13 research outputs found

    Androgens show sex-dependent differences in myelination in immune and non-immune murine models of CNS demyelination

    Get PDF
    Abstract Neuroprotective, anti-inflammatory, and remyelinating properties of androgens are well-characterized in demyelinated male mice and men suffering from multiple sclerosis. However, androgen effects mediated by the androgen receptor (AR), have been only poorly studied in females who make low androgen levels. Here, we show a predominant microglial AR expression in demyelinated lesions from female mice and women with multiple sclerosis, but virtually undetectable AR expression in lesions from male animals and men with multiple sclerosis. In female mice, androgens and estrogens act in a synergistic way while androgens drive microglia response towards regeneration. Transcriptomic comparisons of demyelinated mouse spinal cords indicate that, regardless of the sex, androgens up-regulate genes related to neuronal function integrity and myelin production. Depending on the sex, androgens down-regulate genes related to the immune system in females and lipid catabolism in males. Thus, androgens are required for proper myelin regeneration in females and therapeutic approaches of demyelinating diseases need to consider male-female differences

    Role of Purinergic P2X7 and Apoptosis Fas Receptors in T Lymphocyte Homeostasis and Autoimmune Desease Developpement

    No full text
    Mon Ă©tude a portĂ© sur le rĂŽle du rĂ©cepteur purinergique P2X7 (P2X7R) dans les processus physiopathologiques impliquĂ©s dans le dĂ©veloppement des maladies auto-immunes de type lupique. Les souris MRL/lpr, dĂ©ficientes en rĂ©cepteurs d’apoptose Fas (mutation lpr), dĂ©veloppent spontanĂ©ment ces pathologies suite Ă  l’accumulation lymphocytes T pathogĂ©niques CD4−CD8− (DN) B220+ dans les organes lymphoĂŻdes secondaires. Nous avons observĂ© que ces lymphocytes ont Ă©galement un dĂ©ficit d’expression en P2X7R Ă  leur surface. Cela nous a amenĂ© Ă  postuler que P2X7R pourrait jouer un rĂŽle clĂ© dans l’homĂ©ostasie des lymphocytes T et le dĂ©veloppement du lupus. Afin de vĂ©rifier notre hypothĂšse, nous avons produit des souris C57BL/6J (B6) dĂ©ficientes simultanĂ©ment pour Fas (lpr) et P2X7R (P2X7KO). Ces souris prĂ©sentent une accumulation massive de lymphocytes T DN B220+ et des titres trĂšs Ă©levĂ©s en auto-anticorps et en cytokines proinflammatoires ce qui n’est pas le cas pour les souris B6 simples mutantes lpr ou P2X7KO confirmant pour la premiĂšre fois l’implication de P2X7R dans l’homĂ©ostasie des lymphocytes T, en synergie avec le rĂ©cepteur Fas. Les lymphocytes T DN pathogĂ©niques responsables de la lymphoaccumulation sont issues majoritairement de la sous populations des lymphocytes T CD8+. L’inflammation chronique prĂ©sente chez les souris B6/lpr P2X7KO induit l’activation de l’ensemble des populations lymphocytaires T CD4+ et CD8+ naĂŻves conduisant Ă  l’accumulation de lymphocytes T Effecteurs/MĂ©moires : EM et CM et atteignent parfois le stade exhausted PD1+TIM3+. Ces cellules accumulĂ©es CD4+, CD8+ et DN B220+ ont une capacitĂ© de rĂ©activation rĂ©duite. Ce biais fonctionnel et phĂ©notypique a Ă©tĂ© confirmĂ© en comparant la rĂ©ponse immunitaire adaptative anti-adĂ©novirus entre des souris dĂ©ficientes en Fas et/ou P2X7R. Les rĂ©ponses cellulaires et humorales sont moins importantes