Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 19(7): 1024-1040, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38876109

ABSTRACT

Increasing evidence suggests that the muscle stem cell (MuSC) pool is heterogeneous. In particular, a rare subset of PAX7-positive MuSCs that has never expressed the myogenic regulatory factor MYF5 displays unique self-renewal and engraftment characteristics. However, the scarcity and limited availability of protein markers make the characterization of these cells challenging. Here, we describe the generation of StemRep reporter mice enabling the monitoring of PAX7 and MYF5 proteins based on equimolar levels of dual nuclear fluorescence. High levels of PAX7 protein and low levels of MYF5 delineate a deeply quiescent MuSC subpopulation with an increased capacity for asymmetric division and distinct dynamics of activation, proliferation, and commitment. Aging primarily reduces the MYF5Low MuSCs and skews the stem cell pool toward MYF5High cells with lower quiescence and self-renewal potential. Altogether, we establish the StemRep model as a versatile tool to study MuSC heterogeneity and broaden our understanding of mechanisms regulating MuSC quiescence and self-renewal in homeostatic, regenerating, and aged muscles.


Subject(s)
Aging , Genes, Reporter , Myogenic Regulatory Factor 5 , PAX7 Transcription Factor , Regeneration , Animals , PAX7 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factor 5/genetics , Mice , Aging/metabolism , Stem Cells/metabolism , Stem Cells/cytology , Cell Proliferation , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Cell Differentiation , Mice, Transgenic , Cell Self Renewal
2.
iScience ; 25(7): 104589, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35789856

ABSTRACT

Apelin (Apln) is a myokine that regulates skeletal muscle plasticity and metabolism and declines during aging. Through a yeast one-hybrid transcription factor binding screen, we identified the TEA domain transcription factor 1 (Tead1) as a novel regulator of the Apln promoter. Single-cell analysis of regenerating muscle revealed that the apelin receptor (Aplnr) is enriched in endothelial cells, whereas Tead1 is enriched in myogenic cells. Knock-down of Tead1 stimulates Apln secretion from muscle cells in vitro and myofiber-specific overexpression of Tead1 suppresses Apln secretion in vivo. Apln secretion via Tead1 knock-down in muscle cells stimulates endothelial cell expansion via endothelial Aplnr. In vivo, Apln peptide supplementation enhances endothelial cell expansion while Tead1 muscle overexpression delays endothelial remodeling following muscle injury. Our work describes a novel paracrine crosstalk in which Apln secretion is controlled by Tead1 in myogenic cells and influences endothelial remodeling during muscle repair.

3.
EMBO J ; 41(14): e110655, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35703167

ABSTRACT

Fate decisions in the embryo are controlled by a plethora of microenvironmental interactions in a three-dimensional niche. To investigate whether aspects of this microenvironmental complexity can be engineered to direct myogenic human-induced pluripotent stem cell (hiPSC) differentiation, we here screened murine cell types present in the developmental or adult stem cell niche in heterotypic suspension embryoids. We identified embryonic endothelial cells and fibroblasts as highly permissive for myogenic specification of hiPSCs. After two weeks of sequential Wnt and FGF pathway induction, these three-component embryoids are enriched in Pax7-positive embryonic-like myogenic progenitors that can be isolated by flow cytometry. Myogenic differentiation of hiPSCs in heterotypic embryoids relies on a specialized structural microenvironment and depends on MAPK, PI3K/AKT, and Notch signaling. After transplantation in a mouse model of Duchenne muscular dystrophy, embryonic-like myogenic progenitors repopulate the stem cell niche, reactivate after repeated injury, and, compared to adult human myoblasts, display enhanced fusion and lead to increased muscle function. Altogether, we provide a two-week protocol for efficient and scalable suspension-based 3D derivation of Pax7-positive myogenic progenitors from hiPSCs.


