651
|
Kuswanto W, Burzyn D, Panduro M, Wang KK, Jang YC, Wagers AJ, Benoist C, Mathis D. Poor Repair of Skeletal Muscle in Aging Mice Reflects a Defect in Local, Interleukin-33-Dependent Accumulation of Regulatory T Cells. Immunity 2016; 44:355-67. [PMID: 26872699 DOI: 10.1016/j.immuni.2016.01.009] [Citation(s) in RCA: 378] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/14/2015] [Accepted: 11/17/2015] [Indexed: 12/30/2022]
Abstract
Normal repair of skeletal muscle requires local expansion of a special population of Foxp3(+)CD4(+) regulatory T (Treg) cells. Such cells failed to accumulate in acutely injured muscle of old mice, known to undergo ineffectual repair. This defect reflected reduced recruitment of Treg cells to injured muscle, as well as less proliferation and retention therein. Interleukin-33 (IL-33) regulated muscle Treg cell homeostasis in young mice, and its administration to old mice ameliorated their deficits in Treg cell accumulation and muscle regeneration. The major IL-33-expressing cells in skeletal muscle displayed a constellation of markers diagnostic of fibro/adipogenic progenitor cells and were often associated with neural structures, including nerve fibers, nerve bundles, and muscle spindles, which are stretch-sensitive mechanoreceptors important for proprioception. IL-33(+) cells were more frequent after muscle injury and were reduced in old mice. IL-33 is well situated to relay signals between the nervous and immune systems within the muscle context.
Collapse
Affiliation(s)
- Wilson Kuswanto
- Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Dalia Burzyn
- Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Marisella Panduro
- Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Kathy K Wang
- Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Young Charles Jang
- Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Amy J Wagers
- Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Joslin Diabetes Center, Boston, MA 02215, USA; Paul F Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA 02115, USA
| | - Christophe Benoist
- Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Diane Mathis
- Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
652
|
Snijders T, Smeets JSJ, van Kranenburg J, Kies AK, van Loon LJC, Verdijk LB. Changes in myonuclear domain size do not precede muscle hypertrophy during prolonged resistance-type exercise training. Acta Physiol (Oxf) 2016; 216:231-9. [PMID: 26407634 DOI: 10.1111/apha.12609] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 05/30/2015] [Accepted: 09/20/2015] [Indexed: 11/27/2022]
Abstract
AIM Muscle fibre hypertrophy is accompanied by an increase in myonuclear number, an increase in myonuclear domain size or both. It has been suggested that increases in myonuclear domain size precede myonuclear accretion and subsequent muscle fibre hypertrophy during prolonged exercise training. In this study, we assessed the changes in muscle fibre size, myonuclear and satellite cell content throughout 12 weeks of resistance-type exercise training in young men. METHODS Twenty-two young men (23 ± 1 year) were assigned to a progressive, 12-weeks resistance-type exercise training programme (3 sessions per week). Muscle biopsies from the vastus lateralis muscle were taken before and after 2, 4, 8 and 12 weeks of exercise training. Muscle fibre size, myonuclear content, myonuclear domain size and satellite cell content were assessed by immunohistochemistry. RESULTS Type I and type II muscle fibre size increased gradually throughout the 12 weeks of training (type I: 18 ± 5%, type II: 41 ± 6%, P < 0.01). Myonuclear content increased significantly over time in both the type I (P < 0.01) and type II (P < 0.001) muscle fibres. No changes in type I and type II myonuclear domain size were observed at any time point throughout the intervention. Satellite cell content increased significantly over time in both type I and type II muscle fibres (P < 0.001). CONCLUSION Increases in myonuclear domain size do not appear to drive myonuclear accretion and muscle fibre hypertrophy during prolonged resistance-type exercise training in vivo in humans.
Collapse
Affiliation(s)
- T. Snijders
- NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University; Maastricht the Netherlands
| | - J. S. J. Smeets
- NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University; Maastricht the Netherlands
| | - J. van Kranenburg
- NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University; Maastricht the Netherlands
| | - A. K. Kies
- DSM Biotechnology Center; Applied Biochemistry Department; Delft the Netherlands
| | - L. J. C. van Loon
- NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University; Maastricht the Netherlands
| | - L. B. Verdijk
- NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University; Maastricht the Netherlands
| |
Collapse
|
653
|
Hardy D, Besnard A, Latil M, Jouvion G, Briand D, Thépenier C, Pascal Q, Guguin A, Gayraud-Morel B, Cavaillon JM, Tajbakhsh S, Rocheteau P, Chrétien F. Comparative Study of Injury Models for Studying Muscle Regeneration in Mice. PLoS One 2016; 11:e0147198. [PMID: 26807982 PMCID: PMC4726569 DOI: 10.1371/journal.pone.0147198] [Citation(s) in RCA: 326] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/30/2015] [Indexed: 11/19/2022] Open
Abstract
Background A longstanding goal in regenerative medicine is to reconstitute functional tissus or organs after injury or disease. Attention has focused on the identification and relative contribution of tissue specific stem cells to the regeneration process. Relatively little is known about how the physiological process is regulated by other tissue constituents. Numerous injury models are used to investigate tissue regeneration, however, these models are often poorly understood. Specifically, for skeletal muscle regeneration several models are reported in the literature, yet the relative impact on muscle physiology and the distinct cells types have not been extensively characterised. Methods We have used transgenic Tg:Pax7nGFP and Flk1GFP/+ mouse models to respectively count the number of muscle stem (satellite) cells (SC) and number/shape of vessels by confocal microscopy. We performed histological and immunostainings to assess the differences in the key regeneration steps. Infiltration of immune cells, chemokines and cytokines production was assessed in vivo by Luminex®. Results We compared the 4 most commonly used injury models i.e. freeze injury (FI), barium chloride (BaCl2), notexin (NTX) and cardiotoxin (CTX). The FI was the most damaging. In this model, up to 96% of the SCs are destroyed with their surrounding environment (basal lamina and vasculature) leaving a “dead zone” devoid of viable cells. The regeneration process itself is fulfilled in all 4 models with virtually no fibrosis 28 days post-injury, except in the FI model. Inflammatory cells return to basal levels in the CTX, BaCl2 but still significantly high 1-month post-injury in the FI and NTX models. Interestingly the number of SC returned to normal only in the FI, 1-month post-injury, with SCs that are still cycling up to 3-months after the induction of the injury in the other models. Conclusions Our studies show that the nature of the injury model should be chosen carefully depending on the experimental design and desired outcome. Although in all models the muscle regenerates completely, the trajectories of the regenerative process vary considerably. Furthermore, we show that histological parameters are not wholly sufficient to declare that regeneration is complete as molecular alterations (e.g. cycling SCs, cytokines) could have a major persistent impact.
Collapse
Affiliation(s)
- David Hardy
- Institut Pasteur, Human histopathology and animal models Unit, Infection and Epidemiology Department, Paris, France
- Paris Est University, Créteil, France
| | - Aurore Besnard
- Institut Pasteur, Human histopathology and animal models Unit, Infection and Epidemiology Department, Paris, France
| | - Mathilde Latil
- Institut Pasteur, Human histopathology and animal models Unit, Infection and Epidemiology Department, Paris, France
| | - Grégory Jouvion
- Institut Pasteur, Human histopathology and animal models Unit, Infection and Epidemiology Department, Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Paris France
| | - David Briand
- Institut Pasteur, Human histopathology and animal models Unit, Infection and Epidemiology Department, Paris, France
| | - Cédric Thépenier
- Institut Pasteur, Human histopathology and animal models Unit, Infection and Epidemiology Department, Paris, France
- IRBA, Unité Interactions Hôte-Agents Pathogènes, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Quentin Pascal
- Institut Pasteur, Human histopathology and animal models Unit, Infection and Epidemiology Department, Paris, France
| | - Aurélie Guguin
- Inserm, U955, Plateforme de Cytométrie en Flux, Créteil, France
| | - Barbara Gayraud-Morel
- Institut Pasteur, Stem Cells & Development Unit, Department of Developmental & Stem Cell Biology, Paris, France
| | - Jean-Marc Cavaillon
- Institut Pasteur, Cytokines and Inflammation Unit, Infection and Epidemiology Department, Paris, France
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells & Development Unit, Department of Developmental & Stem Cell Biology, Paris, France
| | - Pierre Rocheteau
- Institut Pasteur, Human histopathology and animal models Unit, Infection and Epidemiology Department, Paris, France
| | - Fabrice Chrétien
- Institut Pasteur, Human histopathology and animal models Unit, Infection and Epidemiology Department, Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Paris France
- Centre Hospitalier Sainte Anne, Laboratoire de Neuropathologie, Paris, France
- * E-mail:
| |
Collapse
|
654
|
Arentson-Lantz EJ, English KL, Paddon-Jones D, Fry CS. Fourteen days of bed rest induces a decline in satellite cell content and robust atrophy of skeletal muscle fibers in middle-aged adults. J Appl Physiol (1985) 2016; 120:965-75. [PMID: 26796754 DOI: 10.1152/japplphysiol.00799.2015] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/20/2016] [Indexed: 01/06/2023] Open
Abstract
Bed rest, a ground-based spaceflight analog, induces robust atrophy of skeletal muscle, an effect that is exacerbated with increasing age. We examined the effect of 14 days of bed rest on skeletal muscle satellite cell content and fiber type atrophy in middle-aged adults, an understudied age demographic with few overt signs of muscle aging that is representative of astronauts who perform long-duration spaceflight. Muscle biopsies were obtained from the vastus lateralis of healthy middle-aged adults [n= 7 (4 male, 3 female); age: 51 ± 1 yr] before (Pre-BR) and after (Post-BR) 14 days of bed rest. Immunohistochemical analyses were used to quantify myosin heavy chain (MyHC) isoform expression, cross-sectional area (CSA), satellite cell and myonuclear content, and capillary density. Peak oxygen consumption, knee extensor strength, and body composition were also measured Pre-BR and Post-BR. Post-BR MyHC type 2a fiber percentage was reduced, and mean CSA decreased in all fiber types (-24 ± 5%;P< 0.05). Satellite cell content was also reduced Post-BR (-39 ± 9%;P< 0.05), and the change in satellite cell content was significantly correlated with the change in mean fiber CSA (r(2)= 0.60;P< 0.05). A decline in capillary density was observed Post-BR (-23 ± 6%;P< 0.05), and Post-BR capillary content was significantly associated with Post-BR peak aerobic capacity (r(2)= 0.59;P< 0.05). A subtle decline in myonuclear content occurred during bed rest (-5 ± 1%;P< 0.05). The rapid maladaptation of skeletal muscle to 14 days of mechanical unloading in middle-aged adults emphasizes the need for robust countermeasures to preserve muscle function in astronauts.
Collapse
Affiliation(s)
- Emily J Arentson-Lantz
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas; Division of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Kirk L English
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas; Division of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Douglas Paddon-Jones
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas; Division of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, Texas; Sealy Center on Aging, University of Texas Medical Branch, Galveston, Texas
| | - Christopher S Fry
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas; Division of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, Texas; Sealy Center on Aging, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
655
|
Pichavant C, Burkholder TJ, Pavlath GK. Decrease of myofiber branching via muscle-specific expression of the olfactory receptor mOR23 in dystrophic muscle leads to protection against mechanical stress. Skelet Muscle 2016; 6:2. [PMID: 26798450 PMCID: PMC4721043 DOI: 10.1186/s13395-016-0077-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/05/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Abnormal branched myofibers within skeletal muscles are commonly found in diverse animal models of muscular dystrophy as well as in patients. Branched myofibers from dystrophic mice are more susceptible to break than unbranched myofibers suggesting that muscles containing a high percentage of these myofibers are more prone to injury. Previous studies showed ubiquitous over-expression of mouse olfactory receptor 23 (mOR23), a G protein-coupled receptor, in wild type mice decreased myofiber branching. Whether mOR23 over-expression specifically in skeletal muscle cells is sufficient to mitigate myofiber branching in dystrophic muscle is unknown. METHODS We created a novel transgenic mouse over-expressing mOR23 specifically in muscle cells and then bred with dystrophic (mdx) mice. Myofiber branching was analyzed in these two transgenic mice and membrane integrity was assessed by Evans blue dye fluorescence. RESULTS mOR23 over-expression in muscle led to a decrease of myofiber branching after muscle regeneration in non-dystrophic mouse muscles and reduced the severity of myofiber branching in mdx mouse muscles. Muscles from mdx mouse over-expressing mOR23 significantly exhibited less damage to eccentric contractions than control mdx muscles. CONCLUSIONS The decrease of myofiber branching in mdx mouse muscles over-expressing mOR23 reduced the amount of membrane damage induced by mechanical stress. These results suggest that modifying myofiber branching in dystrophic patients, while not preventing degeneration, could be beneficial for mitigating some of the effects of the disease process.
