Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.502
Filter
1.
Anim Biotechnol ; 35(1): 2351973, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38753962

ABSTRACT

Vitamin A is an essential nutrient in animals, playing important roles in animal health. In the pig industry, proper supplementation of vitamin A in the feed can improve pork production performance, while deficiency or excessive intake can lead to growth retardation or disease. However, the specific molecular mechanisms through which vitamin A operates on pig skeletal muscle growth as well as muscle stem cell function remain unexplored. Therefore, in this study, we isolated the pig primary skeletal muscle stem cells (pMuSCs) and treated with retinoic acid (RA), the natural metabolite of vitamin A, and then examined the myogenic capacity of pMuSCs via immunostaining, real-time PCR, CCK8 and western-blot analysis. Unexpectedly, the RA caused a significant decrease in the proliferation and differentiation of pMuSCs. Mechanistically, the RA addition induced the activation of retinoic acid receptor gamma (RARγ), which inhibited the myogenesis through the blockage of protein translation of the master myogenic regulator myogenic differentiation 1 gene (MYOD). Specifically, RARγ inactivate AKT kinase (AKT) signalling and lead to dephosphorylation of eukaryotic translation initiation factor 4E binding protein 1 (eIF4EBP1), which in turn repress the eukaryotic translation initiation factor 4E (eIF4E) complex and block mRNA translation of MYOD. Inhibition of AKT could rescue the myogenic defects of RA-treated pMuSCs. Our findings revealed that retinoid acid signalling inhibits the skeletal muscle stem cell proliferation and differentiation in pigs. Therefore, the vitamin A supplement in the feedstuff should be cautiously optimized to avoid the potential adverse consequences on muscle development associated with the excessive levels of retinoic acid.


Subject(s)
Cell Differentiation , Muscle Development , MyoD Protein , Signal Transduction , Tretinoin , Animals , Tretinoin/pharmacology , Swine , Muscle Development/drug effects , Signal Transduction/drug effects , MyoD Protein/genetics , MyoD Protein/metabolism , Cell Differentiation/drug effects , Muscle, Skeletal/drug effects , Receptors, Retinoic Acid/metabolism , Receptors, Retinoic Acid/genetics , Cell Proliferation/drug effects , Protein Biosynthesis/drug effects , Cells, Cultured
2.
Cell Cycle ; 23(5): 573-587, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38701194

ABSTRACT

Myogenic differentiation (MyoD) 1, which is known as a pivotal transcription factor during myogenesis, has been proven dysregulated in several cancers. However, litter is known about the precise role and downstream genes of MyoD1 in gastric cancer (GC) cells. Here, we report that MyoD1 is lowly expressed in primary GC tissues and cells. In our experiments, overexpression of MyoD1 inhibited cell proliferation. Downstream genes of MyoD1 regulation were investigated using RNA-Seq. As a result, 138 up-regulated genes and 20 down-regulated genes and 27 up-regulated lncRNAs and 20 down-regulated lncRNAs were identified in MyoD1 overexpressed MKN-45 cells, which participated in epithelial cell signaling in Helicobacter pylori infection, glycosaminoglycan biosynthesis (keratan sulfate), notch signaling pathway, and others. Among these genes, BIK was directly regulated by MyoD1 in GC cells and inhibited cancer cell proliferation. The BIK knockdown rescued the effects of MyoD1 overexpression on GC cells. In conclusion, MyoD1 inhibited cell proliferation via 158 genes and 47 lncRNAs downstream directly or indirectly that participated in multiple signaling pathways in GC, and among these, MyoD1 promotes BIK transcription by binding to its promoter, then promotes BIK-Bcl2-caspase 3 axis and regulates GC cell apoptosis.


