Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 228
Filter
Add more filters

Publication year range
1.
PLoS Genet ; 20(6): e1010935, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38875306

ABSTRACT

Gene regulatory networks that act upstream of skeletal muscle fate determinants are distinct in different anatomical locations. Despite recent efforts, a clear understanding of the cascade of events underlying the emergence and maintenance of the stem cell pool in specific muscle groups remains unresolved and debated. Here, we invalidated Pitx2 with multiple Cre-driver mice prenatally, postnatally, and during lineage progression. We showed that this gene becomes progressively dispensable for specification and maintenance of the muscle stem (MuSC) cell pool in extraocular muscles (EOMs) despite being, together with Myf5, a major upstream regulator during early development. Moreover, constitutive inactivation of Pax7 postnatally led to a greater loss of MuSCs in the EOMs compared to the limb. Thus, we propose a relay between Pitx2, Myf5 and Pax7 for EOM stem cell maintenance. We demonstrate also that MuSCs in the EOMs adopt a quiescent state earlier that those in limb muscles and do not spontaneously proliferate in the adult, yet EOMs have a significantly higher content of Pax7+ MuSCs per area pre- and post-natally. Finally, while limb MuSCs proliferate in the mdx mouse model for Duchenne muscular dystrophy, significantly less MuSCs were present in the EOMs of the mdx mouse model compared to controls, and they were not proliferative. Overall, our study provides a comprehensive in vivo characterisation of MuSC heterogeneity along the body axis and brings further insights into the unusual sparing of EOMs during muscular dystrophy.


Subject(s)
Homeobox Protein PITX2 , Homeodomain Proteins , Myogenic Regulatory Factor 5 , Oculomotor Muscles , PAX7 Transcription Factor , Transcription Factors , Animals , Humans , Mice , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice, Inbred mdx , Muscle Development/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/growth & development , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Oculomotor Muscles/metabolism , PAX7 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
J Cell Sci ; 135(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35099008

ABSTRACT

Muscle stem (satellite) cells express Pax7, a key transcription factor essential for satellite cell maintenance and adult muscle regeneration. We identify the corepressor transducin-like enhancer of split-4 (TLE4) as a Pax7 interaction partner expressed in quiescent satellite cells under homeostasis. A subset of satellite cells transiently downregulate TLE4 during early time points following muscle injury. We identify these to be activated satellite cells, and that TLE4 downregulation is required for Myf5 activation and myogenic commitment. Our results indicate that TLE4 represses Pax7-mediated Myf5 transcriptional activation by occupying the -111 kb Myf5 enhancer to maintain quiescence. Loss of TLE4 function causes Myf5 upregulation, an increase in satellite cell numbers and altered differentiation dynamics during regeneration. Thus, we have uncovered a novel mechanism to maintain satellite cell quiescence and regulate muscle differentiation mediated by the corepressor TLE4.


Subject(s)
Cell Differentiation , Muscle Development , Muscle, Skeletal , Nuclear Proteins , Repressor Proteins , Cell Differentiation/genetics , Humans , Muscle Development/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/injuries , Muscular Diseases/physiopathology , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , PAX7 Transcription Factor/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Satellite Cells, Skeletal Muscle/cytology
3.
Int J Mol Sci ; 25(8)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38673893

