Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.490
Filter
1.
J Physiol Sci ; 74(1): 51, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39369187

ABSTRACT

Advanced glycation end products (AGEs) are risk factors for various diseases, including sarcopenia. One of the deleterious effects of AGEs is the induction of abnormal reactive oxygen species (ROS) production in skeletal muscle. However, the underlying mechanism remains poorly understood. Therefore, the aim of this study was to elucidate how AGEs induce ROS production in skeletal muscle cells. This study demonstrated that AGEs treatment promoted ROS production in myoblasts and myotubes while PKC inhibitor abolished ROS production by AGEs stimulation. Phosphorylation of p47 phox by kinases such as PKCα is required to form the Nox2 complex, which induces ROS production. In this study, AGEs treatment promoted the phosphorylation of PKCα and p47 phox in myoblasts and myotubes. Our findings suggest that AGEs promote ROS production through the phosphorylation of PKCα and p47 phox in skeletal muscle cells.


Subject(s)
Glycation End Products, Advanced , Muscle Fibers, Skeletal , Muscle, Skeletal , NADPH Oxidases , Reactive Oxygen Species , Glycation End Products, Advanced/metabolism , Reactive Oxygen Species/metabolism , Animals , Mice , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/drug effects , NADPH Oxidases/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Phosphorylation , Myoblasts/metabolism , Myoblasts/drug effects , Protein Kinase C/metabolism , Cell Line , Protein Kinase C-alpha/metabolism , Signal Transduction/physiology , Signal Transduction/drug effects
2.
J Pineal Res ; 76(6): e13009, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39315577

ABSTRACT

Melatonin has been reported to regulate circadian rhythms and have anti-inflammatory characteristics in various inflammatory autoimmune diseases, but its effects in diseases-associated muscle atrophy remain controversial. This study is aimed to determine the evidence of melatonin in rheumatoid arthritis (RA)-related pathological muscle atrophy. We used initially bioinformatics results to show that melatonin regulated significantly the correlation between pro-inflammation and myogenesis in RA synovial fibroblasts (RASF) and myoblasts. The conditioned medium (CM) from melatonin-treated RASF was incubated in myoblasts with growth medium and differentiated medium to investigate the markers of pro-inflammation, atrophy, and myogenesis. We found that melatonin regulated RASF CM-induced pathological muscle pro-inflammation and atrophy in myoblasts and differentiated myocytes through NF-κB signaling pathways. We also showed for the first time that miR-30c-1-3p is negatively regulated by three inflammatory cytokines in human RASF, which is associated with murine-differentiated myocytes. Importantly, oral administration with melatonin in a collagen-induced arthritis (CIA) mouse model also significantly improved arthritic swelling, hind limb grip strength as well as pathological muscle atrophy. In conclusion, our study is the first to demonstrate not only the underlying mechanism whereby melatonin decreases pro-inflammation in RA-induced pathological muscle atrophy but also increases myogenesis in myoblasts and differentiated myocytes.


Subject(s)
Arthritis, Rheumatoid , Fibroblasts , Melatonin , Muscle, Skeletal , Melatonin/pharmacology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/drug therapy , Humans , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Animals , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/drug effects , Inflammation/metabolism , Inflammation/pathology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synovial Membrane/drug effects , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Experimental/drug therapy , Male , Myoblasts/metabolism , Myoblasts/drug effects , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/drug therapy , Mice, Inbred DBA
3.
Mol Med Rep ; 30(5)2024 Nov.
Article in English | MEDLINE | ID: mdl-39301652