Subject(s)
Apoptosis , Cell Proliferation , Gene Expression Regulation, Neoplastic , MyoD Protein , RNA, Long Noncoding , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Humans , Apoptosis/genetics , MyoD Protein/metabolism , MyoD Protein/genetics , Cell Proliferation/genetics , Cell Line, Tumor , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Transcription, Genetic/genetics
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.
Sci Rep ; 14(1): 8500, 2024 04 11.
Article in English | MEDLINE | ID: mdl-38605102

ABSTRACT

Intrauterine growth restriction (IUGR) occurs both in humans and domestic species. It has a particularly high incidence in pigs, and is a leading cause of neonatal morbidity and mortality as well as impaired postnatal growth. A key feature of IUGR is impaired muscle development, resulting in decreased meat quality. Understanding the developmental origins of IUGR, particularly at the molecular level, is important for developing effective strategies to mitigate its economic impact on the pig industry and animal welfare. The aim of this study was to characterise transcriptional profiles in the muscle of growth restricted pig foetuses at different gestational days (GD; gestational length ~ 115 days), focusing on selected genes (related to development, tissue injury and metabolism) that were previously identified as dysregulated in muscle of GD90 fetuses. Muscle samples were collected from the lightest foetus (L) and the sex-matched foetus with weight closest to the litter average (AW) from each of 22 Landrace x Large White litters corresponding to GD45 (n = 6), GD60 (n = 8) or GD90 (n = 8), followed by analyses, using RT-PCR and protein immunohistochemistry, of selected gene targets. Expression of the developmental genes, MYOD, RET and ACTN3 were markedly lower, whereas MSTN expression was higher, in the muscle of L relative to AW littermates beginning on GD45. Levels of all tissue injury-associated transcripts analysed (F5, PLG, KNG1, SELL, CCL16) were increased in L muscle on GD60 and, most prominently, on GD90. Among genes involved in metabolic regulation, KLB was expressed at higher levels in L than AW littermates beginning on GD60, whereas both IGFBP1 and AHSG were higher in L littermates on GD90 but only in males. Furthermore, the expression of genes specifically involved in lipid, hexose sugar or iron metabolism increased or, in the case of UCP3, decreased in L littermates on GD60 (UCP3, APOB, ALDOB) or GD90 (PNPLA3, TF), albeit in the case of ALDOB this only involved females. In conclusion, marked dysregulation of genes with critical roles in development in L foetuses can be observed from GD45, whereas for a majority of transcripts associated with tissue injury and metabolism differences between L and AW foetuses were apparent by GD60 or only at GD90, thus identifying different developmental windows for different types of adaptive responses to IUGR in the muscle of porcine foetuses.


Subject(s)
Fetal Development , Fetal Growth Retardation , Muscle, Skeletal , Swine , Humans , Animals , Male , Female , Swine/genetics , Swine/physiology , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Muscle, Skeletal/metabolism , Gene Expression Regulation, Developmental , Fetal Development/genetics , Transcriptome , Gestational Age , Real-Time Polymerase Chain Reaction , Immunohistochemistry , Fetus/metabolism , Genes, Developmental , MyoD Protein/genetics , MyoD Protein/metabolism , Actinin/genetics , Actinin/metabolism
5.
Nutrients ; 16(2)2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38276564

ABSTRACT

Epicatechin is a polyphenol compound that promotes skeletal muscle differentiation and counteracts the pathways that participate in the degradation of proteins. Several studies present contradictory results of treatment protocols and therapeutic effects. Therefore, the objective of this systematic review was to investigate the current literature showing the molecular mechanism and clinical protocol of epicatechin in muscle atrophy in humans, animals, and myoblast cell-line. The search was conducted in Embase, PubMed/MEDLINE, Cochrane Library, and Web of Science. The qualitative analysis demonstrated that there is a commonness of epicatechin inhibitory action in myostatin expression and atrogenes MAFbx, FOXO, and MuRF1. Epicatechin showed positive effects on follistatin and on the stimulation of factors related to the myogenic actions (MyoD, Myf5, and myogenin). Furthermore, the literature also showed that epicatechin can interfere with mitochondrias' biosynthesis in muscle fibers, stimulation of the signaling pathways of AKT/mTOR protein production, and amelioration of skeletal musculature performance, particularly when combined with physical exercise. Epicatechin can, for these reasons, exhibit clinical applicability due to the beneficial results under conditions that negatively affect the skeletal musculature. However, there is no protocol standardization or enough clinical evidence to draw more specific conclusions on its therapeutic implementation.