ABSTRACT

During embryogenesis, basic fibroblast growth factor (bFGF) is released from neural tube and myotome to promote myogenic fate in the somite, and is routinely used for the culture of adult skeletal muscle (SKM) stem cells (MuSC, called satellite cells). However, the mechanism employed by bFGF to promote SKM lineage and MuSC proliferation has not been analyzed in detail. Furthermore, the question of if the post-translational modification (PTM) of bFGF is important to its stemness-promoting effect has not been answered. In this study, GST-bFGF was expressed and purified from E.coli, which lacks the PTM system in eukaryotes. We found that both GST-bFGF and commercially available bFGF activated the Akt-Erk pathway and had strong cell proliferation effect on C2C12 myoblasts and MuSC. GST-bFGF reversibly compromised the myogenesis of C2C12 myoblasts and MuSC, and it increased the expression of Myf5, Pax3/7, and Cyclin D1 but strongly repressed that of MyoD, suggesting the maintenance of myogenic stemness amid repressed MyoD expression. The proliferation effect of GST-bFGF was conserved in C2C12 over-expressed with MyoD (C2C12-tTA-MyoD), implying its independence of the down-regulation of MyoD. In addition, the repressive effect of GST-bFGF on myogenic differentiation was almost totally rescued by the over-expression of MyoD. Together, these evidences suggest that (1) GST-bFGF and bFGF have similar effects on myogenic cell proliferation and differentiation, and (2) GST-bFGF can promote MuSC stemness and proliferation by differentially regulating MRFs and Pax3/7, (3) MyoD repression by GST-bFGF is reversible and independent of the proliferation effect, and (4) GST-bFGF can be a good substitute for bFGF in sustaining MuSC stemness and proliferation.


Subject(s)
Cell Proliferation , Fibroblast Growth Factor 2 , Muscle Development , MyoD Protein , Myoblasts , Muscle Development/genetics , Animals , Mice , MyoD Protein/metabolism , MyoD Protein/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/genetics , Myoblasts/metabolism , Myoblasts/cytology , Cell Line , PAX7 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , PAX3 Transcription Factor/metabolism , PAX3 Transcription Factor/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factor 5/genetics , Cyclin D1/metabolism , Cyclin D1/genetics , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/cytology , Cell Differentiation , Proto-Oncogene Proteins c-akt/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology
4.
Dev Biol ; 490: 134-143, 2022 10.
Article in English | MEDLINE | ID: mdl-35917935

ABSTRACT

The vertebrate embryonic midline vasculature forms in close proximity to the developing skeletal muscle, which originates in the somites. Angioblasts migrate from bilateral positions along the ventral edge of the somites until they meet at the midline, where they sort and differentiate into the dorsal aorta and the cardinal vein. This migration occurs at the same time that myoblasts in the somites are beginning to differentiate into skeletal muscle, a process which requires the activity of the basic helix loop helix (bHLH) transcription factors Myod and Myf5. Here we examined vasculature formation in myod and myf5 mutant zebrafish. In the absence of skeletal myogenesis, angioblasts migrate normally to the midline but form only the cardinal vein and not the dorsal aorta. The phenotype is due to the failure to activate vascular endothelial growth factor ligand vegfaa expression in the somites, which in turn is required in the adjacent angioblasts for dorsal aorta specification. Myod and Myf5 cooperate with Hedgehog signaling to activate and later maintain vegfaa expression in the medial somites, which is required for angiogenic sprouting from the dorsal aorta. Our work reveals that the early embryonic skeletal musculature in teleosts evolved to organize the midline vasculature during development.


Subject(s)
MyoD Protein , Myogenic Regulatory Factors , Animals , Aorta/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Muscle Proteins/genetics , Muscle, Skeletal , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/genetics , Zebrafish/metabolism
6.
Dev Dyn ; 251(10): 1698-1710, 2022 10.
Article in English | MEDLINE | ID: mdl-35618666

ABSTRACT

BACKGROUND: The turtle carapace is an evolutionary novelty resulting from changes in the processes that build ribs and their associated muscles in most tetrapod species. Turtle embryos have several unique features that might play a role in this process, including the carapacial ridge, a Myf5 gene with shorter coding region that generates an alternative splice variant lacking exon 2, and unusual expression patterns of Lbx1 and HGF. RESULTS: We investigated these turtle-specific expression differences using genetic approaches in mouse embryos. At mid-gestation, mouse embryos producing Myf5 transcripts lacking exon 2 replicated some early properties of turtle somites, but still developed into viable and fertile mice. Extending Lbx1 expression into the hypaxial dermomyotomal lip of trunk somites to mimic the turtle Lbx1 expression pattern, produced fusions in the distal part of the ribs. CONCLUSIONS: Turtle-like Myf5 activity might generate a plastic state in developing trunk somites under which they can either enter carapace morphogenetic routes, possibly triggered by signals from the carapacial ridge, or still engage in the development of a standard tetrapod ribcage in the absence of those signals. In addition, trunk Lbx1 expression might play a later role in the formation of the lateral border of the carapace.


