20 research outputs found

    Immunosuppression by mesenchymal stem cells: mechanisms and clinical applications

    Get PDF
    Mesenchymal stem cells (MSCs) are multipotential nonhematopoietic progenitor cells that are isolated from many adult tissues, in particular from the bone marrow and adipose tissue. Along with their capacity for differentiating into cells of mesodermal lineage, such as adipocytes, osteoblasts and chondrocytes, these cells have also generated great interest for their ability to display immunomodulatory capacities. Indeed, a major breakthrough came with the finding that they are able to induce peripheral tolerance, suggesting they may be used as therapeutic tools in immune-mediated disorders. The present review aims at discussing the current knowledge on the targets and mechanisms of MSC-mediated immunosuppression as well as the potential use of MSCs as modulators of immune responses in a variety of diseases related to alloreactive immunity or autoimmunit

    IL-6-Dependent PGE2 Secretion by Mesenchymal Stem Cells Inhibits Local Inflammation in Experimental Arthritis

    Get PDF
    BACKGROUND: Based on their capacity to suppress immune responses, multipotent mesenchymal stromal cells (MSC) are intensively studied for various clinical applications. Although it has been shown in vitro that the immunomodulatory effect of MSCs mainly occurs through the secretion of soluble mediators, the mechanism is still not completely understood. The aim of the present study was to better understand the mechanisms underlying the suppressive effect of MSCs in vivo, using cells isolated from mice deficient in the production of inducible nitric oxide synthase (iNOS) or interleukin (IL)-6 in the murine model of collagen-induced arthritis. PRINCIPAL FINDINGS: In the present study, we show that primary murine MSCs from various strains of mice or isolated from mice deficient for iNOS or IL-6 exhibit different immunosuppressive potential. The immunomodulatory function of MSCs was mainly attributed to IL-6-dependent secretion of prostaglandin E2 (PGE2) with a minor role for NO. To address the role of these molecules in vivo, we used the collagen-induced arthritis as an experimental model of immune-mediated disorder. MSCs effectively inhibited collagen-induced inflammation during a narrow therapeutic window. In contrast to wild type MSCs, IL-6-deficient MSCs and to a lesser extent iNOS-deficient MSCs were not able to reduce the clinical signs of arthritis. Finally, we show that, independently of NO or IL-6 secretion or Treg cell induction, MSCs modulate the host response by inducing a switch to a Th2 immune response. SIGNIFICANCE: Our data indicate that mscs mediate their immunosuppressive effect via two modes of action: locally, they reduce inflammation through the secretion of anti-proliferative mediators, such as NO and mainly PGE2, and systemically they switch the host response from a Th1/Th17 towards a Th2 immune profile

    Intérêt thérapeutique des cellules souches mésenchymateuses (applications à la régénération du cartilage articulaire et à une immunothérapie de la polyarthrite rhumatoïde)