Subject(s)
Catechin , Animals , Humans , Catechin/pharmacology , Catechin/therapeutic use , Catechin/metabolism , Muscle Fibers, Skeletal , Muscle, Skeletal/metabolism , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , MyoD Protein/metabolism , TOR Serine-Threonine Kinases/metabolism
6.
Exp Physiol ; 108(12): 1531-1547, 2023 12.
Article in English | MEDLINE | ID: mdl-37864311

ABSTRACT

NEW FINDINGS: What is the central question of this study? Does the hormone Klotho affect the myogenic response of muscle cells to mechanical loading or exercise? What is the main finding and its importance? Klotho prevents direct, mechanical activation of genes that regulate muscle differentiation, including genes that encode the myogenic regulatory factor myogenin and proteins in the canonical Wnt signalling pathway. Similarly, elevated levels of klotho expression in vivo prevent the exercise-induced increase in myogenin-expressing cells and reduce exercise-induced activation of the Wnt pathway. These findings demonstrate a new mechanism through which the responses of muscle to the mechanical environment are regulated. ABSTRACT: Muscle growth is influenced by changes in the mechanical environment that affect the expression of genes that regulate myogenesis. We tested whether the hormone Klotho could influence the response of muscle to mechanical loading. Applying mechanical loads to myoblasts in vitro increased RNA encoding transcription factors that are expressed in activated myoblasts (Myod) and in myogenic cells that have initiated terminal differentiation (Myog). However, application of Klotho to myoblasts prevented the loading-induced activation of Myog without affecting loading-induced activation of Myod. This indicates that elevated Klotho inhibits mechanically-induced differentiation of myogenic cells. Elevated Klotho also reduced the transcription of genes encoding proteins involved in the canonical Wnt pathway or their target genes (Wnt9a, Wnt10a, Ccnd1). Because the canonical Wnt pathway promotes differentiation of myogenic cells, these findings indicate that Klotho inhibits the differentiation of myogenic cells experiencing mechanical loading. We then tested whether these effects of Klotho occurred in muscles of mice experiencing high-intensity interval training (HIIT) by comparing wild-type mice and klotho transgenic mice. The expression of a klotho transgene combined with HIIT synergized to tremendously elevate numbers of Pax7+ satellite cells and activated MyoD+ cells. However, transgene expression prevented the increase in myogenin+ cells caused by HIIT in wild-type mice. Furthermore, transgene expression diminished the HIIT-induced activation of the canonical Wnt pathway in Pax7+ satellite cells. Collectively, these findings show that Klotho inhibits loading- or exercise-induced activation of muscle differentiation and indicate a new mechanism through which the responses of muscle to the mechanical environment are regulated.


Subject(s)
Muscles , Satellite Cells, Skeletal Muscle , Animals , Mice , Cell Differentiation , Hormones/metabolism , Muscle Development/genetics , Muscle, Skeletal/metabolism , Muscles/metabolism , MyoD Protein/metabolism , Myogenin/metabolism , Satellite Cells, Skeletal Muscle/metabolism
7.
Cells ; 12(17)2023 08 29.
Article in English | MEDLINE | ID: mdl-37681900

ABSTRACT

Sarcopenia has a high prevalence among the aging population. Sarcopenia is of tremendous socioeconomic importance because it can lead to falls and hospitalization, subsequently increasing healthcare costs while limiting quality of life. In sarcopenic muscle fibers, the E3 ubiquitin ligase F-Box Protein 32 (Fbxo32) is expressed at substantially higher levels, driving ubiquitin-proteasomal muscle protein degradation. As one of the key regulators of muscular equilibrium, the transcription factor Forkhead Box O3 (FOXO3) can increase the expression of Fbxo32, making it a possible target for the regulation of this detrimental pathway. To test this hypothesis, murine C2C12 myoblasts were transduced with AAVs carrying a plasmid for four specific siRNAs against Foxo3. Successfully transduced myoblasts were selected via FACS cell sorting to establish single clone cell lines. Sorted myoblasts were further differentiated into myotubes and stained for myosin heavy chain (MHC) by immunofluorescence. The resulting area was calculated. Myotube contractions were induced by electrical stimulation and quantified. We found an increased Foxo3 expression in satellite cells in human skeletal muscle and an age-related increase in Foxo3 expression in older mice in silico. We established an in vitro AAV-mediated FOXO3 knockdown on protein level. Surprisingly, the myotubes with FOXO3 knockdown displayed a smaller myotube size and a lower number of nuclei per myotube compared to the control myotubes (AAV-transduced with a functionless control plasmid). During differentiation, a lower level of FOXO3 reduced the expression Fbxo32 within the first three days. Moreover, the expression of Myod1 and Myog via ATM and Tp53 was reduced. Functionally, the Foxo3 knockdown myotubes showed a higher contraction duration and time to peak. Early Foxo3 knockdown seems to terminate the initiation of differentiation due to lack of Myod1 expression, and mediates the inhibition of Myog. Subsequently, the myotube size is reduced and the excitability to electrical stimulation is altered.


