Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 489(2): 142-148, 2017 07 22.
Article in English | MEDLINE | ID: mdl-28546002

ABSTRACT

Muscle weakness is one of the most common symptoms in aged individuals and increases risk of mortality. Thus, maintenance of muscle mass is important for inhibiting aging. In this study, we investigated the effect of catechins, polyphenol compounds in green tea, on muscle regeneration. We found that (-)-epicatechin gallate (ECG) and (-)-epigallocatechin-3-gallate (EGCG) activate satellite cells by induction of Myf5 transcription factors. For satellite cell activation, Akt kinase was significantly induced after ECG treatment and ECG-induced satellite cell activation was blocked in the presence of Akt inhibitor. ECG also promotes myogenic differentiation through the induction of myogenic markers, including Myogenin and Muscle creatine kinase (MCK), in satellite and C2C12 myoblast cells. Finally, EGCG administration to mice significantly increased muscle fiber size for regeneration. Taken together, the results suggest that catechins stimulate muscle stem cell activation and differentiation for muscle regeneration.


Subject(s)
Catechin/pharmacology , Muscles/drug effects , Muscles/physiology , Myogenic Regulatory Factor 5/biosynthesis , Regeneration/drug effects , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects , Animals , Catechin/chemistry , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Molecular Conformation , Muscles/cytology , Myogenic Regulatory Factor 5/metabolism , Structure-Activity Relationship
2.
J Shoulder Elbow Surg ; 23(1): 91-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23791493

ABSTRACT

BACKGROUND: The influence of age on rotator cuff function and muscle structure remains poorly understood. We hypothesize that normal aging influences rotator cuff function, muscle structure, and regulatory protein expression in an established rat model of aging. METHODS: Seventeen rats were obtained from the National Institute on Aging. The supraspinatus muscles in 11 middle-aged (12 months old) and 6 old (28 months old) rats were studied for age-related changes in rotator cuff neuromuscular function by in vivo muscle force testing and electromyography (EMG). Changes in muscle structure and molecular changes were assessed with quantitative immunohistochemistry for myogenic determination factor 1 (MyoD) and myogenic factor 5 (Myf5) expression. RESULTS: Old animals revealed significantly decreased peak tetanic muscle force at 0.5 N and 0.7 N preload tension (P < .05). The age of the animal accounted for 20.9% of variance and significantly influenced muscle force (P = .026). Preload tension significantly influenced muscle force production (P < .001) and accounted for 12.7% of total variance. There was regional heterogeneity in maximal compound motor action potential (CMAP) amplitude in the supraspinatus muscle; the proximal portion had a significantly higher CMAP than the middle and distal portions (P < .05). The expression of muscle regulatory factors MyoD and Myf5 was significantly decreased in old animals compared with middle-aged animals (P < .05). CONCLUSIONS: The normal aging process in this rat model significantly influenced contractile strength of the supraspinatus muscle and led to decreased expression of muscle regulatory factors. High preload tensions led to a significant decrease in force production in both middle-aged and old animals.


Subject(s)
Aging/physiology , Rotator Cuff/metabolism , Rotator Cuff/physiopathology , Aging/metabolism , Animals , Electromyography , Immunohistochemistry , Male , Models, Animal , Muscle Contraction/physiology , MyoD Protein/biosynthesis , Myogenic Regulatory Factor 5/biosynthesis , Rats , Rotator Cuff/pathology
3.
Histochem Cell Biol ; 141(3): 289-300, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24186058

ABSTRACT

The embryonic muscles of the axial skeleton and limbs take their origin from the dermomyotomes of the somites. During embryonic myogenesis, muscle precursors delaminate from the dermomyotome giving rise to the hypaxial and epaxial myotome. Mutant studies for myogenic regulatory factors have shown that the development of the hypaxial myotome differs from the formation of the epaxial myotome and that the development of the hypaxial myotome depends on the latter within the trunk region. The transcriptional networks that regulate the transition of proliferative dermomyotomal cells into the predominantly post-mitotic hypaxial myotome, as well as the eventual patterning of the myotome, are not fully understood. Similar transitions occurring during the development of the neural system have been shown to be controlled by the Atonal family of helix-loop-helix transcription factors. Here, we demonstrate that ATOH8, a member of the Atonal family, is expressed in a subset of embryonic muscle cells in the dermomyotome and myotome. Using the RNAi approach, we show that loss of ATOH8 in the lateral somites at the trunk level results in a blockage of differentiation and thus causes cells to be maintained in a predetermined state. Furthermore, we show that ATOH8 is also expressed in cultured C2C12 mouse myoblasts and becomes dramatically downregulated during their differentiation. We propose that ATOH8 plays a role during the transition of myoblasts from the proliferative phase to the differentiation phase and in the regulation of myogenesis in the hypaxial myotome of the trunk.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Body Patterning/genetics , Muscle Development/genetics , Muscle, Skeletal/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Differentiation/genetics , Cell Line , Cell Lineage , Chick Embryo , Down-Regulation , Gene Expression Regulation, Developmental , Mice , Myoblasts/cytology , Myogenic Regulatory Factor 5/biosynthesis , Myogenin/biosynthesis , PAX7 Transcription Factor/biosynthesis , RNA Interference , RNA, Small Interfering , Somites/physiology
4.
Biol Trace Elem Res ; 150(1-3): 166-72, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23054870