Subject(s)
Turtles , Animal Shells , Animals , Biological Evolution , Mice , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Plastics/metabolism , Somites , Turtles/genetics
7.
Proc Biol Sci ; 289(1981): 20220841, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35975445

ABSTRACT

Developmental pathways encompass transcription factors and cis-regulatory elements that interact as transcription factor-regulatory element (TF-RE) units. Independent origins of similar phenotypes likely involve changes in different parts of these units, a hypothesis promisingly tested addressing the evolution of the rib-associated lumbar (RAL) morphotype that characterizes emblematic animals such as snakes and elephants. Previous investigation in these lineages identified a polymorphism in the Homology region 1 [H1] enhancer of the Myogenic factor-5 [Myf5], which interacts with HOX10 proteins to modulate rib development. Here we address the evolution of TF-RE units focusing on independent origins of RAL morphotypes. We compiled an extensive database for H1-Myf5 and HOX10 sequences with two goals: (i) evaluate if the enhancer polymorphism is present in amphibians exhibiting the RAL morphotype and (ii) test a hypothesis of enhanced evolutionary flexibility mediated by TF-RE units, according to which independent origins of the RAL morphotype might involve changes in either component of the interaction unit. We identified the H1-Myf5 polymorphism in lineages that diverged around 340 Ma, including Lissamphibia. Independent origins of the RAL morphotype in Tetrapoda involved sequence variation in either component of the TF-RE unit, confirming that different changes may similarly affect the phenotypic outcome of a given developmental pathway.


Subject(s)
Regulatory Sequences, Nucleic Acid , Transcription Factors , Amphibians/metabolism , Animals , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Snakes/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
8.
PLoS Biol ; 17(2): e3000153, 2019 02.
Article in English | MEDLINE | ID: mdl-30807574

ABSTRACT

The transcriptional mechanisms driving lineage specification during development are still largely unknown, as the interplay of multiple transcription factors makes it difficult to dissect these molecular events. Using a cell-based differentiation platform to probe transcription function, we investigated the role of the key paraxial mesoderm and skeletal myogenic commitment factors-mesogenin 1 (Msgn1), T-box 6 (Tbx6), forkhead box C1 (Foxc1), paired box 3 (Pax3), Paraxis, mesenchyme homeobox 1 (Meox1), sine oculis-related homeobox 1 (Six1), and myogenic factor 5 (Myf5)-in paraxial mesoderm and skeletal myogenesis. From this study, we define a genetic hierarchy, with Pax3 emerging as the gatekeeper between the presomitic mesoderm and the myogenic lineage. By assaying chromatin accessibility, genomic binding and transcription profiling in mesodermal cells from mouse and human Pax3-induced embryonic stem cells and Pax3-null embryonic day (E)9.5 mouse embryos, we identified conserved Pax3 functions in the activation of the skeletal myogenic lineage through modulation of Hedgehog, Notch, and bone morphogenetic protein (BMP) signaling pathways. In addition, we demonstrate that Pax3 molecular function involves chromatin remodeling of its bound elements through an increase in chromatin accessibility and cooperation with sine oculis-related homeobox 4 (Six4) and TEA domain family member 2 (Tead2) factors. To our knowledge, these data provide the first integrated analysis of Pax3 function, demonstrating its ability to remodel chromatin in mesodermal cells from developing embryos and proving a mechanistic footing for the transcriptional hierarchy driving myogenesis.


