Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
In Vitro Cell Dev Biol Anim ; 60(7): 760-770, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38619740

ABSTRACT

Skeletal muscle's regenerative ability is vital for maintaining muscle function, but chronic diseases like Duchenne muscular dystrophy can deplete this capacity. Muscle satellite cells, quiescent in normal situations, are activated during muscle injury, expressing myogenic regulatory factors, and producing myogenic progenitor cells. It was reported that muscle stem cells in primary culture and reserve cells in C2C12 cells express anti-apoptotic protein Bcl-2. Although the role of Bcl-2 expressed in myogenic cells has been thought to be to enhance cell viability, we hypothesized that Bcl-2 may promote the formation of reserve cells. The expression pattern analysis showed the expression of Bcl-2 in undifferentiated mononucleated cells, emphasizing its usefulness as a reserve cell marker and reminding us that cells expressing Bcl-2 have low proliferative potential. Silencing of Bcl-2 by transfection with siRNA decreased cell viability and the number of reserve cells, while overexpression of Bcl-2 not only increases cell viability but also inhibits muscle differentiation and proliferation. These results emphasize dual roles of Bcl-2 in protecting cells from apoptosis and contributing to reserve cell formation by regulating myoblast proliferation and/or differentiation. Overall, the study sheds light on the multifaceted role of Bcl-2 in the maintenance of skeletal muscle regeneration.


Subject(s)
Cell Differentiation , Cell Proliferation , Cell Survival , Muscle Development , Proto-Oncogene Proteins c-bcl-2 , Animals , Mice , Apoptosis/genetics , Cell Line , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/metabolism
2.
FASEB J ; 37(1): e22676, 2023 01.
Article in English | MEDLINE | ID: mdl-36468834

ABSTRACT

The G protein-coupled receptors, GPR43 (free fatty acid receptor 2, FFA2) and GPR41 (free fatty acid receptor 3, FFA3), are activated by short-chain fatty acids produced under various conditions, including microbial fermentation of carbohydrates. Previous studies have implicated this receptor energy homeostasis and immune responses as well as in cell growth arrest and apoptosis. Here, we observed the expression of both receptors in human blood cells and a remarkable enhancement in leukemia cell lines (HL-60, U937, and THP-1 cells) during differentiation. A reporter assay revealed that GPR43 is coupled with Gαi and Gα12/13 and is constitutively active without any stimuli. Specific blockers of GPR43, GLPG0974 and CATPB function as inverse agonists because treatment with these compounds significantly reduces constitutive activity. In HL-60 cells, enhanced expression of GPR43 led to growth arrest through Gα12/13 . In addition, the blockage of GPR43 activity in these cells significantly impaired their adherent properties due to the reduction of adhesion molecules. We further revealed that enhanced GPR43 activity induces F-actin formation. However, the activity of GPR43 did not contribute to butyrate-induced apoptosis in differentiated HL-60 cells because of the ineffectiveness of the inverse agonist on cell death. Collectively, these results suggest that GPR43, which possesses constitutive activity, is crucial for growth arrest, followed by the proper differentiation of leukocytes.


Subject(s)
Fatty Acids, Volatile , Leukocytes , Receptors, Cell Surface , Humans , Fatty Acids, Volatile/metabolism , Leukocytes/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Cell Differentiation , HL-60 Cells
3.
Nat Commun ; 9(1): 2049, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29799007

ABSTRACT

Myotube formation by fusion of myoblasts and subsequent elongation of the syncytia is essential for skeletal muscle formation. However, molecules that regulate myotube formation remain elusive. Here we identify PIEZO1, a mechanosensitive Ca2+ channel, as a key regulator of myotube formation. During myotube formation, phosphatidylserine, a phospholipid that resides in the inner leaflet of the plasma membrane, is transiently exposed to cell surface and promotes myoblast fusion. We show that cell surface phosphatidylserine inhibits PIEZO1 and that the inward translocation of phosphatidylserine, which is driven by the phospholipid flippase complex of ATP11A and CDC50A, is required for PIEZO1 activation. PIEZO1-mediated Ca2+ influx promotes RhoA/ROCK-mediated actomyosin assemblies at the lateral cortex of myotubes, thus preventing uncontrolled fusion of myotubes and leading to polarized elongation during myotube formation. These results suggest that cell surface flip-flop of phosphatidylserine acts as a molecular switch for PIEZO1 activation that governs proper morphogenesis during myotube formation.