Collapse
Affiliation(s)
- Christophe Pichavant
- Department of Pharmacology, Emory University, Atlanta, GA USA ; Present address: Department of Genetics, Stanford University, Stanford, CA USA
| | - Thomas J Burkholder
- School of Applied Physiology, Georgia Institute of Technology, Atlanta, GA USA
| | - Grace K Pavlath
- Department of Pharmacology, Emory University, Atlanta, GA USA ; 1510 Clifton Road, Room 5024, Atlanta, GA 30322 USA
| |
Collapse
|
656
|
Czerwinska AM, Grabowska I, Archacka K, Bem J, Swierczek B, Helinska A, Streminska W, Fogtman A, Iwanicka-Nowicka R, Koblowska M, Ciemerych MA. Myogenic Differentiation of Mouse Embryonic Stem Cells That Lack a Functional Pax7 Gene. Stem Cells Dev 2016; 25:285-300. [PMID: 26649785 PMCID: PMC4761802 DOI: 10.1089/scd.2015.0162] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The transcription factor Pax7 plays a key role during embryonic myogenesis and sustains the proper function of satellite cells, which serve as adult skeletal muscle stem cells. Overexpression of Pax7 has been shown to promote the myogenic differentiation of pluripotent stem cells. However, the effects of the absence of functional Pax7 in differentiating embryonic stem cells (ESCs) have not yet been directly tested. Herein, we studied mouse stem cells that lacked a functional Pax7 gene and characterized the differentiation of these stem cells under conditions that promoted the derivation of myoblasts in vitro. We analyzed the expression of myogenic factors, such as myogenic regulatory factors and muscle-specific microRNAs, in wild-type and mutant cells. Finally, we compared the transcriptome of both types of cells and did not find substantial differences in the expression of genes related to the regulation of myogenesis. As a result, we showed that the absence of functional Pax7 does not prevent the in vitro myogenic differentiation of ESCs.
Collapse
Affiliation(s)
- Areta M Czerwinska
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Iwona Grabowska
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Karolina Archacka
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Joanna Bem
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Barbara Swierczek
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Anita Helinska
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Wladyslawa Streminska
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Anna Fogtman
- 2 Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics , Polish Academy of Sciences, Warsaw, Poland
| | - Roksana Iwanicka-Nowicka
- 2 Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics , Polish Academy of Sciences, Warsaw, Poland .,3 Department of Systems Biology, Faculty of Biology, University of Warsaw , Warsaw, Poland
| | - Marta Koblowska
- 2 Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics , Polish Academy of Sciences, Warsaw, Poland .,3 Department of Systems Biology, Faculty of Biology, University of Warsaw , Warsaw, Poland
| | - Maria A Ciemerych
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| |
Collapse
|
657
|
Zhang C, Ferrari R, Beezhold K, Stearns-Reider K, D'Amore A, Haschak M, Stolz D, Robbins PD, Barchowsky A, Ambrosio F. Arsenic Promotes NF-Κb-Mediated Fibroblast Dysfunction and Matrix Remodeling to Impair Muscle Stem Cell Function. Stem Cells 2016; 34:732-42. [PMID: 26537186 DOI: 10.1002/stem.2232] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 09/14/2015] [Indexed: 12/19/2022]
Abstract
Arsenic is a global health hazard that impacts over 140 million individuals worldwide. Epidemiological studies reveal prominent muscle dysfunction and mobility declines following arsenic exposure; yet, mechanisms underlying such declines are unknown. The objective of this study was to test the novel hypothesis that arsenic drives a maladaptive fibroblast phenotype to promote pathogenic myomatrix remodeling and compromise the muscle stem (satellite) cell (MuSC) niche. Mice were exposed to environmentally relevant levels of arsenic in drinking water before receiving a local muscle injury. Arsenic-exposed muscles displayed pathogenic matrix remodeling, defective myofiber regeneration and impaired functional recovery, relative to controls. When naïve human MuSCs were seeded onto three-dimensional decellularized muscle constructs derived from arsenic-exposed muscles, cells displayed an increased fibrogenic conversion and decreased myogenicity, compared with cells seeded onto control constructs. Consistent with myomatrix alterations, fibroblasts isolated from arsenic-exposed muscle displayed sustained expression of matrix remodeling genes, the majority of which were mediated by NF-κB. Inhibition of NF-κB during arsenic exposure preserved normal myofiber structure and functional recovery after injury, suggesting that NF-κB signaling serves as an important mechanism of action for the deleterious effects of arsenic on tissue healing. Taken together, the results from this study implicate myomatrix biophysical and/or biochemical characteristics as culprits in arsenic-induced MuSC dysfunction and impaired muscle regeneration. It is anticipated that these findings may aid in the development of strategies to prevent or revert the effects of arsenic on tissue healing and, more broadly, provide insight into the influence of the native myomatrix on stem cell behavior.
Collapse
Affiliation(s)
- Changqing Zhang
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ricardo Ferrari
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kevin Beezhold
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kristen Stearns-Reider
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Antonio D'Amore
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Martin Haschak
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Donna Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Paul D Robbins
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, Florida, USA
| | - Aaron Barchowsky
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Physical Therapy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
658
|
Contreras O, Rebolledo DL, Oyarzún JE, Olguín HC, Brandan E. Connective tissue cells expressing fibro/adipogenic progenitor markers increase under chronic damage: relevance in fibroblast-myofibroblast differentiation and skeletal muscle fibrosis. Cell Tissue Res 2016; 364:647-660. [DOI: 10.1007/s00441-015-2343-0] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 12/03/2015] [Indexed: 02/06/2023]
|
659
|
Abstract
Freeze injury is physically induced by exposing skeletal muscle to an extremely cold probe, and results in a robust degenerative and inflammatory response. One unique aspect of freeze injury is that it destroys not only the muscle fiber cells, but also all of the mononuclear cells in the zone of injury. Repair of the muscle is accomplished by satellite cells from outside of the zone of injury, which must migrate in and which may interact with inflammatory cells, hence the length of time before apparent histological recovery of the most damaged zone is typically somewhat longer with freeze injury than with other physical or chemical methods of injury. In this chapter, we present a detailed protocol for the freeze injury of the tibialis anterior (TA) muscle in mouse.
Collapse
|
660
|
Uezumi A, Kasai T, Tsuchida K. Identification, Isolation, and Characterization of Mesenchymal Progenitors in Mouse and Human Skeletal Muscle. Methods Mol Biol 2016; 1460:241-253. [PMID: 27492177 DOI: 10.1007/978-1-4939-3810-0_17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mesenchymal progenitors residing in the muscle interstitial space contribute to pathogeneses such as fat infiltration and fibrosis. Because fat infiltration and fibrosis are hallmarks of diseased muscle, it is important to establish an accurate and reproducible method for isolating mesenchymal progenitors for research on muscle diseases. In this chapter, we describe methods based on fluorescence-activated cell sorting (FACS) to purify mesenchymal progenitors from mouse and human skeletal muscle using the most reliable marker for mesenchymal progenitors, PDGFRα. These methods allow concurrent isolation of the muscle stem cells called satellite cells. The quality of isolated mesenchymal progenitors is confirmed by their remarkable adipogenic potential without myogenic capacity, while purified satellite cells possess robust myogenic activity with no adipogenic potential. Simultaneous isolation of both mesenchymal progenitors and satellite cells from mouse and human tissues provides a powerful platform for studying skeletal muscle regeneration and diseases.
Collapse
Affiliation(s)
- Akiyoshi Uezumi
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan.
| | - Takehiro Kasai
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Kunihiro Tsuchida
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| |
Collapse
|
661
|
Skeletal Muscle Tissue Clearing for LacZ and Fluorescent Reporters, and Immunofluorescence Staining. Methods Mol Biol 2016; 1460:129-40. [PMID: 27492170 DOI: 10.1007/978-1-4939-3810-0_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Skeletal muscle is a highly ordered yet complex tissue containing several cell types that interact with each other in order to maintain structure and homeostasis. It is also a highly regenerative tissue that responds to damage in a highly intricate but stereotypic manner, with distinct spatial and temporal kinetics. Proper examination of this process requires one to look at the three-dimensional orientation of the cellular and subcellular components, which can be accomplished through tissue clearing. While there has been a recent surge of protocols to study biology in whole tissue, it has primarily focused on the nervous system. This chapter describes the workflow for whole mount analysis of murine skeletal muscle for LacZ reporters, fluorescent reporters and immunofluorescence staining. Using this technique, we are able to visualize LacZ reporters more effectively in deep tissue samples, and to perform fluorescent imaging with a depth greater than 1700 μm.
Collapse
|
662
|
Zismanov V, Chichkov V, Colangelo V, Jamet S, Wang S, Syme A, Koromilas A, Crist C. Phosphorylation of eIF2α Is a Translational Control Mechanism Regulating Muscle Stem Cell Quiescence and Self-Renewal. Cell Stem Cell 2016; 18:79-90. [DOI: 10.1016/j.stem.2015.09.020] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 08/21/2015] [Accepted: 09/30/2015] [Indexed: 12/21/2022]
|
663
|
Zhou X, Li M, Huang H, Chen K, Yuan Z, Zhang Y, Nie Y, Chen H, Zhang X, Chen L, Chen Y, Mo D. HMGB2 regulates satellite cell-mediated skeletal muscle regeneration via IGF2BP2. J Cell Sci 2016; 129:4305-4316. [DOI: 10.1242/jcs.189944] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/17/2016] [Indexed: 12/23/2022] Open
Abstract
Although the mechanism underlying modulation of transcription factors in myogenesis has been well elucidated, the function of the transcription cofactors involved in this process remains poorly understood. Here, we identified HMGB2 as an essential nuclear transcriptional co-regulator in myogenesis. HMGB2 was highly expressed in undifferentiated myoblasts and regenerating muscle. Knockdown of HMGB2 inhibited myoblast proliferation and stimulated its differentiation. HMGB2 depletion down-regulated Myf5 and Cyclin A2 on the protein but not mRNA level. In contrast, overexpression of HMGB2 promoted Myf5 and Cyclin A2 protein upregulation. Furthermore, we found that the RNA-binding protein IGF2BP2 is a downstream target of HMGB2, as previously shown for HMGA2. IGF2BP2 binds to mRNAs of Myf5 or Cyclin A2, resulting in translation enhancement or mRNA stabilization, respectively. Notably, overexpression of IGF2BP2 could partially rescue protein levels of Myf5 and Cyclin A2, in response to HMGB2 decrease. Moreover, depletion of HMGB2 in vivo severely attenuated muscle repair; this was due to a decrease in satellite cells. Together, these results highlight the previously undiscovered and critical role of HMGB2-IGF2BP2 axis in myogenesis and muscle regeneration.