Subject(s)
Chromatin Assembly and Disassembly , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Mesoderm/metabolism , Muscle Cells/metabolism , Muscle Development/genetics , PAX3 Transcription Factor/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Line , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Humans , Mesoderm/cytology , Mesoderm/growth & development , Mice , Mice, Transgenic , Muscle Cells/cytology , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , PAX3 Transcription Factor/metabolism , Signal Transduction , T-Box Domain Proteins , TEA Domain Transcription Factors , Trans-Activators/metabolism , Transcription Factors/metabolism
9.
Cell Mol Life Sci ; 78(5): 2157-2167, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32875355

ABSTRACT

Inherited fatty acid oxidation diseases in their mild forms often present as metabolic myopathies. Carnitine Palmitoyl Transferase 2 (CPT2) deficiency, one such prototypical disorder is associated with compromised myotube differentiation. Here, we show that CPT2-deficient myotubes exhibit defects in focal adhesions and redox balance, exemplified by increased SOD2 expression. We document unprecedented alterations in the cellular prion protein PrPC, which directly arise from the failure in CPT2 enzymatic activity. We also demonstrate that the loss of PrPC function in normal myotubes recapitulates the defects in focal adhesion, redox balance and differentiation hallmarks monitored in CPT2-deficient cells. These results are further corroborated by studies performed in muscles from Prnp-/- mice. Altogether, our results unveil a molecular scenario, whereby PrPC dysfunction governed by faulty CPT2 activity may drive aberrant focal adhesion turnover and hinder proper myotube differentiation. Our study adds a novel facet to the involvement of PrPC in diverse physiopathological situations.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , Focal Adhesions/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Diseases/genetics , Prion Proteins/genetics , Animals , Carnitine O-Palmitoyltransferase/deficiency , Cells, Cultured , Focal Adhesions/metabolism , Humans , Mice, Knockout , Muscle Fibers, Skeletal/cytology , Muscular Diseases/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Oxidation-Reduction , Prion Diseases/genetics , Prion Diseases/metabolism , Prion Proteins/deficiency , RNA Interference , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
10.
Anim Biotechnol ; 33(3): 401-407, 2022 Jun.
Article in English | MEDLINE | ID: mdl-32749185

ABSTRACT

This study was conducted to ivnestigate the associations of GH-AluI, STAT5A-AvaI and MYF5-TaqI gene polymorphisms with milk somatic cell count (SCC), electrical conductivity (EC) and pH levels in Holstein dairy cows. For this purpose, 167 blood and 1670 milk samples of 167 Holstein cows in their 2nd lactation were used. There were significant relationships between GH-AluI genotypes and milk EC (p < 0.001) and between STAT5A-AvaI genotypes and milk EC (p = 0.007), but there were not any significant relationships between MYF5 gene polymorphism and the investigated traits (p > 0.05). The greatest EC values were observed in GH-AluI-LV and STAT5A-AvaI-TT-genotyped individuals. Just because of association of EC with mastitis, it was concluded that present GH-AluI and STAT5A-AvaI polymorphisms could be used in further studies to be conducted to improve mastitis resistance and milk quality traits of Holstein dairy cows.


Subject(s)
Cattle Diseases , Growth Hormone/genetics , Mastitis, Bovine , Myogenic Regulatory Factor 5/genetics , STAT5 Transcription Factor/genetics , Animals , Cattle/genetics , Cell Count , Electric Conductivity , Female , Hydrogen-Ion Concentration , Lactation/genetics , Milk , Polymorphism, Genetic/genetics
11.
Dev Biol ; 468(1-2): 133-145, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32768399

