6 research outputs found

    NKG2D and Its Ligand MULT1 Contribute to Disease Progression in a Mouse Model of Multiple Sclerosis

    Get PDF
    NKG2D is an activating receptor expressed on the surface of immune cells including subsets of T lymphocytes. NKG2D binds multiple ligands (NKG2DL) whose expression are differentially triggered in a cell type and stress specific manner. The NKG2D-NKG2DL interaction has been involved in autoimmune disorders but its role in animal models of multiple sclerosis (MS) remains incompletely resolved. Here we show that NKG2D and its ligand MULT1 contribute to the pathobiology of experimental autoimmune encephalomyelitis (EAE). MULT1 protein levels are increased in the central nervous system (CNS) at EAE disease peak; soluble MULT1 is elevated in the cerebrospinal fluid of both active and passive EAE. We establish that such soluble MULT1 enhances effector functions (e.g., IFNγ production) of activated CD8 T lymphocytes from wild type but not from NKG2D-deficient (Klrk1−/−) mice in vitro. The adoptive transfer of activated T lymphocytes from wild type donors induced a significantly reduced EAE disease in Klrk1−/− compared to wild type (Klrk1+/+) recipients. Characterization of T lymphocytes infiltrating the CNS of recipient mice shows that donor (CD45.1) rather than endogenous (CD45.2) CD4 T cells are the main producers of key cytokines (IFNγ, GM-CSF). In contrast, infiltrating CD8 T lymphocytes include mainly endogenous (CD45.2) cells exhibiting effector properties (NKG2D, granzyme B and IFNγ). Our data support the notion that endogenous CD8 T cells contribute to passive EAE pathobiology in a NKG2D-dependent manner. Collectively, our results point to the deleterious role of NKG2D and its MULT1 in the pathobiology of a MS mouse model

    The contribution of NKG2D and its ligands to the pathophysiology of multiple sclerosis

    Full text link
    La sclérose en plaques (SP) est une maladie inflammatoire du système nerveux central (SNC) affectant plus de 2,5 millions de personnes dans le monde. Bien que son étiologie soit inconnue, de nombreuses données supportent la contribution des réponses immunitaires à la maladie. La présence de leucocytes au sein des lésions de démyélinisation est un marqueur neuropathologique de la SP. Les traitements actuels diminuent les exacerbations de la SP mais échouent à arrêter sa progression. Ainsi, il est essentiel d’identifier des nouvelles voies contribuant à la pathologie de la SP. NKG2D est un récepteur co-activateur de cellules effectrices qui participent à la surveillance immunitaire. Cependant, cette voie peut contribuer à l’inflammation et aux lésions tissulaires. Le blocage ou l’élimination du NKG2D réduit la gravité de la maladie dans des modèles animaux de SP. Dans la pathologie humaine, des lymphocytes T CD4+ et CD8+ NKG2D+ sont présents dans les lésions et élevés dans le sang périphérique des patients atteints de la forme cyclique de SP durant une poussée. Au moins un ligand du NKG2D (NKG2DL) est exprimé par des oligodendrocytes dans des lésions et les lymphocytes T CD8+ causent la mort in vitro des oligodendrocytes humains de façon NKG2D dépendante. On ignore encore si d’autres cellules neurales expriment des NKG2DL et sont susceptibles à la reconnaissance par le NKG2D. Dans cette thèse, nous avons investigué la contribution de la voie NKG2D à la pathologie de la SP. La première partie présente la caractérisation de l’expression des NKG2DL au sein du SNC. Nous avons trouvé des niveaux élevés de ULBP4, mais aucun autre ligand, dans les lésions et la matière blanche d’apparence normale chez des patients SP. Nous avons identifié des déclencheurs potentiels de l’expression de ULBP4 par les astrocytes. ULBP4 soluble est détecté dans le liquide céphalo-rachidien, et des essais fonctionnels ont démontré sa capacité à renforcer la sécrétion de cytokines inflammatoires par les lymphocytes T CD8+. Dans la seconde partie, nous avons caractérisé les lymphocytes T exprimant NKG2D dans le sang de patients atteints des formes cycliques et progressives de la SP ainsi que chez des sujets témoins. Bien que nous ayons trouvé des proportions similaires, les lymphocytes T NKG2D+CD4+ de patients SP avaient un phénotype mémoire activé associé aux profils Th1 et Th1/Th17. Les lymphocytes T NKG2D+CD8+ étaient diminués chez les patients atteints de la forme cyclique. Cette population affichait une expression prédominante du granzyme B et manifestait des capacités de dégranulation envers les astrocytes exprimant ULBP4. Finalement, les analyses d’imagerie en temps réel ont révélé un rôle pour NKG2D et son ligand ULBP4 dans les interactions durables entre les astrocytes et les lymphocytes T CD8+ humains. Nos travaux identifient un nouveau mécanisme dans le dialogue entre ces types cellulaires. Globalement, ce projet de thèse présente une caractérisation en profondeur de la voie NKG2D dans la SP. De plus, il fournit de nouvelles preuves quant à l’implication de ULBP4 dans la pathophysiologie de la SP. De nouvelles investigations contribueront à élucider la validité de ULBP4 en tant que cible thérapeutique.Multiple sclerosis (MS) is a neuroinflammatory disease of the central nervous system (CNS) that affects more than 2.5 million people worldwide. Despite its unknown etiology, numerous evidences point to aberrant immune responses that contribute to the typical tissue damage. Indeed, the presence of infiltrating immune cells within the characteristic focal demyelinating lesions is a pathological hallmark of MS. Current treatments, which target the immune system, generally control disease exacerbations, but have failed to stop progression. Therefore, it is essential to identify common immune pathways that contribute to MS pathology. NKG2D is a co-activating receptor of immune cells that plays a critical role in immune surveillance. Nevertheless, aberrant NKG2D-mediated responses can contribute to inflammation and tissue damage. Various studies have implicated the NKG2D pathway in MS. NKG2D blocking or depletion reduced disease severity in various EAE models, a commonly used animal model of MS. In the human pathology, NKG2D+CD4+ and CD8+ T lymphocytes have been found in MS lesions and are upregulated in the peripheral blood of RRMS patients under relapse. Moreover, at least one NKG2DL has been observed in oligodendrocytes from MS lesions, which were found near CD8+ T lymphocytes. Furthermore, in vitro studies have demonstrated NKG2D-dependent killing of human oligodendrocytes by CD8+ T lymphocytes. Whether other neural cells express NKG2DL and can thus be susceptible to NKG2D-mediated recognition was still unknown. In this thesis, we investigated further the contribution of the NKG2D pathway to the pathobiology of MS. The first part of this project consisted in the evaluation of NKG2DL expression within the CNS. We found upregulated levels of ULBP4, and no other NKG2DL, in MS lesions and normal appearing white matter from MS patients. Moreover, we identified potential triggers observed in MS lesions that could impact on ULBP4 expression. Soluble ULBP4 was also found in the cerebrospinal fluid, and functional assays demonstrated its capacity to boost inflammatory cytokines secretion by CD8+ T lymphocytes. In the second part, we performed a deep characterization of CD4+ and CD8+ T lymphocytes expressing NKG2D in blood samples from relapsing-remitting and progressive forms of MS as well as age and sex matched healthy controls. Despite finding similar proportions, NKG2D+CD4+ T lymphocytes from MS patients exhibited an activated memory phenotype associated with Th1 and Th1/Th17 responses. In contrast, NKG2D+CD8+ T lymphocytes were reduced in RRMS patients. This subset displayed a predominant granzyme B expression irrespective of the donors’ group, and exhibited degranulating capacities toward ULBP4-expressing astrocytes. Finally, live imaging analysis revealed a role for NKG2D and its ligand ULBP4 in the establishment of long-lasting interaction between astrocytes and CD8+ T lymphocytes. This provides a new mechanism involved in the dialogue between these cell types. Overall, this thesis project provides a deep characterization of the NKG2D pathway in relapsing-remitting and progressive MS patients. Moreover, it provides new evidence for the involvement of ULBP4, a specific NKG2DL, in the pathophysiology of MS. Further investigations will contribute to elucidate the validity of ULBP4 as a therapeutic target in MS

    Methamphetamine-Induced Toxicity in Indusium Griseum of Mice is Associated with Astro- and Microgliosis

    No full text
    © 2014, Springer Science+Business Media New York. The indusium griseum (IG), a thin layer of gray matter in contact with the dorsal surface of the corpus callosum and the lateral gray matter of the cingulate gyrus, has a common origin with hippocampus and shows similar organization with the dentate gyrus. Although some studies have examined the effect of methamphetamine (METH), an addictive and an illegal psychostimulant on this structure, quantitative effects and possible mechanism of actions of METH in this area are lacking. By applying two different protocols of equivalent METH administration (i.e., a high dose of 1 × 30 mg/kg and a lower and repeated injection dose of 3 × 10 mg/kg) and using a specific silver staining method in mice, we demonstrate that this drug produces degeneration in IG with both protocols, without affecting the dopaminergic system. Moreover, we observed quantitative increases in labeling of GFAP and Iba-1, markers of astro- and microgliosis, respectively, which suggest astrogliosis and microgliosis. Thus, our study provides morphological and semi-quantitative evidence that METH induces neurodegeneration in IG and that this damage is associated with astrogliosis and microgliosis in this area.Peer Reviewe

    Identification of SARS-CoV-2-specific immune alterations in acutely ill patients

    No full text
    Dysregulated immune profiles have been described in symptomatic patients infected with SARS-CoV-2. Whether the reported immune alterations are specific to SARS-CoV-2 infection or also triggered by other acute illnesses remains unclear. We performed flow cytometry analysis on fresh peripheral blood from a consecutive cohort of (a) patients hospitalized with acute SARS-CoV-2 infection, (b) patients of comparable age and sex hospitalized for another acute disease (SARS-CoV-2 negative), and (c) healthy controls. Using both data-driven and hypothesis-driven analyses, we found several dysregulations in immune cell subsets (e.g., decreased proportion of T cells) that were similarly associated with acute SARS-CoV-2 infection and non-COVID-19-related acute illnesses. In contrast, we identified specific differences in myeloid and lymphocyte subsets that were associated with SARS-CoV-2 status (e.g., elevated proportion of ICAM-1+ mature/activated neutrophils, ALCAM+ monocytes, and CD38+CD8+ T cells). A subset of SARS-CoV-2-specific immune alterations correlated with disease severity, disease outcome at 30 days, and mortality. Our data provide an understanding of the immune dysregulation specifically associated with SARS-CoV-2 infection among acute care hospitalized patients. Our study lays the foundation for the development of specific biomarkers to stratify SARS-CoV-2-positive patients at risk of unfavorable outcomes and to uncover candidate molecules to investigate from a therapeutic perspective
    corecore