ABSTRACT

Ginkgolic acid (GA), isolated from the leaves and seed coats of Ginkgo biloba, exerts several biological effects, including antitumor, antibacterial, anti­HIV and anti­inflammatory effects. However, the effects of GA on C2C12 myoblasts remain unclear. The present study assessed cell viability with the MTT assay and evaluated colony formation through crystal violet staining. Flow cytometry was used to analyze apoptosis with Annexin V/7­AAD staining, proliferation with Ki67 staining and cell cycle arrest. Western blotting detected myogenic markers and other relevant proteins. Myotube formation was examined by immunofluorescence, and autophagy was measured using an LC3 antibody­based kit via flow cytometry. The present study showed that treatment of C2C12 cells with GA significantly inhibited their viability and colony formation capacity but did not trigger apoptosis, as indicated by Annexin V/7­AAD staining. However, Ki67 staining indicates that GA exerted dose­dependent antiproliferative effects. Further analysis revealed that GA partially inhibited the growth of C2C12 cells via cell cycle arrest in S phase, highlighting its role in the disruption of cell proliferation. Furthermore, treatment with GA impaired myoblast differentiation, as evidenced by a reduction in the expression of the myogenesis markers, the myosin­heavy chain, myoblast determination protein 1 and myogenin, and suppressed myotube formation. Notably, during C2C12 cell differentiation, GA promoted apoptosis without affecting cell cycle progression or Ki67 expression. Mechanistically, GA could suppress nuclear extracellular signal­regulated kinase phosphorylation, suggesting that it modulates cell proliferation pathways. Moreover, GA triggered autophagy in differentiated C2C12 cells, as confirmed by elevated LC3 II levels. These findings highlight the multifaceted effects of GA on C2C12 cells.


Subject(s)
Apoptosis , Autophagy , Cell Differentiation , Cell Proliferation , Muscle Development , Myoblasts , Salicylates , Animals , Cell Differentiation/drug effects , Mice , Myoblasts/metabolism , Myoblasts/drug effects , Myoblasts/cytology , Cell Proliferation/drug effects , Muscle Development/drug effects , Cell Line , Autophagy/drug effects , Apoptosis/drug effects , Salicylates/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects
4.
Mol Cell Endocrinol ; 593: 112336, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39094930

ABSTRACT

Steroidogenesis occurs locally in peripheral tissues and via adrenal and gonadal glands' biosynthesis. The C2C12 mouse myoblast cell line and rat skeletal muscles harbor a local steroidogenesis pathway for glucocorticoids, and corticosterone is biosynthesized from skeletal muscle cells. However, Cyp11a1 and StAR protein expressions are not observed in C2C12 cells or rat muscular tissues. In this context, this study investigated the relationship between DNA methylation and key steroidogenic genes. Bioinformatics analysis of methylated DNA immune precipitation showed that C2C12 myoblasts and myotubes did not have remarkable DNA methylated regions in the gene-body of Cyp11a1. However, a highly methylated region in the CpG island was detected in the intronic enhancer of Ad4BP/SF-1, known as the transcriptional factor for steroidogenic genes. After C2C12 myoblasts treatment with 5-aza-2-deoxycytidine, the gene expressions of Ad4BP/SF-1, Cyp11a1, and StAR were significantly time- and concentration-dependent upregulated. To clarify the contribution of Ad4BP/SF-1 on Cyp11a1 and StAR transcripts, we silenced Ad4BP/SF-1 during the 5-aza-2-deoxycytidine treatment in C2C12 myoblasts, resulting in significant suppression of both Cyp11a1 and StAR. Additionally, pregnenolone levels in the supernatants of C2C12 cells were enhanced by 5-aza-2-deoxycytidine treatment, whereas pregnenolone production by C2C12 myoblasts was significantly suppressed by Ad4BP/SF-1 knockdown. These results indicate that DNA methylation of Ad4BP/SF-1 might be involved in the downregulation of steroidogenic genes, such as Cyp11a1 and StAR in C2C12 myoblasts.


Subject(s)
Cholesterol Side-Chain Cleavage Enzyme , CpG Islands , DNA Methylation , Myoblasts , Phosphoproteins , Animals , Mice , Rats , Azacitidine/pharmacology , Cell Line , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , CpG Islands/genetics , Decitabine/pharmacology , DNA Methylation/genetics , DNA Methylation/drug effects , Gene Expression Regulation/drug effects , Myoblasts/metabolism , Myoblasts/drug effects , Phosphoproteins/metabolism , Phosphoproteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Pharmacol Res ; 208: 107376, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39216837