Subject(s)
Induced Pluripotent Stem Cells , Animals , Cell Differentiation , Endothelial Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Muscle Development , Phosphatidylinositol 3-Kinases/metabolism , Stem Cell Niche
4.
Trends Cell Biol ; 31(7): 556-568, 2021 07.
Article in English | MEDLINE | ID: mdl-33674167

ABSTRACT

Muscle stem cells (MuSCs) are tissue-resident stem cells required for growth and repair of skeletal muscle, that are otherwise maintained in a cell-cycle-arrested state called quiescence. While quiescence was originally believed to be a state of cellular inactivity, increasing evidence suggests that quiescence is dynamically regulated and contributes to stemness, the long-term capacity to maintain regenerative functions. Here, we review the current understanding of MuSC quiescence and highlight recently discovered molecular markers, which differentiate depth of quiescence and influence self-renewal capacity. We also discuss how quiescent MuSCs integrate paracrine factors from their niche and dynamically regulate cell signaling, metabolism and proteostasis as they anticipate physiological needs, and how perturbing these cues during aging impairs muscle regeneration.


Subject(s)
Myoblasts , Stem Cells , Cell Division , Muscle, Skeletal
5.
Cell Stem Cell ; 24(3): 433-446.e7, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30686765

ABSTRACT

Research on age-related regenerative failure of skeletal muscle has extensively focused on the phenotypes of muscle stem cells (MuSCs). In contrast, the impact of aging on regulatory cells in the MuSC niche remains largely unexplored. Here, we demonstrate that aging impairs the function of mouse fibro-adipogenic progenitors (FAPs) and thereby indirectly affects the myogenic potential of MuSCs. Using transcriptomic profiling, we identify WNT1 Inducible Signaling Pathway Protein 1 (WISP1) as a FAP-derived matricellular signal that is lost during aging. WISP1 is required for efficient muscle regeneration and controls the expansion and asymmetric commitment of MuSCs through Akt signaling. Transplantation of young FAPs or systemic treatment with WISP1 restores the myogenic capacity of MuSCs in aged mice and rescues skeletal muscle regeneration. Our work establishes that loss of WISP1 from FAPs contributes to MuSC dysfunction in aged skeletal muscles and demonstrates that this mechanism can be targeted to rejuvenate myogenesis.


Subject(s)
Adipocytes/metabolism , Aging/metabolism , CCN Intercellular Signaling Proteins/metabolism , Muscle, Skeletal/metabolism , Proto-Oncogene Proteins/metabolism , Stem Cells/metabolism , Adipocytes/cytology , Adipogenesis , Animals , CCN Intercellular Signaling Proteins/deficiency , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/cytology , Proto-Oncogene Proteins/deficiency , Stem Cells/cytology
6.
Methods Mol Biol ; 1556: 51-102, 2017.
Article in English | MEDLINE | ID: mdl-28247345

ABSTRACT

Multinucleated myofibers, the functional contractile units of adult skeletal muscle, harbor mononuclear Pax7+ myogenic progenitors on their surface between the myofiber basal lamina and plasmalemma. These progenitors, known as satellite cells, are the primary myogenic stem cells in adult muscle. This chapter describes our laboratory protocols for isolating, culturing, and immunostaining intact myofibers from mouse skeletal muscle as a means for studying satellite cell dynamics. The first protocol discusses myofiber isolation from the flexor digitorum brevis (FDB) muscle. These short myofibers are plated in dishes coated with PureCol collagen (formerly known as Vitrogen) and maintained in a mitogen-poor medium (± supplemental growth factors). Employing such conditions, satellite cells remain at the surface of the parent myofiber while synchronously undergoing a limited number of proliferative cycles and rapidly differentiate. The second protocol discusses the isolation of longer myofibers from the extensor digitorum longus (EDL) muscle. These EDL myofibers are routinely plated individually as adherent myofibers in wells coated with Matrigel and maintained in a mitogen-rich medium, conditions in which satellite cells migrate away from the parent myofiber, proliferate extensively, and generate numerous differentiating progeny. Alternatively, these EDL myofibers can be plated as non-adherent myofibers in uncoated wells and maintained in a mitogen-poor medium (± supplemental growth factors), conditions that retain satellite cell progeny at the myofiber niche similar to the FDB myofiber cultures. However, the adherent myofiber format is our preferred choice for monitoring satellite cells in freshly isolated (Time 0) myofibers. We conclude this chapter by promoting the Nestin-GFP transgenic mouse as an efficient tool for direct analysis of satellite cells in isolated myofibers. While satellite cells have been often detected by their expression of the Pax7 protein or the Myf5nLacZ knockin reporter (approaches that are also detailed herein), the Nestin-GFP reporter distinctively permits quantification of satellite cells in live myofibers, which enables linking initial Time 0 numbers and subsequent performance upon culturing. We additionally point out to the implementation of the Nestin-GFP transgene for monitoring other selective cell lineages as illustrated by GFP expression in capillaries, endothelial tubes and neuronal cells. Myofibers from other types of muscles, such as diaphragm, masseter, and extraocular, can also be isolated and analyzed using protocols described herein. Collectively, this chapter provides essential tools for studying satellite cells in their native position and their interplay with the parent myofiber.


Subject(s)
Cell Separation/methods , Immunophenotyping/methods , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Animals , Biomarkers , Cell Culture Techniques , Cell Differentiation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence , Muscle Fibers, Skeletal/ultrastructure , Nestin/genetics , Nestin/metabolism , Phenotype , Primary Cell Culture , Satellite Cells, Skeletal Muscle/ultrastructure
7.
Biol Open ; 5(10): 1351-1361, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27744291

ABSTRACT

Refilins (RefilinA and RefilinB) are members of a novel family of Filamin binding proteins that function as molecular switches to conformationally alter the Actin filament network into bundles. We show here that Refilins are extremely labile proteins. An N-terminal PEST/DSG(X)2-4S motif mediates ubiquitin-independent rapid degradation. A second degradation signal is localized within the C-terminus. Only RefilinB is protected from rapid degradation by an auto-inhibitory domain that masks the PEST/DSG(X)2-4S motif. Dual regulation of RefilinA and RefilinB stability was confirmed in rat brain NG2 precursor cells (polydendrocyte). Using loss- and gain-of-function approaches we show that in these cells, and in U373MG cells, Refilins contribute to the dynamics of lamellipodium protrusion by catalysing Actin bundle formation within the lamella Actin network. These studies extend the Actin bundling function of the Refilin-Filamin complex to dynamic regulation of cell membrane remodelling.

8.
Methods Mol Biol ; 1460: 101-27, 2016.
Article in English | MEDLINE | ID: mdl-27492169

ABSTRACT

The extraocular muscles (EOMs) comprise a group of highly specialized skeletal muscles controlling eye movements. Although a number of unique features of EOMs including their sparing in Duchenne muscular dystrophy have drawn a continuous interest, knowledge about these hard to reach muscles is still limited. The goal of this chapter is to provide detailed methods for the isolation and histological analysis of mouse EOMs. We first introduce in brief the basic anatomy and established nomenclature of the extraocular primary and accessory muscles. We then provide a detailed description with step-by-step images of our procedure for isolating (and subsequently cryosectioning) EOMs while preserving the integrity of their original structural organization. Next, we present several useful histological protocols frequently used by us, including: (1) a method for highlighting the general organization of periocular tissue, using the MyoD(Cre) × R26(mTmG) reporter mouse that elegantly distinguishes muscle (MyoD(Cre)-driven GFP(+)) from the non-myogenic constituents (Tomato(+)); (2) analysis by H&E staining, allowing for example, detection of the pathological features of the dystrophin-null phenotype in affected limb and diaphragm muscles that are absent in EOMs; (3) detection of the myogenic progenitors (i.e., satellite cells) in their native position underneath the myofiber basal lamina using Pax7/laminin double immunostaining. The EOM tissue harvesting procedure described here can also be adapted for isolating and studying satellite cells and other cell types. Overall, the methods described in this chapter should provide investigators the necessary tools for entering the EOM research field and contribute to a better understanding of this highly specialized muscle group and its complex micro-anatomy.


Subject(s)
Immunohistochemistry , Oculomotor Muscles/cytology , Satellite Cells, Skeletal Muscle/cytology , Animals , Disease Models, Animal , Dystrophin/metabolism , Immunohistochemistry/methods , Laminin/metabolism , Mice , Mice, Transgenic , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Oculomotor Muscles/metabolism , Satellite Cells, Skeletal Muscle/metabolism
9.
FEBS J ; 283(9): 1653-68, 2016 05.
Article in English | MEDLINE | ID: mdl-26881702

ABSTRACT

Regeneration of skeletal muscles is required throughout life to ensure optimal performance. Therefore, a better understanding of the resident cells involved in muscle repair is essential. Muscle repair relies on satellite cells (SCs), the resident myogenic progenitors, but also involves the contribution of interstitial cells including fibro/adipocyte progenitors (FAPs). To elucidate the role of the fibroblast growth factor (FGF) signaling in these two cell populations, we previously analyzed freshly isolated cells for their FGF receptor (FGFR) signature. Transcript analysis of the four Fgfr genes revealed distinct expression profiles for SCs and FAPs, raising the possibility that these two cell types have different FGF-mediated processes. Here, we pursued this hypothesis exploring the role of the Klotho genes, whose products are known to function as FGFR co-receptors for the endocrine FGF subfamily. Isolated SC and FAP populations were analyzed in culture, exhibiting spontaneous myogenic or adipogenic differentiation, respectively. αKlotho expression was not detected in either population. ßKlotho expression, while not detected in SCs, was strongly upregulated in FAPs entering adipogenic differentiation, coinciding with expression of a panel of adipogenic genes and preceding the appearance of intracellular lipid droplets. Overexpression of ßKlotho in mouse cell line models enhanced adipogenesis in NIH3T3 fibroblasts but had no effect on C2C12 myogenic cells. Our study supports a pro-adipogenic role for ßKlotho in skeletal muscle fibro/adipogenesis and calls for further research on involvement of the FGF-FGFR-ßKlotho axis in the fibro/adipogenic infiltration associated with functional deterioration of skeletal muscle in aging and muscular dystrophy.


Subject(s)
Adipocytes/metabolism , Fibroblast Growth Factors/genetics , Fibroblasts/metabolism , Membrane Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Satellite Cells, Skeletal Muscle/metabolism , Adipocytes/cytology , Adipogenesis/genetics , Animals , Cell Differentiation , Cell Line , Fibroblast Growth Factors/metabolism , Fibroblasts/cytology , Gene Expression Regulation , Klotho Proteins , Lipid Droplets , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Satellite Cells, Skeletal Muscle/cytology , Signal Transduction
10.
Front Aging Neurosci ; 7: 85, 2015.
Article in English | MEDLINE | ID: mdl-26074812

ABSTRACT

Skeletal muscle satellite cells (SCs) are Pax7(+) myogenic stem cells that reside between the basal lamina and the plasmalemma of the myofiber. In mature muscles, SCs are typically quiescent, but can be activated in response to muscle injury. Depending on the magnitude of tissue trauma, SCs may divide minimally to repair subtle damage within individual myofibers or produce a larger progeny pool that forms new myofibers in cases of overt muscle injury. SC transition through proliferation, differentiation and renewal is governed by the molecular blueprint of the cells as well as by the extracellular milieu at the SC niche. In particular, the role of the fibroblast growth factor (FGF) family in regulating SCs during growth and aging is well recognized. Of the several FGFs shown to affect SCs, FGF1, FGF2, and FGF6 proteins have been documented in adult skeletal muscle. These prototypic paracrine FGFs transmit their mitogenic effect through the FGFRs, which are transmembrane tyrosine kinase receptors. Using the mouse model, we show here that of the four Fgfr genes, only Fgfr1 and Fgfr4 are expressed at relatively high levels in quiescent SCs and their proliferating progeny. To further investigate the role of FGFR1 in adult myogenesis, we have employed a genetic (Cre/loxP) approach for myogenic-specific (MyoD(Cre)-driven) ablation of Fgfr1. Neither muscle histology nor muscle regeneration following cardiotoxin-induced injury were overtly affected in Fgfr1-ablated mice. This suggests that FGFR1 is not obligatory for SC performance in this acute muscle trauma model, where compensatory growth factor/cytokine regulatory cascades may exist. However, the SC mitogenic response to FGF2 is drastically repressed in isolated myofibers prepared from Fgfr1-ablated mice. Collectively, our study indicates that FGFR1 is important for FGF-mediated proliferation of SCs and its mitogenic role is not compensated by FGFR4 that is also highly expressed in SCs.