ABSTRACT

The biological function of selenium (Se) is mainly elicited through Se-containing proteins. Selenoprotein W (SelW), one member of the selenoprotein family, is essential for the normal function of the skeletal muscle system. To investigate the possible relationship of Se in the process of differentiation in chicken myoblasts and the expression of SelW, the cultured chicken embryonic myoblasts were incubated with sodium selenite at different concentrations for 72 h, and then the mRNA levels of SelW and myogenic regulatory factors (MRFs) in myoblasts were determined at 12, 24, 48, and 72 h, respectively. Furthermore, the correlation between SelW mRNA expression and MRF mRNA expression was assessed. The results showed that the sodium selenite medium enhanced the mRNA expression of SelW, Myf-5, MRF4, and myogenin in chicken myoblasts. The mRNA expression levels of MRFs were significantly correlated with those of SelW at 24, 48, and 72 h. These data demonstrate that Se is involved in the differentiation of chicken embryonic myoblasts, and SelW showed correlation with MRFs.


Subject(s)
Myoblasts/metabolism , Myogenic Regulatory Factor 5/biosynthesis , Myogenic Regulatory Factors/biosynthesis , Myogenin/biosynthesis , Selenium/metabolism , Selenoprotein W/biosynthesis , Up-Regulation , Animals , Animals, Inbred Strains , Avian Proteins/biosynthesis , Avian Proteins/genetics , Avian Proteins/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chick Embryo , Muscle Development , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Myoblasts/cytology , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Myogenin/genetics , Myogenin/metabolism , Osmolar Concentration , RNA, Messenger/metabolism , Selenoprotein W/genetics , Selenoprotein W/metabolism , Sodium Selenite/metabolism , Time Factors
5.
J Proteomics ; 75(9): 2601-10, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22465714

ABSTRACT

Both p300 and ß-catenin are transcriptional activators and phosphoproteins, and play a central role in Wnt/ß-catenin-dependent transcriptional regulation. The minimum ß-catenin binding domain of p300 has been mapped to the N-terminus 1-111 amino acids. Here, we performed phosphoproteomic analysis for the critical binding region using LC-MS/MS approach to investigate potential phosphosites that may affect the binding affinity. By implementing TiO(2)-based phosphopeptide affinity purification followed by LC-MS/MS analysis with both collision-induced dissociation (CID) and electron transfer dissociation (ETD) methods, two unique phosphosites Ser12 and Ser89 were identified, of which, phosphorylation at Ser12 is novel. Functional studies aided by site-directed mutagenesis, co-immunoprecipitation and mammalian two-hybrid assay have concluded that phosphorylation at Ser12 critically mediates the binding ability of p300 with ß-catenin. Further studies utilizing specific MAPK inhibitors suggest that the p38 MAPK activation is the upstream signal required for Ser12 phosphorylation. The transcriptional roles of p300/ß-catenin complex in myoblast differentiation are discussed.


Subject(s)
E1A-Associated p300 Protein/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Cell Differentiation , Humans , Immunoprecipitation , Myoblasts/cytology , Myogenic Regulatory Factor 5/biosynthesis , Phosphorylation , Serine/metabolism , Tandem Mass Spectrometry , Two-Hybrid System Techniques , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
6.
J Biol Chem ; 287(18): 14524-34, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22399301

ABSTRACT

We previously showed in vitro that calcium entry through Trpc1 ion channels regulates myoblast migration and differentiation. In the present work, we used primary cell cultures and isolated muscles from Trpc1(-/-) and Trpc1(+/+) murine model to investigate the role of Trpc1 in myoblast differentiation and in muscle regeneration. In these models, we studied regeneration consecutive to cardiotoxin-induced muscle injury and observed a significant hypotrophy and a delayed regeneration in Trpc1(-/-) muscles consisting in smaller fiber size and increased proportion of centrally nucleated fibers. This was accompanied by a decreased expression of myogenic factors such as MyoD, Myf5, and myogenin and of one of their targets, the developmental MHC (MHCd). Consequently, muscle tension was systematically lower in muscles from Trpc1(-/-) mice. Importantly, the PI3K/Akt/mTOR/p70S6K pathway, which plays a crucial role in muscle growth and regeneration, was down-regulated in regenerating Trpc1(-/-) muscles. Indeed, phosphorylation of both Akt and p70S6K proteins was decreased as well as the activation of PI3K, the main upstream regulator of the Akt. This effect was independent of insulin-like growth factor expression. Akt phosphorylation also was reduced in Trpc1(-/-) primary myoblasts and in control myoblasts differentiated in the absence of extracellular Ca(2+) or pretreated with EGTA-AM or wortmannin, suggesting that the entry of Ca(2+) through Trpc1 channels enhanced the activity of PI3K. Our results emphasize the involvement of Trpc1 channels in skeletal muscle development in vitro and in vivo, and identify a Ca(2+)-dependent activation of the PI3K/Akt/mTOR/p70S6K pathway during myoblast differentiation and muscle regeneration.