Subject(s)
Forkhead Box Protein O3 , MyoD Protein , Myogenin , Quality of Life , Sarcopenia , Aged , Animals , Humans , Mice , Forkhead Box Protein O3/genetics , Muscle Fibers, Skeletal , Muscle, Skeletal , Myoblasts , Myogenin/metabolism , MyoD Protein/metabolism
8.
J Cell Physiol ; 238(11): 2638-2650, 2023 11.
Article in English | MEDLINE | ID: mdl-37683043

ABSTRACT

Skeletal muscle regeneration is a crucial physiological process that occurs in response to injury or disease. As an important transcriptome surveillance system that regulates tissue development, the role of nonsense-mediated mRNA decay (NMD) in muscle regeneration remains unclear. Here, we found that NMD inhibits myoblast differentiation by targeting the phosphoinositide-3-kinase regulatory subunit 5 gene, which leads to the suppression of the transcriptional activity of myogenic differentiation (MyoD), a key regulator of myoblast differentiation. This disruption of MyoD transcriptional activity subsequently affects the expression levels of myogenin and myosin heavy chain, crucial markers of myoblast differentiation. Additionally, through up-frameshift protein 1 knockdown experiments, we observed that inhibiting NMD can accelerate muscle regeneration in vivo. These findings highlight the potential of NMD as a novel therapeutic target for the treatment of muscle-related injuries and diseases.


Subject(s)
Myoblasts , Nonsense Mediated mRNA Decay , Animals , Male , Mice , Cell Differentiation/genetics , Cell Line , Mice, Inbred C57BL , Muscle Development/genetics , Muscle, Skeletal/metabolism , Muscles , Myoblasts/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenin/genetics , Myogenin/metabolism , Nonsense Mediated mRNA Decay/genetics
9.
Epigenetics ; 18(1): 2237789, 2023 12.
Article in English | MEDLINE | ID: mdl-37506369

ABSTRACT

Long non-coding RNAs (lncRNAs) are involved in the process of muscle cell differentiation and play an important role. Previous studies have shown that lncRNA-MEG3 promotes the differentiation of porcine skeletal muscle satellite cells (PSCs), but the regulatory mechanism of MEG3 interaction with target protein has not been well studied. We demonstrated that MEG3 can bind dihydrolipoamide succinyltransferase (DLST) by RNA pull down and RIP-qPCR. Subsequently, knockdown and overexpression experiments showed that DLST promotes PSCs differentiation. Rescue experiments showed that the expression of DLST protein was significantly increased with MEG3 overexpression and decreased with MEG3 knockdown, while its mRNA expression was not changed. Furthermore, we have successfully predicted and validated that the transcription factor myogenic differentiation (MYOD) binds to the MEG3 core promoter though utilizing chromatin immunoprecipitation (CHIP) and luciferase reporter assays. The results indicated that MYOD acts as a transcription factor of MEG3 to promote MEG3 transcription. Knockdown of MEG3 in vivo indicated that MEG3 is involved in skeletal muscle regeneration. It is concluded that MYOD acts as a transcription factor to induce MEG3 expression. MEG3 acts as a molecular scaffold to bind and promote DLST protein expression. This paper provides a new molecular mechanism for MEG3 to promote the differentiation of PSCs.


