7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Myeloid‐derived suppressor cells support remyelination in a murine model of multiple sclerosis by promoting oligodendrocyte precursor cell survival, proliferation, and differentiation

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The most frequent variant of multiple sclerosis (MS) is the relapsing–remitting form, characterized by symptomatic phases followed by periods of total/partial recovery. Hence, it is possible that these patients can benefit from endogenous agents that control the inflammatory process and favor spontaneous remyelination. In this context, there is increasing interest in the role of myeloid‐derived suppressor cells (MDSCs) during the clinical course of experimental autoimmune encephalomyelitis (EAE). MDSCs speed up infiltrated T‐cell anergy and apoptosis. In different animal models of MS, a milder disease course is related to higher presence/density of MDSCs in the periphery, and smaller demyelinated lesions in the central nervous system (CNS). These observations lead us to wonder whether MDSCs might not only exert an anti‐inflammatory effect but might also have direct influence on oligodendrocyte precursor cells (OPCs) and remyelination. In the present work, we reveal for the first time the relationship between OPCs and MDSCs in EAE, relationship that is guided by the distance from the inflammatory core. We describe the effects of MDSCs on survival, proliferation, as well as potent promoters of OPC differentiation toward mature phenotypes. We show for the first time that osteopontin is remarkably present in the analyzed secretome of MDSCs. The ablation of this cue from MDSCs‐secretome demonstrates that osteopontin is the main MDSC effector on these oligodendroglial cells. These data highlight a crucial pathogenic interaction between innate immunity and the CNS, opening ways to develop MDSC‐ and/or osteopontin‐based therapies to promote effective myelin preservation and repair in MS patients.

          Main Points

          • The abundance of myeloid‐derived suppressor cells (MDSCs) correlates with the density of oligodendrocyte precursor cells (OPCs) in experimental autoimmune encephalomyelitis demyelinating lesions.

          • MDSC secretome favors OPC survival, proliferation, and differentiation toward myelinating phenotypes.

          • Osteopontin is crucial in MDSCs secretome to act on OPC biology.

          Related collections

          Most cited references82

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards

          Myeloid-derived suppressor cells (MDSC) are a heterogeneous population expanded in cancer and other chronic inflammatory conditions. Here the authors identify the challenges and propose a set of minimal reporting guidelines for mouse and human MDSC.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Competing waves of oligodendrocytes in the forebrain and postnatal elimination of an embryonic lineage.

            The developmental origin of oligodendrocyte progenitors (OLPs) in the forebrain has been controversial. We now show, by Cre-lox fate mapping in transgenic mice, that the first OLPs originate in the medial ganglionic eminence (MGE) and anterior entopeduncular area (AEP) in the ventral forebrain. From there, they populate the entire embryonic telencephalon including the cerebral cortex before being joined by a second wave of OLPs from the lateral and/or caudal ganglionic eminences (LGE and CGE). Finally, a third wave arises within the postnatal cortex. When any one population is destroyed at source by the targeted expression of diphtheria toxin, the remaining cells take over and the mice survive and behave normally, with a normal complement of oligodendrocytes and myelin. Thus, functionally redundant populations of OLPs compete for space in the developing brain. Notably, the embryonic MGE- and AEP-derived population is eliminated during postnatal life, raising questions about the nature and purpose of the competition.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination.

              The lack of therapies for progressive multiple sclerosis highlights the need to understand the regenerative process of remyelination that can follow CNS demyelination. This involves an innate immune response consisting of microglia and macrophages, which can be polarized to distinct functional phenotypes: pro-inflammatory (M1) and anti-inflammatory or immunoregulatory (M2). We found that a switch from an M1- to an M2-dominant response occurred in microglia and peripherally derived macrophages as remyelination started. Oligodendrocyte differentiation was enhanced in vitro with M2 cell conditioned media and impaired in vivo following intra-lesional M2 cell depletion. M2 cell densities were increased in lesions of aged mice in which remyelination was enhanced by parabiotic coupling to a younger mouse and in multiple sclerosis lesions that normally show remyelination. Blocking M2 cell-derived activin-A inhibited oligodendrocyte differentiation during remyelination in cerebellar slice cultures. Thus, our results indicate that M2 cell polarization is essential for efficient remyelination and identify activin-A as a therapeutic target for CNS regeneration.
                Bookmark

                Author and article information

                Contributors
                fdecastro@cajal.csic.es
                dclemente@sescam.jccm.es
                Journal
                Glia
                Glia
                10.1002/(ISSN)1098-1136
                GLIA
                Glia
                John Wiley & Sons, Inc. (Hoboken, USA )
                0894-1491
                1098-1136
                20 November 2020
                April 2021
                : 69
                : 4 ( doiID: 10.1002/glia.v69.4 )
                : 905-924
                Affiliations
                [ 1 ] Instituto Cajal‐CSIC Madrid Spain
                [ 2 ] Grupo de Neuroinmuno‐Reparación Hospital Nacional de Parapléjicos‐SESCAM Toledo Spain
                Author notes
                [*] [* ] Correspondence

                Fernando de Castro, Grupo de Neurobiología del Desarrollo‐GNDe, Instituto Cajal‐CSIC, Avda. Dr Arce 37, 28002 Madrid, Spain.

                Email: fdecastro@ 123456cajal.csic.es

                Diego Clemente, Grupo de Neuroinmuno‐Reparación, Hospital Nacional de Parapléjicos, SESCAM, Finca “La Peraleda” s/n, 45071 Toledo, Spain.

                Email: dclemente@ 123456sescam.jccm.es

                Carolina Melero‐Jerez and Beatriz Fernández‐Gómez have equally contributed to this work.

                Diego Clemente and Fernando de Castro are both co‐corresponding authors and have equally contributed to this work.

                Author information
                https://orcid.org/0000-0001-9039-889X
                https://orcid.org/0000-0003-4829-9497
                https://orcid.org/0000-0002-7018-8032
                Article
                GLIA23936
                10.1002/glia.23936
                7894183
                33217041
                b465d656-7911-455e-b5b9-155115ca13c6
                © 2020 The Authors. GLIA published by Wiley Periodicals LLC.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.

                History
                : 01 April 2020
                : 05 November 2020
                : 06 November 2020
                Page count
                Figures: 10, Tables: 1, Pages: 20, Words: 14061
                Funding
                Funded by: AELEM
                Funded by: ADEMTO
                Funded by: ATORDEM
                Funded by: Consejo Superior de Investigaciones Científicas , open-funder-registry 10.13039/501100003339;
                Award ID: 2019AEP033
                Award ID: CSIC‐2015201023
                Award ID: LINKA20268
                Award ID: PID2019‐109858RB‐100
                Funded by: Fondation pour l'Aide à la Recherche sur la Sclérose en Plaques , open-funder-registry 10.13039/100007379;
                Funded by: Instituto de Salud Carlos III , open-funder-registry 10.13039/501100004587;
                Award ID: PI15‐00963
                Award ID: PI18‐00357
                Award ID: RD12‐0032‐12
                Award ID: RD16‐0015‐0019
                Funded by: Spanish Ministerio de Economía, Industria y Competitividad‐MINEICO
                Award ID: SAF2015‐72325‐EXP
                Award ID: SAF2012‐40023
                Award ID: SAF2016‐77575‐R
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                April 2021
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.9.7 mode:remove_FC converted:19.02.2021

                Neurosciences
                eae,myelin,myelination,neural repair,neuroimmunology,neuropathology,neuroregeneration,opc,osteopontin

                Comments

                Comment on this article