Subject(s)
Cell Differentiation/physiology , Myoblasts, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Regeneration/physiology , Signal Transduction/physiology , TRPC Cation Channels/metabolism , Animals , Calcium/metabolism , Enzyme Activation/physiology , Gene Expression Regulation/physiology , Mice , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , MyoD Protein/biosynthesis , MyoD Protein/genetics , Myoblasts, Skeletal/cytology , Myogenic Regulatory Factor 5/biosynthesis , Myogenic Regulatory Factor 5/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , TRPC Cation Channels/genetics
7.
Am J Physiol Cell Physiol ; 302(11): C1621-31, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22460713

ABSTRACT

The chemokine-like receptor-1 (CMKLR1) is a G protein-coupled receptor that is activated by chemerin, a secreted plasma leukocyte attractant and adipokine. Previous studies identified that CMKLR1 is expressed in skeletal muscle in a stage-specific fashion during embryogenesis and in adult mice; however, its function in skeletal muscle remains unclear. Based on the established function of CMKLR1 in cell migration and differentiation, we investigated the hypothesis that CMKLR1 regulates the differentiation of myoblasts into myotubes. In C(2)C(12) mouse myoblasts, CMKLR1 expression increased threefold with differentiation into multinucleated myotubes. Decreasing CMKLR1 expression by adenoviral-delivered small-hairpin RNA (shRNA) impaired the differentiation of C(2)C(12) myoblasts into mature myotubes and reduced the mRNA expression of myogenic regulatory factors myogenin and MyoD while increasing Myf5 and Mrf4. At embryonic day 12.5 (E12.5), CMKLR1 knockout (CMKLR1(-/-)) mice appeared developmentally delayed and displayed significantly lower wet weights and a considerably diminished myotomal component of somites as revealed by immunolocalization of myosin heavy chain protein compared with wild-type (CMKLR1(+/+)) mouse embryos. These changes were associated with increased Myf5 and decreased MyoD protein expression in the somites of E12.5 CMKLR1(-/-) mouse embryos. Adult male CMKLR1(-/-) mice had significantly reduced bone-free lean mass and weighed less than the CMKLR1(+/+) mice. We conclude that CMKLR1 is essential for myogenic differentiation of C(2)C(12) cells in vitro, and the CMKLR1 null mice have a subtle skeletal muscle deficit beginning from embryonic life that persists during postnatal life.


Subject(s)
Muscle Cells/metabolism , Muscle Development , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Receptors, G-Protein-Coupled/metabolism , Absorptiometry, Photon , Animals , Cell Differentiation , Cells, Cultured , Male , Mice , Mice, Knockout , Muscle Cells/physiology , Muscle Fibers, Skeletal/physiology , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscle, Skeletal/embryology , Muscle, Skeletal/physiology , MyoD Protein/biosynthesis , MyoD Protein/genetics , Myogenic Regulatory Factor 5/biosynthesis , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, Chemokine
8.
Pediatr Surg Int ; 27(6): 579-82, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21258934

ABSTRACT

PURPOSE: The nitrofen model of congenital diaphragmatic hernia (CDH) is widely used to investigate the pathogenesis of CDH. However, the exact pathomechanism of the diaphragmatic defect is still unclear. Diaphragmatic muscularization represents the last stage of diaphragmatic development. Myogenic differentiation 1 (MyoD) and myogenic factor 5 (Myf5) play a crucial role in muscularization. MyoD(-/-) : Myf5(+/-) mutant mice show reduced diaphragmatic size, whereas MyoD(+/-) : Myf5(-/-) mutants have normal diaphragms. We designed this study to investigate diaphragmatic gene expression of MyoD and Myf5 in the nitrofen CDH model. METHODS: Pregnant rats received nitrofen or vehicle on day 9 of gestation (D9), followed by cesarean section on D18 and D21. Fetal diaphragms (n = 40) were micro-dissected and divided into CDH group and controls. MyoD and Myf5 mRNA-expression were determined using Real-time PCR. Immunohistochemistry was performed to evaluate protein expression of MyoD and Myf5. RESULTS: Relative diaphragmatic mRNA expression levels and immunoreactivity of MyoD were decreased in the CDH group on D18 and D21. Myf 5 mRNA and protein expression were not altered in the CDH group. CONCLUSION: This is the first study showing that MyoD expression is selectively decreased in the diaphragm muscle in the nitrofen model of CDH.