Collapse
Affiliation(s)
- Xingyu Zhou
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Mingsen Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Huaxing Huang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Keren Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zhuning Yuan
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Ying Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yaping Nie
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Hu Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xumeng Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Luxi Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
664
|
Fu X, Zhu M, Zhang S, Foretz M, Viollet B, Du M. Obesity Impairs Skeletal Muscle Regeneration Through Inhibition of AMPK. Diabetes 2016; 65:188-200. [PMID: 26384382 PMCID: PMC4686944 DOI: 10.2337/db15-0647] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 09/10/2015] [Indexed: 12/18/2022]
Abstract
Obesity is increasing rapidly worldwide and is accompanied by many complications, including impaired muscle regeneration. The obese condition is known to inhibit AMPK activity in multiple tissues. We hypothesized that the loss of AMPK activity is a major reason for hampered muscle regeneration in obese subjects. We found that obesity inhibits AMPK activity in regenerating muscle, which was associated with impeded satellite cell activation and impaired muscle regeneration. To test the mediatory role of AMPKα1, we knocked out AMPKα1 and found that both proliferation and differentiation of satellite cells are reduced after injury and that muscle regeneration is severely impeded, reminiscent of hampered muscle regeneration seen in obese subjects. Transplanted satellite cells with AMPKα1 deficiency had severely impaired myogenic capacity in regenerating muscle fibers. We also found that attenuated muscle regeneration in obese mice is rescued by AICAR, a drug that specifically activates AMPK, but AICAR treatment failed to improve muscle regeneration in obese mice with satellite cell-specific AMPKα1 knockout, demonstrating the importance of AMPKα1 in satellite cell activation and muscle regeneration. In summary, AMPKα1 is a key mediator linking obesity and impaired muscle regeneration, providing a convenient drug target to facilitate muscle regeneration in obese populations.
Collapse
Affiliation(s)
- Xing Fu
- Washington Center for Muscle Biology, Department of Animal Sciences and Department of Pharmaceutical Sciences, Washington State University, Pullman, WA
| | - Meijun Zhu
- School of Food Science, Washington State University, Pullman, WA
| | - Shuming Zhang
- School of Food Science, Washington State University, Pullman, WA
| | - Marc Foretz
- INSERM U1016, Institut Cochin, Paris, France Université Paris Descartes, Sorbonne Paris Cité, Paris, France CNRS UMR 8104, Paris, France
| | - Benoit Viollet
- INSERM U1016, Institut Cochin, Paris, France Université Paris Descartes, Sorbonne Paris Cité, Paris, France CNRS UMR 8104, Paris, France
| | - Min Du
- Washington Center for Muscle Biology, Department of Animal Sciences and Department of Pharmaceutical Sciences, Washington State University, Pullman, WA
| |
Collapse
|
665
|
Owens DJ, Sharples AP, Polydorou I, Alwan N, Donovan T, Tang J, Fraser WD, Cooper RG, Morton JP, Stewart C, Close GL. A systems-based investigation into vitamin D and skeletal muscle repair, regeneration, and hypertrophy. Am J Physiol Endocrinol Metab 2015; 309:E1019-31. [PMID: 26506852 DOI: 10.1152/ajpendo.00375.2015] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/19/2015] [Indexed: 01/08/2023]
Abstract
Skeletal muscle is a direct target for vitamin D. Observational studies suggest that low 25[OH]D correlates with functional recovery of skeletal muscle following eccentric contractions in humans and crush injury in rats. However, a definitive association is yet to be established. To address this gap in knowledge in relation to damage repair, a randomised, placebo-controlled trial was performed in 20 males with insufficient concentrations of serum 25(OH)D (45 ± 25 nmol/l). Prior to and following 6 wk of supplemental vitamin D3 (4,000 IU/day) or placebo (50 mg of cellulose), participants performed 20 × 10 damaging eccentric contractions of the knee extensors, with peak torque measured over the following 7 days of recovery. Parallel experimentation using isolated human skeletal muscle-derived myoblast cells from biopsies of 14 males with low serum 25(OH)D (37 ± 11 nmol/l) were subjected to mechanical wound injury, which enabled corresponding in vitro studies of muscle repair, regeneration, and hypertrophy in the presence and absence of 10 or 100 nmol 1α,25(OH)2D3. Supplemental vitamin D3 increased serum 25(OH)D and improved recovery of peak torque at 48 h and 7 days postexercise. In vitro, 10 nmol 1α,25(OH)2D3 improved muscle cell migration dynamics and resulted in improved myotube fusion/differentiation at the biochemical, morphological, and molecular level together with increased myotube hypertrophy at 7 and 10 days postdamage. Together, these preliminary data are the first to characterize a role for vitamin D in human skeletal muscle regeneration and suggest that maintaining serum 25(OH)D may be beneficial for enhancing reparative processes and potentially for facilitating subsequent hypertrophy.
Collapse
Affiliation(s)
- Daniel J Owens
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Adam P Sharples
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Ioanna Polydorou
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Nura Alwan
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Timothy Donovan
- Department of Sport and Exercise Sciences, Glyndwr University, Plas Coch Campus, Wrexham, United Kingdom
| | - Jonathan Tang
- Faculty of Medicine and Health Science, Norwich Medical School, University of East Anglia, Norwich, United Kingdom; and
| | - William D Fraser
- Faculty of Medicine and Health Science, Norwich Medical School, University of East Anglia, Norwich, United Kingdom; and
| | - Robert G Cooper
- Medical Research Council-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing, University of Liverpool, Liverpool, United Kingdom
| | - James P Morton
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Claire Stewart
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Graeme L Close
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, United Kingdom;
| |
Collapse
|
666
|
Rocheteau P, Chatre L, Briand D, Mebarki M, Jouvion G, Bardon J, Crochemore C, Serrani P, Lecci PP, Latil M, Matot B, Carlier PG, Latronico N, Huchet C, Lafoux A, Sharshar T, Ricchetti M, Chrétien F. Sepsis induces long-term metabolic and mitochondrial muscle stem cell dysfunction amenable by mesenchymal stem cell therapy. Nat Commun 2015; 6:10145. [PMID: 26666572 PMCID: PMC4682118 DOI: 10.1038/ncomms10145] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 11/09/2015] [Indexed: 02/08/2023] Open
Abstract
Sepsis, or systemic inflammatory response syndrome, is the major cause of critical illness resulting in admission to intensive care units. Sepsis is caused by severe infection and is associated with mortality in 60% of cases. Morbidity due to sepsis is complicated by neuromyopathy, and patients face long-term disability due to muscle weakness, energetic dysfunction, proteolysis and muscle wasting. These processes are triggered by pro-inflammatory cytokines and metabolic imbalances and are aggravated by malnutrition and drugs. Skeletal muscle regeneration depends on stem (satellite) cells. Herein we show that mitochondrial and metabolic alterations underlie the sepsis-induced long-term impairment of satellite cells and lead to inefficient muscle regeneration. Engrafting mesenchymal stem cells improves the septic status by decreasing cytokine levels, restoring mitochondrial and metabolic function in satellite cells, and improving muscle strength. These findings indicate that sepsis affects quiescent muscle stem cells and that mesenchymal stem cells might act as a preventive therapeutic approach for sepsis-related morbidity. Sepsis patients often develop muscle atrophy that can last for years. Here the authors show in a mouse model that sepsis causes long-term impairment of the satellite cells, affecting mitochondrial function and energy metabolism, and that injection of mesenchymal stem cells restores satellite cell metabolism and muscle regeneration.
Collapse
Affiliation(s)
- P Rocheteau
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, 75724 cedex15, Paris, France
| | - L Chatre
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Stem Cells and Development, 75724 cedex15, Paris, France.,Team Stability of Nuclear and Mitochondrial DNA, CNRS UMR 3525, 75724 cedex15, Paris, France
| | - D Briand
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, 75724 cedex15, Paris, France
| | - M Mebarki
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, 75724 cedex15, Paris, France
| | - G Jouvion
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, 75724 cedex15, Paris, France
| | - J Bardon
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, 75724 cedex15, Paris, France
| | - C Crochemore
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Stem Cells and Development, 75724 cedex15, Paris, France.,Team Stability of Nuclear and Mitochondrial DNA, CNRS UMR 3525, 75724 cedex15, Paris, France
| | - P Serrani
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, 75724 cedex15, Paris, France
| | - P P Lecci
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, 75724 cedex15, Paris, France
| | - M Latil
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, 75724 cedex15, Paris, France
| | - B Matot
- NMR Laboratory, Institute of Myology, Paris 75013, France.,CEA, I2BM, MIRCen, NMR Laboratory, Paris 75013, France
| | - P G Carlier
- NMR Laboratory, Institute of Myology, Paris 75013, France.,CEA, I2BM, MIRCen, NMR Laboratory, Paris 75013, France
| | - N Latronico
- Anesthesia and Reanimation Department, Department of Surgery, University of Brescia, Brescia 25121, Italy
| | - C Huchet
- INSERM UMR1087/ CNRS UMR6291, Institut du Thorax, Therassay, Université de Nantes, Faculté des Sciences et des Techniques, F44322 Nantes 44000, France
| | - A Lafoux
- INSERM UMR1087/ CNRS UMR6291, Institut du Thorax, Therassay, Université de Nantes, Faculté des Sciences et des Techniques, F44322 Nantes 44000, France
| | - T Sharshar
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, 75724 cedex15, Paris, France.,Service de réanimation médico-chirurgicale adulte, Hôpital Raymond Poincaré, Garches 92380, France.,Université Versailles Saint Quentin, Versailles 78000, France.,TRIGGERSEP, F-CRIN Network, Versailles 78000, France
| | - M Ricchetti
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Stem Cells and Development, 75724 cedex15, Paris, France.,Team Stability of Nuclear and Mitochondrial DNA, CNRS UMR 3525, 75724 cedex15, Paris, France
| | - F Chrétien
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, 75724 cedex15, Paris, France.,TRIGGERSEP, F-CRIN Network, Versailles 78000, France.,Laboratoire de Neuropathologie, Centre Hospitalier Sainte Anne, Paris 75014, France.,Paris Descartes University, Sorbonne Paris Cité, Paris 75006, France
| |
Collapse
|
667
|
Pawlikowski B, Pulliam C, Betta ND, Kardon G, Olwin BB. Pervasive satellite cell contribution to uninjured adult muscle fibers. Skelet Muscle 2015; 5:42. [PMID: 26668715 PMCID: PMC4677447 DOI: 10.1186/s13395-015-0067-1] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 11/04/2015] [Indexed: 12/22/2022] Open
Abstract
Background Adult skeletal muscle adapts to functional needs, maintaining consistent numbers of myonuclei and stem cells. Although resident muscle stem cells or satellite cells are required for muscle growth and repair, in uninjured muscle, these cells appear quiescent and metabolically inactive. To investigate the satellite cell contribution to myofibers in adult uninjured skeletal muscle, we labeled satellite cells by inducing a recombination of LSL-tdTomato in Pax7CreER mice and scoring tdTomato+ myofibers as an indicator of satellite cell fusion. Results Satellite cell fusion into myofibers plateaus postnatally between 8 and 12 weeks of age, reaching a steady state in hindlimb muscles, but in extra ocular or diaphragm muscles, satellite cell fusion is maintained at postnatal levels irrespective of the age assayed. Upon recombination and following a 2-week chase in 6-month-old mice, tdTomato-labeled satellite cells fused into myofibers as 20, 50, and 80 % of hindlimb, extra ocular, and diaphragm myofibers, respectively, were tdTomato+. Satellite cells contribute to uninjured myofibers either following a cell division or directly without an intervening cell division. Conclusions The frequency of satellite cell fusion into the skeletal muscle fibers is greater than previously estimated, suggesting an important functional role for satellite cell fusion into adult myofibers and a requirement for active maintenance of satellite cell numbers in uninjured skeletal muscle.