Subject(s)
Cell Differentiation , Cell Membrane/metabolism , Ion Channels/metabolism , Muscle Fibers, Skeletal/metabolism , Phosphatidylserines/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Membrane/genetics , Humans , Ion Channels/genetics , Mice , Muscle Fibers, Skeletal/cytology
4.
Exp Cell Res ; 333(2): 228-237, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25773777

ABSTRACT

Skeletal muscle stem cells named muscle satellite cells are normally quiescent but are activated in response to various stimuli, such as injury and overload. Activated satellite cells enter the cell cycle and proliferate to produce a large number of myogenic progenitor cells, and these cells then differentiate and fuse to form myofibers. Zinc is one of the essential elements in the human body, and has multiple roles, including cell growth and DNA synthesis. However, the role of zinc in myogenic cells is not well understood, and is the focus of this study. We first examined the effects of zinc on differentiation of murine C2C12 myoblasts and found that zinc promoted proliferation, with an increased number of cells incorporating EdU, but inhibited differentiation with reduced myogenin expression and myotube formation. Furthermore, we used the C2C12 reserve cell model of myogenic quiescence to investigate the role of zinc on activation of myogenic cells. The number of reserve cells incorporating BrdU was increased by zinc in a dose dependent manner, with the number dramatically further increased using a combination of zinc and insulin. Akt and extracellular signal-regulated kinase (ERK) are downstream of insulin signaling, and both were phosphorylated after zinc treatment. The zinc/insulin combination-induced activation involved the phosphoinositide 3-kinase (PI3K)/Akt and ERK cascade. We conclude that zinc promotes activation and proliferation of myogenic cells, and this activation requires phosphorylation of PI3K/Akt and ERK as part of the signaling cascade.


Subject(s)
Cell Proliferation , Satellite Cells, Skeletal Muscle/physiology , Zinc/physiology , Animals , Cell Line , Insulin/physiology , MAP Kinase Signaling System , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism
5.
Am J Pathol ; 184(11): 3094-104, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25174878

ABSTRACT

Duchenne muscular dystrophy is a lethal X-linked disease with no effective treatment. Progressive muscle degeneration, increased macrophage infiltration, and ectopic calcification are characteristic features of the mdx mouse, a murine model of Duchenne muscular dystrophy. Because dietary phosphorus/phosphate consumption is increasing and adverse effects of phosphate overloading have been reported in several disease conditions, we examined the effects of dietary phosphorus intake in mdx mice phenotypes. On weaning, control and mdx mice were fed diets containing 0.7, 1.0, or 2.0 g phosphorus per 100 g until they were 90 days old. Dystrophic phenotypes were evaluated in cryosections of quadriceps and tibialis anterior muscles, and maximal forces and voluntary activity were measured. Ectopic calcification was analyzed by electron microscopy to determine the cells initially responsible for calcium deposition in skeletal muscle. Dietary phosphorus overload dramatically exacerbated the dystrophic phenotypes of mdx mice by increasing inflammation associated with infiltration of M1 macrophages. In contrast, minimal muscle necrosis and inflammation were observed in exercised mdx mice fed a low-phosphorus diet, suggesting potential beneficial therapeutic effects of lowering dietary phosphorus intake on disease progression. To our knowledge, this is the first report showing that dietary phosphorus intake directly affects muscle pathological characteristics of mdx mice. Dietary phosphorus overloading promoted dystrophic disease progression in mdx mice, whereas restricting dietary phosphorus intake improved muscle pathological characteristics and function.


Subject(s)
Calcinosis/pathology , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Phosphorus, Dietary/administration & dosage , Animals , Calcinosis/metabolism , Calcium/metabolism , Dose-Response Relationship, Drug , Dystrophin/genetics , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Phenotype
6.
Exp Cell Res ; 326(1): 112-24, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24960577

ABSTRACT

Skeletal muscle can regenerate repeatedly due to the presence of resident stem cells, called satellite cells. Because satellite cells are usually quiescent, they must be activated before participating in muscle regeneration in response to stimuli such as injury, overloading, and stretch. Although satellite cell activation is a crucial step in muscle regeneration, little is known of the molecular mechanisms controlling this process. Recent work showed that the bioactive lipid sphingosine-1-phosphate (S1P) plays crucial roles in the activation, proliferation, and differentiation of muscle satellite cells. We investigated the role of growth factors in S1P-mediated satellite cell activation. We found that epidermal growth factor (EGF) in combination with insulin induced proliferation of quiescent undifferentiated mouse myoblast C2C12 cells, which are also known as reserve cells, in serum-free conditions. Sphingosine kinase activity increased when reserve cells were stimulated with EGF. Treatment of reserve cells with the D-erythro-N,N-dimethylsphingosine, Sphingosine Kinase Inhibitor, or siRNA duplexes specific for sphingosine kinase 1, suppressed EGF-induced C2C12 activation. We also present the evidence showing the S1P receptor S1P2 is involved in EGF-induced reserve cell activation. Moreover, we demonstrated a combination of insulin and EGF promoted activation of satellite cells on single myofibers in a manner dependent on SPHK and S1P2. Taken together, our observations show that EGF-induced satellite cell activation is mediated by S1P and its receptor.