Subject(s)
Diaphragm/embryology , Gene Expression Regulation, Developmental , Hernias, Diaphragmatic, Congenital , MyoD Protein/genetics , Myogenic Regulatory Factor 5/genetics , Pregnancy, Animal , RNA, Messenger/genetics , Animals , Animals, Newborn , Diaphragm/metabolism , Disease Models, Animal , Female , Hernia, Diaphragmatic/chemically induced , Hernia, Diaphragmatic/genetics , Hernia, Diaphragmatic/metabolism , Immunohistochemistry , MyoD Protein/biosynthesis , Myogenic Regulatory Factor 5/biosynthesis , Phenyl Ethers/toxicity , Polymerase Chain Reaction , Pregnancy , Rats , Rats, Sprague-Dawley
9.
J Biol Chem ; 286(12): 10649-59, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21252233

ABSTRACT

R-spondins (RSPOs) are a recently characterized family of secreted proteins that activate WNT/ß-catenin signaling. In this study, we investigated the potential roles of the RSPO proteins during myogenic differentiation. Overexpression of the Rspo1 gene or administration of recombinant RSPO2 protein enhanced mRNA and protein expression of a basic helix-loop-helix (bHLH) class myogenic determination factor, MYF5, in both C2C12 myoblasts and primary satellite cells, whereas MYOD or PAX7 expression was not affected. RSPOs also promoted myogenic differentiation and induced hypertrophic myotube formation in C2C12 cells. In addition, Rspo2 and Rspo3 gene knockdown by RNA interference significantly compromised MYF5 expression, myogenic differentiation, and myotube formation. Furthermore, Myf5 expression was reduced in the developing limbs of mouse embryos lacking the Rspo2 gene. Finally, we demonstrated that blocking of WNT/ß-catenin signaling by DKK1 or a dominant-negative form of TCF4 reversed MYF5 expression, myogenic differentiation, and hypertrophic myotube formation induced by RSPO2, indicating that RSPO2 exerts its activity through the WNT/ß-catenin signaling pathway. Our results provide strong evidence that RSPOs are key positive regulators of skeletal myogenesis acting through the WNT/ß-catenin signaling pathway.


Subject(s)
Embryo, Mammalian/embryology , Intercellular Signaling Peptides and Proteins/biosynthesis , Muscle Development/physiology , Muscle Fibers, Skeletal/metabolism , Thrombospondins/biosynthesis , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Gene Expression Regulation, Developmental/physiology , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Mutant Strains , Muscle Fibers, Skeletal/cytology , Myogenic Regulatory Factor 5/biosynthesis , Myogenic Regulatory Factor 5/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Thrombospondins/genetics , Wnt Proteins/genetics , beta Catenin/genetics
10.
J Biol Chem ; 286(8): 6855-64, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21159776

ABSTRACT

Myf5 is a myogenic regulatory factor that functions in myogenesis. An intronic microRNA, miR-In300, located within zebrafish myf5 intron I, has been reported to silence myf5 through the targeting of dickkopf-3-related gene (dkk3r). However, the molecular mechanism underlying the control of myf5 expression by dkk3r is unknown. By injecting dkk3r-specific morpholino-oligonucleotide (dkk3r-MO) to knock down Dkk3r, we found that the phosphorylated p38a protein was reduced. Knockdown of p38a resulted in malformed somites and reduced myf5 transcripts, which photocopied the defects induced by injection of dkk3r-MO. To block the MAPK pathway, phosphorylation of p38 was inhibited by introduction of SB203580, which caused the down-regulation of myf5 expression. The GFP signal was dramatically decreased in somites when we injected p38a-MO into embryos derived from transgenic line Tg(myf5(80K):GFP), in which the GFP was driven by the myf5 promoter. Although these p38a-MO-induced defects were rescued by co-injection with p38a mRNA, they were not rescued with p38a mRNA containing a mutation at the phosphorylation domain. Moreover, overexpression of Smad2 or Smad3a enhanced myf5 expression, but the defects induced by the dominant negative form of either Smad2 or Smad3a equaled those of embryos injected with either dkk3r-MO or p38a-MO. These results support the involvement of Smad2·Smad3a in p38a mediation. Overexpression of Smad4 enabled the rescue of myf5 defects in the dkk3r-MO-injected embryos, but knockdown of either dkk3r or p38a caused Smad4 protein to lose stability. Therefore, we concluded that Dkk3r regulates p38a phosphorylation to maintain Smad4 stability, in turn enabling the Smad2·Smad3a·Smad4 complex to form and activate the myf5 promoter.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Myogenic Regulatory Factor 5/biosynthesis , Smad4 Protein/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Imidazoles/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Myogenic Regulatory Factor 5/genetics , Oligonucleotides/pharmacology , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Stability , Pyridines/pharmacology , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Smad4 Protein/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
11.
PLoS One ; 4(10): e7482, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19829708

