20
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The EIIIA domain from astrocyte‐derived fibronectin mediates proliferation of oligodendrocyte progenitor cells following CNS demyelination

      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

          Central nervous system remyelination by oligodendrocyte progenitor cells (OPCs) ultimately fails in the majority of multiple sclerosis (MS) lesions. Remyelination benefits from transient expression of factors that promote migration and proliferation of OPCs, which may include fibronectin (Fn). Fn is present in demyelinated lesions in two major forms; plasma Fn (pFn), deposited following blood‐brain barrier disruption, and cellular Fn, synthesized by resident glial cells and containing alternatively spliced domains EIIIA and EIIIB. Here, we investigated the distinctive roles that astrocyte‐derived Fn (aFn) and pFn play in remyelination. We used an inducible Cre‐lox recombination strategy to selectively remove pFn, aFn or both from mice, and examined the impact on remyelination of toxin‐induced demyelinated lesions of spinal cord white matter. This approach revealed that astrocytes are a major source of Fn in demyelinated lesions. Furthermore, following aFn conditional knockout, the number of OPCs recruited to the demyelinated lesion decreased significantly, whereas OPC numbers were unaltered following pFn conditional knockout. However, remyelination completed normally following conditional knockout of aFn and pFn. Both the EIIIA and EIIIB domains of aFn were expressed following demyelination, and in vitro assays demonstrated that the EIIIA domain of aFn mediates proliferation of OPCs, but not migration. Therefore, although the EIIIA domain from aFn mediates OPC proliferation, aFn is not essential for successful remyelination. Since previous findings indicated that astrocyte‐derived Fn aggregates in chronic MS lesions inhibit remyelination, aFn removal may benefit therapeutic strategies to promote remyelination in MS. GLIA 2015;63:242–256

          Main Points

          • We investigated the roles of cellular and plasma fibronectin in remyelination, and found that astrocyte‐derived fibronectin mediates oligodendrocyte progenitor cell proliferation in a toxin‐induced demyelination model, likely via the EIIIA domain.

          Related collections

          Most cited references58

          • Record: found
          • Abstract: found
          • Article: not found

          Dysregulation of the Wnt pathway inhibits timely myelination and remyelination in the mammalian CNS.

          The progressive loss of CNS myelin in patients with multiple sclerosis (MS) has been proposed to result from the combined effects of damage to oligodendrocytes and failure of remyelination. A common feature of demyelinated lesions is the presence of oligodendrocyte precursors (OLPs) blocked at a premyelinating stage. However, the mechanistic basis for inhibition of myelin repair is incompletely understood. To identify novel regulators of OLP differentiation, potentially dysregulated during repair, we performed a genome-wide screen of 1040 transcription factor-encoding genes expressed in remyelinating rodent lesions. We report that approximately 50 transcription factor-encoding genes show dynamic expression during repair and that expression of the Wnt pathway mediator Tcf4 (aka Tcf7l2) within OLPs is specific to lesioned-but not normal-adult white matter. We report that beta-catenin signaling is active during oligodendrocyte development and remyelination in vivo. Moreover, we observed similar regulation of Tcf4 in the developing human CNS and lesions of MS. Data mining revealed elevated levels of Wnt pathway mRNA transcripts and proteins within MS lesions, indicating activation of the pathway in this pathological context. We show that dysregulation of Wnt-beta-catenin signaling in OLPs results in profound delay of both developmental myelination and remyelination, based on (1) conditional activation of beta-catenin in the oligodendrocyte lineage in vivo and (2) findings from APC(Min) mice, which lack one functional copy of the endogenous Wnt pathway inhibitor APC. Together, our findings indicate that dysregulated Wnt-beta-catenin signaling inhibits myelination/remyelination in the mammalian CNS. Evidence of Wnt pathway activity in human MS lesions suggests that its dysregulation might contribute to inefficient myelin repair in human neurological disorders.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Defects in mesoderm, neural tube and vascular development in mouse embryos lacking fibronectin.