dans les souris B6/lpr P2X7KO que B6, B6-P2X7KO. Ces rĂ©ponses antivirales sont intermĂ©diaires dans les souris B6/lpr. L’ensemble de ces rĂ©sultats renforcent notre hypothĂšse sur le rĂŽle synergique des rĂ©cepteurs Fas et P2X7R dans l’homĂ©ostasie des lymphocytes T. Le taux d’apoptose induit par l’activation des rĂ©cepteurs Fas ou P2X7R sĂ©parĂ©ment est moins important dans les lymphocytes T CD8+ par rapport au lymphocytes T CD4+. La synergie Fas-P2X7R serait donc nĂ©cessaire pour l’homĂ©ostasie des lymphocytes T CD8+. Afin de prĂ©ciser les mĂ©canismes Ă  l’origine de la maladie et d’identifier l’influence de chaque rĂ©cepteur sur l’expression des loci de susceptibilitĂ©, nous avons sĂ©quencĂ© les ARNm exprimĂ©s dans la rate et les ganglions lymphatiques des souris MRL/lpr avant et aprĂšs le dĂ©veloppement de l’auto-immunitĂ© ainsi que chez les souris B6, B6/lpr, B6 P2X7KO et B6/lpr P2X7KO.My project aims to determine the role of the purinergic receptor P2X7 (P2X7R) in the pathophysiological processes involved in the development of autoimmune lupus-like syndrome. MRL/lpr mice, deficient for the cell death receptor Fas (lpr mutation), spontaneously develop this pathology following the accumulation of pathogenic B220+CD4−CD8− (DN) T lymphocytes in secondary lymphoid organs. We have observed that these lymphocytes are also deficient in P2X7R cell surface expression. This led us to hypothesize that P2X7R could play a key role in T cell homeostasis and lupus development. To test our hypothesis, we produced B6 mice deficient for both Fas (lpr) and P2X7R (P2X7KO). These mice, but not single mutant B6 mice (lpr or P2X7KO), develop a massive accumulation of DN B220+ T lymphocytes and high levels of autoantibodies and proinflammatory cytokines, confirming for the first time the involvement of P2X7R in T-cell homeostasis. I have found that the pathogenic DN T lymphocytes are predominantly derived from the CD8+ T lymphocyte subpopulation. Chronic inflammation in B6/lpr P2X7KO mice induces the activation of the whole CD4+ and CD8+ naĂŻve T lymphocyte subpopulations leading to the accumulation of Effector/Memory and exhausted T lymphocytes. Accumulated T-cells lose the ability to be reactivated. To confirm these results, I compared the adaptive immune response against adenovirus between mice deficient for Fas (lpr mutation), P2X7R-deficient mice or both receptors. The cellular and the humoral responses were lower in the B6/lpr-P2X7KO mouse strain compared to B6, B6-P2X7KO and B6/lpr mouse strains. The antiviral immune response in the B6/lpr mice was lower than in B6 and B6-P2X7KO mice. These results reinforce our hypothesis about the synergistic role of both receptors in the maintaining of T cell homeostasis. Ours results suggest that Fas and P2X7R play their synergistic role in T-cell homeostasis. In collaboration with a team from the University of Taiwan, we sequenced the mRNAs expressed in the spleen and lymph nodes of MRL/lpr mice before and after the onset of the diseases as well as in the B6, B6/lpr, B6 P2X7KO and B6/lpr P2X7KO mouse strains in order to better understand the mechanism triggering the disease and to identify the role of each receptor on the expression of the susceptibility loci

    RÎle des récepteurs purinergiques P2X7 et d'apoptose Fas dans l'homéostasie des lymphocytes T et le développement des maladies auto-immunes.