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD) is a dominant disease linked to contractions of the D4Z4 repeat array in 4q35. We have previously identified a double homeobox gene (DUX4) within each D4Z4 unit that encodes a transcription factor expressed in FSHD but not control myoblasts. DUX4 and its target genes contribute to the global dysregulation of gene expression observed in FSHD. We have now characterized the homologous DUX4c gene mapped 42 kb centromeric of the D4Z4 repeat array. It encodes a 47-kDa protein with a double homeodomain identical to DUX4 but divergent in the carboxyl-terminal region. DUX4c was detected in primary myoblast extracts by Western blot with a specific antiserum, and was induced upon differentiation. The protein was increased about 2-fold in FSHD versus control myotubes but reached 2-10-fold induction in FSHD muscle biopsies. We have shown by Western blot and by a DNA-binding assay that DUX4c over-expression induced the MYF5 myogenic regulator and its DNA-binding activity. DUX4c might stabilize the MYF5 protein as we detected their interaction by co-immunoprecipitation. In keeping with the known role of Myf5 in myoblast accumulation during mouse muscle regeneration DUX4c over-expression activated proliferation of human primary myoblasts and inhibited their differentiation. Altogether, these results suggested that DUX4c could be involved in muscle regeneration and that changes in its expression could contribute to the FSHD pathology.


Subject(s)
Gene Expression Regulation , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , Myoblasts/cytology , Myogenic Regulatory Factor 5/biosynthesis , Myogenic Regulatory Factor 5/genetics , Up-Regulation , Animals , Biopsy , Cell Proliferation , HeLa Cells , Humans , Mice , Models, Genetic , Muscles/pathology , Muscular Dystrophy, Facioscapulohumeral/pathology , Protein Structure, Tertiary
12.
Anat Histol Embryol ; 38(4): 305-10, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19614631

ABSTRACT

We examined expression of four important members of myogenic regulatory factors (MRFs) in the myoblasts both at mRNA and protein levels, which were subjected to mechanical stretching in in vitro condition. Our results showed that MyoD expression existed both in the stretch and in the control group at all time periods of the mechanical stimulus. Myf-5 expressed only at early stage of the stretch group. Although mRNA and protein expressions of myogenin and MRF4 were detected both in the stretch and in the control group at 12 h after the stretching, their expressions were only shown in the stretch group at 24 h after the mechanical stimulus. However, at 36 and 48 h, none of the MRFs examined except MyoD appeared in both groups. Our results suggest that the MRFs are up-regulated upon mechanical stimulus and each member plays a different major role for either proliferation or differentiation of the myoblasts.


Subject(s)
MyoD Protein/biosynthesis , Myoblasts/physiology , Myogenic Regulatory Factor 5/biosynthesis , Myogenic Regulatory Factors/biosynthesis , Myogenin/biosynthesis , Animals , Blotting, Western , Cell Count , Cell Line , Cell Proliferation , Gene Expression , Mice , Microscopy, Phase-Contrast , MyoD Protein/genetics , Myoblasts/cytology , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factors/genetics , Myogenin/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Stress, Mechanical
13.
Eur J Appl Physiol ; 101(4): 427-36, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17641910

ABSTRACT

Evidence indicates that repeated-bouts of eccentric exercise (EE) do not exacerbate the extent of muscle damage indices, as compared to a single-bout. We hypothesized that molecular adaptations, under repeated-bouts of EE, would include suppression of muscle repair inhibitory factors such as myostatin and up-regulation of muscle repair positive regulatory factors such as myogenic regulatory factors (MRFs). Fifteen males were recruited for this study. The exercise group (n=9) successfully completed six sets of 15 reps of maximum voluntary eccentric contractions, for six consecutive days, using a dynamometer (Multicont-II). Blood and muscle biopsy samples were obtained from each subject 1 week prior to exercise, 2 days post the first training session, and 24 h after the last training session. Gene expression levels were determined using real-time RT-PCR. Blood samples were analyzed for creatine kinase (CK) and lactate-dehydrogenase (LDH) activity. Repeated-bouts of EE induced a large down-regulation of myostatin mRNA (-73%) which persisted throughout the study. The responses of MRFs were mild. At day 3 only myogenin increased significantly (1.9 fold) while MyoD decreased by 45%. Surprisingly, at day 7, despite the presence of muscle damage indices, all MRFs returned to the pre-exercise levels. The results of the present study showed that repeated-bouts of EE, for six consecutive days, dramatically decreased Myostatin mRNA expression but impaired the expression patterns of MRFs such that, with the exception of myogenin that showed a moderate non-sustained increase, MyoD and MYf5 response was minimal.


Subject(s)
Exercise/physiology , Gene Expression/physiology , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Transforming Growth Factor beta/biosynthesis , Adult , Cell Differentiation/physiology , Cell Proliferation , Creatine Kinase/metabolism , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Humans , Ki-67 Antigen/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Muscle Contraction/physiology , Muscle Strength/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , MyoD Protein/biosynthesis , MyoD Protein/genetics , Myogenic Regulatory Factor 5/biosynthesis , Myogenic Regulatory Factor 5/genetics , Myostatin , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/physiology , p21-Activated Kinases/biosynthesis , p21-Activated Kinases/genetics
14.
Proc Natl Acad Sci U S A ; 104(27): 11310-5, 2007 Jul 03.
Article in English | MEDLINE | ID: mdl-17592144

ABSTRACT

Myf5, a member of the myogenic regulatory factor family, plays a major role in determining myogenic cell fate at the onset of skeletal muscle formation in the embryo. Spatiotemporal control of its expression during development requires multiple enhancer elements spread over >100 kb at the Myf5 locus. Transcription in embryonic limbs is regulated by a 145-bp element located at -57.5 kb from the Myf5 gene. In the present study we show that Myf5 expression is severely impaired in the limb buds of Six1(-/-) and Six1(-/-)Six4(-/+) mouse mutants despite the presence of myogenic progenitor cells. The 145-bp regulatory element contains a sequence that binds Six1 and Six4 in electromobility shift assays in vitro and in chromatin immunoprecipitation assays with embryonic extracts. We further show that Six1 is able to transactivate a reporter gene under the control of this sequence. In vivo functionality of the Six binding site is demonstrated by transgenic analysis. Mutation of this site impairs reporter gene expression in the limbs and in mature somites where the 145-bp regulatory element is also active. Six1/4 therefore regulate Myf5 transcription, together with Pax3, which was previously shown to be required for the activity of the 145-bp element. Six homeoproteins, which also directly regulate the myogenic differentiation gene Myogenin and lie genetically upstream of Pax3, thus control hypaxial myogenesis at multiple levels.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/physiology , Myogenic Regulatory Factor 5/biosynthesis , Myogenic Regulatory Factor 5/genetics , Trans-Activators/physiology , Animals , Base Sequence , Cells, Cultured , Chick Embryo , Homeodomain Proteins/genetics , Limb Buds , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Myogenic Regulatory Factor 5/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/physiology , Trans-Activators/deficiency , Trans-Activators/genetics
15.
Article in English | MEDLINE | ID: mdl-17395511

ABSTRACT

The cDNA of myogenic factor (Myf5) was isolated from sea perch (Lateolabrax japonicus) using Reverse-transcription Polymerase Chain Reaction (RT-PCR) and rapid amplification of cDNA ends (RACE). The 5' flanking sequence of the cDNA contains a TATA box, GC box, CAAT box, several E box sites and muscle-specific regulatory elements determined by genome walking. The Myf5 gene consists of 3 exons and 2 introns. The open reading frame was found to code a protein with 238 amino-acid residues, containing the conserved basic helix-loop-helix domain (bHLH). RT-PCR indicated the Myf5 was highly expressed in muscle, and weakly expressed in brain, eyes, spleen, gill, liver, kidney, intestine and heart. In early embryonic stages, Myf5 mRNA transcripts are highly detectable in the early gastrula stage while decreasing up to a low level at the late gastrula stage, subsequently greatly increased up to the highest level in the somites stage, then gradually decreases from the tail-bud stage to 15 d larvae after hatching, but they are still detectable. Further, Myf5 mRNA was expressed in several sea perch cell lines such as LJES1, LJHK, LJH-1, LJH-2, LJS, LJL, although its expression level varied greatly among different cell lines.


