11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Distinct metabolic states govern skeletal muscle stem cell fates during prenatal and postnatal myogenesis

      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

          During growth, homeostasis and regeneration, stem cells are exposed to different energy demands. Here, we characterise the metabolic pathways that mediate the commitment and differentiation of mouse skeletal muscle stem cells, and how their modulation can influence the cell state. We show that quiescent satellite stem cells have low energetic demands and perturbed oxidative phosphorylation during ageing, which is also the case for cells from post-mortem tissues. We show also that myogenic fetal cells have distinct metabolic requirements compared to those proliferating during regeneration, with the former displaying a low respiration demand relying mostly on glycolysis. Furthermore, we show distinct requirements for peroxisomal and mitochondrial fatty acid oxidation (FAO) in myogenic cells. Compromising peroxisomal but not mitochondrial FAO promotes early differentiation of myogenic cells. Acute muscle injury and pharmacological block of peroxisomal and mitochondrial FAO expose differential requirements for these organelles during muscle regeneration. Taken together, these observations indicate that changes in myogenic cell state lead to significant alterations in metabolic requirements. In addition, perturbing specific metabolic pathways impacts on myogenic cell fates and the regeneration process.

          Abstract

          Summary: Distinct energy metabolism pathways act during mouse skeletal muscle stem cell commitment and differentiation in different physiological states.

          Related collections

          Most cited references54

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

          Autophagy maintains stemness by preventing senescence.

          During ageing, muscle stem-cell regenerative function declines. At advanced geriatric age, this decline is maximal owing to transition from a normal quiescence into an irreversible senescence state. How satellite cells maintain quiescence and avoid senescence until advanced age remains unknown. Here we report that basal autophagy is essential to maintain the stem-cell quiescent state in mice. Failure of autophagy in physiologically aged satellite cells or genetic impairment of autophagy in young cells causes entry into senescence by loss of proteostasis, increased mitochondrial dysfunction and oxidative stress, resulting in a decline in the function and number of satellite cells. Re-establishment of autophagy reverses senescence and restores regenerative functions in geriatric satellite cells. As autophagy also declines in human geriatric satellite cells, our findings reveal autophagy to be a decisive stem-cell-fate regulator, with implications for fostering muscle regeneration in sarcopenia.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche.

            Bone marrow transplantation is the primary therapy for numerous hematopoietic disorders. The efficiency of bone marrow transplantation depends on the function of long-term hematopoietic stem cells (LT-HSCs), which is markedly influenced by their hypoxic niche. Survival in this low-oxygen microenvironment requires significant metabolic adaptation. Here, we show that LT-HSCs utilize glycolysis instead of mitochondrial oxidative phosphorylation to meet their energy demands. We used flow cytometry to identify a unique low mitochondrial activity/glycolysis-dependent subpopulation that houses the majority of hematopoietic progenitors and LT-HSCs. Finally, we demonstrate that Meis1 and Hif-1alpha are markedly enriched in LT-HSCs and that Meis1 regulates HSC metabolism through transcriptional activation of Hif-1alpha. These findings reveal an important transcriptional network that regulates HSC metabolism. Copyright 2010 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Hematopoietic stem cell: self-renewal versus differentiation.

              The mammalian blood system, containing more than 10 distinct mature cell types, stands on one specific cell type, hematopoietic stem cell (HSC). Within the system, only HSCs possess the ability of both multipotency and self-renewal. Multipotency is the ability to differentiate into all functional blood cells. Self-renewal is the ability to give rise to HSC itself without differentiation. Since mature blood cells (MBCs) are predominantly short-lived, HSCs continuously provide more differentiated progenitors while properly maintaining the HSC pool size throughout life by precisely balancing self-renewal and differentiation. Thus, understanding the mechanisms of self-renewal and differentiation of HSC has been a central issue. In this review, we focus on the hierarchical structure of the hematopoietic system, the current understanding of microenvironment and molecular cues regulating self-renewal and differentiation of adult HSCs, and the currently emerging systems approaches to understand HSC biology. © 2010 John Wiley & Sons, Inc.
                Bookmark

                Author and article information

                Journal
                J Cell Sci
                J. Cell. Sci
                JCS
                joces
                Journal of Cell Science
                The Company of Biologists Ltd
                0021-9533
                1477-9137
                15 July 2018
                27 July 2018
                27 July 2018
                : 131
                : 14
                : jcs212977
                Affiliations
                [1 ]Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur , Paris 75015, France
                [2 ]CNRS UMR 3738, Institut Pasteur , Paris 75015, France
                [3 ]Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli “Federico II” , Via S. Pansini 5, 80131 Napoli, Italy
                [4 ]Stem Cells and Development, Team Stability of Nuclear and Mitochondrial DNA, Department of Developmental & Stem Cell Biology, Institut Pasteur , Paris 75015, France
                Author notes
                [*]

                Present address: Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, University of Copenhagen, 3B Blegdamsvej, DK-2200 Copenhagen N, Denmark.

                []Author for correspondence ( shahragim.tajbakhsh@ 123456pasteur.fr )
                Author information
                http://orcid.org/0000-0001-5746-9211
                http://orcid.org/0000-0003-1809-7202
                Article
                JCS212977
                10.1242/jcs.212977
                6080609
                30054310
                390646fa-578e-4d13-b33f-39ff7d4bd4bc
                © 2018. Published by The Company of Biologists Ltd

                This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.

                History
                : 8 November 2017
                : 18 April 2018
                Funding
                Funded by: Core, http://dx.doi.org/10.13039/100006441;
                Funded by: Institut Pasteur, http://dx.doi.org/10.13039/501100003762;
                Funded by: Centre National pour la Recherche Scientifique, http://dx.doi.org/10.13039/501100006319;
                Funded by: Agence Nationale de la Recherche, http://dx.doi.org/10.13039/501100001665;
                Award ID: ANR-10-LABX- 73
                Funded by: European Research Council, http://dx.doi.org/10.13039/100010663;
                Award ID: 332893
                Categories
                Research Article

                Cell biology
                skeletal muscle stem cells,metabolic state,peroxisome,mitochondria,regeneration,ageing
                Cell biology
                skeletal muscle stem cells, metabolic state, peroxisome, mitochondria, regeneration, ageing

                Comments

                Comment on this article