Subject(s)
MyoD Protein , RNA, Long Noncoding , Satellite Cells, Skeletal Muscle , Animals , Cell Differentiation/genetics , DNA Methylation , MyoD Protein/genetics , MyoD Protein/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Swine , Transcription Factors/genetics
10.
Methods Mol Biol ; 2640: 259-276, 2023.
Article in English | MEDLINE | ID: mdl-36995601

ABSTRACT

The bHLH transcription factor MyoD is a master regulator of myogenic differentiation, and its sustained expression in fibroblasts suffices to differentiate them into muscle cells. MyoD expression oscillates in activated muscle stem cells of developing, postnatal and adult muscle under various conditions: when the stem cells are dispersed in culture, when they remain associated with single muscle fibers, or when they reside in muscle biopsies. The oscillatory period is around 3 h and thus much shorter than the cell cycle or circadian rhythm. Unstable MyoD oscillations and long periods of sustained MyoD expression are observed when stem cells undergo myogenic differentiation. The oscillatory expression of MyoD is driven by the oscillatory expression of the bHLH transcription factor Hes1 that periodically represses MyoD. Ablation of the Hes1 oscillator interferes with stable MyoD oscillations and leads to prolonged periods of sustained MyoD expression. This interferes with the maintenance of activated muscle stem cells and impairs muscle growth and repair. Thus, oscillations of MyoD and Hes1 control the balance between the proliferation and differentiation of muscle stem cells. Here, we describe time-lapse imaging methods using luciferase reporters, which can monitor dynamic MyoD gene expression in myogenic cells.


Subject(s)
Muscle Fibers, Skeletal , MyoD Protein , MyoD Protein/genetics , MyoD Protein/metabolism , Muscle Fibers, Skeletal/metabolism , Stem Cells , Cell Differentiation , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Muscle, Skeletal/metabolism
11.
Int J Legal Med ; 137(3): 875-886, 2023 May.
Article in English | MEDLINE | ID: mdl-36797435

ABSTRACT

From the perspective of forensic wound age estimation, experiments related to skeletal muscle regeneration after injury have rarely been reported. Here, we examined the time-dependent expression patterns of multiple biomarkers associated with satellite cell fate, including the transcription factor paired box 7 (Pax7), myoblast determination protein (MyoD), myogenin, and insulin-like growth factor (IGF-1), using immunohistochemistry, western blotting, and quantitative real-time PCR in contused skeletal muscle. An animal model of skeletal muscle contusion was established in 30 Sprague-Dawley male rats, and another five rats were employed as non-contused controls. Morphometrically, the data obtained from the numbers of Pax7 + , MyoD + , and myogenin + cells were highly correlated with the wound age. Pax7, MyoD, myogenin, and IGF-1 expression patterns were upregulated after injury at both the mRNA and protein levels. Pax7, MyoD, and myogenin protein expression levels confirmed the results of the morphometrical analysis. Additionally, the relative quantity of IGF-1 protein > 0.92 suggested a wound age of 3 to 7 days. The relative quantity of Pax7 mRNA > 2.44 also suggested a wound age of 3 to 7 days. Relative quantities of Myod1, Myog, and Igf1 mRNA expression > 2.78, > 7.80, or > 3.13, respectively, indicated a wound age of approximately 3 days. In conclusion, the expression levels of Pax7, MyoD, myogenin, and IGF-1 were upregulated in a time-dependent manner during skeletal muscle wound healing, suggesting the potential for using them as candidate biomarkers for wound age estimation in skeletal muscle.