11.
Dev Biol ; 397(1): 31-44, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25236433

ABSTRACT

Extraocular muscles (EOMs) are highly specialized skeletal muscles that originate from the head mesoderm and control eye movements. EOMs are uniquely spared in Duchenne muscular dystrophy and animal models of dystrophin deficiency. Specific traits of myogenic progenitors may be determinants of this preferential sparing, but very little is known about the myogenic cells in this muscle group. While satellite cells (SCs) have long been recognized as the main source of myogenic cells in adult muscle, most of the knowledge about these cells comes from the prototypic limb muscles. In this study, we show that EOMs, regardless of their distinctive Pax3-negative lineage origin, harbor SCs that share a common signature (Pax7(+), Ki67(-), Nestin-GFP(+), Myf5(nLacZ+), MyoD-positive lineage origin) with their limb and diaphragm somite-derived counterparts, but are remarkably endowed with a high proliferative potential as revealed in cell culture assays. Specifically, we demonstrate that in adult as well as in aging mice, EOM SCs possess a superior expansion capacity, contributing significantly more proliferating, differentiating and renewal progeny than their limb and diaphragm counterparts. These robust growth and renewal properties are maintained by EOM SCs isolated from dystrophin-null (mdx) mice, while SCs from muscles affected by dystrophin deficiency (i.e., limb and diaphragm) expand poorly in vitro. EOM SCs also retain higher performance in cell transplantation assays in which donor cells were engrafted into host mdx limb muscle. Collectively, our study provides a comprehensive picture of EOM myogenic progenitors, showing that while these cells share common hallmarks with the prototypic SCs in somite-derived muscles, they distinctively feature robust growth and renewal capacities that warrant the title of high performance myo-engines and promote consideration of their properties for developing new approaches in cell-based therapy to combat skeletal muscle wasting.


Subject(s)
Dystrophin/physiology , Gene Expression Regulation, Developmental , Muscle, Skeletal/embryology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/cytology , Stem Cells/cytology , Animals , Cell Lineage , Cell Proliferation , Cell Separation , Cell Transplantation , Disease Models, Animal , Dystrophin/deficiency , Extremities/embryology , Female , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Transgenic , Muscular Dystrophy, Duchenne/genetics
12.
Dev Biol ; 385(2): 366-79, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-23969310

ABSTRACT

Extraocular muscles (EOM) represent a unique muscle group that controls eye movements and originates from head mesoderm, while the more typically studied body and limb muscles are somite-derived. Aiming to investigate myogenic progenitors (satellite cells) in EOM versus limb and diaphragm of adult mice, we have been using flow cytometry in combination with myogenic-specific Cre-loxP lineage marking for cell isolation. While analyzing cells from the EOM of mice that harbor Myf5(Cre)-driven GFP expression, we identified in addition to the expected GFP(+) myogenic cells (presumably satellite cells), a second dominant GFP(+) population distinguished as being Sca1(+), non-myogenic, and exhibiting a fibro/adipogenic potential. This unexpected population was not only unique to EOM compared to the other muscles but also specific to the Myf5(Cre)-driven reporter when compared to the MyoD(Cre) driver. Histological studies of periocular tissue preparations demonstrated the presence of Myf5(Cre)-driven GFP(+) cells in connective tissue locations adjacent to the muscle masses, including cells in the vasculature wall. These vasculature-associated GFP(+) cells were further identified as mural cells based on the presence of the specific XLacZ4 transgene. Unlike the EOM satellite cells that originate from a Pax3-negative lineage, these non-myogenic Myf5(Cre)-driven GFP(+) cells appear to be related to cells of a Pax3-expressing origin, presumably derived from the neural crest. In all, our lineage tracing based on multiple reporter lines has demonstrated that regardless of common ancestral expression of Myf5, there is a clear distinction between periocular myogenic and non-myogenic cell lineages according to their mutually exclusive antecedence of MyoD and Pax3 gene activity.


Subject(s)
Eye , Myogenic Regulatory Factor 5/genetics , Satellite Cells, Skeletal Muscle/metabolism , Stem Cells/cytology , Animals , Cell Lineage , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , Immunohistochemistry , Male , Mice , Satellite Cells, Skeletal Muscle/cytology
13.
FEBS J ; 280(17): 4063-73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23464362

ABSTRACT

Satellite cells, the myogenic progenitors located at the myofibre surface, are essential for the repair of adult skeletal muscle. There is ample evidence for an age-linked decline in the number of satellite cells and performance in limb muscles. Hence, an effective means of activating and expanding the satellite cell pool may enhance muscle maintenance and reduce the impact of age-associated muscle deterioration (sarcopaenia). Accordingly, in the present study, we explored the beneficial effects of endurance exercise on satellite cells in young and old mice. Animals were subjected to an 8-week moderate-intensity treadmill-running approach that does not inflict apparent muscle damage (0° inclination, 11.5 m·min(-1) for 30 min·day(-1) , 6 days·week(-1) ). Myofibres of extensor digitorum longus muscles were then isolated from exercised and sedentary mice and used for monitoring the number of satellite cells, as well as for harvesting individual satellite cells for clonal growth assays. We specifically focused on satellite cell pools of single myofibres, with the view that daily wear of muscles probably affects individual myofibres rather than causing overall muscle damage. We found an expansion of the satellite cell pool in the exercised groups compared to the sedentary groups, with the same increase (~ 1.6-fold) in both ages. The results of the present study are in agreement with our findings obtained using rat gastrocnemius, indicating the consistent effect of exercise on satellite cell expansion in limb muscles. The experimental paradigm established in the present study is useful for investigating satellite cell dynamics at the myofibre niche, as well as for broader investigations of the impact of physiologically and pathologically relevant factors on adult myogenesis.


Subject(s)
Exercise Test , Muscle Development/physiology , Muscle Fibers, Skeletal/cytology , Running/physiology , Satellite Cells, Skeletal Muscle/cytology , Stem Cells/cytology , Animals , Cell Proliferation , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intermediate Filament Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Fibers, Skeletal/physiology , Nerve Tissue Proteins/genetics , Nestin , Promoter Regions, Genetic/genetics , Satellite Cells, Skeletal Muscle/physiology , Stem Cells/physiology
14.
J Histochem Cytochem ; 60(9): 638-57, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22723526

ABSTRACT

The thoracic diaphragm is a unique skeletal muscle composed of costal, crural, and central tendon domains. Although commonly described in medical textbooks, newer insights into the diaphragm cell composition are scarce. Here, using reporter mice, combined with gene expression analyses of whole tissues and primary cultures, we compared the diaphragm domains and their myogenic progenitors (i.e., Pax3/7 satellite cells). The outcomes of these analyses underscore the similarities between the myogenic aspects of the costal and crural domains. Expression levels of all myogenic genes examined (except Pax3) were strongly affected in mdx (dystrophin-null) mice and accompanied by an increase in fibrosis- and adiposity-related gene expression. Cell culture studies further indicated the presence of a non-myogenic Pax3-expressing population, potentially related to vascular mural cells. We additionally investigated the diaphragm vasculature. XLacZ4 and Sca1-GFP transgenes allowed a fine definition of the arterial and microvasculature network based on reporter expression in mural cells and capillary endothelium, respectively. We also provide insights into the organization of the diaphragm venous system, especially apparent in the central tendon and exhibiting arcades lined with fat-containing cells. The novel information in this "contemporary atlas" can be further explored in the context of diaphragm pathology and genetic disorders.


Subject(s)
Diaphragm , Paired Box Transcription Factors/metabolism , Adiposity/genetics , Animals , Arteries/metabolism , Atlases as Topic , Diaphragm/anatomy & histology , Diaphragm/blood supply , Diaphragm/metabolism , Dystrophin/genetics , Endothelium, Vascular/metabolism , Female , Fibrosis/genetics , Gene Expression Profiling , Genes, Reporter , Mice , Mice, Inbred mdx , Mice, Transgenic , Microvessels/metabolism , PAX3 Transcription Factor , Primary Cell Culture , Tendons/blood supply , Tendons/metabolism , Veins/metabolism
15.
J Biol Chem ; 285(17): 12670-83, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20139084

ABSTRACT

Calpain 3 is a calcium-dependent cysteine protease that is primarily expressed in skeletal muscle and is implicated in limb girdle muscular dystrophy type 2A. To date, its best characterized function is located within the sarcomere, but this protease is found in other cellular compartments, which suggests that it exerts multiple roles. Here, we present evidence that calpain 3 is involved in the myogenic differentiation process. In the course of in vitro culture of myoblasts to fully differentiated myotubes, a population of quiescent undifferentiated "reserve cells" are maintained. These reserve cells are closely related to satellite cells responsible for adult muscle regeneration. In the present work, we observe that reserve cells express higher levels of endogenous Capn3 mRNA than proliferating myoblasts. We show that calpain 3 participates in the establishment of the pool of reserve cells by decreasing the transcriptional activity of the key myogenic regulator MyoD via proteolysis independently of the ubiquitin-proteasome degradation pathway. Our results identify calpain 3 as a potential new player in the muscular regeneration process by promoting renewal of the satellite cell compartment.


Subject(s)
Calpain/metabolism , Cell Differentiation , Down-Regulation , Muscle Development , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , MyoD Protein/metabolism , Myoblasts/metabolism , Calpain/genetics , Cell Line , Humans , Muscle Proteins/genetics , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , MyoD Protein/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Regeneration/genetics , Satellite Cells, Skeletal Muscle/metabolism , Transcription, Genetic/genetics , Ubiquitin/metabolism
16.
Exp Cell Res ; 316(1): 115-25, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19651121

ABSTRACT

The reduced regenerative potential of muscle fibres, most likely due to a decreased number and/or function of satellite cells, could play a significant role in the progression of muscle ageing. Accumulation of reactive oxygen species has been clearly correlated to sarcopenia and could contribute to the impairment of satellite cell function. In this work we have investigated the effect of oxidative stress generated by hydrogen peroxide in cultured human skeletal muscle satellite cells. We specifically focused on the activity and regulation of calpains. These calcium-dependent proteases are known to regulate many transduction pathways including apoptosis and play a critical role in satellite cell function. In our experimental conditions, which induce an increase in calcium concentration, protein oxidation and apoptotic cell death, a significant up-regulation of calpain expression and activity were observed and ATP synthase, a major component of the respiratory chain, was identified as a calpain target. Interestingly we were able to protect the cells from these H(2)O(2)-induced effects and prevent calpain up-regulation with a natural antioxidant extracted from pine bark (Oligopin). These data strongly suggest that oxidative stress could impair satellite cell functionality via calpain-dependent pathways and that an antioxidant such as Oligopin could prevent apoptosis and calpain activation.


Subject(s)
Calcium Signaling/physiology , Calpain/metabolism , Myoblasts/metabolism , Oxidative Stress/physiology , Up-Regulation/physiology , Antioxidants/pharmacology , Apoptosis/drug effects , Calcium Signaling/drug effects , Calpain/antagonists & inhibitors , Calpain/genetics , Cell Survival/drug effects , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Cytoplasm/metabolism , Cytoplasmic Structures/metabolism , Dipeptides/pharmacology , Flavonoids/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Humans , Hydrogen Peroxide/pharmacology , Mitochondrial Proton-Translocating ATPases/metabolism , Myoblasts/drug effects , Myoblasts/enzymology , Oxidative Stress/drug effects , Phenols/pharmacology , Pinus/chemistry , Plant Extracts/pharmacology , Polyphenols , Protein Carbonylation/drug effects , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...