ABSTRACT

The importance of skeletal muscle for rib development and patterning in the mouse embryo has not been resolved, largely because different experimental approaches have yielded disparate results. In this study, we utilize both gene knockouts and muscle cell ablation approaches to re-visit the extent to which rib growth and patterning are dependent on developing musculature. Consistent with previous studies, we show that rib formation is highly dependent on the MYOD family of myogenic regulatory factors (MRFs), and demonstrate that the extent of rib formation is gene-, allele-, and dosage-dependent. In the absence of Myf5 and MyoD, one allele of Mrf4 is sufficient for extensive rib growth, although patterning is abnormal. Under conditions of limiting MRF dosage, MyoD is identified as a positive regulator of rib patterning, presumably due to improved intercostal muscle development. In contrast to previous muscle ablation studies, we show that diphtheria toxin subunit A (DTA)-mediated ablation of muscle progenitors or differentiated muscle, using MyoDiCre or HSA-Cre drivers, respectively, profoundly disrupts rib development. Further, a comparison of three independently derived Rosa26-based DTA knockin alleles demonstrates that the degree of rib perturbations in MyoDiCre/+/DTA embryos is markedly dependent on the DTA allele used, and may in part explain discrepancies with previous findings. The results support the conclusion that the extent and quality of rib formation is largely dependent on the dosage of Myf5 and Mrf4, and that both early myotome-sclerotome interactions, as well as later muscle-rib interactions, are important for proper rib growth and patterning.


Subject(s)
Body Patterning , Muscle, Skeletal/embryology , Ribs/embryology , Alleles , Animals , Gonadotropin-Releasing Hormone/analogs & derivatives , Mice, Transgenic , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism
12.
Am J Hum Genet ; 103(1): 115-124, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29887215

ABSTRACT

MYF5 is member of the Myc-like basic helix-loop-helix transcription factor family and, in cooperation with other myogenic regulatory factors MYOD and MYF5, is a key regulator of early stages of myogenesis. Here, we report three consanguineous families with biallelic homozygous loss-of-function mutations in MYF5 who define a clinical disorder characterized by congenital ophthalmoplegia with scoliosis and vertebral and rib anomalies. The clinical phenotype overlaps strikingly with that reported in several Myf5 knockout mouse models. Affected members of two families share a haploidentical region that contains a homozygous 10 bp frameshift mutation in exon 1 of MYF5 (c.23_32delAGTTCTCACC [p.Gln8Leufs∗86]) predicted to undergo nonsense-mediated decay. Affected members of the third family harbor a homozygous missense change in exon 1 of MYF5 (c.283C>T [p.Arg95Cys]). Using in vitro assays, we show that this missense mutation acts as a loss-of-function allele by impairing MYF5 DNA binding and nuclear localization. We performed whole-genome sequencing in one affected individual with the frameshift mutation and did not identify additional rare variants in the haploidentical region that might account for differences in severity among the families. These data support the direct role of MYF5 in rib, spine, and extraocular muscle formation in humans.


Subject(s)
Mutation/genetics , Myogenic Regulatory Factor 5/genetics , Ophthalmoplegia/genetics , Ribs/abnormalities , Spine/abnormalities , Alleles , Amino Acid Sequence , Anal Canal/abnormalities , Animals , DNA-Binding Proteins/genetics , Esophagus/abnormalities , Exons/genetics , Female , Heart Defects, Congenital , Humans , Kidney/abnormalities , Limb Deformities, Congenital , Male , Mice, Knockout , MyoD Protein/genetics , Phenotype , Sequence Alignment , Trachea/abnormalities , Whole Genome Sequencing/methods
13.
Anim Biotechnol ; 32(1): 121-129, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32564663

ABSTRACT

This study determined the association between Myf5, CAST and MSTN genes and fattening performance traits in 228 (Holstein = 103 and Brown = 125) cattle breeds. Classification and regression tree (CART) was used to determine association between genes and fattening performance. The allele frequencies of Holstein cattle in terms of Myf5, CAST and MSTN genes were A:0.30, B:0.70; A:0.43, G:0.56; A:0.97, B:0.03, respectively, whereas in the Brown Swiss cattle were A:0.36, B:0.64; A:0.65, G:0.34; A:0.88, B:0.12 respectively. The Myf5, CAST, and MSTN genes were found to be in the Hardy-Weinberg equilibrium (p > 0.05) in both of the cattle breeds. As a result, the association between Myf5, CAST and MSTN genes and fattening performance traits were found to be significant according to CART analysis.