Subject(s)
Contusions , Satellite Cells, Skeletal Muscle , Rats , Animals , Male , Myogenin/genetics , Myogenin/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Rats, Sprague-Dawley , Muscle, Skeletal/metabolism , Contusions/metabolism , Biomarkers/metabolism , RNA, Messenger/metabolism , Satellite Cells, Skeletal Muscle/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism
12.
J Biol Chem ; 299(3): 102978, 2023 03.
Article in English | MEDLINE | ID: mdl-36739949

ABSTRACT

The mitochondrial phospholipid cardiolipin (CL) is critical for numerous essential biological processes, including mitochondrial dynamics and energy metabolism. Mutations in the CL remodeling enzyme TAFAZZIN cause Barth syndrome, a life-threatening genetic disorder that results in severe physiological defects, including cardiomyopathy, skeletal myopathy, and neutropenia. To study the molecular mechanisms whereby CL deficiency leads to skeletal myopathy, we carried out transcriptomic analysis of the TAFAZZIN-knockout (TAZ-KO) mouse myoblast C2C12 cell line. Our data indicated that cardiac and muscle development pathways are highly decreased in TAZ-KO cells, consistent with a previous report of defective myogenesis in this cell line. Interestingly, the muscle transcription factor myoblast determination protein 1 (MyoD1) is significantly repressed in TAZ-KO cells and TAZ-KO mouse hearts. Exogenous expression of MyoD1 rescued the myogenesis defects previously observed in TAZ-KO cells. Our data suggest that MyoD1 repression is caused by upregulation of the MyoD1 negative regulator, homeobox protein Mohawk, and decreased Wnt signaling. Our findings reveal, for the first time, that CL metabolism regulates muscle differentiation through MyoD1 and identify the mechanism whereby MyoD1 is repressed in CL-deficient cells.


Subject(s)
Barth Syndrome , Cardiolipins , MyoD Protein , Animals , Mice , Acyltransferases/genetics , Barth Syndrome/genetics , Barth Syndrome/metabolism , Cardiolipins/genetics , Cardiolipins/metabolism , Mice, Knockout , Muscles/metabolism , Transcription Factors/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism
13.
Gene ; 849: 146907, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36174904

ABSTRACT

The flavanol (-)-epicatechin has exercise-mimetic properties. Besides, several miRNAs play a role in modulating the adaptation of the muscle to different training protocols. However, notwithstanding all information, few studies aimed to determine if (-)-epicatechin can modify the expression of miRNAs related to skeletal muscle development and regeneration. Mice were treated for fifteen days by oral gavage with the flavanol (-)-epicatechin. After treatment, the quadriceps of the mice was dissected, and total RNA was extracted. The expression level of miR-133, -204, -206, -223, -486, and -491 was analyzed by qRT-PCR. We also used bioinformatic analysis to predict the participation of these miRNAs in different skeletal muscle signal transduction pathways. Additionally, we analyzed the level of the myogenic proteins MyoD and myogenin by Western blot and measured the cross-sectional area of muscle fibers stained with E&H. (-)-Epicatechin upregulated the expression of miR-133, -204, -206, -223, and -491 significantly, which was associated with an increase in the level of the myogenic proteins MyoD and Myogenin and an augment in the fiber size. The bioinformatics analysis showed that the studied miRNAs might participate in different signal transduction pathways related to muscle development and adaptation. Our results showed that (-)-epicatechin upregulated miRNAs that participate in skeletal exercise muscle adaptation, induced muscle hypertrophy, and increased the level of myogenic proteins MyoD and MyoG.


Subject(s)
Catechin , MicroRNAs , Mice , Animals , Myogenin/genetics , Myogenin/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Catechin/pharmacology , Muscle, Skeletal/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Differentiation
14.
Commun Biol ; 5(1): 1201, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36352000

ABSTRACT

Muscle stem cells (MuSCs) are essential for skeletal muscle development and regeneration, ensuring muscle integrity and normal function. The myogenic proliferation and differentiation of MuSCs are orchestrated by a cascade of transcription factors. In this study, we elucidate the specific role of transcription factor 12 (Tcf12) in muscle development and regeneration based on loss-of-function studies. Muscle-specific deletion of Tcf12 cause muscle weight loss owing to the reduction of myofiber size during development. Inducible deletion of Tcf12 specifically in adult MuSCs delayed muscle regeneration. The examination of MuSCs reveal that Tcf12 deletion resulted in cell-autonomous defects during myogenesis and Tcf12 is necessary for proper myogenic gene expression. Mechanistically, TCF12 and MYOD work together to stabilise chromatin conformation and sustain muscle cell fate commitment-related gene and chromatin architectural factor expressions. Altogether, our findings identify Tcf12 as a crucial regulator of MuSCs chromatin remodelling that regulates muscle cell determination and participates in skeletal muscle development and regeneration.