Subject(s)
Fish Proteins/genetics , Gene Expression Regulation/physiology , Myogenic Regulatory Factor 5/genetics , Perches/genetics , 5' Untranslated Regions/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Exons/genetics , Fish Proteins/biosynthesis , Introns/genetics , Molecular Sequence Data , Myogenic Regulatory Factor 5/biosynthesis , Open Reading Frames/genetics , Organ Specificity/physiology , Perches/metabolism , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Dev Biol ; 290(2): 359-72, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16386728

ABSTRACT

Myf5, one of the basic helix-loop-helix transcription factors, controls muscle differentiation and is expressed in somites during early embryogenesis. However, the transcription factors bound to the cis-elements of myf5 are poorly understood. In this study, we used the yeast one-hybrid assay and found that Forkhead box d3 (Foxd3) interacted specifically with the -82/-62 cassette, a key element directing somite-specific expression of myf5. The dual-luciferase assay revealed that the expression of Foxd3 potently transactivated the myf5 promoter. Knocking down foxd3 with morpholino oligonucleotide (MO) resulted in a dramatic down-regulation of myf5 in somites and adaxial cells but not in the presomitic mesoderm. On the other hand, myod expression remained unchanged in foxd3 morphants. Foxd3 mediation of myf5 expression is stage-dependent, maintaining myf5 expression in the somites and adaxial cells during the 7- to 18-somite stage. Furthermore, in the pax3 morphant, the expression of foxd3 was down-regulated greatly and the expression of myf5 was similar to that of the foxd3 morphant. Co-injection of foxd3 mRNA and pax3-MO1 greatly restored the expression of myf5 in the somites and adaxial cells, suggesting that pax3 induces foxd3 expression, which then induces the expression of myf5. This report is the first study to show that Foxd3, a well-known regulator in neural crest development, is also involved in myf5 regulation.


Subject(s)
Forkhead Transcription Factors/physiology , Gene Expression Regulation, Developmental , Myogenic Regulatory Factor 5/biosynthesis , Somites/metabolism , Zebrafish Proteins/physiology , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cloning, Molecular , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Forkhead Transcription Factors/metabolism , In Situ Hybridization , Luciferases/metabolism , Mesoderm/metabolism , Models, Biological , Neural Crest/embryology , Oligonucleotides/chemistry , PAX3 Transcription Factor , Paired Box Transcription Factors/metabolism , Plasmids/metabolism , Protein Binding , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Time Factors , Transcriptional Activation , Two-Hybrid System Techniques , Zebrafish , Zebrafish Proteins/metabolism
17.
Dev Biol ; 288(1): 73-86, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16248994

ABSTRACT

The p38 MAPK signaling pathway is essential for skeletal muscle differentiation in tissue culture models. We demonstrate a novel role for p38 MAPK in myogenesis during early Xenopus laevis development. Interfering with p38 MAPK causes distinct defects in myogenesis. The initial expression of Myf5 is selectively blocked, while expression of MyoD is unaffected. Expression of a subset of muscle structural genes is reduced. Convergent extension movements are prevented and segmentation of the paraxial mesoderm is delayed, probably due to the failure of cells to withdraw from the cell cycle. Myotubes are properly formed; however, at later stages, they begin to degenerate, and the boundaries between somites disappear. Significant apoptotic cell death occurs in most parts of the somites. The ventral body wall muscle derived from migratory progenitor cells of the ventral somite region is poorly formed. Our data indicate that the developmental defects caused by p38alpha-knockdown were mediated by the loss of XMyf5 expression. Thus, this study identifies a specific intracellular pathway in which p38 MAPK and Myf5 proteins regulate a distinct myogenic program.


Subject(s)
Muscle Development/physiology , Muscle, Skeletal/embryology , Muscle, Skeletal/enzymology , Myogenic Regulatory Factor 5/biosynthesis , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Apoptosis/physiology , Body Patterning/physiology , Cell Cycle/physiology , Muscle, Skeletal/cytology , MyoD Protein/biosynthesis , MyoD Protein/genetics , Myogenic Regulatory Factor 5/genetics , Phenotype , Somites/cytology , Somites/enzymology , Somites/physiology , Xenopus laevis
18.
Dev Growth Differ ; 47(7): 483-92, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16179075

ABSTRACT

To investigate the roles played by MyoD in the terminal differentiation of satellite cell-derived myoblasts, the effect of antisense inhibition of MyoD expression was examined in bovine adult myoblast culture, in which inhibition treatment was limited to the terminal differentiation phase. MyoD antisense oligonucleotide DNA (AS-mD) suppressed the formation of multinucleated myotubes in the cell culture. Myotube formation was suppressed even when AS-mD treatment was limited to the period preceding the onset of myotube formation. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis revealed that treatment with AS-mD suppressed the expression of myosin heavy chain embryonic isoform and troponin T isoforms at 4 days after the induction of differentiation. AS-mD also suppressed the expression of MRF4, but did not alter the expression of either Myf5 or myogenin, in contrast to previous results using mouse cells possessing MyoD(-/-) genetic background. These findings suggest that MyoD controls myogenesis but not Myf5 or myogenin mRNA expression during the terminal differentiation phase. Furthermore, among the alpha4, alpha5, alpha6, and alpha7 integrins, alpha4, alpha5, and alpha7 integrin expression was suppressed by AS-mD treatment, in parallel with the suppression of myotube formation, which suggests that MyoD controls myotube formation by regulating the expression of alpha4, alpha5, and alpha7 integrins.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation/physiology , MyoD Protein/biosynthesis , Myogenic Regulatory Factor 5/biosynthesis , Myogenin/biosynthesis , Satellite Cells, Skeletal Muscle/physiology , Animals , Cattle , Cell Differentiation/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Integrins/biosynthesis , Integrins/genetics , Muscle Development/drug effects , Muscle Development/physiology , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/physiology , MyoD Protein/genetics , Myogenic Regulatory Factor 5/genetics , Myogenin/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Reverse Transcriptase Polymerase Chain Reaction/methods , Satellite Cells, Skeletal Muscle/cytology
19.
Development ; 132(19): 4211-22, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16120642