Subject(s)
Calcium-Binding Proteins/genetics , Cattle/genetics , Myogenic Regulatory Factor 5/genetics , Myostatin/genetics , Polymorphism, Genetic/genetics , Animals , Fats/metabolism , Gene Frequency/genetics , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length
14.
Genomics ; 112(4): 2804-2812, 2020 07.
Article in English | MEDLINE | ID: mdl-32220486

ABSTRACT

Myogenic factor 5 plays actively roles in the regulation of myogenesis. The aims of this study are to identify the evolution information of MYF5 protein among 10 domestic and mammalian animals, to uncover the expression patterns of MYF5 gene in calves and adults of Qinchuan cattle, and to expose the genetic variants of the MYF5 gene and explore its effect on cattle growth traits and beef quality traits in Qinchuan cattle. The bioinformatics results showed that the MYF5 proteins highly conserved in different mammalian or domestic animals apart from chicken. The expression level of MYF5 gene in the heart, muscle, lung, large intestine and liver was greater than that of other tissues. PCR amplicons sequencing identified four novel SNPs at g.5738A>G, g.5785C>T and g.5816A>G in the 3rd exon region and g.6535A>G in the 3' UTR. Genotypic frequencies of g.5785C>T was harshly deviated from the HWE (P < .05). Genetic diversity was low or intermediate for the four SNPs and those SNPs were in the weak linkage disequilibrium. Association analysis results indicated g.5785C>T, g.5816A>G and g.6535A>G significant effect on growth performance and beef quality traits of Qinchuan cattle. H1H3 diplotype had greater body size and better beef quality. All the results implicate that the MYF5 gene might be applied as a promising candidate gene in Qinchuan cattle breeding.


Subject(s)
Cattle/genetics , Meat , Myogenic Regulatory Factor 5/genetics , Amino Acid Motifs , Animals , Cattle/growth & development , Cattle/metabolism , Female , Genotype , Haplotypes , Linkage Disequilibrium , Myogenic Regulatory Factor 5/chemistry , Myogenic Regulatory Factor 5/classification , Myogenic Regulatory Factor 5/metabolism , Phylogeny , Polymorphism, Single Nucleotide , Protein Domains , RNA, Messenger/metabolism , Sequence Alignment
15.
Int J Mol Sci ; 22(17)2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34502190

ABSTRACT

Inductors of myogenic stem cell differentiation attract attention, as they can be used to treat myodystrophies and post-traumatic injuries. Functionalization of fullerenes makes it possible to obtain water-soluble derivatives with targeted biochemical activity. This study examined the effects of the phosphonate C60 fullerene derivatives on the expression of myogenic transcription factors and myogenic differentiation of human mesenchymal stem cells (MSCs). Uptake of the phosphonate C60 fullerene derivatives in human MSCs, intracellular ROS visualization, superoxide scavenging potential, and the expression of myogenic, adipogenic, and osteogenic differentiation genes were studied. The prolonged MSC incubation (within 7-14 days) with the C60 pentaphoshonate potassium salt promoted their differentiation towards the myogenic lineage. The transcription factors and gene expressions determining myogenic differentiation (MYOD1, MYOG, MYF5, and MRF4) increased, while the expression of osteogenic differentiation factors (BMP2, BMP4, RUNX2, SPP1, and OCN) and adipogenic differentiation factors (CEBPB, LPL, and AP2 (FABP4)) was reduced or did not change. The stimulation of autophagy may be one of the factors contributing to the increased expression of myogenic differentiation genes in MSCs. Autophagy may be caused by intracellular alkalosis and/or short-term intracellular oxidative stress.