Subject(s)
Chromatin , MyoD Protein , MyoD Protein/genetics , MyoD Protein/metabolism , Chromatin/genetics , Chromatin/metabolism , Muscle, Skeletal/metabolism , Muscle Development/genetics , Myoblasts
15.
Int J Mol Sci ; 23(21)2022 Nov 06.
Article in English | MEDLINE | ID: mdl-36362402

ABSTRACT

Lamina-associated polypeptide 1 (LAP1) is a ubiquitously expressed inner nuclear membrane protein encoded by TOR1AIP1, and presents as two isoforms in humans, LAP1B and LAP1C. While loss of both isoforms results in a multisystemic progeroid-like syndrome, specific loss of LAP1B causes muscular dystrophy and cardiomyopathy, suggesting that LAP1B has a critical role in striated muscle. To gain more insight into the molecular pathophysiology underlying muscular dystrophy caused by LAP1B, we established a patient-derived fibroblast line that was transdifferentiated into myogenic cells using inducible MyoD expression. Compared to the controls, we observed strongly reduced myogenic differentiation and fusion potentials. Similar defects were observed in the C2C12 murine myoblasts carrying loss-of-function LAP1A/B mutations. Using RNA sequencing, we found that, despite MyoD overexpression and efficient cell cycle exit, transcriptional reprogramming of the LAP1B-deficient cells into the myogenic lineage is impaired with delayed activation of MYOG and muscle-specific genes. Gene set enrichment analyses suggested dysregulations of protein metabolism, extracellular matrix, and chromosome organization. Finally, we found that the LAP1B-deficient cells exhibit nuclear deformations, such as an increased number of micronuclei and altered morphometric parameters. This study uncovers the phenotypic and transcriptomic changes occurring during myoconversion of patient-derived LAP1B-deficient fibroblasts and provides a useful resource to gain insights into the mechanisms implicated in LAP1B-associated nuclear envelopathies.


Subject(s)
Muscular Dystrophies , Nuclear Envelope , Animals , Humans , Mice , Cell Differentiation/genetics , Fibroblasts/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle Development/genetics , Muscular Dystrophies/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Nuclear Envelope/metabolism , Protein Isoforms/metabolism
16.
Res Vet Sci ; 152: 569-578, 2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36191510

ABSTRACT

This study aims to explore the functional role of Myoz2 in myoblast differentiation, and elucidate the potential factors interact with Myoz2 in promoter transcriptional regulation. The temporal-spatial expression results showed that the bovine Myoz2 gene was highest expressed in longissimus dorsi, and in individual growth stages and myoblast differentiation stages. Knockdown of Myoz2 inhibited the differentiation of myoblast, and negative effect of MyoD, MyoG, MyH and MEF2A expression on mRNA levels. Subsequently, the promoter region of bovine Myoz2 gene with 1.7 Kb sequence was extracted, and then it was set as eight series of deleted fragments, which were ligated into pGL3-basic to detect core promoter regions of Myoz2 gene in myoblasts and myotubes. Transcription factors MyoD and MyoG were identified as important cis-acting elements in the core promoter region (-159/+1). Also, it was highly conserved in different species based on dual-luciferase analysis and multiple sequence alignment analysis, respectively. Furthermore, a chromatin immunoprecipitation (ChIP) analysis combined with site-directed mutation and siRNA interference and overexpression confirmed that the combination of MyoD and MyoG occurred in region -159/+1, and played an important role in the regulation of bovine Myoz2 gene. These findings explored the regulatory network mechanism of Myoz2 gene during the development of bovine skeletal muscle.


Subject(s)
MyoD Protein , Myoblasts , Cattle , Animals , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/physiology , Promoter Regions, Genetic , Gene Expression Regulation , Transcription Factors/metabolism , Cell Differentiation/genetics , Muscle Development/genetics
17.
Nucleic Acids Res ; 50(18): 10733-10755, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36200826

ABSTRACT

Long noncoding RNAs (lncRNAs) play important roles in the spatial and temporal regulation of muscle development and regeneration. Nevertheless, the determination of their biological functions and mechanisms underlying muscle regeneration remains challenging. Here, we identified a lncRNA named lncMREF (lncRNA muscle regeneration enhancement factor) as a conserved positive regulator of muscle regeneration among mice, pigs and humans. Functional studies demonstrated that lncMREF, which is mainly expressed in differentiated muscle satellite cells, promotes myogenic differentiation and muscle regeneration. Mechanistically, lncMREF interacts with Smarca5 to promote chromatin accessibility when muscle satellite cells are activated and start to differentiate, thereby facilitating genomic binding of p300/CBP/H3K27ac to upregulate the expression of myogenic regulators, such as MyoD and cell differentiation. Our results unravel a novel temporal-specific epigenetic regulation during muscle regeneration and reveal that lncMREF/Smarca5-mediated epigenetic programming is responsible for muscle cell differentiation, which provides new insights into the regulatory mechanism of muscle regeneration.


Subject(s)
RNA, Long Noncoding , Adenosine Triphosphatases , Animals , Cell Differentiation , Cell Line , Chromatin/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic , Humans , Mice , Muscle Development , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Regeneration , Swine
18.
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
19.
Biochem Biophys Res Commun ; 621: 88-93, 2022 09 17.
Article in English | MEDLINE | ID: mdl-35810596

ABSTRACT

The basic helix-loop-helix (bHLH) family is one of the most conserved transcription factor families that plays an important role in regulating cell growth, differentiation and tissue development. Typically, members of this family form homo- or heterodimers to recognize specific motifs and activate transcription. MyoD is a vital transcription factor that regulates muscle cell differentiation. However, it is necessary for MyoD to form a heterodimer with E-proteins to activate transcription. Even though the crystal structure of the MyoD homodimer has been determined, the structure of the MyoD heterodimer in complex with the E-box protein remains unclear. In this study, we determined the crystal structure of the bHLH domain of the MyoD-E47 heterodimer at 2.05 Å. Our structural analysis revealed that MyoD interacts with E47 through a hydrophobic interface. Moreover, we confirmed that heterodimerization could enhance the binding affinity of MyoD to E-box sequences. Our results provide new structural insights into the heterodimer of MyoD and E-box protein, suggesting the molecular mechanism of transcription activation of MyoD upon binding to E-box protein.


Subject(s)
DNA-Binding Proteins , MyoD Protein , DNA-Binding Proteins/metabolism , Helix-Loop-Helix Motifs , MyoD Protein/metabolism , Protein Binding , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factors/metabolism
20.
Int J Mol Sci ; 23(13)2022 Jun 23.
Article in English | MEDLINE | ID: mdl-35805999

ABSTRACT

Epinephelus coioides is a fish species with high economic value due to its delicious meat, high protein content, and rich fatty acid nutrition. It has become a high-economic fish in southern parts of China and some other Southeast Asian countries. In this study, the myostatin nucleic acid vaccine was constructed and used to immunize E. coioides. The results from body length and weight measurements indicated the myostatin nucleic acid vaccine promoted E. coioides growth performance by increasing muscle fiber size. The results from RT-qPCR analysis showed that myostatin nucleic acid vaccine upregulated the expression of myod, myog and p21 mRNA, downregulated the expression of smad3 and mrf4 mRNA. This preliminary study is the first report that explored the role of myostatin in E. coioides and showed positive effects of autologous nucleic acid vaccine on the muscle growth of E. coioides. Further experiments with increased numbers of animals and different doses are needed for its application to E. coiodes aquaculture production.


Subject(s)
Muscle Fibers, Skeletal , Myostatin , Perciformes , Animals , Body Weight , Fishes , Gene Expression Regulation , Muscle Fibers, Skeletal/physiology , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Myogenin/genetics , Myogenin/metabolism , Myostatin/genetics , Myostatin/immunology , Nucleic Acid-Based Vaccines/administration & dosage , Nucleic Acid-Based Vaccines/immunology , Perciformes/growth & development , Perciformes/physiology , Smad3 Protein/genetics , Smad3 Protein/metabolism , Vaccination , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...