Elsevier

Experimental Cell Research

Volume 314, Issue 7, 15 April 2008, Pages 1553-1565
Experimental Cell Research

Research Article
Neural cell adhesion molecule (NCAM) marks adult myogenic cells committed to differentiation

https://doi.org/10.1016/j.yexcr.2008.01.021Get rights and content

Abstract

Although recent advances in broad-scale gene expression analysis have dramatically increased our knowledge of the repertoire of mRNAs present in multiple cell types, it has become increasingly clear that examination of the expression, localization, and associations of the encoded proteins will be critical for determining their functional significance. In particular, many signaling receptors, transducers, and effectors have been proposed to act in higher-order complexes associated with physically distinct areas of the plasma membrane. Adult muscle stem cells (satellite cells) must, upon injury, respond appropriately to a wide range of extracellular stimuli: the role of such signaling scaffolds is therefore a potentially important area of inquiry. To address this question, we first isolated detergent-resistant membrane fractions from primary satellite cells, then analyzed their component proteins using liquid chromatography–tandem mass spectrometry. Transmembrane and juxtamembrane components of adhesion-mediated signaling pathways made up the largest group of identified proteins; in particular, neural cell adhesion molecule (NCAM), a multifunctional cell-surface protein that has previously been associated with muscle regeneration, was significant. Immunohistochemical analysis revealed that not only is NCAM localized to discrete areas of the plasma membrane, it is also a very early marker of commitment to terminal differentiation. Using flow cytometry, we have sorted physically homogeneous myogenic cultures into proliferating and differentiating fractions based solely upon NCAM expression.

Introduction

Skeletal muscle is a terminally differentiated tissue consisting of ordered arrays of multinucleated, contractile myofibers. In vertebrates, skeletal muscle is formed during fetal and postnatal development by differentiation of previously specified myoblasts, which irreversibly exit the cell cycle and become committed myocytes. This transition is accompanied by changes in gene expression, growth factor responsiveness and structural protein production (reviewed in [1]). Upon differentiation myocytes subsequently fuse with each other or with existing myotubes to produce contractile syncytial myofibers [2].

In adult vertebrates myogenesis is believed to be primarily carried out by satellite cells, the somatic stem cells responsible for in vivo maintenance and regeneration of skeletal muscle tissue [3], [4]. These cells, which comprise a very small (1–6%) fraction of total muscle-associated nuclei, are defined anatomically by their position between the basement membrane and the sarcolemma of differentiated muscle fibers [3], [5], [6]. In response to injury, otherwise mitotically quiescent satellite cells become activated and proliferate extensively. The resulting population of adult myoblasts will then transit to the site of injury and differentiate into myocytes to replace the damaged myofibers, either by fusion with each other to form new muscle fibers or by fusing into existing post-mitotic muscle fibers [7], [8]. While the satellite cell compartment is repopulated following completion of a cycle of acute regeneration, it remains unclear what the exact cellular source(s) of these new quiescent cells may be: evidence exists for satellite cell self-renewal, either by asymmetric division [9] or stochastic events [10], as well as possible contributions from muscle-associated mesenchymal stem cell populations [9], [11].

The extracellular milieu encountered by newly-activated satellite cells requires that they detect and respond appropriately to a diverse array of rapidly changing stimuli. In addition to the damaged host muscle, local signaling sources would include coincidently damaged connective tissue, vasculature and nervous tissue, as well as infiltrating cells of the immune system [3]. Local extracellular signals would also be expected to vary with time after the initial injury. Thus, critical roles have been demonstrated for many soluble factors and matrix/adhesion molecules in the muscle tissue during satellite cell-mediated muscle repair [12], [13], [14], and there is a significant amount of ongoing investigation into the signaling pathways that function in satellite cells during regeneration.

An area that has not yet been addressed with respect to satellite cell signaling is the possible involvement of higher-order signaling complexes, such as those that have been proposed to assemble in membrane ‘rafts’. Membrane rafts, also known as lipid rafts, are small (10–200 nm), cholesterol and sphingolipid enriched membranes that function to compartmentalize cellular processes [15], [16]. These regions of the plasma membrane play important roles in intracellular protein transport, membrane fusion and transcytosis; they have also been proposed to act as platforms for assembly of signaling complexes, cell-surface antigens and adhesion molecules. Cellular events commonly associated with membrane raft complexes include cell activation, polarization and signaling [17], [18]. In other adult stem cells (i.e., hematopoietic stem cells) membrane rafts are critical for cell cycle regulation and survival [19], [20], however very little is known about signaling pathways mediated by membrane rafts in satellite cells.