Subject(s)
Fullerenes/pharmacology , Mesenchymal Stem Cells/drug effects , Muscle Development , Autophagy , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , MyoD Protein/genetics , Myogenic Regulatory Factor 5/genetics , Myogenin/genetics , Reactive Oxygen Species/metabolism
16.
Cell Mol Life Sci ; 76(4): 777-789, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30448891

ABSTRACT

Thoracic aorta perivascular adipose tissue (T-PVAT) has critical roles in regulating vascular homeostasis. However, the developmental characteristics and cellular lineage of adipocyte in the T-PVAT remain unclear. We show that T-PVAT contains three long strip-shaped fat depots, anterior T-PVAT (A-T-PVAT), left lateral T-PVAT (LL-T-PVAT), and right lateral T-PVAT (RL-T-PVAT). A-T-PVAT displays a distinct transcriptional profile and developmental origin compared to the two lateral T-PVATs (L-T-PVAT). Lineage tracing studies indicate that A-T-PVAT adipocytes are primarily derived from SM22α+ progenitors, whereas L-T-PVAT contains both SM22α+ and Myf5+ cells. We also show that L-T-PVAT contains more UCP1+ brown adipocytes than A-T-PVAT, and L-T-PVAT exerts a greater relaxing effect on aorta than A-T-PVAT. Angiotensin II-infused hypertensive mice display greater macrophage infiltration into A-T-PVAT than L-T-PVAT. These combined results indicate that L-T-PVAT has a distinct development from A-T-PVAT with different cellular lineage, and suggest that L-T-PVAT and A-T-PVAT have different physiological and pathological functions.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Aorta, Thoracic/metabolism , Gene Expression Profiling/methods , Adipose Tissue/cytology , Adipose Tissue/growth & development , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Gene Ontology , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Stem Cells/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
17.
Int J Mol Sci ; 21(6)2020 Mar 19.
Article in English | MEDLINE | ID: mdl-32204424

ABSTRACT

Skeletal muscle injuries in competitive sports cause lengthy absences of athletes from tournaments. This is of tremendous competitive and economic relevance for both the athletes and their respective clubs. Therapy for structural muscle lesions aims to promote regeneration and fast-track return-to-play. A common clinical treatment strategy for muscle injuries is the intramuscular injection of calf blood compound and the homeopathic drug, Tr14. Although the combination of these two agents was reported to reduce recovery time, the regulatory mechanism whereby this occurs remains unknown. In this in vivo study, we selected a rat model of mechanical muscle injury to investigate the effect of this combination therapy on muscle regeneration. Gene expression analysis and histological images revealed that this combined intramuscular injection for muscle lesions can enhance the expression of pro-myogenic genes and proteins and accelerate muscle regeneration. These findings are novel and depict the positive effects of calf blood compound and the homeopathic drug, Tr14, which are utilized in the field of Sports medicine.


Subject(s)
Heme/analogs & derivatives , Minerals/pharmacology , Muscle, Skeletal/drug effects , Plant Extracts/pharmacology , Regeneration/drug effects , Animals , Athletic Injuries/physiopathology , Athletic Injuries/prevention & control , Gene Expression/drug effects , Heme/administration & dosage , Heme/pharmacology , Homeopathy , Humans , Injections, Intramuscular , Male , Minerals/administration & dosage , Models, Animal , Muscle, Skeletal/injuries , Muscle, Skeletal/physiopathology , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Plant Extracts/administration & dosage , Rats, Wistar , Regeneration/genetics , Wound Healing/drug effects , Wound Healing/physiology
18.
Semin Cell Dev Biol ; 72: 19-32, 2017 12.
Article in English | MEDLINE | ID: mdl-29127046

ABSTRACT

Discovery of the myogenic regulatory factor family of transcription factors MYF5, MYOD, Myogenin and MRF4 was a seminal step in understanding specification of the skeletal muscle lineage and control of myogenic differentiation during development. These factors are also involved in specification of the muscle satellite cell lineage, which becomes the resident stem cell compartment inadult skeletal muscle. While MYF5, MYOD, Myogenin and MRF4 have subtle roles in mature muscle, they again play a crucial role in directing satellite cell function to regenerate skeletal muscle: linking the genetic control of developmental and regenerative myogenesis. Here, I review the role of the myogenic regulatory factors in developing and mature skeletal muscle, satellite cell specification and muscle regeneration.