    No full text
    Les pathologies ostéoarticulaires dont les plus fréquentes sont la polyarthrite rhumatoïde (PR) et l'arthrose représentent un problème majeur de santé publique. Ces maladies sont caractérisées par la dégradation du cartilage articulaire suite à l'inflammation locale ou l'usure des articulations, respectivement. En absence de régénération spontanée du cartilage, les traitements utilisés reposent essentiellement sur des interventions chirurgicales ne permettant pas une réparation efficace à long terme. C'est pourquoi, des approches innovantes d'immunothérapies visant à inhiber la réponse inflammatoire ou d'ingénierie tissulaire pour régénérer le tissu détruit sont envisagés. Elles reposent notamment sur l'utilisation de cellules souches mésenchymateuses (CSM) qui possèdent des caractéristiques indispensables à la mise en place de stratégies de thérapie cellulaire, notamment leur taux élevé d'amplification ex vivo, leurs proporiétés immunosuppressives et leur potentiel de différenciation. L'objectif de mon projet de thèse a consisté à valider l'intérêt thérapeutique des CSM dans 2 applications de thérapie cellulaire : une approche d'immunothérapie de la PR et une approche d'ingénierie du cartilage articulaire. La première stratégie a permis de démontrer que les CSM sont capables d'inhiber les signes cliniques de l'arthrite expérimentale dans une fenêtre restreinte d'application. Dans ce modèle, les mécanismes d'immunosuppression mis en place par les CSM consistent essentiellement à orienter la réponse immunitaire vers un profil Th2. La seconde stratégie a consisté à tester un nouveau biomatériau, les microsphères phramacologiquement actives (PAM) pour une ingénierie du cartilage. Nous avons montré l'intérêt des PAM pour délivrer progressivement du TGF-b3 et induire spécifiquement in vitro et in vivo la différenciation des CSM en chondrocytesRheumatoid arthrisits and osteoarthrisits are the most prominent osteao-articular diseases representing a major public health problem. Degradation of articular cartilage il the common characteristic of these pathologies which occur following local inflammation or loss of chrondrocyte anabolic activity, respectively. In absence of spontaneous regeneration of this issue, treatments are primarily based on surgical techniques which do not allow effective repair on the long term. That's the reason why, innovative cell therapy approaches are currently being developed in order to inhibit inflammatory responses or to induce cartilage repair using tissues engineering strategies. These approaches rely on the use of mesenchymal stem cells (MSC) which may represent an intersting source of cells thanks to their high rate of expansion ex vivo, their immunosuppressive properties and their differentiation potential. The aim of my PhD project consisted in validating the therapeutic potential of MSC in two cell therapy approaches : a first strategy aiming at modulatin inflammation in RA and a second one for articular cartilage repair. Indeed, the first strategy allowed demonstrating that MSC are able to inhibit the clinical signs of collagen-induced arthrisis when injected during a restricted window of application. In this model, MSC-mediated immunosuppression was shown to act locally, via the secretion of anti-inflammatory mediators and systemically, by the switch of the lost response from a Th1 to a Th2 immune profil. In the second strategy, we evaluated the interest of a new scaffold, pharmacologically active microcarriers (PAM), for cartilage engineering. We showed that PAM are efficient scaffolds to release progressively the TGF-b3 growth factor and can induce specifically in vitro and in vivo differentiation pf MSC into chrondrocytesMONTPELLIER-BU Médecine UPM (341722108) / SudocMONTPELLIER-BU Médecine (341722104) / SudocSudocFranceF

    Phenotypic and functional characterization of primary murine MSCs.

    No full text
    <p>A) Immunophenotype of bone marrow-derived MSCs isolated from C57BL/6 and DBA/1 mice. Cells were stained with antibodies against the indicated antigens and analysed by flow cytometry. Representative histograms are shown in black and the respective isotype controls are shown as grey line and are representative of three experiments. B) Differentiation potential of murine MSCs. The differentiation in the 3 lineages is shown by up-regulation of specific markers by RT-qPCR and specific staining. Osteogenesis is characterized by the expression of osteocalcin (OC), alkaline phosphatase (AP) and Alizarin Red S positive staining in differentiation medium (lower panel) versus proliferative medium (upper panel). Adipogenesis is characterized by the expression of peroxysome proliferator-activated receptor (PPAR)-γ; fatty acid binding protein 4 (aP2) and Oil Red O positive staining in differentiation medium (lower panel) versus proliferative medium (upper panel). Chondrogenesis is characterized by the expression of collagen II (Col II) and aggrecan (Agg) and anti-aggrecan positive staining on pellet sections (lower panel) versus unstained control section (upper panel). Results are representative of three independent experiments. C) Immunosuppressive potential of murine C57BL/6 (left panel) and DBA/1 (right panel) MSCs. Allogeneic splenocytes were incubated for three days with ConA without or with various ratios of MSCs. Results are expressed as the percentage of ConA-induced proliferation which was assigned the value of 100%± SEM (n = 3; ***: p<0.005).</p

    In vitro evaluation of the immunosuppressive potential of MSCs by quantification of soluble mediators.