In this study, we attempted to isolate and characterize plasma membrane proteins expressed by primary mouse satellite cells, with the goal of prospectively identifying additional signaling pathways that may impinge upon satellite cell activity. Using liquid chromatograpy–tandem mass spectrometry, we identified classes of transmembrane and membrane-associated proteins present in freshly isolated murine satellite cells and enriched in detergent-resistant membrane domains. While surprisingly few of the expected transmembrane receptors were detected above the reliability threshold, multiple proteins associated with adhesion-mediated signaling were identified. Several have not previously been connected with myogenesis, although many have; a significant subset has also been reported to act via membrane raft complexes. One such protein, neural cell adhesion molecule (NCAM), was found to be present and enriched in the detergent-resistant membrane fraction, and was selected for further study.

When examined in heterogeneous populations of adult myoblasts and myocytes, we found NCAM expression to be coincident with the earliest detectable markers of commitment to differentiation. In order to unequivocally differentiate between proliferation-competent myoblasts and committed myocytes, it is common to assay for expression of differentiation-associated proteins such as myogenin, p21, or muscle structural proteins. However, all of these proteins are cytoplasmic or nuclear, and it would be desirable to determine cell status by assaying for a cell-surface epitope, allowing analysis of living unfixed cells. To date, no such marker has been reported. Here we show that by indirect immunohistochemistry, NCAM labels only nonproliferating cells that, based on their expression of differentiation markers including myogenin and muscle creatine kinase have committed to differentiation. We also use NCAM expression to separate a heterogeneous population of adult myoblasts into proliferating and differentiated fractions, as confirmed by expression of either proliferation or pro-myogenesis proteins. This molecular tool therefore represents a novel, non-terminal assay for the early identification and sorting of committed myocytes from heterogeneous populations derived from primary mouse satellite cells.

Section snippets

Primary satellite cell isolation and culture

Mouse satellite cells were isolated and cultured as described previously [21]. Briefly, muscle was dissected from the hind limbs of adult female mice (B6D2F1; Jackson labs) between 80 and 130 days of age. Muscles from both legs were minced and digested in 400 U/ml collagenase type I (Worthington) diluted in Ham's F-12 medium (Invitrogen). The resulting cells were collected and pre-plated on gelatin-coated (0.66%) petri dishes in growth medium [Ham's F-12 (Gibco), 15% horse serum (Equitech) and

Detergent-resistant membrane domain proteins of primary satellite cells

The light buoyant density and other physical properties of membrane raft domains facilitate their isolation based on insolubility in cold nonionic detergents [25]. To determine what proteins are partitioned into the detergent-resistant membrane domains (DRMs), we used cold Triton X-100 insolubility and flotation over a discontinuous sucrose gradient to isolate them from cultures amplified from primary murine satellite cells. We then used liquid chromatography–tandem mass spectrometry to

Conclusions and future directions

Since the initial observation of NCAM expression on rat primary satellite cells, the role of NCAM during myogenesis has been elusive [12], [55]. In muscle, NCAM exists in multiple isoforms, which arise from alternative splicing of a single gene [61], [67], [68]. NCAM is also subject to several forms of post-translational modification: polysialylation of residues in the immunoglobulin domains is commonly found during development and has recently been shown to increase membrane repulsion and to

Acknowledgments

This work was supported by grants to DDWC from the University of Missouri Research Board and the Muscular Dystrophy Association.

References (75)

  • N.J. Lennon et al.

    Dysferlin interacts with Annexins A1 and A2 and mediates sarcolemmal wound-healing

    J. Biol. Chem.

    (2003)
  • F. Galbiati et al.

    Caveolin-3 null mice show a loss of caveolae, changes in the microdomain distribution of the dystrophin-glycoprotein complex, and t-tubule abnormalities

    J. Biol. Chem.

    (2001)
  • Q. Eastman et al.

    Regulation of LEF-1/TCF transcription factors by Wnt and other signals

    Curr. Opin. Cell Biol.

    (1999)
  • F. Galbiati et al.

    Caveolin-1 expression inhibits Wnt/beta-catenin/Lef-1 signaling by recruiting beta-catenin to caveolae membrane domains

    J. Biol. Chem.

    (2000)
  • C.S. Mermelstein et al.