Subject(s)
Epidermal Growth Factor/pharmacology , Lysophospholipids/pharmacology , Muscle, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Sphingosine/analogs & derivatives , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Proliferation , Cells, Cultured , Mice , Muscle, Skeletal/cytology , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sphingosine/pharmacology
7.
Cell Struct Funct ; 35(1): 63-71, 2010.
Article in English | MEDLINE | ID: mdl-20460816

ABSTRACT

Satellite cells are muscle-resident stem cells, which are located beneath the basement membrane of myofibers. Because the number of satellite cells is normally constant, there must be a tight regulation of satellite cell activation and self-renewal. However, the molecular mechanisms involved in satellite cell maintenance are largely unknown, and thus have become the subject of extensive study these days. Although RNA interference with a small interfering RNA has been widely used to investigate the role of specific gene products, inefficient knockdown of Grb2 expression occurred in quiescent reserve cells, a model for quiescent satellite cells, by ordinary transfection protocol. In this study we report that pretreatment with trypsin greatly enhanced siRNA delivery into quiescent reserve cells, resulting in efficient silencing of Grb2 expression. By applying a combination of Grb2-silencing and protein kinase C inhibitors, we demonstrated that extracellular signal-regulated kinase (ERK) phosphorylation induced with fibroblast growth factor 2 (FGF2) was dependent on both Grb2 and protein kinase C (PKC) with different kinetics. We concluded that the PKC-mediated pathway contributes to rapid initiation and termination of ERK phosphorylation, while the Grb2-mediated pathway contributes to delayed and sustained ERK phosphorylation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/pharmacology , GRB2 Adaptor Protein/metabolism , Protein Kinase C/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Animals , Cell Line, Tumor , GRB2 Adaptor Protein/genetics , Gene Knockdown Techniques , Mice , Phosphorylation , Protein Kinase C/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction
8.
Cell Struct Funct ; 34(2): 77-88, 2009.
Article in English | MEDLINE | ID: mdl-19622873

ABSTRACT

Ectopic calcification occurs in the skeletal muscle of mdx mice, a dystrophin-deficient animal model of Duchenne muscular dystrophy. The purpose of this study was to clarify the mechanism of the calcification. The calcified deposits were identified as hydroxyapatite, a crystallized form of calcium phosphate, and the serum inorganic phosphate (Pi) level in the mdx mice was approximately 1.4 times higher than that in the normal B10 mice, suggesting that Pi plays a critical role in the ectopic calcification. When C2C12 mouse myoblasts were cultured under high-Pi conditions, myogenic differentiation was retarded while the expression of osteogenic markers such as osteocalcin and Runx2 were upregulated. This was followed by the generation of calcium deposition. Moreover, ectopic calcification reduced to an undetectable level in most of the mdx mice fed a Pi-reduced diet. We therefore conclude that the Pi-induced osteogenesis of muscle cells is responsible for ectopic calcification in the skeletal muscle of mdx mice.


Subject(s)
Calcinosis/blood , Hydroxyapatites/blood , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/blood , Ossification, Heterotopic/blood , Phosphates/blood , Animals , Calcinosis/metabolism , Calcinosis/pathology , Cell Line , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Hydroxyapatites/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Muscle, Skeletal/drug effects , Muscle, Skeletal/ultrastructure , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/pathology , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Osteocalcin/metabolism , Phosphates/metabolism
9.
J Cell Biol ; 174(2): 245-53, 2006 Jul 17.
Article in English | MEDLINE | ID: mdl-16847102

ABSTRACT

Adult skeletal muscle is able to repeatedly regenerate because of the presence of satellite cells, a population of stem cells resident beneath the basal lamina that surrounds each myofiber. Little is known, however, of the signaling pathways involved in the activation of satellite cells from quiescence to proliferation, a crucial step in muscle regeneration. We show that sphingosine-1-phosphate induces satellite cells to enter the cell cycle. Indeed, inhibiting the sphingolipid-signaling cascade that generates sphingosine-1-phosphate significantly reduces the number of satellite cells able to proliferate in response to mitogen stimulation in vitro and perturbs muscle regeneration in vivo. In addition, metabolism of sphingomyelin located in the inner leaflet of the plasma membrane is probably the main source of sphingosine-1-phosphate used to mediate the mitogenic signal. Together, our observations show that sphingolipid signaling is involved in the induction of proliferation in an adult stem cell and a key component of muscle regeneration.