Collapse
Affiliation(s)
- Bradley Pawlikowski
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80039 USA
| | - Crystal Pulliam
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80039 USA
| | - Nicole Dalla Betta
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80039 USA
| | - Gabrielle Kardon
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80039 USA
| | - Bradley B Olwin
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80039 USA
| |
Collapse
|
668
|
Marino S, Di Foggia V. Invited Review: Polycomb group genes in the regeneration of the healthy and pathological skeletal muscle. Neuropathol Appl Neurobiol 2015; 42:407-22. [PMID: 26479276 DOI: 10.1111/nan.12290] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 10/14/2015] [Accepted: 10/19/2015] [Indexed: 12/21/2022]
Abstract
The polycomb group (PcG) proteins are epigenetic repressors required during key developmental processes, such as maintenance of cell identity and stem cell differentiation. To exert their repressive function, PcG proteins assemble on chromatin into multiprotein complexes, known as polycomb repressive complex 1 and 2. In this review, we will focus on the role and mode of function of PcG proteins in the development and regeneration of the skeletal muscle, both in normal and pathological conditions and we will discuss the emerging concept of modulation of their expression to enhance the muscle-specific regenerative process for patient benefit.
Collapse
Affiliation(s)
- S Marino
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - V Di Foggia
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
669
|
Webster MT, Manor U, Lippincott-Schwartz J, Fan CM. Intravital Imaging Reveals Ghost Fibers as Architectural Units Guiding Myogenic Progenitors during Regeneration. Cell Stem Cell 2015; 18:243-52. [PMID: 26686466 DOI: 10.1016/j.stem.2015.11.005] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/24/2015] [Accepted: 11/06/2015] [Indexed: 12/16/2022]
Abstract
How resident stem cells and their immediate progenitors rebuild tissues of pre-injury organization and size for proportional regeneration is not well understood. Using 3D, time-lapse intravital imaging for direct visualization of the muscle regeneration process in live mice, we report that extracellular matrix remnants from injured skeletal muscle fibers, "ghost fibers," govern muscle stem/progenitor cell behaviors during proportional regeneration. Stem cells were immobile and quiescent without injury whereas their activated progenitors migrated and divided after injury. Unexpectedly, divisions and migration were primarily bi-directionally oriented along the ghost fiber longitudinal axis, allowing for spreading of progenitors throughout ghost fibers. Re-orienting ghost fibers impacted myogenic progenitors' migratory paths and division planes, causing disorganization of regenerated muscle fibers. We conclude that ghost fibers are autonomous, architectural units necessary for proportional regeneration after tissue injury. This finding reinforces the need to fabricate bioengineered matrices that mimic living tissue matrices for tissue regeneration therapy.
Collapse
Affiliation(s)
- Micah T Webster
- Department of Embryology, Carnegie Institution of Washington, 3520 San Martin Drive, Baltimore, MD 21218, USA
| | - Uri Manor
- Cell Biology and Metabolism Branch, Eunice Kennedy Shriver National Institutes of Child Health and Human Development, National Institute of Health, Building 35A, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Jennifer Lippincott-Schwartz
- Cell Biology and Metabolism Branch, Eunice Kennedy Shriver National Institutes of Child Health and Human Development, National Institute of Health, Building 35A, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Chen-Ming Fan
- Department of Embryology, Carnegie Institution of Washington, 3520 San Martin Drive, Baltimore, MD 21218, USA.
| |
Collapse
|
670
|
|
671
|
Dubois V, Simitsidellis I, Laurent MR, Jardi F, Saunders PTK, Vanderschueren D, Claessens F. Enobosarm (GTx-024) Modulates Adult Skeletal Muscle Mass Independently of the Androgen Receptor in the Satellite Cell Lineage. Endocrinology 2015; 156:4522-33. [PMID: 26393303 DOI: 10.1210/en.2015-1479] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Androgens increase skeletal muscle mass, but their clinical use is hampered by a lack of tissue selectivity and subsequent side effects. Selective androgen receptor modulators elicit muscle-anabolic effects while only sparingly affecting reproductive tissues. The selective androgen receptor modulator, GTx-024 (enobosarm), is being investigated for cancer cachexia, sarcopenia, and muscle wasting diseases. Here we investigate the role of muscle androgen receptor (AR) in the anabolic effect of GTx-024. In mice lacking AR in the satellite cell lineage (satARKO), the weight of the androgen-sensitive levator ani muscle was lower but was decreased further upon orchidectomy. GTx-024 was as effective as DHT in restoring levator ani weights to sham levels. Expression of the muscle-specific, androgen-responsive genes S-adenosylmethionine decarboxylase and myostatin was decreased by orchidectomy and restored by GTx-024 and DHT in control mice, whereas the expression was low and unaffected by androgen status in satARKO. In contrast, insulin-like growth factor 1Ea expression was not different between satARKO and control muscle, decreased upon castration, and was restored by DHT and GTx-024 in both genotypes. These data indicate that GTx-024 does not selectively modulate AR in the satellite cell lineage and that cells outside this lineage remain androgen responsive in satARKO muscle. Indeed, residual AR-positive cells were present in satARKO muscle, coexpressing the fibroblast-lineage marker vimentin. AR positive, muscle-resident fibroblasts could therefore be involved in the indirect effects of androgens on muscle. In conclusion, both DHT and GTx-024 target AR pathways in the satellite cell lineage, but cells outside this lineage also contribute to the anabolic effects of androgens.
Collapse
Affiliation(s)
- Vanessa Dubois
- Molecular Endocrinology Laboratory (V.D., M.R.L., F.C.), Department of Cellular and Molecular Medicine, Department of Gerontology and Geriatrics (M.R.L.), and Clinical and Experimental Endocrinology (F.J., D.V.), Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; and Medical Research Council Centre for Inflammation Research (I.S., P.T.K.S.), University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Ioannis Simitsidellis
- Molecular Endocrinology Laboratory (V.D., M.R.L., F.C.), Department of Cellular and Molecular Medicine, Department of Gerontology and Geriatrics (M.R.L.), and Clinical and Experimental Endocrinology (F.J., D.V.), Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; and Medical Research Council Centre for Inflammation Research (I.S., P.T.K.S.), University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Michaël R Laurent
- Molecular Endocrinology Laboratory (V.D., M.R.L., F.C.), Department of Cellular and Molecular Medicine, Department of Gerontology and Geriatrics (M.R.L.), and Clinical and Experimental Endocrinology (F.J., D.V.), Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; and Medical Research Council Centre for Inflammation Research (I.S., P.T.K.S.), University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Ferran Jardi
- Molecular Endocrinology Laboratory (V.D., M.R.L., F.C.), Department of Cellular and Molecular Medicine, Department of Gerontology and Geriatrics (M.R.L.), and Clinical and Experimental Endocrinology (F.J., D.V.), Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; and Medical Research Council Centre for Inflammation Research (I.S., P.T.K.S.), University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Philippa T K Saunders
- Molecular Endocrinology Laboratory (V.D., M.R.L., F.C.), Department of Cellular and Molecular Medicine, Department of Gerontology and Geriatrics (M.R.L.), and Clinical and Experimental Endocrinology (F.J., D.V.), Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; and Medical Research Council Centre for Inflammation Research (I.S., P.T.K.S.), University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Dirk Vanderschueren
- Molecular Endocrinology Laboratory (V.D., M.R.L., F.C.), Department of Cellular and Molecular Medicine, Department of Gerontology and Geriatrics (M.R.L.), and Clinical and Experimental Endocrinology (F.J., D.V.), Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; and Medical Research Council Centre for Inflammation Research (I.S., P.T.K.S.), University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Frank Claessens
- Molecular Endocrinology Laboratory (V.D., M.R.L., F.C.), Department of Cellular and Molecular Medicine, Department of Gerontology and Geriatrics (M.R.L.), and Clinical and Experimental Endocrinology (F.J., D.V.), Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; and Medical Research Council Centre for Inflammation Research (I.S., P.T.K.S.), University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| |
Collapse
|
672
|
Jackson JR, Kirby TJ, Fry CS, Cooper RL, McCarthy JJ, Peterson CA, Dupont-Versteegden EE. Reduced voluntary running performance is associated with impaired coordination as a result of muscle satellite cell depletion in adult mice. Skelet Muscle 2015; 5:41. [PMID: 26579218 PMCID: PMC4647638 DOI: 10.1186/s13395-015-0065-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/02/2015] [Indexed: 11/10/2022] Open
Abstract
Background Satellite cells, or muscle stem cells, have been thought to be responsible for all muscle plasticity, but recent studies using genetically modified mouse models that allow for the conditional ablation of satellite cells have challenged this dogma. Results have confirmed the absolute requirement of satellite cells for muscle regeneration but surprisingly also showed that they are not required for adult muscle growth. While the function of satellite cells in muscle growth and regeneration is becoming better defined, their role in the response to aerobic activity remains largely unexplored. The purpose of the current study was to assess the involvement of satellite cells in response to aerobic exercise by evaluating the effect of satellite cell depletion on wheel running performance. Results Four-month-old female Pax7/DTA mice (n = 8–12 per group) were satellite cell depleted via tamoxifen administration; at 6 months of age, mice either remained sedentary or were provided with running wheels for 8 weeks. Plantaris muscles were significantly depleted of Pax7+cells (≥90 % depleted), and 8 weeks of wheel running did not result in an increase in Pax7+ cells, or in myonuclear accretion. Interestingly, satellite cell-depleted animals ran ~27 % less distance and were 23 % slower than non-depleted animals. Wheel running was associated with elevated succinate dehydrogenase activity, muscle vascularization, lipid accumulation, and a significant shift toward more oxidative myosin heavy chain isoforms, as well as an increase in voltage dependent anion channel abundance, a marker of mitochondrial density. Importantly, these changes were independent of satellite cell content. Interestingly, depletion of Pax7+ cells from intra- as well as extrafusal muscle fibers resulted in atrophy of intrafusal fibers, thickening of muscle spindle-associated extracellular matrix, and a marked reduction of functional outcomes including grip strength, gait fluidity, and balance, which likely contributed to the impaired running performance. Conclusions Depletion of Pax7-expressing cells in muscle resulted in reduced voluntary wheel running performance, without affecting markers of aerobic adaptation; however, their absence may perturb proprioception via disruption of muscle spindle fibers resulting in loss of gross motor coordination, indicating that satellite cells have a yet unexplored role in muscle function. Electronic supplementary material The online version of this article (doi:10.1186/s13395-015-0065-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Janna R Jackson
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, KY USA ; Center for Muscle Biology, University of Kentucky, Lexington, KY USA
| | - Tyler J Kirby
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA ; Center for Muscle Biology, University of Kentucky, Lexington, KY USA ; Present address: Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY USA
| | - Christopher S Fry
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, KY USA ; Center for Muscle Biology, University of Kentucky, Lexington, KY USA ; Present address: Department of Nutrition and Metabolism, School of Health Professions, University of Texas Medical Branch, Galveston, TX USA
| | - Robin L Cooper
- Center for Muscle Biology, University of Kentucky, Lexington, KY USA ; Department of Biology, University of Kentucky, Lexington, KY USA
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA ; Center for Muscle Biology, University of Kentucky, Lexington, KY USA
| | - Charlotte A Peterson
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, KY USA ; Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA ; Center for Muscle Biology, University of Kentucky, Lexington, KY USA
| | - Esther E Dupont-Versteegden
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, KY USA ; Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA ; Center for Muscle Biology, University of Kentucky, Lexington, KY USA
| |
Collapse
|
673
|
Orlandi D, Corazza A, Arcidiacono A, Messina C, Serafini G, Sconfienza LM, Silvestri E. Ultrasound-guided procedures to treat sport-related muscle injuries. Br J Radiol 2015; 89:20150484. [PMID: 26562097 DOI: 10.1259/bjr.20150484] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Ultrasound is well known as a low-cost, radiation-free and effective imaging technique to guide percutaneous procedures. The lower limb muscles represent a good target to perform such procedures under ultrasound guidance, thus allowing for clear and precise visualization of the needle during the whole procedure. The knowledge of guidelines and technical aspects is mandatory to act in the most safe and accurate way on target tissues that can be as small as a few millimetres. This review will focus above the local treatments of traumatic lower limb muscle injuries described in literature, focusing on new and promising approaches, such as platelet-rich plasma treatment of muscle tears in athletes. For each procedure, a brief how-to-do practical guide will be provided, emphasizing precautions and tricks based on day-by-day experience that may help to improve the outcome of percutaneous ultrasound-guided procedures around the lower limb muscles.