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked recessive progressive degenerative disease of skeletal muscle, characterized by intramuscular inflammation, muscle regeneration disorder and replacement of muscle with fibroadipose tissue. DMD is caused by the absence of normal dystrophy. Impaired self-renew ability and limited differentiation capacity of satellite cells are proved as main reasons for muscle regeneration failure. The deficiency of estrogen impedes the process of muscle regeneration. However, the role of estrogen receptor ß (ERß) in muscle regeneration is still unclear. This study aims to investigate the role and the pharmacological effect of ERß activation on muscle regeneration in mdx mice. This study showed that mRNA levels of ERß and myogenic-related genes both witnessed increasing trends in dystrophic context. Our results revealed that treatment with selective ERß agonist (DPN, diarylpropionitrile) significantly increased myogenic differentiation 1 (MyoD-1) level and promoted muscle regeneration in mdx mice. Similarly, in mdx mice with muscle-specific estrogen receptor α (ERα) ablation, DPN treatment still promoted muscle regeneration. Moreover, we demonstrated that myoblasts differentiation was accompanied by raised nuclear accumulation of ERß. DPN treatment augmented the nuclear accumulation of ERß and, thus, contributed to myotubes formation. One important finding was that forkhead box O3A (FOXO3A), as a pivotal transcription factor in Myod-1 transcription, participated in the ERß-promoted muscle regeneration. Overall, we offered an interesting explanation about the crucial role of ERß during myogenesis.


Subject(s)
Estrogen Receptor beta , Forkhead Box Protein O3 , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal , Muscular Dystrophy, Duchenne , MyoD Protein , Nitriles , Propionates , Regeneration , Animals , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrogen Receptor beta/agonists , MyoD Protein/genetics , MyoD Protein/metabolism , Regeneration/drug effects , Forkhead Box Protein O3/metabolism , Forkhead Box Protein O3/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nitriles/pharmacology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Mice , Propionates/pharmacology , Male , Muscle Development/drug effects , Cell Nucleus/metabolism , Cell Nucleus/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Cell Differentiation/drug effects
6.
Life Sci ; 354: 122941, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39098595

ABSTRACT

AIMS: Study of the role of mitochondria-generated reactive oxygen species (mtROS) and mitochondrial polarization in mitochondrial fragmentation at the initial stages of myogenesis. MAIN METHODS: Mitochondrial morphology, Drp1 protein phosphorylation, mitochondrial electron transport chain components content, mtROS and mitochondrial lipid peroxidation levels, and mitochondrial polarization were evaluated on days 1 and 2 of human MB135 myoblasts differentiation. A mitochondria-targeted antioxidant SkQ1 was used to elucidate the effect of mtROS on mitochondria. KEY FINDINGS: In immortalized human MB135 myoblasts, mitochondrial fragmentation began on day 1 of differentiation before the myoblast fusion. This fragmentation was preceded by dephosphorylation of p-Drp1 (Ser-637). On day 2, an increase in the content of some mitochondrial proteins was observed, indicating mitochondrial biogenesis stimulation. Furthermore, we found that myogenic differentiation, even on day 1, was accompanied both by an increased production of mtROS, and lipid peroxidation of the inner mitochondrial membrane. SkQ1 blocked these effects and partially reduced the level of mitochondrial fragmentation, but did not affect the dephosphorylation of p-Drp1 (Ser-637). Importantly, mitochondrial fragmentation at early stages of MB135 differentiation was not accompanied by depolarization, as an important stimulus for mitochondrial fragmentation. SIGNIFICANCE: Mitochondrial fragmentation during early myogenic differentiation depends on mtROS production rather than mitochondrial depolarization. SkQ1 only partially inhibited mitochondrial fragmentation, without significant effects on mitophagy or early myogenic differentiation.