Subject(s)
Cell Cycle , Lysophospholipids/metabolism , Satellite Cells, Skeletal Muscle/cytology , Signal Transduction , Sphingomyelins/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Membrane/metabolism , Cell Proliferation , DNA/biosynthesis , Lysophospholipids/biosynthesis , Mice , Mice, Inbred C57BL , Muscle, Skeletal/physiology , Regeneration , Sphingosine/biosynthesis , Sphingosine/metabolism
10.
J Cell Sci ; 119(Pt 9): 1824-32, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16608873

ABSTRACT

Skeletal muscle growth and regeneration are attributed to satellite cells - muscle stem cells resident beneath the basal lamina that surrounds each myofibre. Quiescent satellite cells express the transcription factor Pax7 and when activated, coexpress Pax7 with MyoD. Most then proliferate, downregulate Pax7 and differentiate. By contrast, others maintain Pax7 but lose MyoD and return to a state resembling quiescence. Here we show that Pax7 is able to drive transcription in quiescent and activated satellite cells, and continues to do so in those cells that subsequently cease proliferation and withdraw from immediate differentiation. We found that constitutive expression of Pax7 in satellite-cell-derived myoblasts did not affect MyoD expression or proliferation. Although maintained expression of Pax7 delayed the onset of myogenin expression it did not prevent, and was compatible with, myogenic differentiation. Constitutive Pax7 expression in a Pax7-null C2C12 subclone increased the proportion of cells expressing MyoD, showing that Pax7 can act genetically upstream of MyoD. However these Pax7-null cells were unable to differentiate into normal myotubes in the presence of Pax7. Therefore Pax7 may be involved in maintaining proliferation and preventing precocious differentiation, but does not promote quiescence.


Subject(s)
Muscle, Skeletal/cytology , MyoD Protein/metabolism , PAX7 Transcription Factor/metabolism , Satellite Cells, Skeletal Muscle/physiology , Transcription, Genetic , Animals , Cell Differentiation/physiology , Cell Fusion , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Mice , MyoD Protein/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Satellite Cells, Skeletal Muscle/cytology
11.
J Histochem Cytochem ; 54(4): 375-84, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16400000

ABSTRACT

Satellite cells are responsible for postnatal growth, hypertrophy, and regeneration of skeletal muscle. They are normally quiescent, and must be activated to fulfill these functions, yet little is known of how this is regulated. As a first step in determining the role of lipids in this process, we examined the dynamics of sphingomyelin in the plasma membrane. Sphingomyelin contributes to caveolae/lipid rafts, which act to concentrate signaling molecules, and is also a precursor of several bioactive lipids. Proliferating or differentiated C2C12 muscle cells did not bind lysenin, a sphingomyelin-specific binding protein, but noncycling reserve cells did. Quiescent satellite cells also bound lysenin, revealing high levels of sphingomyelin in their plasma membranes. On activation, however, the levels of sphingomyelin drop, so that lysenin did not label proliferating satellite cells. Although most satellite cell progeny differentiate, others stop cycling, maintain Pax7, downregulate MyoD, and escape immediate differentiation. Importantly, many of these Pax7-positive/MyoD-negative cells also regained lysenin binding on their surface, showing that the levels of sphingomyelin had again increased. Our observations show that quiescent satellite cells are characterized by high levels of sphingomyelin in their plasma membranes and that lysenin provides a novel marker of myogenic quiescence.


Subject(s)
Satellite Cells, Skeletal Muscle/physiology , Sphingomyelins/metabolism , Animals , Blotting, Western , Cell Membrane/metabolism , Cell Proliferation , Cells, Cultured , Immunohistochemistry , Indicators and Reagents , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Proteins/chemistry , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Sphingomyelins/chemistry , Toxins, Biological
12.
J Cell Biol ; 166(3): 347-57, 2004 Aug 02.
Article in English | MEDLINE | ID: mdl-15277541

ABSTRACT

Growth, repair, and regeneration of adult skeletal muscle depends on the persistence of satellite cells: muscle stem cells resident beneath the basal lamina that surrounds each myofiber. However, how the satellite cell compartment is maintained is unclear. Here, we use cultured myofibers to model muscle regeneration and show that satellite cells adopt divergent fates. Quiescent satellite cells are synchronously activated to coexpress the transcription factors Pax7 and MyoD. Most then proliferate, down-regulate Pax7, and differentiate. In contrast, other proliferating cells maintain Pax7 but lose MyoD and withdraw from immediate differentiation. These cells are typically located in clusters, together with Pax7-ve progeny destined for differentiation. Some of the Pax7+ve/MyoD-ve cells then leave the cell cycle, thus regaining the quiescent satellite cell phenotype. Significantly, noncycling cells contained within a cluster can be stimulated to proliferate again. These observations suggest that satellite cells either differentiate or switch from terminal myogenesis to maintain the satellite cell pool.


Subject(s)
Cell Differentiation/physiology , Satellite Cells, Skeletal Muscle/physiology , Animals , Down-Regulation , Genes, Reporter , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , MyoD Protein/metabolism , PAX7 Transcription Factor
SELECTION OF CITATIONS
SEARCH DETAIL