Collapse
Affiliation(s)
- Davide Orlandi
- 1 Department of Radiology, Genoa University, Genova, Italy
| | - Angelo Corazza
- 1 Department of Radiology, Genoa University, Genova, Italy
| | | | - Carmelo Messina
- 2 Department of Radiology, IRCCS Policlinico San Donato, Milano, Italy
| | - Giovanni Serafini
- 3 Department of Radiology, Ospedale Santa Corona, Pietra Ligure, Savona, Italy
| | - Luca M Sconfienza
- 2 Department of Radiology, IRCCS Policlinico San Donato, Milano, Italy.,4 Department of Biomedical Sciences for Health, Università degli Studi di Milano, Italy
| | - Enzo Silvestri
- 5 Department of Radiology, Ospedale Evangelico Internazionale, Genoa, Italy
| |
Collapse
|
674
|
Biressi S, Miyabara EH, Gopinath SD, Carlig PMM, Rando TA. A Wnt-TGFβ2 axis induces a fibrogenic program in muscle stem cells from dystrophic mice. Sci Transl Med 2015; 6:267ra176. [PMID: 25520397 DOI: 10.1126/scitranslmed.3008411] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We have previously observed that Wnt signaling activates a fibrogenic program in adult muscle stem cells, called satellite cells, during aging. We genetically labeled satellite cells in a mouse model of Duchenne muscular dystrophy to follow their fate during the progression of the disease. We observed that a fraction of satellite cells had a reduced myogenic potential and showed enhanced expression of profibrotic genes compared to age-matched controls. By combining in vitro and in vivo results, we found that expression of transforming growth factor-β2 (TGFβ2) was induced in response to elevated canonical Wnt signaling in dystrophic muscles and that the resulting increase in TGFβ activity affected the behavior of satellite cells in an autocrine or paracrine fashion. Indeed, pharmacological inhibition of the TGFβ pathway in vivo reduced the fibrogenic characteristics of satellite cells. These studies shed new light on the cellular and molecular mechanisms responsible for stem cell dysfunction in dystrophic muscle and may contribute to the development of more effective and specific therapeutic approaches for the prevention of muscle fibrosis.
Collapse
Affiliation(s)
- Stefano Biressi
- Paul F. Glenn Laboratories for the Biology of Aging and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elen H Miyabara
- Paul F. Glenn Laboratories for the Biology of Aging and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA. Anatomy Department, Institute of Biomedical Sciences, University of São Paulo, 2415 Lineu Prestes Avenue, São Paulo, São Paulo 05508-000, Brazil
| | - Suchitra D Gopinath
- Paul F. Glenn Laboratories for the Biology of Aging and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Poppy M M Carlig
- Paul F. Glenn Laboratories for the Biology of Aging and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas A Rando
- Paul F. Glenn Laboratories for the Biology of Aging and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA. Neurology Service, VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA 94304, USA.
| |
Collapse
|
675
|
Blau HM, Cosgrove BD, Ho ATV. The central role of muscle stem cells in regenerative failure with aging. Nat Med 2015; 21:854-62. [PMID: 26248268 DOI: 10.1038/nm.3918] [Citation(s) in RCA: 311] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 07/10/2015] [Indexed: 02/07/2023]
Abstract
Skeletal muscle mass, function, and repair capacity all progressively decline with aging, restricting mobility, voluntary function, and quality of life. Skeletal muscle repair is facilitated by a population of dedicated muscle stem cells (MuSCs), also known as satellite cells, that reside in anatomically defined niches within muscle tissues. In adult tissues, MuSCs are retained in a quiescent state until they are primed to regenerate damaged muscle through cycles of self-renewal divisions. With aging, muscle tissue homeostasis is progressively disrupted and the ability of MuSCs to repair injured muscle markedly declines. Until recently, this decline has been largely attributed to extrinsic age-related alterations in the microenvironment to which MuSCs are exposed. However, as highlighted in this Perspective, recent reports show that MuSCs also progressively undergo cell-intrinsic alterations that profoundly affect stem cell regenerative function with aging. A more comprehensive understanding of the interplay of stem cell-intrinsic and extrinsic factors will set the stage for improving cell therapies capable of restoring tissue homeostasis and enhancing muscle repair in the aged.
Collapse
Affiliation(s)
- Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Andrew T V Ho
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
676
|
Comparative study of muscle regeneration following cardiotoxin and glycerol injury. Ann Anat 2015; 202:18-27. [DOI: 10.1016/j.aanat.2015.07.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 07/27/2015] [Accepted: 07/28/2015] [Indexed: 01/02/2023]
|
677
|
Lemos DR, Eisner C, Hopkins CI, Rossi FMV. Skeletal muscle-resident MSCs and bone formation. Bone 2015; 80:19-23. [PMID: 26103092 DOI: 10.1016/j.bone.2015.06.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/28/2015] [Accepted: 06/17/2015] [Indexed: 12/11/2022]
Abstract
Recent research has highlighted the importance of bone and muscle interactions during development and regeneration. There still remains, however, a large gap in the current understanding of the cells and mechanisms involved in this interplay. In particular, how muscle-derived cells, specifically mesenchymal stromal cells (MSCs), can impact bone regeneration or lead to pathologic ectopic bone formation is unclear. Here, a review is given of the evidence supporting the contribution of muscle-derived MSC to bone regeneration and suggesting a critical role for the inflammatory milieu. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Dario R Lemos
- Biomedical Research Centre, The University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Faculty of Medicine, The University of British Columbia, 317-2194 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Christine Eisner
- Biomedical Research Centre, The University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Faculty of Medicine, The University of British Columbia, 317-2194 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Claudia I Hopkins
- Biomedical Research Centre, The University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Faculty of Medicine, The University of British Columbia, 317-2194 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Fabio M V Rossi
- Biomedical Research Centre, The University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Faculty of Medicine, The University of British Columbia, 317-2194 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
678
|
Schaaf GJ, van Gestel TJM, Brusse E, Verdijk RM, de Coo IFM, van Doorn PA, van der Ploeg AT, Pijnappel WWMP. Lack of robust satellite cell activation and muscle regeneration during the progression of Pompe disease. Acta Neuropathol Commun 2015; 3:65. [PMID: 26510925 PMCID: PMC4625612 DOI: 10.1186/s40478-015-0243-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/08/2015] [Indexed: 01/01/2023] Open
Abstract
Introduction Muscle stem cells termed satellite cells are essential for muscle regeneration. A central question in many neuromuscular disorders is why satellite cells are unable to prevent progressive muscle wasting. We have analyzed muscle fiber pathology and the satellite cell response in Pompe disease, a metabolic myopathy caused by acid alpha-glucosidase deficiency and lysosomal glycogen accumulation. Pathology included muscle fiber vacuolization, loss of cross striation, and immune cell infiltration. Results The total number of Pax7-positive satellite cells in muscle biopsies from infantile, childhood onset and adult patients (with different ages and disease severities) were indistinguishable from controls, indicating that the satellite cell pool is not exhausted in Pompe disease. Pax7/Ki67 double stainings showed low levels of satellite cell proliferation similar to controls, while MyoD and Myogenin stainings showed undetectable satellite cell differentiation. Muscle regenerative activity monitored with expression of embryonic Myosin Heavy Chain was weak in the rapidly progressing classic infantile form and undetectable in the more slowly progressive childhood and adult onset disease including in severely affected patients. Conclusions These results imply that ongoing muscle wasting in Pompe disease may be explained by insufficient satellite cell activation and muscle regeneration. The preservation of the satellite cell pool may offer a venue for the development of novel treatment strategies directed towards the activation of endogenous satellite cells. Electronic supplementary material The online version of this article (doi:10.1186/s40478-015-0243-x) contains supplementary material, which is available to authorized users.
Collapse
|
679
|
Le Roux I, Konge J, Le Cam L, Flamant P, Tajbakhsh S. Numb is required to prevent p53-dependent senescence following skeletal muscle injury. Nat Commun 2015; 6:8528. [PMID: 26503169 PMCID: PMC4639798 DOI: 10.1038/ncomms9528] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 09/01/2015] [Indexed: 12/29/2022] Open
Abstract
Regeneration relies on coordinated action of multiple cell types to reconstitute the damaged tissue. Here we inactivate the endocytic adaptor protein Numb in skeletal muscle stem cells prior to chronic or severe muscle injury in mice. We observe two types of senescence in regenerating muscle; a transient senescence in non-myogenic cells of control and Numb mutant mice that partly depends on INK4a/ARF activity, and a persistent senescence in myogenic cells lacking Numb. The senescence levels of Numb-deficient muscle is reduced to wild type levels by an anti-oxidant treatment or p53 ablation, resulting in functional rescue of the regenerative potential in Numb mutants. Ex vivo experiments suggest that Numb-deficient senescent cells recruit macrophages to sustain inflammation and drive fibrosis, two hallmarks of the impaired muscle regeneration in Numb mutants. These findings provide insights into previously reported developmental and oncogenic senescence that are also differentially regulated by p53.
Collapse
Affiliation(s)
- Isabelle Le Roux
- Department of Developmental and Stem Cell Biology, Stem Cells and Development, CNRS URA 2578, Institut Pasteur, 25 rue du Dr Roux, Paris 75015, France
| | - Julie Konge
- Department of Developmental and Stem Cell Biology, Stem Cells and Development, CNRS URA 2578, Institut Pasteur, 25 rue du Dr Roux, Paris 75015, France
| | - Laurent Le Cam
- Molecular Basis of Carcinogenesis, Institut de Recherche en Cancérologie de Montpellier, 208 rue des Apothicaires, Montpellier, cedex 5 34298, France
| | - Patricia Flamant
- Human Histopathology and Animal Models, Institut Pasteur, 25 rue du Dr Roux, Paris 75015, France
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell Biology, Stem Cells and Development, CNRS URA 2578, Institut Pasteur, 25 rue du Dr Roux, Paris 75015, France
| |
Collapse
|
680
|
Gumucio JP, Sugg KB, Mendias CL. TGF-β superfamily signaling in muscle and tendon adaptation to resistance exercise. Exerc Sport Sci Rev 2015; 43:93-9. [PMID: 25607281 DOI: 10.1249/jes.0000000000000041] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Numerous studies in muscle and tendon have identified a central role of the transforming growth factor-β (TGF-β) superfamily of cytokines in the regulation of extracellular matrix growth and remodeling, protein degradation, and cell proliferation and differentiation. We provide a novel framework for TGF-β and myostatin signaling in controlling the coordinated adaptation of both skeletal muscle and tendon tissue to resistance training.
Collapse
Affiliation(s)
- Jonathan P Gumucio
- Departments of 1Orthopaedic Surgery, 2Molecular and Integrative Physiology, and 3Surgery, Section of Plastic Surgery, University of Michigan Medical School, Ann Arbor, MI
| | | | | |
Collapse
|
681
|
Snijders T, Nederveen JP, McKay BR, Joanisse S, Verdijk LB, van Loon LJC, Parise G. Satellite cells in human skeletal muscle plasticity. Front Physiol 2015; 6:283. [PMID: 26557092 PMCID: PMC4617172 DOI: 10.3389/fphys.2015.00283] [Citation(s) in RCA: 227] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/23/2015] [Indexed: 01/06/2023] Open
Abstract
Skeletal muscle satellite cells are considered to play a crucial role in muscle fiber maintenance, repair and remodeling. Our knowledge of the role of satellite cells in muscle fiber adaptation has traditionally relied on in vitro cell and in vivo animal models. Over the past decade, a genuine effort has been made to translate these results to humans under physiological conditions. Findings from in vivo human studies suggest that satellite cells play a key role in skeletal muscle fiber repair/remodeling in response to exercise. Mounting evidence indicates that aging has a profound impact on the regulation of satellite cells in human skeletal muscle. Yet, the precise role of satellite cells in the development of muscle fiber atrophy with age remains unresolved. This review seeks to integrate recent results from in vivo human studies on satellite cell function in muscle fiber repair/remodeling in the wider context of satellite cell biology whose literature is largely based on animal and cell models.