    No full text
    <p>A) Inhibition of T cell proliferation following co-culture with MSCs. Allogeneic splenocytes (10<sup>5</sup> cells) were incubated for three days with ConA and when indicated, 5×10<sup>4</sup> wildtype (wt) C57Bl/6 or MSCs isolated from iNOS<sup>−/−</sup>, IL-6<sup>−/−</sup> knock-out C57Bl/6 mice or DBA/1 MSCs were added. Results are expressed as the percentage of ConA-induced proliferation which was assigned the value of 100% ± SEM (n = 3). B) Quantification of NO<sub>2</sub> secretion using a modified Griess reagent. C) Quantification of IL-6 secretion by ELISA. D) Quantification of PGE<sub>2</sub> secretion by ELISA. Levels of the various soluble mediators were quantified in supernatants of the proliferative assays performed in A) after stimulation with ConA. Parallel assays were performed on cells incubated in the same conditions with the addition of inhibitors specific for NO<sub>2</sub> (L-NAME) or PGE<sub>2</sub> (indomethacin; Indo). Results are expressed as the mean of 3 independent experiments ± SEM; p values referred to ConA-activated samples (black histograms), if not indicated (*: p<0.05).</p

    Immunosuppression is dependent of the injection time of primary MSCs in experimental arthritis.

    No full text
    <p>A) Evaluation of the increase of the paw swelling of CIA control mice or mice injected with syngeneic D1 MSCs at various time points. Results are expressed as mean increase ± SEM and are representative of three independent experiments. B) Incidence of arthritis evaluating the percentage of mice with a clinical score >1 (see experimental procedures). C) Radiological score evaluated after X-ray examination on hind paws from mice after euthanasia as assessed in experimental procedures. Results are expressed as the mean ± SEM. D) Histological score evaluated on paraffin sections of hind paws stained by hematoxylin-eosin as assessed in experimental procedures. Results are expressed as the mean ± SEM. E) Representative photomicrographs of joints from CIA control mice (left), or mice injected with MSCs on d18 and d24 (right). Magnification ×5. F) Evaluation of the paw swelling in CIA following injection of D1 MSCs after disease onset. Results are representative of two independent experiments (Statistical analysis showed not significance between groups).</p

    Immunosuppressive effect of iNOS<sup>−/−</sup> and IL-6<sup>−/−</sup> MSCs in experimental arthritis.

    No full text
    <p>A) Evaluation of the severity of arthritis by measurement of the increase of the paw swelling of control mice (CIA) or mice injected with wt MSCs, iNOS<sup>−/−</sup> or IL-6<sup>−/−</sup> MSCs at d18 and d24 (see arrows). Results are expressed as mean increase ± SEM and representative of three independent experiments (*: p<0.05 between wt MSC-treated group and CIA group; #: p≤0.05 between wt MSC-treated group and IL-6<sup>−/−</sup> MSC-treated group). B) T-cell proliferation assay using splenocytes (10<sup>5</sup> cells) from the various treated mice incubated with 1, 5 or 25 µg/ml of bCII. C) Determination of the ratio of the bCII-specific IgG1/IgG2a by ELISA in mouse sera. Results are expressed as mean ± SEM. D) Quantification of IL-6 by ELISA in the mouse sera. E) Quantification of cytokines secreted by 2×10<sup>6</sup> splenocytes cultured <i>in vitro</i> without activation (dashed histograms) or stimulated with 10 µg/ml bCII (black histograms) for 24 h (IFN-γ and IL-17) or 48 h (others) by ELISA. Results are expressed as the mean ± SEM (*: p<0.05; **: p<0.01; ***: p<0.005; NS: not significant).</p

    Induction of a Th2 cellular response after treatment with MSCs.

    No full text
    <p>A) Percentage of CD4<sup>+</sup>CD25<sup>+</sup>Foxp3<sup>+</sup> Treg cells in individual spleen, pooled draining lymph nodes (DLN) and individual peripheral blood mononuclear cells (PBMC) from control CIA or MSC-treated mice at day 31. FACS analyses show CD25<sup>+</sup>Foxp3<sup>+</sup> cells gated on CD4<sup>+</sup> T cells in the splenocyte population. B) Percentage of CD3<sup>+</sup>CD8<sup>+</sup>CD28<sup>−</sup> Treg cells in spleen, DLN and PBMC from control CIA or treated mice at day 31. For DLN, cells isolated from all mice in each treated group were pooled resulting in absence of SEM. Results are shown as representative dot plots for 1 mouse per group of mice (upper panel) and histograms in lower panel (% ± SEM), representative of three independent experiments of FACS analyses. C) Relative expression of mRNA level of GATA-3 and Foxp3 transcription factors to GAPDH mRNA in splenocytes using the formula 2<sup>−ΔCt</sup>. Results are expressed as the mean ± SEM (NS: not significant).</p
    corecore