    Wnt/beta-catenin pathway activation and myogenic differentiation are induced by cholesterol depletion

    Differentiation

    (2007)
  • R. Simson et al.

    Structural mosaicism on the submicron scale in the plasma membrane

    Biophys. J.

    (1998)
  • M.D. Grounds

    Towards understanding skeletal muscle regeneration

    Pathol. Res. Pract.

    (1991)
  • D.D. Cornelison et al.

    Syndecan-3 and syndecan-4 specifically mark skeletal muscle satellite cells and are implicated in satellite cell maintenance and muscle regeneration

    Dev. Biol.

    (2001)
  • C. Dubois et al.

    Expression of Ncam and its polysialylated isoforms during Mdx mouse muscle regeneration and in-vitro myogenesis

    Neuromuscul. Disord.

    (1994)
  • C.P. Johnson et al.

    Direct evidence that neural cell adhesion molecule (NCAM) polysialylation increases intermembrane repulsion and abrogates adhesion

    J. Biol. Chem.

    (2005)
  • M. Suzuki et al.

    Polysialic acid and mucin type o-glycans on the neural cell adhesion molecule differentially regulate myoblast fusion

    J. Biol. Chem.

    (2003)
  • C.A. Charlton et al.

    Neural cell adhesion molecule (NCAM) and myoblast fusion

    Dev. Biol.

    (2000)
  • B. Christ et al.

    Limb muscle development

    Int. J. Dev. Biol.

    (2002)
  • T.J. Hawke et al.

    Myogenic satellite cells: physiology to molecular biology

    J. Appl. Physiol.

    (2001)
  • X. Shi et al.

    Muscle stem cells in development, regeneration, and disease

    Genes Dev.

    (2006)
  • A. Mauro

    Satellite cell of skeletal muscle fibers

    J. Biophys. Biochem. Cytol.

    (1961)
  • R. Bischoff

    Analysis of muscle regeneration using single myofibers in culture

    Med. Sci. Sports Exerc.

    (1989)
  • M.D. Grounds

    Age-associated changes in the response of skeletal muscle cells to exercise and regeneration

  • E. Schultz et al.

    Skeletal muscle satellite cells

    Rev. Physiol., Biochem. Pharmacol.

    (1994)
  • J.C. Angello et al.

    Skeletal muscle satellite cells: timelapse videomicroscopic evidence that renewal is stochastic

    BAM

    (1996)
  • G. Ferrari et al.

    Muscle regeneration by bone marrow-derived myogenic progenitors

    Science

    (1988)
  • R.S. Krauss et al.

    Close encounters: regulation of vertebrate skeletal myogenesis by cell–cell contact

    J. Cell Sci.

    (2005)
  • S.B. Charge et al.

    Cellular and molecular regulation of muscle regeneration

    Physiol. Rev.

    (2004)
  • D.A. Brown et al.

    Functions of lipid rafts in biological membranes

    Annu. Rev. Cell Dev. Biol.

    (1998)
  • K. Simons et al.

    Lipid rafts and signal transduction

    Nat. Rev., Mol. Cell Biol.

    (2000)
  • R.G. Parton et al.

    Lipid rafts and caveolae as portals for endocytosis: new insights and common mechanisms

    Traffic

    (2003)
  • S. Yamazaki et al.

    Cytokine signals modulated via lipid rafts mimic niche signals and induce hibernation in hematopoietic stem cells

    EMBO J.

    (2006)
  • Cited by (70)

    • Muscle stem cells and rotator cuff injury

      2021, JSES Reviews, Reports, and Techniques
    • Heterocellular molecular contacts in the mammalian stem cell niche

      2018, European Journal of Cell Biology
      Citation Excerpt :

      NCAM is expressed mainly on M-Cad positive SatCs, in microdomains of the plasma membrane on the same side of the cell, facing the adult myofiber (Snijders et al., 2015). NCAM has been detected in non-proliferating SatCs committed to differentiation (Capkovic et al., 2008), as well as by myoblasts, myotubes, and muscle fibers during development and regeneration, where most probably favor cell-cell fusion by heterophilic interactions (Suzuki et al., 2003). However, NCAM-null mice exhibit no defects during muscle development and primary myoblast fusion, probably due to other compensating adhesion molecules (Charlton et al., 2000).

    View all citing articles on Scopus
    View full text