Subject(s)
Cell Differentiation , Lipid Peroxidation , Mitochondria , Myoblasts , Reactive Oxygen Species , Humans , Reactive Oxygen Species/metabolism , Cell Differentiation/drug effects , Myoblasts/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Lipid Peroxidation/drug effects , Muscle Development/physiology , Muscle Development/drug effects , Membrane Potential, Mitochondrial/drug effects , Dynamins/metabolism , Phosphorylation , Cell Line
7.
Elife ; 132024 Aug 28.
Article in English | MEDLINE | ID: mdl-39196610

ABSTRACT

Senescent cells are characterized by multiple features such as increased expression of senescence-associated ß-galactosidase activity (SA ß-gal) and cell cycle inhibitors such as p21 or p16. They accumulate with tissue damage and dysregulate tissue homeostasis. In the context of skeletal muscle, it is known that agents used for chemotherapy such as Doxorubicin (Doxo) cause buildup of senescent cells, leading to the inhibition of tissue regeneration. Senescent cells influence the neighboring cells via numerous secreted factors which form the senescence-associated secreted phenotype (SASP). Lipids are emerging as a key component of SASP that can control tissue homeostasis. Arachidonic acid-derived lipids have been shown to accumulate within senescent cells, specifically 15d-PGJ2, which is an electrophilic lipid produced by the non-enzymatic dehydration of the prostaglandin PGD2. This study shows that 15d-PGJ2 is also released by Doxo-induced senescent cells as an SASP factor. Treatment of skeletal muscle myoblasts with the conditioned medium from these senescent cells inhibits myoblast fusion during differentiation. Inhibition of L-PTGDS, the enzyme that synthesizes PGD2, diminishes the release of 15d-PGJ2 by senescent cells and restores muscle differentiation. We further show that this lipid post-translationally modifies Cys184 of HRas in C2C12 mouse skeletal myoblasts, causing a reduction in the localization of HRas to the Golgi, increased HRas binding to Ras Binding Domain (RBD) of RAF Kinase (RAF-RBD), and activation of cellular Mitogen Activated Protein (MAP) kinase-Extracellular Signal Regulated Kinase (Erk) signaling (but not the Akt signaling). Mutating C184 of HRas prevents the ability of 15d-PGJ2 to inhibit the differentiation of muscle cells and control the activity of HRas. This work shows that 15d-PGJ2 released from senescent cells could be targeted to restore muscle homeostasis after chemotherapy.


Subject(s)
Cell Differentiation , Cellular Senescence , Myoblasts , Prostaglandin D2 , Proto-Oncogene Proteins p21(ras) , Animals , Mice , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Prostaglandin D2/pharmacology , Cellular Senescence/drug effects , Myoblasts/metabolism , Myoblasts/drug effects , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Cell Differentiation/drug effects , Senescence-Associated Secretory Phenotype , Cell Line , Doxorubicin/pharmacology
8.
Phytomedicine ; 134: 155964, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39168012

ABSTRACT

BACKGROUND: High glucose levels are a primary cause of diabetes-associated cellular dysfunction and tissue damage. Muscles are the key insulin target organ and therefore, have a high level of sensitivity to hyperglycemia. Our previous study revealed that 20(S)-ginsenoside Rg3 (S-Rg3) is a monomer with a good myogenic differentiation effect in ginsenoside. Furthermore, it can alleviate dexamethasone-induced muscle atrophy by protecting mitochondrial function. However, whether S-Rg3 is effective for diabetic-induced muscle atrophy has not been reported. PURPOSE: This study aimed to investigate the protective effect of S-Rg3 on diabetic-induced muscle atrophy. METHODS: C2C12 myoblasts, Drosophila, and mice were used as model systems, and the protective effect of S-Rg3 on diabetes was evaluated by assessing the levels of glucose and lipids. Furthermore, H&E, toluidine blue, Giemsa, and immunofluorescence staining were performed to detect the effects of S-Rg3 on muscle atrophy and myogenic differentiation. Moreover, the effects of S-Rg3 on mitochondrial morphology and function were also evaluated by electron microscopy, flow cytometry, and Seahorse. In addition, the underlying pathways of S-Rg3 effects were detected by Western blot. The related inhibitors and gene mutations in Drosophila were used for validation. RESULTS: The analysis of diabetic mice model fed with a high-fat diet (HFD) and high glucose (HG) revealed that in the injured C2C12 myoblasts, S-Rg3 treatment significantly reduced the levels of triglycerides and glucose. Furthermore, it promoted the differentiation of myoblasts and inhibited mitochondrial dysfunction. In the Drosophila HG and HFD diabetic model, S-Rg3 reduced triglyceride and trehalose levels, increased climbing distance values, promoted myoblasts differentiation, preserved mitochondrial function, and inhibited muscle atrophy. Mechanistically, the beneficial effects of S-Rg3 were at least partially associated with the phosphorylation of AMPK and FoxO3 together with the inhibition of Smad3 phosphorylation, this pathway was validated by the UAS-AMPKα-RNAi Drosophila model. CONCLUSION: In summary, this study revealed mechanistic insights into how S-Rg3 protects against diabetes-associated muscle atrophy in cells, Drosophila, and mice.


Subject(s)
Cell Differentiation , Diabetes Mellitus, Experimental , Ginsenosides , Mitochondria , Muscular Atrophy , Myoblasts , Animals , Ginsenosides/pharmacology , Mice , Myoblasts/drug effects , Muscular Atrophy/drug therapy , Muscular Atrophy/prevention & control , Cell Differentiation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Mitochondria/drug effects , Mitochondria/metabolism , Male , Cell Line , Mice, Inbred C57BL , Drosophila , Drosophila melanogaster/drug effects
9.
J Cachexia Sarcopenia Muscle ; 15(5): 1834-1849, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38982896

ABSTRACT

BACKGROUND: Skeletal muscle injury is one of the most common sports injuries; if not properly treated or not effective rehabilitation treatment after injury, it can be transformed into chronic cumulative injury. Curcumin, an herbal ingredient, has been found to promote skeletal muscle injury repair and regeneration. The Wnt5a pathway is related to the expression of myogenic regulatory factors, and Ca2+ promotes the differentiation and fusion process of myoblasts. This study explored the effect and mechanism of curcumin on myoblast differentiation during the repair and regeneration of injured skeletal muscle and its relationship with the Wnt5a pathway and Ca2+ channel. METHODS: Myogenic differentiation of C2C12 cells was induced with 2% horse serum, and a mouse (male, 10 weeks old) model of acute skeletal muscle injury was established using cardiotoxin (20 µL). In addition, we constructed a Wnt5a knockdown C2C12 cell model and a Wnt5a knockout mouse model. Besides, curcumin was added to the cell culture solution (80 mg/L) and fed to the mice (50 mg/kg). Fluorescence microscopy was used to determine the concentration of Ca2+. Western blot and RT-qPCR were used to detect the protein and mRNA levels of Wnt5a, CaN, NFAT2, MyoD, Myf5, Pax7, and Myogenin. The expression levels of MyoD, Myf5, Myogenin, MHC, Desmin, and NFAT2 were detected using immunofluorescence techniques. In addition, MyoD expression was observed using immunohistochemistry, and morphological changes in mouse muscle tissue were observed using HE staining. RESULTS: During myoblast differentiation and muscle regeneration, Wnt5a expression was upregulated (P < 0.001) and the Wnt5a signalling pathway was activated. Wnt5a overexpression promoted the expression of MyoD, Myf5, Myogenin, MHC, and Desmin (P < 0.05), and conversely, knockdown of Wnt5a inhibited their expression (P < 0.001). The Wnt5a pathway mediated the opening of Ca2+ channels, regulated the expression levels of CaN, NFAT2, MyoD, Myf5, Myogenin, MHC, and Desmin (P < 0.01) and promoted the differentiation of C2C12 myoblasts and the repair and regeneration of injured skeletal muscle. The expression of Wnt5a, CaN, NFAT2, MyoD, Myogenin, Myf5, and MHC in C2C12 myoblast was significantly increased after curcumin intervention (P < 0.05); however, their expression decreased significantly after knocking down Wnt5a on the basis of curcumin intervention (P < 0.05). Similarly, in Wnt5a knockout mice, the promotion of muscle regeneration by curcumin was significantly attenuated. CONCLUSIONS: Curcumin can activate the Wnt5a signalling pathway and mediate the opening of Ca2+ channels to accelerate the myogenic differentiation of C2C12 cells and the repair and regeneration of injured skeletal muscle.