Collapse
Affiliation(s)
- Tim Snijders
- Department of Kinesiology and Medical Physics and Applied Radiation Sciences, McMaster University Hamilton, ON, Canada ; Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Maastricht, Netherlands
| | - Joshua P Nederveen
- Department of Kinesiology and Medical Physics and Applied Radiation Sciences, McMaster University Hamilton, ON, Canada
| | - Bryon R McKay
- Department of Kinesiology and Medical Physics and Applied Radiation Sciences, McMaster University Hamilton, ON, Canada
| | - Sophie Joanisse
- Department of Kinesiology and Medical Physics and Applied Radiation Sciences, McMaster University Hamilton, ON, Canada
| | - Lex B Verdijk
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Maastricht, Netherlands
| | - Luc J C van Loon
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Maastricht, Netherlands
| | - Gianni Parise
- Department of Kinesiology and Medical Physics and Applied Radiation Sciences, McMaster University Hamilton, ON, Canada
| |
Collapse
|
682
|
Mizuno S, Yoda M, Shimoda M, Tohmonda T, Okada Y, Toyama Y, Takeda S, Nakamura M, Matsumoto M, Horiuchi K. A Disintegrin and Metalloprotease 10 (ADAM10) Is Indispensable for Maintenance of the Muscle Satellite Cell Pool. J Biol Chem 2015; 290:28456-28464. [PMID: 26453297 DOI: 10.1074/jbc.m115.653477] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Indexed: 12/20/2022] Open
Abstract
Satellite cells (SCs) are muscle-specific stem cells that are essential for the regeneration of damaged muscles. Although SCs have a robust capacity to regenerate myofibers, the number of SCs decreases with aging, leading to insufficient recovery after muscle injury. We herein show that ADAM10 (a disintegrin and metalloprotease 10), a membrane-bound proteolytic enzyme with a critical role in Notch processing (S2 cleavage), is essential for the maintenance of SC quiescence. We generated mutant mice in which ADAM10 in SCs can be conditionally abrogated by tamoxifen injection. Tamoxifen-treated mutant mice did not show any apparent defects and grew normally under unchallenged conditions. However, these mice showed a nearly complete loss of muscle regeneration after chemically induced muscle injury. In situ hybridization and flow cytometric analyses revealed that the mutant mice had significantly less SCs compared with wild type controls. Of note, we found that inactivation of ADAM10 in SCs severely compromised Notch signaling and led to dysregulated myogenic differentiation, ultimately resulting in deprivation of the SC pool in vivo. Taken together, the present findings underscore the role of ADAM10 as an indispensable component of Notch signaling in SCs and for maintaining the SC pool.
Collapse
Affiliation(s)
- Sakiko Mizuno
- Departments of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masaki Yoda
- Anti-aging Orthopedic Research, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masayuki Shimoda
- Pathology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Takahide Tohmonda
- Anti-aging Orthopedic Research, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yasunori Okada
- Pathology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yoshiaki Toyama
- Departments of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
| | - Masaya Nakamura
- Departments of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Morio Matsumoto
- Departments of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Keisuke Horiuchi
- Departments of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan; Anti-aging Orthopedic Research, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
683
|
Messi ML, Li T, Wang ZM, Marsh AP, Nicklas B, Delbono O. Resistance Training Enhances Skeletal Muscle Innervation Without Modifying the Number of Satellite Cells or their Myofiber Association in Obese Older Adults. J Gerontol A Biol Sci Med Sci 2015; 71:1273-80. [PMID: 26447161 DOI: 10.1093/gerona/glv176] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/17/2015] [Indexed: 11/14/2022] Open
Abstract
Studies in humans and animal models provide compelling evidence for age-related skeletal muscle denervation, which may contribute to muscle fiber atrophy and loss. Skeletal muscle denervation seems relentless; however, long-term, high-intensity physical activity appears to promote muscle reinnervation. Whether 5-month resistance training (RT) enhances skeletal muscle innervation in obese older adults is unknown. This study found that neural cell-adhesion molecule, NCAM+ muscle area decreased with RT and was inversely correlated with muscle strength. NCAM1 and RUNX1 gene transcripts significantly decreased with the intervention. Type I and type II fiber grouping in the vastus lateralis did not change significantly but increases in leg press and knee extensor strength inversely correlated with type I, but not with type II, fiber grouping. RT did not modify the total number of satellite cells, their number per area, or the number associated with specific fiber subtypes or innervated/denervated fibers. Our results suggest that RT has a beneficial impact on skeletal innervation, even when started late in life by sedentary obese older adults.
Collapse
Affiliation(s)
- María Laura Messi
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine and J Paul Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tao Li
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine and J Paul Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhong-Min Wang
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine and J Paul Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Anthony P Marsh
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, North Carolina
| | - Barbara Nicklas
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine and J Paul Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine and J Paul Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina.
| |
Collapse
|
684
|
Randolph ME, Pavlath GK. A muscle stem cell for every muscle: variability of satellite cell biology among different muscle groups. Front Aging Neurosci 2015; 7:190. [PMID: 26500547 PMCID: PMC4595652 DOI: 10.3389/fnagi.2015.00190] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/21/2015] [Indexed: 12/22/2022] Open
Abstract
The human body contains approximately 640 individual skeletal muscles. Despite the fact that all of these muscles are composed of striated muscle tissue, the biology of these muscles and their associated muscle stem cell populations are quite diverse. Skeletal muscles are affected differentially by various muscular dystrophies (MDs), such that certain genetic mutations specifically alter muscle function in only a subset of muscles. Additionally, defective muscle stem cells have been implicated in the pathology of some MDs. The biology of muscle stem cells varies depending on the muscles with which they are associated. Here we review the biology of skeletal muscle stem cell populations of eight different muscle groups. Understanding the biological variation of skeletal muscles and their resident stem cells could provide valuable insight into mechanisms underlying the susceptibility of certain muscles to myopathic disease.
Collapse
|
685
|
MacKrell JG, Yaden BC, Bullock H, Chen K, Shetler P, Bryant HU, Krishnan V. Molecular targets of androgen signaling that characterize skeletal muscle recovery and regeneration. NUCLEAR RECEPTOR SIGNALING 2015; 13:e005. [PMID: 26457071 PMCID: PMC4599140 DOI: 10.1621/nrs.13005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 09/05/2015] [Indexed: 01/25/2023]
Abstract
The high regenerative capacity of adult skeletal muscle relies on a self-renewing depot of adult stem cells, termed muscle satellite cells (MSCs). Androgens, known mediators of overall body composition and specifically skeletal muscle mass, have been shown to regulate MSCs. The possible overlapping function of androgen regulation of muscle growth and MSC activation has not been carefully investigated with regards to muscle regeneration.Therefore, the aim of this study was to examine coinciding androgen-mediated genetic changes in an in vitro MSC model and clinically relevant in vivo models. A gene signature was established via microarray analysis for androgen-mediated MSC engagement and highlighted several markers including follistatin (FST), IGF-1, C-X-C chemokine receptor 4 (CXCR4), hepatocyte growth factor (HGF) and glucocorticoid receptor (GR). In an in vivo muscle atrophy model, androgen re-supplementation significantly increased muscle size and expression of IGF-1, FST, and HGF, while significantly decreasing expression of GR. Biphasic gene expression profiles over the 7-day re-supplementation period identified temporal androgen regulation of molecular targets involved in satellite cell engagement into myogenesis. In a muscle injury model, removal of androgens resulted in delayed muscle recovery and regeneration. Modifications in the androgen signaling gene signature, along with reduced Pax7 and MyoD expression, suggested that limited MSC activation and increased inflammation contributed to the delayed regeneration. However, enhanced MSC activation in the androgen-deplete mouse injury model was driven by an androgen receptor (AR) agonist. These results provide novel in vitro and in vivo evidence describing molecular targets of androgen signaling, while also increasing support for translational use of AR agonists in skeletal muscle recovery and regeneration.
Collapse
Affiliation(s)
- James G MacKrell
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Benjamin C Yaden
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Heather Bullock
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Keyue Chen
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Pamela Shetler
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Henry U Bryant
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Venkatesh Krishnan
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| |
Collapse
|
686
|
Grasman JM, Zayas MJ, Page RL, Pins GD. Biomimetic scaffolds for regeneration of volumetric muscle loss in skeletal muscle injuries. Acta Biomater 2015. [PMID: 26219862 DOI: 10.1016/j.actbio.2015.07.038] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Skeletal muscle injuries typically result from traumatic incidents such as combat injuries where soft-tissue extremity injuries are present in one of four cases. Further, about 4.5 million reconstructive surgical procedures are performed annually as a result of car accidents, cancer ablation, or cosmetic procedures. These combat- and trauma-induced skeletal muscle injuries are characterized by volumetric muscle loss (VML), which significantly reduces the functionality of the injured muscle. While skeletal muscle has an innate repair mechanism, it is unable to compensate for VML injuries because large amounts of tissue including connective tissue and basement membrane are removed or destroyed. This results in a significant need to develop off-the-shelf biomimetic scaffolds to direct skeletal muscle regeneration. Here, the structure and organization of native skeletal muscle tissue is described in order to reveal clear design parameters that are necessary for scaffolds to mimic in order to successfully regenerate muscular tissue. We review the literature with respect to the materials and methodologies used to develop scaffolds for skeletal muscle tissue regeneration as well as the limitations of these materials. We further discuss the variety of cell sources and different injury models to provide some context for the multiple approaches used to evaluate these scaffold materials. Recent findings are highlighted to address the state of the field and directions are outlined for future strategies, both in scaffold design and in the use of different injury models to evaluate these materials, for regenerating functional skeletal muscle. STATEMENT OF SIGNIFICANCE Volumetric muscle loss (VML) injuries result from traumatic incidents such as those presented from combat missions, where soft-tissue extremity injuries are represented in one of four cases. These injuries remove or destroy large amounts of skeletal muscle including the basement membrane and connective tissue, removing the structural, mechanical, and biochemical cues that usually direct its repair. This results in a significant need to develop off-the-shelf biomimetic scaffolds to direct skeletal muscle regeneration. In this review, we examine current strategies for the development of scaffold materials designed for skeletal muscle regeneration, highlighting advances and limitations associated with these methodologies. Finally, we identify future approaches to enhance skeletal muscle regeneration.
Collapse
|
687
|
Abstract
The developmental mechanisms that control head muscle formation are distinct from those that operate in the trunk. Head and neck muscles derive from various mesoderm populations in the embryo and are regulated by distinct transcription factors and signaling molecules. Throughout the last decade, developmental, and lineage studies in vertebrates and invertebrates have revealed the peculiar nature of the pharyngeal mesoderm that forms certain head muscles and parts of the heart. Studies in chordates, the ancestors of vertebrates, revealed an evolutionarily conserved cardiopharyngeal field that progressively facilitates the development of both heart and craniofacial structures during vertebrate evolution. This ancient regulatory circuitry preceded and facilitated the emergence of myogenic cell types and hierarchies that exist in vertebrates. This chapter summarizes studies related to the origins, signaling circuits, genetics, and evolution of the head musculature, highlighting its heterogeneous characteristics in all these aspects, with a special focus on the FGF-ERK pathway. Additionally, we address the processes of head muscle regeneration, and the development of stem cell-based therapies for treatment of muscle disorders.
Collapse
Affiliation(s)
- Inbal Michailovici
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Eigler
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Tzahor
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
688
|
Buckingham M, Relaix F. PAX3 and PAX7 as upstream regulators of myogenesis. Semin Cell Dev Biol 2015; 44:115-25. [PMID: 26424495 DOI: 10.1016/j.semcdb.2015.09.017] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/23/2015] [Indexed: 10/23/2022]
Abstract
Like other subclasses within the PAX transcription factor family, PAX3 and PAX7 play important roles in the emergence of a number of different tissues during development. PAX3 regulates neural crest and, together with its orthologue PAX7, is also expressed in parts of the central nervous system. In this chapter we will focus on their role in skeletal muscle. Both factors are key regulators of myogenesis where Pax3 plays a major role during early skeletal muscle formation in the embryo while Pax7 predominates during post-natal growth and muscle regeneration in the adult. We review the expression and functions of these factors in the myogenic context. We also discuss mechanistic aspects of PAX3/7 function and modulation of their activity by interaction with other proteins, as well as the post-transcriptional and transcriptional regulation of their expression.