            To examine the role of fibronectin in vivo, we have generated mice in which the fibronectin gene is inactivated. Heterozygotes have one half normal levels of plasma fibronectin, yet appear normal. When homozygous, the mutant allele causes early embryonic lethality, proving that fibronectin is required for embryogenesis. However, homozygous mutant embryos implant and initiate gastrulation normally including extensive mesodermal movement. Neural folds also form but the mutant embryos subsequently display shortened anterior-posterior axes, deformed neural tubes and severe defects in mesodermally derived tissues. Notochord and somites are absent; the heart and embryonic vessels are variable and deformed, and the yolk sac, extraembryonic vasculature and amnion are also defective. These abnormalities can be interpreted as arising from fundamental deficits in mesodermal migration, adhesion, proliferation or differentiation as a result of the absence of fibronectin. The nature of these embryonic defects leads to reevaluation of suggested roles for fibronectin during early development based on results obtained in vitro and in embryos of other species.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Plasma and cellular fibronectin: distinct and independent functions during tissue repair

              Fibronectin (FN) is a ubiquitous extracellular matrix (ECM) glycoprotein that plays vital roles during tissue repair. The plasma form of FN circulates in the blood, and upon tissue injury, is incorporated into fibrin clots to exert effects on platelet function and to mediate hemostasis. Cellular FN is then synthesized and assembled by cells as they migrate into the clot to reconstitute damaged tissue. The assembly of FN into a complex three-dimensional matrix during physiological repair plays a key role not only as a structural scaffold, but also as a regulator of cell function during this stage of tissue repair. FN fibrillogenesis is a complex, stepwise process that is strictly regulated by a multitude of factors. During fibrosis, there is excessive deposition of ECM, of which FN is one of the major components. Aberrant FN-matrix assembly is a major contributing factor to the switch from normal tissue repair to misregulated fibrosis. Understanding the mechanisms involved in FN assembly and how these interplay with cellular, fibrotic and immune responses may reveal targets for the future development of therapies to regulate aberrant tissue-repair processes.
                Bookmark

                Author and article information

                Journal
                Glia
                Glia
                10.1002/(ISSN)1098-1136
                GLIA
                Glia
                John Wiley and Sons Inc. (Hoboken )
                0894-1491
                1098-1136
                25 August 2014
                February 2015
                : 63
                : 2 ( doiID: 10.1002/glia.v63.2 )
                : 242-256
                Affiliations
                [ 1 ] Department of Cell BiologyUniversity of Groningen, University Medical Center Groningen The Netherlands
                [ 2 ] Wellcome Trust‐Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences University of Cambridge United Kingdom
                Author notes
                [*] [* ]Address correspondence to Chao Zhao, Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences; University of Cambridge, Clifford Allbutt Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0AH, United Kingdom. E‐mail: cz213@ 123456cam.ac.uk (or) Wia Baron, Department of Cell Biology, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands. E‐mail: w.baron@ 123456umcg.nl
                Article
                GLIA22748
                10.1002/glia.22748
                4737254
                25156142
                a6f0e5f9-556d-4dd7-8816-cbd2d1970f18
                © 2014 The Authors. Glia Published by Wiley Periodicals, Inc.

                This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 12 June 2014
                : 11 August 2014
                Page count
                Pages: 15
                Funding
                Funded by: Netherlands Organization of Scientific Research
                Funded by: Dutch MS Research Foundation
                Funded by: UK MS Society
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                glia22748
                February 2015
                Converter:WILEY_ML3GV2_TO_NLMPMC version:4.8.6 mode:remove_FC converted:19.04.2016

                Neurosciences
                fibronectin,astrocyte,oligodendrocyte,remyelination,multiple sclerosis
                Neurosciences
                fibronectin, astrocyte, oligodendrocyte, remyelination, multiple sclerosis

                Comments

                Comment on this article