ABSTRACT

Fibroblast growth factors (Fgfs) have long been implicated in regulating vertebrate skeletal muscle differentiation, but their precise role(s) in vivo remain unclear. Here, we show that Fgf8 signalling in the somite is required for myod expression and terminal differentiation of a subset of fast muscle cells in the zebrafish lateral somite. In the absence of Fgf8, lateral somite cells transiently express myf5 but fail to make muscle and remain in a dermomyotome-like state characterised by pax3 and meox expression. Slow muscle fibres form and commence normal migration in the absence of Fgf8, but fail to traverse the expanded undifferentiated lateral somite. The Fgf8-independent residual population of medial fast muscle fibres is not Hedgehog dependent. However, Fgf8-independent medial fast muscle precursors are lacking in floatinghead mutants, suggesting that they require another ventral midline-derived signal. We conclude that Fgf8 drives terminal differentiation of a specific population of lateral muscle precursor cells within the early somite.


Subject(s)
Fibroblast Growth Factor 8/physiology , Muscle Fibers, Fast-Twitch/physiology , Myoblasts/physiology , Somites/physiology , Zebrafish/physiology , Animals , Body Patterning/physiology , Cell Differentiation , Embryo, Nonmammalian/metabolism , Hedgehog Proteins , Muscle Fibers, Fast-Twitch/cytology , MyoD Protein/biosynthesis , Myoblasts/metabolism , Myogenic Regulatory Factor 5/biosynthesis , Myogenin/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/biosynthesis , Signal Transduction , Trans-Activators/physiology , Zebrafish/embryology , Zebrafish Proteins/biosynthesis
20.
Hum Mol Genet ; 13(22): 2829-40, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15385444

ABSTRACT

Formation and remodeling of the pharyngeal arches play central roles in craniofacial development. TBX1, encoding a T-box-containing transcription factor, is the major candidate gene for del22q11.2 (DiGeorge or velo-cardio-facial) syndrome, characterized by craniofacial defects, thymic hypoplasia, cardiovascular anomalies, velopharyngeal insufficiency and skeletal muscle hypotonia. Tbx1 is expressed in pharyngeal mesoderm, which gives rise to branchiomeric skeletal muscles of the head and neck. Although the genetic control of craniofacial muscle development is known to involve pathways distinct from those operational in the trunk, the regulation of branchiomeric myogenesis has remained enigmatic. Here we show that branchiomeric muscle development is severely perturbed in Tbx1 mutant mice. In the absence of Tbx1, the myogenic determination genes Myf5 and MyoD fail to be normally activated in pharyngeal mesoderm. Unspecified precursor cells expressing genes encoding the transcriptional repressors Capsulin and MyoR are present in the mandibular arch of Tbx1 mutant embryos. Sporadic activation of Myf5 and MyoD in these precursor cells results in the random presence or absence of hypoplastic mandibular arch-derived muscles at later developmental stages. Tbx1 is also required for normal expression of Tlx1 and Fgf10 in pharyngeal mesoderm, in addition to correct neural crest cell patterning in the mandibular arch. Tbx1 therefore regulates the onset of branchiomeric myogenesis and controls normal mandibular arch development, including robust transcriptional activation of myogenic determination genes. While no abnormalities in branchiomeric myogenesis were detected in Tbx1(+/-) mice, reduced TBX1 levels may contribute to pharyngeal hypotonia in del22q11.2 patients.


Subject(s)
Branchial Region/embryology , Mandible/embryology , Muscle Development/genetics , Pharyngeal Muscles/embryology , T-Box Domain Proteins/genetics , Animals , Branchial Region/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Fibroblast Growth Factor 10/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/biosynthesis , Humans , Mandible/metabolism , Mesoderm/metabolism , Mice , Mice, Mutant Strains , Mutation , MyoD Protein/biosynthesis , Myogenic Regulatory Factor 5/biosynthesis , Pharyngeal Muscles/metabolism , T-Box Domain Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...