    No full text
    My project aims to determine the role of the purinergic receptor P2X7 (P2X7R) in the pathophysiological processes involved in the development of autoimmune lupus-like syndrome. MRL/lpr mice, deficient for the cell death receptor Fas (lpr mutation), spontaneously develop this pathology following the accumulation of pathogenic B220+CD4−CD8− (DN) T lymphocytes in secondary lymphoid organs. We have observed that these lymphocytes are also deficient in P2X7R cell surface expression. This led us to hypothesize that P2X7R could play a key role in T cell homeostasis and lupus development. To test our hypothesis, we produced B6 mice deficient for both Fas (lpr) and P2X7R (P2X7KO). These mice, but not single mutant B6 mice (lpr or P2X7KO), develop a massive accumulation of DN B220+ T lymphocytes and high levels of autoantibodies and proinflammatory cytokines, confirming for the first time the involvement of P2X7R in T-cell homeostasis. I have found that the pathogenic DN T lymphocytes are predominantly derived from the CD8+ T lymphocyte subpopulation. Chronic inflammation in B6/lpr P2X7KO mice induces the activation of the whole CD4+ and CD8+ naĂŻve T lymphocyte subpopulations leading to the accumulation of Effector/Memory and exhausted T lymphocytes. Accumulated T-cells lose the ability to be reactivated. To confirm these results, I compared the adaptive immune response against adenovirus between mice deficient for Fas (lpr mutation), P2X7R-deficient mice or both receptors. The cellular and the humoral responses were lower in the B6/lpr-P2X7KO mouse strain compared to B6, B6-P2X7KO and B6/lpr mouse strains. The antiviral immune response in the B6/lpr mice was lower than in B6 and B6-P2X7KO mice. These results reinforce our hypothesis about the synergistic role of both receptors in the maintaining of T cell homeostasis. Ours results suggest that Fas and P2X7R play their synergistic role in T-cell homeostasis. In collaboration with a team from the University of Taiwan, we sequenced the mRNAs expressed in the spleen and lymph nodes of MRL/lpr mice before and after the onset of the diseases as well as in the B6, B6/lpr, B6 P2X7KO and B6/lpr P2X7KO mouse strains in order to better understand the mechanism triggering the disease and to identify the role of each receptor on the expression of the susceptibility loci.Mon Ă©tude a portĂ© sur le rĂŽle du rĂ©cepteur purinergique P2X7 (P2X7R) dans les processus physiopathologiques impliquĂ©s dans le dĂ©veloppement des maladies auto-immunes de type lupique. Les souris MRL/lpr, dĂ©ficientes en rĂ©cepteurs d’apoptose Fas (mutation lpr), dĂ©veloppent spontanĂ©ment ces pathologies suite Ă  l’accumulation lymphocytes T pathogĂ©niques CD4−CD8− (DN) B220+ dans les organes lymphoĂŻdes secondaires. Nous avons observĂ© que ces lymphocytes ont Ă©galement un dĂ©ficit d’expression en P2X7R Ă  leur surface. Cela nous a amenĂ© Ă  postuler que P2X7R pourrait jouer un rĂŽle clĂ© dans l’homĂ©ostasie des lymphocytes T et le dĂ©veloppement du lupus. Afin de vĂ©rifier notre hypothĂšse, nous avons produit des souris C57BL/6J (B6) dĂ©ficientes simultanĂ©ment pour Fas (lpr) et P2X7R (P2X7KO). Ces souris prĂ©sentent une accumulation massive de lymphocytes T DN B220+ et des titres trĂšs Ă©levĂ©s en auto-anticorps et en cytokines proinflammatoires ce qui n’est pas le cas pour les souris B6 simples mutantes lpr ou P2X7KO confirmant pour la premiĂšre fois l’implication de P2X7R dans l’homĂ©ostasie des lymphocytes T, en synergie avec le rĂ©cepteur Fas. Les lymphocytes T DN pathogĂ©niques responsables de la lymphoaccumulation sont issues majoritairement de la sous populations des lymphocytes T CD8+. L’inflammation chronique prĂ©sente chez les souris B6/lpr P2X7KO induit l’activation de l’ensemble des populations lymphocytaires T CD4+ et CD8+ naĂŻves conduisant Ă  l’accumulation de lymphocytes T Effecteurs/MĂ©moires : EM et CM et atteignent parfois le stade exhausted PD1+TIM3+. Ces cellules accumulĂ©es CD4+, CD8+ et DN B220+ ont une capacitĂ© de rĂ©activation rĂ©duite. Ce biais fonctionnel et phĂ©notypique a Ă©tĂ© confirmĂ© en comparant la rĂ©ponse immunitaire adaptative anti-adĂ©novirus entre des souris dĂ©ficientes en Fas et/ou P2X7R. Les rĂ©ponses cellulaires et humorales sont moins importantes dans les souris B6/lpr P2X7KO que B6, B6-P2X7KO. Ces rĂ©ponses antivirales sont intermĂ©diaires dans les souris B6/lpr. L’ensemble de ces rĂ©sultats renforcent notre hypothĂšse sur le rĂŽle synergique des rĂ©cepteurs Fas et P2X7R dans l’homĂ©ostasie des lymphocytes T. Le taux d’apoptose induit par l’activation des rĂ©cepteurs Fas ou P2X7R sĂ©parĂ©ment est moins important dans les lymphocytes T CD8+ par rapport au lymphocytes T CD4+. La synergie Fas-P2X7R serait donc nĂ©cessaire pour l’homĂ©ostasie des lymphocytes T CD8+. Afin de prĂ©ciser les mĂ©canismes Ă  l’origine de la maladie et d’identifier l’influence de chaque rĂ©cepteur sur l’expression des loci de susceptibilitĂ©, nous avons sĂ©quencĂ© les ARNm exprimĂ©s dans la rate et les ganglions lymphatiques des souris MRL/lpr avant et aprĂšs le dĂ©veloppement de l’auto-immunitĂ© ainsi que chez les souris B6, B6/lpr, B6 P2X7KO et B6/lpr P2X7KO