Collapse
Affiliation(s)
- Margaret Buckingham
- Department of Developmental and Stem Cell Biology, CNRS URA 2578, Institut Pasteur, 28 rue du Dr Roux, 75015 Paris, France.
| | - Frédéric Relaix
- INSERM U955 IMRB, Team 10, 94000 Creteil, France; UPEC Paris Est-Creteil University, Faculty of Medicine, F-94000 Creteil, France; Etablissement Français du Sang, 94017 Creteil, France; Université Paris Est, Ecole Nationale Veterinaire d'Alfort, 94700 Maison Alfort, France.
| |
Collapse
|
689
|
Aguilar CA, Shcherbina A, Ricke DO, Pop R, Carrigan CT, Gifford CA, Urso ML, Kottke MA, Meissner A. In vivo Monitoring of Transcriptional Dynamics After Lower-Limb Muscle Injury Enables Quantitative Classification of Healing. Sci Rep 2015; 5:13885. [PMID: 26381351 PMCID: PMC4585378 DOI: 10.1038/srep13885] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 08/07/2015] [Indexed: 01/07/2023] Open
Abstract
Traumatic lower-limb musculoskeletal injuries are pervasive amongst athletes and the military and typically an individual returns to activity prior to fully healing, increasing a predisposition for additional injuries and chronic pain. Monitoring healing progression after a musculoskeletal injury typically involves different types of imaging but these approaches suffer from several disadvantages. Isolating and profiling transcripts from the injured site would abrogate these shortcomings and provide enumerative insights into the regenerative potential of an individual's muscle after injury. In this study, a traumatic injury was administered to a mouse model and healing progression was examined from 3 hours to 1 month using high-throughput RNA-Sequencing (RNA-Seq). Comprehensive dissection of the genome-wide datasets revealed the injured site to be a dynamic, heterogeneous environment composed of multiple cell types and thousands of genes undergoing significant expression changes in highly regulated networks. Four independent approaches were used to determine the set of genes, isoforms, and genetic pathways most characteristic of different time points post-injury and two novel approaches were developed to classify injured tissues at different time points. These results highlight the possibility to quantitatively track healing progression in situ via transcript profiling using high- throughput sequencing.
Collapse
Affiliation(s)
- Carlos A. Aguilar
- Massachusetts Institute of Technology - Lincoln Laboratory, Lexington, MA 02127, USA
| | - Anna Shcherbina
- Massachusetts Institute of Technology - Lincoln Laboratory, Lexington, MA 02127, USA
| | - Darrell O. Ricke
- Massachusetts Institute of Technology - Lincoln Laboratory, Lexington, MA 02127, USA
| | - Ramona Pop
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, Harvard Stem Cell Institute, Cambridge, MA 02138, Dept. of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Christopher T. Carrigan
- United States Army Institute of Environmental Medicine - Military Performance Division, Natick, MA 01760, USA.
| | - Casey A. Gifford
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, Harvard Stem Cell Institute, Cambridge, MA 02138, Dept. of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Maria L. Urso
- United States Army Institute of Environmental Medicine - Military Performance Division, Natick, MA 01760, USA.
| | - Melissa A. Kottke
- United States Army Institute of Environmental Medicine - Military Performance Division, Natick, MA 01760, USA.
| | - Alexander Meissner
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, Harvard Stem Cell Institute, Cambridge, MA 02138, Dept. of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
690
|
Saera-Vila A, Kasprick DS, Junttila TL, Grzegorski SJ, Louie KW, Chiari EF, Kish PE, Kahana A. Myocyte Dedifferentiation Drives Extraocular Muscle Regeneration in Adult Zebrafish. Invest Ophthalmol Vis Sci 2015; 56:4977-93. [PMID: 26230763 DOI: 10.1167/iovs.14-16103] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The purpose of this study was to characterize the injury response of extraocular muscles (EOMs) in adult zebrafish. METHODS Adult zebrafish underwent lateral rectus (LR) muscle myectomy surgery to remove 50% of the muscle, followed by molecular and cellular characterization of the tissue response to the injury. RESULTS Following myectomy, the LR muscle regenerated an anatomically correct and functional muscle within 7 to 10 days post injury (DPI). Following injury, the residual muscle stump was replaced by a mesenchymal cell population that lost cell polarity and expressed mesenchymal markers. Next, a robust proliferative burst repopulated the area of the regenerating muscle. Regenerating cells expressed myod, identifying them as myoblasts. However, both immunofluorescence and electron microscopy failed to identify classic Pax7-positive satellite cells in control or injured EOMs. Instead, some proliferating nuclei were noted to express mef2c at the very earliest point in the proliferative burst, suggesting myonuclear reprogramming and dedifferentiation. Bromodeoxyuridine (BrdU) labeling of regenerating cells followed by a second myectomy without repeat labeling resulted in a twice-regenerated muscle broadly populated by BrdU-labeled nuclei with minimal apparent dilution of the BrdU signal. A double-pulse experiment using BrdU and 5-ethynyl-2'-deoxyuridine (EdU) identified double-labeled nuclei, confirming the shared progenitor lineage. Rapid regeneration occurred despite a cell cycle length of 19.1 hours, whereas 72% of the regenerating muscle nuclei entered the cell cycle by 48 hours post injury (HPI). Dextran lineage tracing revealed that residual myocytes were responsible for muscle regeneration. CONCLUSIONS EOM regeneration in adult zebrafish occurs by dedifferentiation of residual myocytes involving a muscle-to-mesenchyme transition. A mechanistic understanding of myocyte reprogramming may facilitate novel approaches to the development of molecular tools for targeted therapeutic regeneration in skeletal muscle disorders and beyond.
Collapse
|
691
|
Abstract
The abundance and cross-linking of intramuscular connective tissue contributes to the background toughness of meat, and is thus undesirable. Connective tissue is mainly synthesized by intramuscular fibroblasts. Myocytes, adipocytes and fibroblasts are derived from a common pool of progenitor cells during the early embryonic development. It appears that multipotent mesenchymal stem cells first diverge into either myogenic or non-myogenic lineages; non-myogenic mesenchymal progenitors then develop into the stromal-vascular fraction of skeletal muscle wherein adipocytes, fibroblasts and derived mesenchymal progenitors reside. Because non-myogenic mesenchymal progenitors mainly undergo adipogenic or fibrogenic differentiation during muscle development, strengthening progenitor proliferation enhances the potential for both intramuscular adipogenesis and fibrogenesis, leading to the elevation of both marbling and connective tissue content in the resulting meat product. Furthermore, given the bipotent developmental potential of progenitor cells, enhancing their conversion to adipogenesis reduces fibrogenesis, which likely results in the overall improvement of marbling (more intramuscular adipocytes) and tenderness (less connective tissue) of meat. Fibrogenesis is mainly regulated by the transforming growth factor (TGF) β signaling pathway and its regulatory cascade. In addition, extracellular matrix, a part of the intramuscular connective tissue, provides a niche environment for regulating myogenic differentiation of satellite cells and muscle growth. Despite rapid progress, many questions remain in the role of extracellular matrix on muscle development, and factors determining the early differentiation of myogenic, adipogenic and fibrogenic cells, which warrant further studies.
Collapse
|
692
|
Joanisse S, McKay BR, Nederveen JP, Scribbans TD, Gurd BJ, Gillen JB, Gibala MJ, Tarnopolsky M, Parise G. Satellite cell activity, without expansion, after nonhypertrophic stimuli. Am J Physiol Regul Integr Comp Physiol 2015; 309:R1101-11. [PMID: 26333785 DOI: 10.1152/ajpregu.00249.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/27/2015] [Indexed: 11/22/2022]
Abstract
The purpose of the present studies was to determine the effect of various nonhypertrophic exercise stimuli on satellite cell (SC) pool activity in human skeletal muscle. Previously untrained men and women (men: 29 ± 9 yr and women: 29 ± 2 yr, n = 7 each) completed 6 wk of very low-volume high-intensity sprint interval training. In a separate study, recreationally active men (n = 16) and women (n = 3) completed 6 wk of either traditional moderate-intensity continuous exercise (n = 9, 21 ± 4 yr) or low-volume sprint interval training (n = 10, 21 ± 2 yr). Muscle biopsies were obtained from the vastus lateralis before and after training. The fiber type-specific SC response to training was determined, as was the activity of the SC pool using immunofluorescent microscopy of muscle cross sections. Training did not induce hypertrophy, as assessed by muscle cross-sectional area, nor did the SC pool expand in any group. However, there was an increase in the number of active SCs after each intervention. Specifically, the number of activated (Pax7(+)/MyoD(+), P ≤ 0.05) and differentiating (Pax7(-)/MyoD(+), P ≤ 0.05) SCs increased after each training intervention. Here, we report evidence of activated and cycling SCs that may or may not contribute to exercise-induced adaptations while the SC pool remains constant after three nonhypertrophic exercise training protocols.
Collapse
Affiliation(s)
- Sophie Joanisse
- Departments of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Bryon R McKay
- Departments of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Joshua P Nederveen
- Departments of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Trisha D Scribbans
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada
| | - Brendon J Gurd
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada
| | - Jenna B Gillen
- Departments of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Martin J Gibala
- Departments of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Mark Tarnopolsky
- Department of Pediatrics and Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Gianni Parise
- Departments of Kinesiology, McMaster University, Hamilton, Ontario, Canada; Medical Physics and Applied Radiation Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
693
|
Shan T, Zhang P, Liang X, Bi P, Yue F, Kuang S. Lkb1 is indispensable for skeletal muscle development, regeneration, and satellite cell homeostasis. Stem Cells 2015; 32:2893-907. [PMID: 25069613 DOI: 10.1002/stem.1788] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 06/14/2014] [Accepted: 06/19/2014] [Indexed: 12/17/2022]
Abstract
Serine/threonine kinase 11, commonly known as liver kinase b1 (Lkb1), is a tumor suppressor that regulates cellular energy metabolism and stem cell function. Satellite cells are skeletal muscle resident stem cells that maintain postnatal muscle growth and repair. Here, we used MyoD(Cre)/Lkb1(flox/flox) mice (called MyoD-Lkb1) to delete Lkb1 in embryonic myogenic progenitors and their descendant satellite cells and myofibers. The MyoD-Lkb1 mice exhibit a severe myopathy characterized by central nucleated myofibers, reduced mobility, growth retardation, and premature death. Although tamoxifen-induced postnatal deletion of Lkb1 in satellite cells using Pax7(CreER) mice bypasses the developmental defects and early death, Lkb1 null satellite cells lose their regenerative capacity cell-autonomously. Strikingly, Lkb1 null satellite cells fail to maintain quiescence in noninjured resting muscles and exhibit accelerated proliferation but reduced differentiation kinetics. At the molecular level, Lkb1 limits satellite cell proliferation through the canonical AMP-activated protein kinase/mammalian target of rapamycin pathway, but facilitates differentiation through phosphorylation of GSK-3β, a key component of the WNT signaling pathway. Together, these results establish a central role of Lkb1 in muscle stem cell homeostasis, muscle development, and regeneration.
Collapse
Affiliation(s)
- Tizhong Shan
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | | | | | | | | | | |
Collapse
|
694
|
Rapid release of growth factors regenerates force output in volumetric muscle loss injuries. Biomaterials 2015; 72:49-60. [PMID: 26344363 DOI: 10.1016/j.biomaterials.2015.08.047] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 08/18/2015] [Accepted: 08/25/2015] [Indexed: 11/21/2022]
Abstract
A significant challenge in the design and development of biomaterial scaffolds is to incorporate mechanical and biochemical cues to direct organized tissue growth. In this study, we investigated the effect of hepatocyte growth factor (HGF) loaded, crosslinked fibrin (EDCn-HGF) microthread scaffolds on skeletal muscle regeneration in a mouse model of volumetric muscle loss (VML). The rapid, sustained release of HGF significantly enhanced the force production of muscle tissue 60 days after injury, recovering more than 200% of the force output relative to measurements recorded immediately after injury. HGF delivery increased the number of differentiating myoblasts 14 days after injury, and supported an enhanced angiogenic response. The architectural morphology of microthread scaffolds supported the ingrowth of nascent myofibers into the wound site, in contrast to fibrin gel implants which did not support functional regeneration. Together, these data suggest that EDCn-HGF microthreads recapitulate several of the regenerative cues lost in VML injuries, promote remodeling of functional muscle tissue, and enhance the functional regeneration of skeletal muscle. Further, by strategically incorporating specific biochemical factors and precisely tuning the structural and mechanical properties of fibrin microthreads, we have developed a powerful platform technology that may enhance regeneration in other axially aligned tissues.
Collapse
|
695
|
Liu W, Wei-LaPierre L, Klose A, Dirksen RT, Chakkalakal JV. Inducible depletion of adult skeletal muscle stem cells impairs the regeneration of neuromuscular junctions. eLife 2015; 4. [PMID: 26312504 PMCID: PMC4579298 DOI: 10.7554/elife.09221] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/26/2015] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle maintenance depends on motor innervation at neuromuscular junctions (NMJs). Multiple mechanisms contribute to NMJ repair and maintenance; however muscle stem cells (satellite cells, SCs), are deemed to have little impact on these processes. Therefore, the applicability of SC studies to attenuate muscle loss due to NMJ deterioration as observed in neuromuscular diseases and aging is ambiguous. We employed mice with an inducible Cre, and conditionally expressed DTA to deplete or GFP to track SCs. We found SC depletion exacerbated muscle atrophy and type transitions connected to neuromuscular disruption. Also, elevated fibrosis and further declines in force generation were specific to SC depletion and neuromuscular disruption. Fate analysis revealed SC activity near regenerating NMJs. Moreover, SC depletion aggravated deficits in reinnervation and post-synaptic morphology at regenerating NMJs. Therefore, our results propose a mechanism whereby further NMJ and skeletal muscle decline ensues upon SC depletion and neuromuscular disruption. DOI:http://dx.doi.org/10.7554/eLife.09221.001 New muscle fibers are made throughout our lives to replace those that have been damaged by normal wear and tear, and to meet new physical demands. These new muscle fibers develop from a pool of muscle stem cells. To create and maintain fully working muscles, nerve cells called motor neurons must also properly attach to the muscle fibers. These nerve cells transmit messages from the brain that tell the muscles what to do. If the muscle-nerve connections do not form correctly, or are severed, muscles can waste away. This may occur as part of a neuromuscular disease, and also happens to some extent as a normal part of aging. It was thought that muscle stem cells do not affect how the muscle-nerve connections form. By studying genetically engineered mice, Liu et al. now show that this is not the case. These mice had modifications to their muscle stem cells that allowed the number of these cells to be artificially reduced, and some cells also produced a fluorescent protein that allowed them to be tracked. Surgically severing some of the muscle-nerve connections in the mice triggered the rebuilding of the connections, but also weakened the muscles and caused some disease-related changes in the muscle tissue. During the healing process, the muscle stem cells are active near the regenerating connections. Reducing the number of muscle stem cells in the mice while these broken connections were healing further weakened the muscles. Closer inspection of the muscle-nerve connections also revealed poorer quality connections were formed in the stem-cell deficient mice. Further study of how stem cells help to form strong nerve-muscle connections may allow scientists to develop new treatments for age- or disease-related muscle loss. DOI:http://dx.doi.org/10.7554/eLife.09221.002
Collapse
Affiliation(s)
- Wenxuan Liu
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States
| | - Lan Wei-LaPierre
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, United States
| | - Alanna Klose
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, United States
| | - Joe V Chakkalakal
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States
| |
Collapse
|
696
|
Bustos F, de la Vega E, Cabezas F, Thompson J, Cornelison DDW, Olwin BB, Yates JR, Olguín HC. NEDD4 Regulates PAX7 Levels Promoting Activation of the Differentiation Program in Skeletal Muscle Precursors. Stem Cells 2015; 33:3138-51. [PMID: 26304770 DOI: 10.1002/stem.2125] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023]
Abstract
The transcription factor Pax7 regulates skeletal muscle stem cell (satellite cells) specification and maintenance through various mechanisms, including repressing the activity of the muscle regulatory factor MyoD. Hence, Pax7-to-MyoD protein ratios can determine maintenance of the committed-undifferentiated state or activation of the differentiation program. Pax7 expression decreases sharply in differentiating myoblasts but is maintained in cells (re)acquiring quiescence, yet the mechanisms regulating Pax7 levels based on differentiation status are not well understood. Here we show that Pax7 levels are directly regulated by the ubiquitin-ligase Nedd4. Our results indicate that Nedd4 is expressed in quiescent and activated satellite cells, that Nedd4 and Pax7 physically interact during early muscle differentiation-correlating with Pax7 ubiquitination and decline-and that Nedd4 loss of function prevented this effect. Furthermore, even transient nuclear accumulation of Nedd4 induced a drop in Pax7 levels and precocious muscle differentiation. Consequently, we propose that Nedd4 functions as a novel Pax7 regulator, which activity is temporally and spatially controlled to modulate the Pax7 protein levels and therefore satellite cell fate.
Collapse
Affiliation(s)
- Francisco Bustos
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eduardo de la Vega
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Cabezas
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - James Thompson
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA
| | - D D W Cornelison
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Bradley B Olwin
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA
| | - Hugo C Olguín
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
697
|
Padilla S, Sánchez M, Orive G, Anitua E. Towards a correct timing and dosage in PRP applications. Injury 2015; 46:1697-8. [PMID: 25920374 DOI: 10.1016/j.injury.2015.03.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/05/2015] [Indexed: 02/02/2023]
Affiliation(s)
- S Padilla
- Foundation Eduardo Anitua, Vitoria, Spain
| | - M Sánchez
- Arthroscopy Surgery Unit, Hospital Vithas San José, Vitoria, Spain
| | - G Orive
- Foundation Eduardo Anitua, Vitoria, Spain.
| | - E Anitua
- Foundation Eduardo Anitua, Vitoria, Spain; Biotechnology Institute (BTI), Vitoria, Spain
| |
Collapse
|
698
|
Chakroun I, Yang D, Girgis J, Gunasekharan A, Phenix H, Kærn M, Blais A. Genome-wide association between Six4, MyoD, and the histone demethylase Utx during myogenesis. FASEB J 2015; 29:4738-55. [PMID: 26229056 DOI: 10.1096/fj.15-277053] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/20/2015] [Indexed: 01/14/2023]
Abstract
Adult skeletal muscles can regenerate after injury, due to the presence of satellite cells, a quiescent population of myogenic progenitor cells. Once activated, satellite cells repair the muscle damage by undergoing myogenic differentiation. The myogenic regulatory factors (MRFs) coordinate the process of progenitor differentiation in cooperation with other families of transcription factors (TFs). The Six1 and Six4 homeodomain TFs are expressed in developing and adult muscle and Six1 is critical for embryonic and adult myogenesis. However, the lack of a muscle developmental phenotype in Six4-null mice, which has been attributed to compensation by other Six family members, has discouraged further assessment of the role of Six4 during adult muscle regeneration. By employing genome-wide approaches to address the function of Six4 during adult skeletal myogenesis, we have identified a core set of muscle genes coordinately regulated in adult muscle precursors by Six4 and the MRF MyoD. Throughout the genome of differentiating adult myoblasts, the cooperation between Six4 and MyoD is associated with chromatin repressive mark removal by Utx, a demethylase of histone H3 trimethylated at lysine 27. Among the genes coordinately regulated by Six4 and MyoD are several genes critical for proper in vivo muscle regeneration, implicating a role of Six4 in this process. Using in vivo RNA interference of Six4, we expose an uncompensated function of this TF during muscle regeneration. Together, our results reveal a role for Six4 during adult muscle regeneration and suggest a widespread mechanism of cooperation between Six4 and MyoD.
Collapse
Affiliation(s)
- Imane Chakroun
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Dabo Yang
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - John Girgis
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Atchayaa Gunasekharan
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Hilary Phenix
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Mads Kærn
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Alexandre Blais
- *Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; and Department of Biochemistry, Microbiology, and Immunology and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
699
|
Abstract
The Parkes Weber syndrome is a congenital vascular malformation, characterized by varicose veins, arterio-venous fistulas and overgrown limbs. No broadly accepted animal model of Parkes Weber syndrome has been described. We created side-to-side arterio-venous fistula between common femoral vessels with proximal non-absorbable ligature on common femoral vein limiting the enlargement of the vein diameter in Wistar rats. Contralateral limb was sham operated. Invasive blood pressure measurements in both iliac and inferior cava veins were performed in rats 30 days after fistula creation. Tight circumference and femoral bone length were measured. Histopathology and morphology of soleus muscle, extensor digitorum longus muscle, and the common femoral vessel were analyzed. 30 days following arterio-venous fistula creation, a statistically significant elevation of blood pressure in common iliac vein and limb overgrowth was observed. Limb enlargement was caused by muscle overgrowth, varicose veins formation and bone elongation. Arterio-venous fistula with proximal outflow limitation led to significant increase of femoral vein circumference and venous wall thickness. Our study indicates that the described rat model mimics major clinical features characteristic for the human Parkes Weber syndrome: presence of arterio-venous fistula, venous hypertension and dilatation, varicose veins formation, and the limb hypertrophy. We reveal that limb overgrowth is caused by bone elongation, muscle hypertrophy, and venous dilatation. The newly established model will permit detailed studies on the mechanisms underlying the disease and on the efficacy of novel therapeutic strategies for the Parkes Weber syndrome treatment.
Collapse
|
700
|
Moresi V, Marroncelli N, Adamo S. New insights into the epigenetic control of satellite cells. World J Stem Cells 2015; 7:945-955. [PMID: 26240681 PMCID: PMC4515437 DOI: 10.4252/wjsc.v7.i6.945] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 02/12/2015] [Accepted: 05/11/2015] [Indexed: 02/06/2023] Open
Abstract
Epigenetics finely tunes gene expression at a functional level without modifying the DNA sequence, thereby contributing to the complexity of genomic regulation. Satellite cells (SCs) are adult muscle stem cells that are important for skeletal post-natal muscle growth, homeostasis and repair. The understanding of the epigenome of SCs at different stages and of the multiple layers of the post-transcriptional regulation of gene expression is constantly expanding. Dynamic interactions between different epigenetic mechanisms regulate the appropriate timing of muscle-specific gene expression and influence the lineage fate of SCs. In this review, we report and discuss the recent literature about the epigenetic control of SCs during the myogenic process from activation to proliferation and from their commitment to a muscle cell fate to their differentiation and fusion to myotubes. We describe how the coordinated activities of the histone methyltransferase families Polycomb group (PcG), which represses the expression of developmentally regulated genes, and Trithorax group, which antagonizes the repressive activity of the PcG, regulate myogenesis by restricting gene expression in a time-dependent manner during each step of the process. We discuss how histone acetylation and deacetylation occurs in specific loci throughout SC differentiation to enable the time-dependent transcription of specific genes. Moreover, we describe the multiple roles of microRNA, an additional epigenetic mechanism, in regulating gene expression in SCs, by repressing or enhancing gene transcription or translation during each step of myogenesis. The importance of these epigenetic pathways in modulating SC activation and differentiation renders them as promising targets for disease interventions. Understanding the most recent findings regarding the epigenetic mechanisms that regulate SC behavior is useful from the perspective of pharmacological manipulation for improving muscle regeneration and for promoting muscle homeostasis under pathological conditions.
Collapse
|