Subject(s)
Muscle, Skeletal/metabolism , MyoD Protein/genetics , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factors/genetics , Myogenin/genetics , Satellite Cells, Skeletal Muscle/metabolism , Animals , Gene Expression Regulation, Developmental , Muscle Development/genetics , Muscle, Skeletal/embryology , Muscle, Skeletal/growth & development , Regeneration/genetics
19.
Int J Sports Med ; 40(12): 803-809, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31476781

ABSTRACT

The use of injections to treat structural muscle injuries is controversially discussed. In our controlled in vitro study, we investigated the biological impact of Actovegin and Traumeel alone and in combination on primary human skeletal muscle cells. Cells were characterized by immunofluorescence staining for myogenic factor 5 (Myf5) and MyoD, and cultured with or without Actovegin and / or Traumeel. The effects of these agents were assayed by cell viability and gene expression of the specific markers MyoD, Myf5, neural adhesion molecule (NCAM), and CD31. Myotube formation was determined by myosin staining. Neither Actovegin nor Traumeel showed toxic effects or influenced cell viability significantly. High volumes of Actovegin down-regulated gene expression of NCAM after 3 days but had no effect on MyoD, Myf5, and CD31 gene expression. High volumes of Traumeel inhibited MyoD gene expression after 3 days, whereas after 7 days MyoD expression was significantly up-regulated. The combination of both agents did not significantly influence cell viability or gene expression. This is the first study demonstrating that Actovegin and Traumeel potentially modulate human skeletal muscle cells. The relevance of these in vitro findings has to be highlighted in further in vivo studies.


Subject(s)
Cell Differentiation/drug effects , Heme/analogs & derivatives , Minerals/pharmacology , Muscle Fibers, Skeletal/physiology , Plant Extracts/pharmacology , Adult , Aged , CD56 Antigen/drug effects , CD56 Antigen/genetics , Cell Survival , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , Heme/pharmacology , Humans , Male , Middle Aged , MyoD Protein/drug effects , MyoD Protein/genetics , Myogenic Regulatory Factor 5/drug effects , Myogenic Regulatory Factor 5/genetics , Platelet Endothelial Cell Adhesion Molecule-1/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/genetics
20.
J Cell Sci ; 129(22): 4305-4316, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27672022

ABSTRACT

Although the mechanism underlying modulation of transcription factors in myogenesis has been well elucidated, the function of the transcription cofactors involved in this process remains poorly understood. Here, we identified HMGB2 as an essential nuclear transcriptional co-regulator in myogenesis. HMGB2 was highly expressed in undifferentiated myoblasts and regenerating muscle. Knockdown of HMGB2 inhibited myoblast proliferation and stimulated its differentiation. HMGB2 depletion downregulated Myf5 and cyclin A2 at the protein but not mRNA level. In contrast, overexpression of HMGB2 promoted Myf5 and cyclin A2 protein upregulation. Furthermore, we found that the RNA-binding protein IGF2BP2 is a downstream target of HMGB2, as previously shown for HMGA2. IGF2BP2 binds to mRNAs of Myf5 or cyclin A2, resulting in translation enhancement or mRNA stabilization, respectively. Notably, overexpression of IGF2BP2 could partially rescue protein levels of Myf5 and cyclin A2, in response to HMGB2 decrease. Moreover, depletion of HMGB2 in vivo severely attenuated muscle repair; this was due to a decrease in satellite cells. Taken together, these results highlight the previously undiscovered and crucial role of the HMGB2-IGF2BP2 axis in myogenesis and muscle regeneration.


Subject(s)
HMGB2 Protein/metabolism , Muscle, Skeletal/metabolism , RNA-Binding Proteins/metabolism , Regeneration , Satellite Cells, Skeletal Muscle/metabolism , Animals , Animals, Newborn , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line , Cell Nucleus/metabolism , Cell Proliferation , Cyclin A2/genetics , Cyclin A2/metabolism , Male , Mice, Inbred C57BL , Muscle Development , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL