Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.664
Filter
1.
FASEB J ; 38(14): e23841, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39051762

ABSTRACT

Skeletal muscles undergo robust regeneration upon injury, and infiltrating immune cells play a major role in not only clearing damaged tissues but also regulating the myogenic process through secreted cytokines. Chemokine C-C motif ligand 8 (Ccl8), along with Ccl2 and Ccl7, has been reported to mediate inflammatory responses to suppress muscle regeneration. Ccl8 is also expressed by muscle cells, but a role of the muscle cell-derived Ccl8 in myogenesis has not been reported. In this study, we found that knockdown of Ccl8, but not Ccl2 or Ccl7, led to increased differentiation of C2C12 myoblasts. Analysis of existing single-cell transcriptomic datasets revealed that both immune cells and muscle stem cells (MuSCs) in regenerating muscles express Ccl8, with the expression by MuSCs at a much lower level, and that the temporal patterns of Ccl8 expression were different in MuSCs and macrophages. To probe a function of muscle cell-derived Ccl8 in vivo, we utilized a mouse system in which Cas9 was expressed in Pax7+ myogenic progenitor cells (MPCs) and Ccl8 gene editing was induced by AAV9-delivered sgRNA. Depletion of Ccl8 in Pax7+ MPCs resulted in accelerated muscle regeneration after barium chloride-induced injury in both young and middle-aged mice, and intramuscular administration of a recombinant Ccl8 reversed the phenotype. Accelerated regeneration was also observed when Ccl8 was depleted in Myf5+ or MyoD+ MPCs by similar approaches. Our results suggest that muscle cell-derived Ccl8 plays a unique role in regulating the initiation of myogenic differentiation during injury-induced muscle regeneration.


Subject(s)
Cell Differentiation , Chemokine CCL8 , Muscle Development , Muscle, Skeletal , Myoblasts , Regeneration , Animals , Mice , Regeneration/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Muscle, Skeletal/injuries , Muscle Development/physiology , Chemokine CCL8/metabolism , Chemokine CCL8/genetics , Myoblasts/metabolism , Myoblasts/physiology , Mice, Inbred C57BL , Cell Line , Male , Chemokine CCL7/metabolism , Chemokine CCL7/genetics , Macrophages/metabolism
2.
PLoS One ; 19(7): e0305907, 2024.
Article in English | MEDLINE | ID: mdl-39052586

ABSTRACT

The mechanisms governing gene regulation in domestic Yuzhong pigeon breast muscle development remain largely elusive. Here, we conducted a comparative analysis using Iso-seq and RNA-seq data from domestic Yuzhong pigeons and European meat pigeons to uncover signaling pathways and genes involved in breast muscle development. The Iso-seq data from domestic Yuzhong pigeons yielded 131,377,075 subreads, resulting in 16,587 non-redundant high-quality full-length transcripts post-correction. Furthermore, utilizing pfam, CPC, PLEK, and CPAT, we predicted 5575, 4973, 2333, and 4336 lncRNAs, respectively. Notably, several genes potentially implicated in breast muscle development were identified, including tropomyosin beta chain, myosin regulatory light chain 2, and myosin binding protein C. KEGG enrichment analysis revealed critical signaling pathways in breast muscle development, spanning carbon metabolism, biosynthesis of amino acids, glycolysis/gluconeogenesis, estrogen signaling, PI3K-AKT signaling, protein processing in the endoplasmic reticulum, oxidative phosphorylation, pentose phosphate pathway, fructose and mannose metabolism, and tight junctions. These findings offer insights into the biological processes driving breast muscle development in domestic Yuzhong pigeon, contributing to our understanding of this complex phenomenon.


Subject(s)
Columbidae , Muscle Development , RNA-Seq , Animals , Columbidae/genetics , Columbidae/growth & development , Columbidae/metabolism , Muscle Development/genetics , Signal Transduction/genetics , Sequence Analysis, RNA , RNA, Long Noncoding/genetics
3.
Theranostics ; 14(10): 3963-3983, 2024.
Article in English | MEDLINE | ID: mdl-38994033

ABSTRACT

Piezo1, a mechanosensitive ion channel, has emerged as a key player in translating mechanical stimuli into biological signaling. Its involvement extends beyond physiological and pathological processes such as lymphatic vessel development, axon growth, vascular development, immunoregulation, and blood pressure regulation. The musculoskeletal system, responsible for structural support, movement, and homeostasis, has recently attracted attention regarding the significance of Piezo1. This review aims to provide a comprehensive summary of the current research on Piezo1 in the musculoskeletal system, highlighting its impact on bone formation, myogenesis, chondrogenesis, intervertebral disc homeostasis, tendon matrix cross-linking, and physical activity. Additionally, we explore the potential of targeting Piezo1 as a therapeutic approach for musculoskeletal disorders, including osteoporosis, muscle atrophy, intervertebral disc degeneration, and osteoarthritis.


Subject(s)
Ion Channels , Musculoskeletal Diseases , Humans , Ion Channels/metabolism , Animals , Musculoskeletal Diseases/metabolism , Musculoskeletal System/metabolism , Chondrogenesis/physiology , Mechanotransduction, Cellular , Osteogenesis/physiology , Muscle Development
4.
FASEB J ; 38(14): e23808, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-38994637

ABSTRACT

Muscle development is a multistep process regulated by diverse gene networks, and circRNAs are considered novel regulators mediating myogenesis. Here, we systematically analyzed the role and underlying regulatory mechanisms of circRBBP7 in myoblast proliferation and differentiation. Results showed that circRBBP7 has a typical circular structure and encodes a 13 -kDa protein. By performing circRBBP7 overexpression and RNA interference, we found that the function of circRBBP7 was positively correlated with the proliferation and differentiation of myoblasts. Using RNA sequencing, we identified 1633 and 532 differentially expressed genes (DEGs) during myoblast proliferation or differentiation, respectively. The DEGs were found mainly enriched in cell cycle- and skeletal muscle development-related pathways, such as the MDM2/p53 and PI3K-Akt signaling pathways. Further co-IP and IF co-localization analysis revealed that VEGFR-1 is a target of circRBBP7 in myoblasts. qRT-PCR and WB analysis further confirmed the positive correlation between VEGFR-1 and circRBBP7. Moreover, we found that in vivo transfection of circRBBP7 into injured muscle tissues significantly promoted the regeneration and repair of myofibers in mice. Therefore, we speculate that circRBBP7 may affect the activity of MDM2 by targeting VEGFR-1, altering the expression of muscle development-related genes by mediating p53 degradation, and ultimately promoting myoblast development and muscle regeneration. This study provides essential evidence that circRBBP7 can serve as a potential target for myogenesis regulation and a reference for the application of circRBBP7 in cattle genetic breeding and muscle injury treatment.


Subject(s)
Cell Differentiation , Cell Proliferation , Muscle Development , Myoblasts , RNA, Circular , Animals , Male , Mice , Cell Line , Mice, Inbred C57BL , Muscle Development/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Myoblasts/metabolism , Myoblasts/cytology , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Circular/genetics , RNA, Circular/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics
5.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000437

ABSTRACT

Ubiquitin C-terminal hydrolase L1 (UCHL1) is a deubiquitinating enzyme originally found in the brain. Our previous work revealed that UCHL1 was also expressed in skeletal muscle and affected myoblast differentiation and metabolism. In this study, we further tested the role of UCHL1 in myogenesis and muscle regeneration following muscle ischemia-reperfusion (IR) injury. In the C2C12 myoblast, UCHL1 knockdown upregulated MyoD and myogenin and promoted myotube formation. The skeletal muscle-specific knockout (smKO) of UCHL1 increased muscle fiber sizes in young mice (1 to 2 months old) but not in adult mice (3 months old). In IR-injured hindlimb muscle, UCHL1 was upregulated. UCHL1 smKO ameliorated tissue damage and injury-induced inflammation. UCHL1 smKO also upregulated myogenic factors and promoted functional recovery in IR injury muscle. Moreover, UCHL1 smKO increased Akt and Pink1/Parkin activities. The overall results suggest that skeletal muscle UCHL1 is a negative factor in skeletal muscle development and recovery following IR injury and therefore is a potential therapeutic target to improve muscle regeneration and functional recovery following injuries.


Subject(s)
Mice, Knockout , Muscle Development , Muscle, Skeletal , Ubiquitin Thiolesterase , Animals , Male , Mice , Cell Differentiation , Cell Line , Mice, Inbred C57BL , Muscle Development/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/injuries , Myoblasts/metabolism , Regeneration , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Female
6.
Mol Biol Rep ; 51(1): 840, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39042282

ABSTRACT

BACKGROUND: MiR-486-5p has been identified as a crucial regulator of the PI3K/AKT signalling pathway, which plays a significant role in skeletal muscle development. Its host gene, sANK1, is also essential for skeletal muscle development. However, the understanding of porcine miR-486-5p and sANK1 has been limited. METHODS AND RESULTS: In this study, PCR analyses revealed a positive correlation between the expression of miR-486-5p and sANK1 in the longissimus dorsi muscle of the Bama mini-pig and Landrace-pig, as well as during myoblast differentiation. Furthermore, the expression of miR-486-5p/sANK1 was higher in the Bama mini-pig compared to the Landrace-pig. There was a total of 18 single nucleotide polymorphisms (SNP) present in the sANK1 promoter region. Among these SNPs, 14 of them resulted in alterations in transcription factor binding sites (TFBs). Additionally, the promoter fluorescence assay demonstrated that the activity of the sANK1 promoter derived from the Bama mini-pig was significantly higher compared to Landrace-pig. It is worth noting that ten regulatory SNPs have the potential to influence the activity of the sANK1 promoter. A nuclear mutation A-G located at position - 401 (relative to the transcription start site) in the Bama mini-pig was identified, which creates a putative TFB motif for MyoD. CONCLUSIONS: The findings presented in this study offer fundamental molecular knowledge and expression patterns of miR-486-5p/sANK1, which can be valuable for gaining a deeper understanding of the gene's involvement in porcine skeletal muscle development, and meat quality.


Subject(s)
MicroRNAs , Muscle, Skeletal , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Swine/genetics , Muscle, Skeletal/metabolism , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Muscle Development/genetics , Cell Differentiation/genetics , Myoblasts/metabolism , Gene Expression Regulation/genetics , Binding Sites , MyoD Protein/genetics , MyoD Protein/metabolism , Signal Transduction/genetics
7.
Sci Rep ; 14(1): 16422, 2024 07 16.
Article in English | MEDLINE | ID: mdl-39013963

ABSTRACT

Interactions between tissues and cell types, mediated by cytokines or direct cell-cell exchanges, regulate growth. To determine whether mature adipocytes influence the in vitro growth of trout mononucleated muscle cells, we developed an indirect coculture system, and showed that adipocytes (5 × 106 cells/well) derived from perivisceral adipose tissue increased the proliferation (BrdU-positive cells) of the mononucleated muscle cells (26% vs. 39%; p < 0.001) while inhibiting myogenic differentiation (myosin+) (25% vs. 15%; p < 0.001). Similar effects were obtained with subcutaneous adipose tissue-derived adipocytes, although requiring more adipocytes (3 × 107 cells/well vs. 5 × 106 cells/well). Conditioned media recapitulated these effects, stimulating proliferation (31% vs. 39%; p < 0.001) and inhibiting myogenic differentiation (32 vs. 23%; p < 0.001). Adipocytes began to reduce differentiation after 24 h, whereas proliferation stimulation was observed after 48 h. While adipocytes did not change pax7+ and myoD1/2+ percentages, they reduced myogenin+ cells showing inhibition from early differentiation stage. Finally, adipocytes increased BrdU+ cells in the Pdgfrα+ population but not in the myoD+ one. Collectively, our results demonstrate that trout adipocytes promote fibro-adipocyte precursor proliferation while inhibiting myogenic cells differentiation in vitro, suggesting the key role of adipose tissue in regulating fish muscle growth.


Subject(s)
Adipocytes , Adipogenesis , Cell Differentiation , Cell Proliferation , Muscle Development , Animals , Adipocytes/cytology , Coculture Techniques , Cells, Cultured , Trout , Culture Media, Conditioned/pharmacology
8.
Sci Rep ; 14(1): 16474, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014195

ABSTRACT

Males and females have long shown disparities in body weight and height; yet, the underlying mechanisms influencing growth and development remain unclear. Male and female Zhedong White Geese (ZDW) geese have long been selected for large body size and egg production, respectively. This led to a large difference in body weight between males and females, making them a unique model for studying the effects of sex on growth and development. This study aimed to elucidate these mechanisms by comparing the transcriptomes of muscle and pituitary tissues in male and female ZDW geese to identify the critical genes responsible for the effects of sex on growth performance. Our analysis revealed 1101 differentially expressed genes (DEGs) in leg musculature (507 upregulated, 594 downregulated), 773 DEGs in breast musculature (311 upregulated, 462 downregulated), and 517 DEGs in the pituitary gland (281 upregulated, 236 downregulated) between male and female geese. These DEGs were significantly enriched in gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways associated with endocrine metabolism (e.g., hormonal activities), muscle formation (e.g., sarcomere and myofibril), and bone formation (e.g., bone morphogenesis and cartilage formation). The upregulated genes in males were enriched in KEGG pathways involving nutrient digestion and absorption (vitamin and protein), as well as the secretion of digestive juices (gastric acid and bile). Through protein-protein interaction analyses, we also observed high-density gene networks related to muscle fiber development, calcium ion metabolism, mitochondrial respiratory chain, and bone development. Therefore, our multi-tissue transcriptome analysis provides a deeper understanding of the complex and systematic gender-driven effects on growth and development in geese. IGF1, GHRHR, and NCAPG-LCORL and pathways related to myogenesis might play vital roles in gender differences before hormones exert their effect.


Subject(s)
Geese , Muscle Development , Transcriptome , Animals , Female , Male , Geese/genetics , Geese/growth & development , Muscle Development/genetics , Gene Expression Profiling , Muscle, Skeletal/metabolism , Muscle, Skeletal/growth & development , Gene Ontology
9.
Commun Biol ; 7(1): 910, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39068219

ABSTRACT

Breast muscle growth rate and intramuscular fat (IMF) content show apparent differences between fast-growing broilers and slow-growing indigenous chickens. However, the underlying genetic basis of these phenotypic characteristics remains elusive. In this study, we investigate the dynamic alterations of three-dimensional genome architecture and chromatin accessibility in breast muscle across four key developmental stages from embryo to starter chick in Arbor Acres (AA) broilers and Yufen (YF) indigenous chickens. The limited breed-specifically up-regulated genes (Bup-DEGs) are embedded in breed-specific A compartment, while a majority of the Bup-DEGs involving myogenesis and adipogenesis are regulated by the breed-specific TAD reprogramming. Chromatin loops allow distal accessible regions to interact with myogenic genes, and those loops share an extremely low similarity between chicken with different growth rate. Moreover, AA-specific loop interactions promote the expression of 40 Bup-DEGs, such as IGF1, which contributes to myofiber hypertrophy. YF-specific loop interactions or distal accessible regions lead to increased expression of 5 Bup-DEGs, including PIGO, PEMT, DHCR7, TMEM38B, and DHDH, which contribute to IMF deposition. These results help elucidate the regulation of breast muscle growth and IMF deposition in chickens.


Subject(s)
Chickens , Chromatin , Muscle Development , Phenotype , Animals , Chickens/genetics , Chickens/growth & development , Chromatin/metabolism , Chromatin/genetics , Muscle Development/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/growth & development , Pectoralis Muscles/metabolism , Pectoralis Muscles/growth & development , Chick Embryo , Gene Expression Regulation, Developmental
10.
Sci Rep ; 14(1): 15678, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977785

ABSTRACT

Aging and lack of exercise are the most important etiological factors for muscle loss. We hypothesized that new factors that contribute to muscle loss could be identified from ones commonly altered in expression in aged and exercise-limited skeletal muscles. Mouse gastrocnemius muscles were subjected to mass spectrometry-based proteomic analysis. The muscle proteomes of hindlimb-unloaded and aged mice were compared to those of exercised and young mice, respectively. C1qbp expression was significantly upregulated in the muscles of both hindlimb-unloaded and aged mice. In vitro myogenic differentiation was not affected by altering intracellular C1qbp expression but was significantly suppressed upon recombinant C1qbp treatment. Additionally, recombinant C1qbp repressed the protein level but not the mRNA level of NFATc1. NFATc1 recruited the transcriptional coactivator p300, leading to the upregulation of acetylated histone H3 levels. Furthermore, NFATc1 silencing inhibited p300 recruitment, downregulated acetylated histone H3 levels, and consequently suppressed myogenic differentiation. The expression of C1qbp was inversely correlated with that of NFATc1 in the gastrocnemius muscles of exercised or hindlimb-unloaded, and young or aged mice. These findings demonstrate a novel role of extracellular C1qbp in suppressing myogenesis by inhibiting the NFATc1/p300 complex. Thus, C1qbp can serve as a novel therapeutic target for muscle loss.


Subject(s)
Muscle Development , Muscle, Skeletal , NFATC Transcription Factors , Animals , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , Muscle Development/genetics , Mice , Muscle, Skeletal/metabolism , Cell Differentiation , Histones/metabolism , Male , Mice, Inbred C57BL , Acetylation
11.
Zool Res ; 45(4): 951-960, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39021083

ABSTRACT

Tumor necrosis factor α (TNFα) exhibits diverse biological functions; however, its regulatory roles in myogenesis are not fully understood. In the present study, we explored the function of TNFα in myoblast proliferation, differentiation, migration, and myotube fusion in primary myoblasts and C2C12 cells. To this end, we constructed TNFα muscle-conditional knockout ( TNFα-CKO) mice and compared them with flox mice to assess the effects of TNFα knockout on skeletal muscles. Results indicated that TNFα-CKO mice displayed phenotypes such as accelerated muscle development, enhanced regenerative capacity, and improved exercise endurance compared to flox mice, with no significant differences observed in major visceral organs or skeletal structure. Using label-free proteomic analysis, we found that TNFα-CKO altered the distribution of several muscle development-related proteins, such as Hira, Casz1, Casp7, Arhgap10, Gas1, Diaph1, Map3k20, Cfl2, and Igf2, in the nucleus and cytoplasm. Gene set enrichment analysis (GSEA) further revealed that TNFα deficiency resulted in positive enrichment in oxidative phosphorylation and MyoD targets and negative enrichment in JAK-STAT signaling. These findings suggest that TNFα-CKO positively regulates muscle growth and development, possibly via these newly identified targets and pathways.


Subject(s)
Mice, Knockout , Muscle Development , Muscle, Skeletal , Regeneration , Tumor Necrosis Factor-alpha , Animals , Muscle Development/physiology , Mice , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Cell Line , Cell Differentiation , Myoblasts/metabolism , Myoblasts/physiology
12.
Biomed Res ; 45(4): 173-177, 2024.
Article in English | MEDLINE | ID: mdl-39010193

ABSTRACT

Sarcopenia is a common complication of chronic kidney disease (CKD) and has a detrimental effect on prognosis. Previous studies have explored the role of secondary calciprotein particles (CPP2) in determining the progression of complications and poor outcomes in patients with CKD. However, no study has demonstrated that CPP2 impairs skeletal myogenesis. Our study revealed that CPP2 exposure inhibits skeletal myogenesis by suppressing myotube formation and expression of skeletal muscle-specific myosin heavy chain and actin in human primary myoblasts. Moreover, CPP2 exposure altered the expression patterns of lineage-determinative transcription factors responsible for regulating myotube differentiation marker genes. This study first demonstrated that CPP2 interferes with myoblast differentiation and myotube formation in vitro.


Subject(s)
Cell Differentiation , Muscle Development , Myoblasts , Humans , Myoblasts/metabolism , Myoblasts/cytology , Cells, Cultured , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Gene Expression Regulation
13.
J Agric Food Chem ; 72(28): 15530-15540, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38963795

ABSTRACT

The skeletal muscle is the major muscle tissue in animals, and its production is subject to a complex and strict regulation. The proliferation and differentiation of myoblasts are important factors determining chicken muscle development. Circular RNAs (circRNAs) are endogenous RNAs that are widely present in various tissues of organisms. Recent studies have shown that circRNA plays key roles in the development of skeletal muscles. The solute carrier (SLC) family functions in the transport of metabolites such as amino acids, glucose, nucleotides, and essential nutrients and is widely involved in various basic physiological metabolic processes within the body. In this study, we have cloned a novel chicken circular RNA circSLC2A13 generated from the solute carrier family 2 member 13 gene (SLC2A13). Also, circSLC2A1 was confirmed by sequencing verification, RNase R treatment, and reverse transcription analysis. Currently, our results show that circSLC2A13 promoted the proliferation and differentiation of chicken myoblasts. The double luciferase reporter system revealed that circSLC2A13 regulated the proliferation and differentiation of myoblasts by competitive binding with miR-34a-3p. In addition, results indicated that circSLC2A13 acts as a miR-34a-3p sponge to relieve its inhibitory effect on the target SMAD3 gene. In summary, this study found that chicken circSLC2A13 can bind to miR-34a-3p and weaken its inhibitory effect on the SMAD family member 3 gene (SMAD3), thereby promoting the proliferation and differentiation of myoblasts. This study laid foundations for broiler industry and muscle development research.


Subject(s)
Cell Differentiation , Cell Proliferation , Chickens , MicroRNAs , Muscle Development , Muscle, Skeletal , Myoblasts , RNA, Circular , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Chickens/genetics , Chickens/growth & development , Chickens/metabolism , Muscle Development/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/growth & development , Myoblasts/metabolism , Myoblasts/cytology
14.
Magnes Res ; 37(1): 1-11, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39077819

ABSTRACT

A physiological concentration of magnesium (Mg) is essential for optimal skeletal muscle function. Indeed, Mg plays a crucial role during the differentiation process (myogenesis), in muscle fiber composition, muscle contraction and performance. This narrative review describes in detail the relevance of Mg in skeletal muscle, highlighting the importance of adequate Mg intake to ensure optimal skeletal muscle cell function and performance in individuals of all ages.


Subject(s)
Magnesium , Muscle Development , Muscle, Skeletal , Magnesium/metabolism , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Animals
15.
Nat Commun ; 15(1): 6307, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39060228

ABSTRACT

Rhabdomyosarcoma (RMS) is a pediatric tumor that resembles undifferentiated muscle cells; yet the extent to which cell state heterogeneity is shared with human development has not been described. Using single-cell/nucleus RNA sequencing from patient tumors, patient-derived xenografts, primary in vitro cultures, and cell lines, we identify four dominant muscle-lineage cell states: progenitor, proliferative, differentiated, and ground cells. We stratify these RMS cells/nuclei along the continuum of human muscle development and show that they share expression patterns with fetal/embryonal myogenic precursors rather than postnatal satellite cells. Fusion-negative RMS (FN-RMS) have a discrete stem cell hierarchy that recapitulates fetal muscle development and contain therapy-resistant FN-RMS progenitors that share transcriptomic similarity with bipotent skeletal mesenchymal cells. Fusion-positive RMS have tumor-acquired cells states, including a neuronal cell state, that are not found in myogenic development. This work identifies previously underappreciated cell state heterogeneity including unique treatment-resistant and tumor-acquired cell states that differ across RMS subtypes.


Subject(s)
Gene Expression Profiling , Rhabdomyosarcoma , Single-Cell Analysis , Transcriptome , Humans , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/metabolism , Single-Cell Analysis/methods , Animals , Gene Expression Profiling/methods , Cell Line, Tumor , Mice , Child , Drug Resistance, Neoplasm/genetics , Cell Differentiation , Muscle Development/genetics , Gene Expression Regulation, Neoplastic
16.
Sci Rep ; 14(1): 15696, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977909

ABSTRACT

As the largest organ in the human body, skeletal muscle is essential for breathing support, movement initiation, and maintenance homeostasis. It has been shown that programmed cell death (PCD), which includes autophagy, apoptosis, and necrosis, is essential for the development of skeletal muscle. A novel form of PCD called ferroptosis is still poorly understood in relation to skeletal muscle. In this study, we observed that the activation of ferroptosis significantly impeded the differentiation of C2C12 myoblasts into myotubes and concurrently suppressed the expression of OTUB1, a crucial deubiquitinating enzyme. OTUB1-silenced C2C12 mouse myoblasts were used to investigate the function of OTUB1 in ferroptosis. The results show that OTUB1 knockdown in vitro significantly increased C2C12 ferroptosis and inhibited myogenesis. Interestingly, the induction of ferroptosis resulting from OTUB1 knockdown was concomitant with the activation of autophagy. Furthermore, OTUB1 interacted with the P62 protein and stabilized its expression by deubiquitinating it, thereby inhibiting autophagy-dependent ferroptosis and promoting myogenesis. All of these findings demonstrate the critical role that OTUB1 plays in controlling ferroptosis, and we suggest that focusing on the OTUB1-P62 axis may be a useful tactic in the treatment and prevention of disorders involving the skeletal muscle.


Subject(s)
Autophagy , Cell Differentiation , Cysteine Endopeptidases , Ferroptosis , Muscle Development , Muscle Fibers, Skeletal , Myoblasts , Animals , Mice , Muscle Fibers, Skeletal/metabolism , Ferroptosis/genetics , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Myoblasts/metabolism , Myoblasts/cytology , Cell Line , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Ubiquitination , Humans , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics
17.
FASEB J ; 38(13): e23797, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38963344

ABSTRACT

The role of N-glycosylation in the myogenic process remains poorly understood. Here, we evaluated the impact of N-glycosylation inhibition by Tunicamycin (TUN) or by phosphomannomutase 2 (PMM2) gene knockdown, which encodes an enzyme essential for catalyzing an early step of the N-glycosylation pathway, on C2C12 myoblast differentiation. The effect of chronic treatment with TUN on tibialis anterior (TA) and extensor digitorum longus (EDL) muscles of WT and MLC/mIgf-1 transgenic mice, which overexpress muscle Igf-1Ea mRNA isoform, was also investigated. TUN-treated and PMM2 knockdown C2C12 cells showed reduced ConA, PHA-L, and AAL lectin binding and increased ER-stress-related gene expression (Chop and Hspa5 mRNAs and s/uXbp1 ratio) compared to controls. Myogenic markers (MyoD, myogenin, and Mrf4 mRNAs and MF20 protein) and myotube formation were reduced in both TUN-treated and PMM2 knockdown C2C12 cells. Body and TA weight of WT and MLC/mIgf-1 mice were not modified by TUN treatment, while lectin binding slightly decreased in the TA muscle of WT (ConA and AAL) and MLC/mIgf-1 (ConA) mice. The ER-stress-related gene expression did not change in the TA muscle of WT and MLC/mIgf-1 mice after TUN treatment. TUN treatment decreased myogenin mRNA and increased atrogen-1 mRNA, particularly in the TA muscle of WT mice. Finally, the IGF-1 production and IGF1R signaling pathways activation were reduced due to N-glycosylation inhibition in TA and EDL muscles. Decreased IGF1R expression was found in TUN-treated C2C12 myoblasts which was associated with lower IGF-1-induced IGF1R, AKT, and ERK1/2 phosphorylation compared to CTR cells. Chronic TUN-challenge models can help to elucidate the molecular mechanisms through which diseases associated with aberrant N-glycosylation, such as Congenital Disorders of Glycosylation (CDG), affect muscle and other tissue functions.


Subject(s)
Cell Differentiation , Endoplasmic Reticulum Chaperone BiP , Muscle, Skeletal , Myoblasts , Receptor, IGF Type 1 , Signal Transduction , Tunicamycin , Animals , Mice , Glycosylation , Myoblasts/metabolism , Endoplasmic Reticulum Chaperone BiP/metabolism , Tunicamycin/pharmacology , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/genetics , Muscle, Skeletal/metabolism , Muscle Development/physiology , Cell Line , Mice, Transgenic , Endoplasmic Reticulum Stress , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/genetics
18.
Biomed Pharmacother ; 177: 117041, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964182

ABSTRACT

The modification of RNA through the N6-methyladenosine (m6A) has emerged as a growing area of research due to its regulatory role in gene expression and various biological processes regulating the expression of genes. m6A RNA methylation is a post-transcriptional modification that is dynamic and reversible and found in mRNA, tRNA, rRNA, and other non-coding RNA of most eukaryotic cells. It is executed by special proteins known as "writers," which initiate methylation; "erasers," which remove methylation; and "readers," which recognize it and regulate the expression of the gene. Modification by m6A regulates gene expression by affecting the splicing, translation, stability, and localization of mRNA. Aging causes molecular and cellular damage, which forms the basis of most age-related diseases. The decline in skeletal muscle mass and functionality because of aging leads to metabolic disorders and morbidities. The inability of aged muscles to regenerate and repair after injury poses a great challenge to the geriatric populace. This review seeks to explore the m6A epigenetic regulation in the myogenesis and regeneration processes in skeletal muscle as well as the progress made on the m6A epigenetic regulation of aging skeletal muscles.


Subject(s)
Adenosine , Aging , Epigenesis, Genetic , Muscle, Skeletal , Humans , Aging/genetics , Aging/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Muscle, Skeletal/metabolism , Transcriptome , Muscle Development/genetics , Methylation
19.
Genes (Basel) ; 15(7)2024 Jun 23.
Article in English | MEDLINE | ID: mdl-39062608

ABSTRACT

Skeletal muscle is a complex organ essential for locomotion, posture, and metabolic health. This review explores our current knowledge of Mustn1, particularly in the development and function of skeletal muscle. Mustn1 expression originates from Pax7-positive satellite cells in skeletal muscle, peaks during around the third postnatal month, and is crucial for muscle fiber differentiation, fusion, growth, and regeneration. Clinically, Mustn1 expression is potentially linked to muscle-wasting conditions such as muscular dystrophies. Studies have illustrated that Mustn1 responds dynamically to injury and exercise. Notably, ablation of Mustn1 in skeletal muscle affects a broad spectrum of physiological aspects, including glucose metabolism, grip strength, gait, peak contractile strength, and myofiber composition. This review summarizes our current knowledge of Mustn1's role in skeletal muscle and proposes future research directions, with a goal of elucidating the molecular function of this regulatory gene.


Subject(s)
Muscle, Skeletal , Muscle, Skeletal/metabolism , Humans , Animals , Muscle Development/genetics , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/physiology
20.
Cell Mol Life Sci ; 81(1): 310, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39066929

ABSTRACT

Anatomically connected bones and muscles determine movement of the body. Forces exerted on muscles are then turned to bones to promote osteogenesis. The crosstalk between muscle and bone has been identified as mechanotransduction previously. In addition to the mechanical features, bones and muscles are also secretory organs which interact closely with one another through producing myokines and osteokines. Moreover, besides the mechanical features, other factors, such as nutrition metabolism, physiological rhythm, age, etc., also affect bone-muscle crosstalk. What's more, osteogenesis and myogenesis within motor system occur almost in parallel. Pathologically, defective muscles are always detected in bone associated diseases and induce the osteopenia, inflammation and abnormal bone metabolism, etc., through biomechanical or biochemical coupling. Hence, we summarize the study findings of bone-muscle crosstalk and propose potential strategies to improve the skeletal or muscular symptoms of certain diseases. Altogether, functional improvement of bones or muscles is beneficial to each other within motor system.


Subject(s)
Bone and Bones , Muscle, Skeletal , Humans , Bone and Bones/metabolism , Bone and Bones/pathology , Muscle, Skeletal/metabolism , Animals , Osteogenesis/physiology , Mechanotransduction, Cellular , Muscle Development
SELECTION OF CITATIONS
SEARCH DETAIL