyon

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Farhan S. Cyprian

École normale supérieure de Lyon

View shared research outputs
Researchain Logo
Decentralizing Knowledge