    Hypercholesterolemia Negatively Regulates P2X7-Induced Cellular Function in CD4<sup>+</sup> and CD8<sup>+</sup> T-Cell Subsets from B6 Mice Fed a High-Fat Diet

    No full text
    We have previously showed that plasma membrane cholesterol and GM1 ganglioside content are responsible for the opposite sensitivity of mouse leukemic T cells to ATP. We also reported that the sensitivity of CD4+ and CD8+ T cells to ATP depends on their stage of differentiation. Here, we show that CD4+ and CD8+ T cells from B6 mice express different levels of membrane GM1 and P2X7 but similar levels of cholesterol. Thus, in CD4+ T cells, membrane cholesterol content negatively correlated with ATP/P2X7-induced CD62L shedding but positively correlated with pore formation, phosphatidylserine externalization, and cell death. By contrast, in CD8+ T cells, cholesterol, GM1, and P2X7 levels negatively correlated with all these ATP/P2X7-induced cellular responses. The relationship between cholesterol and P2X7-induced cellular responses was confirmed by modulating cholesterol levels either ex vivo or through a high-fat diet. Membrane cholesterol enrichment ex vivo led to a significant reduction in all P2X7-induced cellular responses in T cells. Importantly, diet-induced hypercholesterolemia in B6 mice was also associated with decreased sensitivity to ATP in CD4+ and CD8+ T cells, highlighting the relationship between cholesterol intake and the amplitudes of P2X7-induced cellular responses in T cells

    Coopération fonctionnelle des voies de signalisation Hedgehog et des androgÚnes au cours de la myélinisation développementale et réparatrice

    No full text
    International audienceAbstract Hedgehog morphogens control fundamental cellular processes during tissue development and regeneration. In the central nervous system (CNS), Hedgehog signaling has been implicated in oligodendrocyte and myelin production, where it functions in a concerted manner with other pathways. Since androgen receptor (AR) plays a key role in establishing the sexual phenotype of myelin during development and is required for spontaneous myelin regeneration in the adult CNS, we hypothesized the existence of a possible coordination between Hedgehog and androgen signals in oligodendrocyte and myelin production. Here, we report complementary activities of both pathways during early postnatal oligodendrogenesis further revealing that persistent Hedgehog signaling activation impedes myelin production. The data also uncover prominent pro‐myelinating activity of testosterone and involvement of AR in the control of neural stem cell commitment toward the oligodendroglial lineage. In the context of CNS demyelination, we provide evidence for the functional cooperation of the pathways leading to acceleration of myelin regeneration that might be related to their respective role on microglial and astroglial responses, higher preservation of axonal integrity, lower neuroinflammation, and functional improvement of animals in an immune model of CNS demyelination. Strong decreases of deleterious cytokines in the CNS (GM‐CSF, TNF‐α, IL‐17A) and spleen (IL‐2, IFN‐γ) stand as unique features of the combined drugs while the potent therapeutic activity of testosterone on peripheral immune cells contributes to increase tolerogenic CD11c + dendritic cells, reduce the clonal expansion of conventional CD4 + T cells and increase CD4 + Foxp3 + regulatory T cells. Altogether, these data might open promising perspectives for demyelinating diseases

    Variations in Cellular Responses of Mouse T Cells to Adenosine-5'-Triphosphate Stimulation Do Not Depend on P2X7 Receptor Expression Levels but on Their Activation and Differentiation Stage

    No full text
    International audienceA previous report has shown that regulatory T cells (Treg) were markedly more sensitive to adenosine-5'-triphosphate (ATP) than conventional T cells (Tconv). Another one has shown that Tregs and CD45RBlowTconvs, but not CD45RBhighTconvs, displayed similar high sensitivity to ATP. We have previously reported that CD45RBlowTconvs expressing B220/CD45RABC molecules in a pre-apoptotic stage are resistant to ATP stimulation due to the loss of P2X7 receptor (P2X7R) membrane expression. To gain a clearer picture on T-cell sensitivity to ATP, we have quantified four different cellular activities triggered by ATP in mouse T cells at different stages of activation/differentiation, in correlation with levels of P2X7R membrane expression. P2X7R expression significantly increases on Tconvs during differentiation from naive CD45RBhighCD44lowto effector/memory CD45RBlowCD44highstage. Maximum levels of upregulation are reached on recently activated CD69+naive and memory Tconvs. Ectonucleotidases CD39 and CD73 expression levels increase in parallel with those of P2X7R. Recently activated CD69+CD45RBhighCD44lowTconvs, although expressing high levels of P2X7R, fail to cleave homing receptor CD62L after ATP treatment, but efficiently form pores and externalize phosphatidylserine (PS). In contrast, naive CD45RBhighCD44lowTconvs cleave CD62L with high efficiency although they express a lower level of P2X7, thus suggesting that P2X7R levels are not a limiting factor for signaling ATP-induced cellular responses. Contrary to common assumption, P2X7R-mediated cellular activities in mouse Tconvs are not triggered in an all-or-none manner, but depend on their stage of activation/differentiation. Compared to CD45RBlowTconvs, CD45RBlowFoxp3+Tregs show significantly higher levels of P2X7R membrane expression and of sensitivity to ATP as evidenced by their high levels of CD62L shedding, pore formation and PS externalization observed after ATP treatment. In summary, the different abilities of ATP-treated Tconvs to form pore or cleave CD62L depending on their activation and differentiation state suggests that P2X7R signaling varies according to the physiological role of T convs during antigen activation in secondary lymphoid organs or trafficking to inflammatory sites

    Variations in Cellular Responses of Mouse T Cells to Adenosine-5â€Č-Triphosphate Stimulation Do Not Depend on P2X7 Receptor Expression Levels but on Their Activation and Differentiation Stage

    No full text
    A previous report has shown that regulatory T cells (Treg) were markedly more sensitive to adenosine-5â€Č-triphosphate (ATP) than conventional T cells (Tconv). Another one has shown that Tregs and CD45RBlow Tconvs, but not CD45RBhigh Tconvs, displayed similar high sensitivity to ATP. We have previously reported that CD45RBlow Tconvs expressing B220/CD45RABC molecules in a pre-apoptotic stage are resistant to ATP stimulation due to the loss of P2X7 receptor (P2X7R) membrane expression. To gain a clearer picture on T-cell sensitivity to ATP, we have quantified four different cellular activities triggered by ATP in mouse T cells at different stages of activation/differentiation, in correlation with levels of P2X7R membrane expression. P2X7R expression significantly increases on Tconvs during differentiation from naive CD45RBhighCD44low to effector/memory CD45RBlowCD44high stage. Maximum levels of upregulation are reached on recently activated CD69+ naive and memory Tconvs. Ectonucleotidases CD39 and CD73 expression levels increase in parallel with those of P2X7R. Recently activated CD69+ CD45RBhighCD44low Tconvs, although expressing high levels of P2X7R, fail to cleave homing receptor CD62L after ATP treatment, but efficiently form pores and externalize phosphatidylserine (PS). In contrast, naive CD45RBhighCD44low Tconvs cleave CD62L with high efficiency although they express a lower level of P2X7, thus suggesting that P2X7R levels are not a limiting factor for signaling ATP-induced cellular responses. Contrary to common assumption, P2X7R-mediated cellular activities in mouse Tconvs are not triggered in an all-or-none manner, but depend on their stage of activation/differentiation. Compared to CD45RBlow Tconvs, CD45RBlowFoxp3+ Tregs show significantly higher levels of P2X7R membrane expression and of sensitivity to ATP as evidenced by their high levels of CD62L shedding, pore formation and PS externalization observed after ATP treatment. In summary, the different abilities of ATP-treated Tconvs to form pore or cleave CD62L depending on their activation and differentiation state suggests that P2X7R signaling varies according to the physiological role of T convs during antigen activation in secondary lymphoid organs or trafficking to inflammatory sites

    DataSheet_1_P2X7 purinergic receptor plays a critical role in maintaining T-cell homeostasis and preventing lupus pathogenesis.docx

    No full text
    The severe lymphoproliferative and lupus diseases developed by MRL/lpr mice depend on interactions between the Faslpr mutation and MRL genetic background. Thus, the Faslpr mutation causes limited disease in C57BL/6 mice. We previously found that accumulating B220+ CD4–CD8– double negative (DN) T cells in MRL/lpr mice show defective P2X7 receptor ( P2X7)-induced cellular functions, suggesting that P2X7 contributes to T-cell homeostasis, along with Fas. Therefore, we generated a B6/lpr mouse strain (called B6/lpr-p2x7KO) carrying homozygous P2X7 knockout alleles. B6/lpr-p2x7KO mice accumulated high numbers of FasL-expressing B220+ DN T cells of CD45RBhighCD44high effector/memory CD8+ T-cell origin and developed severe lupus, characterized by leukocyte infiltration into the tissues, high levels of IgG anti-dsDNA and rheumatoid factor autoantibodies, and marked cytokine network dysregulation. B6/lpr-p2x7KO mice also exhibited a considerably reduced lifespan. P2X7 is therefore a novel regulator of T-cell homeostasis, of which cooperation with Fas is critical to prevent lymphoaccumulation and autoimmunity.</p

    Image_1.tif

    No full text
    <p>A previous report has shown that regulatory T cells (Treg) were markedly more sensitive to adenosine-5â€Č-triphosphate (ATP) than conventional T cells (Tconv). Another one has shown that Tregs and CD45RB<sup>low</sup> Tconvs, but not CD45RB<sup>high</sup> Tconvs, displayed similar high sensitivity to ATP. We have previously reported that CD45RB<sup>low</sup> Tconvs expressing B220/CD45RABC molecules in a pre-apoptotic stage are resistant to ATP stimulation due to the loss of P2X7 receptor (P2X7R) membrane expression. To gain a clearer picture on T-cell sensitivity to ATP, we have quantified four different cellular activities triggered by ATP in mouse T cells at different stages of activation/differentiation, in correlation with levels of P2X7R membrane expression. P2X7R expression significantly increases on Tconvs during differentiation from naive CD45RB<sup>high</sup>CD44<sup>low</sup> to effector/memory CD45RB<sup>low</sup>CD44<sup>high</sup> stage. Maximum levels of upregulation are reached on recently activated CD69<sup>+</sup> naive and memory Tconvs. Ectonucleotidases CD39 and CD73 expression levels increase in parallel with those of P2X7R. Recently activated CD69<sup>+</sup> CD45RB<sup>high</sup>CD44<sup>low</sup> Tconvs, although expressing high levels of P2X7R, fail to cleave homing receptor CD62L after ATP treatment, but efficiently form pores and externalize phosphatidylserine (PS). In contrast, naive CD45RB<sup>high</sup>CD44<sup>low</sup> Tconvs cleave CD62L with high efficiency although they express a lower level of P2X7, thus suggesting that P2X7R levels are not a limiting factor for signaling ATP-induced cellular responses. Contrary to common assumption, P2X7R-mediated cellular activities in mouse Tconvs are not triggered in an all-or-none manner, but depend on their stage of activation/differentiation. Compared to CD45RB<sup>low</sup> Tconvs, CD45RB<sup>low</sup>Foxp3<sup>+</sup> Tregs show significantly higher levels of P2X7R membrane expression and of sensitivity to ATP as evidenced by their high levels of CD62L shedding, pore formation and PS externalization observed after ATP treatment. In summary, the different abilities of ATP-treated Tconvs to form pore or cleave CD62L depending on their activation and differentiation state suggests that P2X7R signaling varies according to the physiological role of T convs during antigen activation in secondary lymphoid organs or trafficking to inflammatory sites.</p
    corecore