Subject(s)
Cell Differentiation , Curcumin , Muscle, Skeletal , Myoblasts , Regeneration , Wnt-5a Protein , Animals , Wnt-5a Protein/metabolism , Mice , Cell Differentiation/drug effects , Regeneration/drug effects , Myoblasts/metabolism , Myoblasts/drug effects , Curcumin/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Calcium Channels/metabolism , Male , Muscle Development/drug effects , Signal Transduction/drug effects , Cell Line
10.
J Proteome Res ; 23(8): 3444-3459, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39024330

ABSTRACT

Ferroptosis adversely affects the viability, differentiation, and metabolic integrity of C2C12 myoblasts, contributing to the decline in skeletal muscle health. The intricate mechanisms behind this process are not fully understood. In this study, we induced ferroptosis in myoblasts using targeted inducers and found a marked decrease in specific redox metabolites, particularly taurine. Taurine supplementation effectively reversed the deleterious effects of ferroptosis, significantly increased cellular glutathione levels, reduced MDA and ROS levels, and rejuvenated impaired myogenic differentiation. Furthermore, taurine downregulated HO-1 expression and decreased intracellular Fe2+ levels, thereby stabilizing the labile iron pool. Using NMR metabolomic analysis, we observed that taurine profoundly promoted glycerophospholipid metabolism, which is critical for cell membrane repair, and enhanced mitochondrial bioenergetics, thereby increasing the energy reserves essential for muscle satellite cell regeneration. These results suggest that taurine is a potent ferroptosis inhibitor that attenuates key drivers of this process, strengthens oxidative defenses, and improves redox homeostasis. This combined effect protects cells from ferroptosis-induced damage. This study highlights the potential of taurine as a valuable ferroptosis inhibitor that protects skeletal muscle from ferroptosis-induced damage and provides a basis for therapeutic strategies to rejuvenate and facilitate the regeneration of aging skeletal muscle.


Subject(s)
Ferroptosis , Homeostasis , Iron , Myoblasts , Oxidation-Reduction , Taurine , Taurine/pharmacology , Ferroptosis/drug effects , Oxidation-Reduction/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/cytology , Iron/metabolism , Animals , Mice , Homeostasis/drug effects , Cell Line , Reactive Oxygen Species/metabolism , Cell Differentiation/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Glutathione/metabolism , Oxidative Stress/drug effects , Glycerophospholipids/metabolism
11.
Cell Death Dis ; 15(7): 470, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956034

ABSTRACT

The present study aims to develop and characterize a controlled-release delivery system for protein therapeutics in skeletal muscle regeneration following an acute injury. The therapeutic protein, a membrane-GPI anchored protein called Cripto, was immobilized in an injectable hydrogel delivery vehicle for local administration and sustained release. The hydrogel was made of poly(ethylene glycol)-fibrinogen (PEG-Fibrinogen, PF), in the form of injectable microspheres. The PF microspheres exhibited a spherical morphology with an average diameter of approximately 100 micrometers, and the Cripto protein was uniformly entrapped within them. The release rate of Cripto from the PF microspheres was controlled by tuning the crosslinking density of the hydrogel, which was varied by changing the concentration of poly(ethylene glycol) diacrylate (PEG-DA) crosslinker. In vitro experiments confirmed a sustained-release profile of Cripto from the PF microspheres for up to 27 days. The released Cripto was biologically active and promoted the in vitro proliferation of mouse myoblasts. The therapeutic effect of PF-mediated delivery of Cripto in vivo was tested in a cardiotoxin (CTX)-induced muscle injury model in mice. The Cripto caused an increase in the in vivo expression of the myogenic markers Pax7, the differentiation makers eMHC and Desmin, higher numbers of centro-nucleated myofibers and greater areas of regenerated muscle tissue. Collectively, these results establish the PF microspheres as a potential delivery system for the localized, sustained release of therapeutic proteins toward the accelerated repair of damaged muscle tissue following acute injuries.


Subject(s)
Delayed-Action Preparations , Muscle, Skeletal , Polyethylene Glycols , Animals , Muscle, Skeletal/metabolism , Muscle, Skeletal/injuries , Muscle, Skeletal/drug effects , Mice , Polyethylene Glycols/chemistry , Microspheres , Fibrinogen/metabolism , Hydrogels/chemistry , Regeneration/drug effects , Myoblasts/metabolism , Myoblasts/drug effects , Humans , Cell Proliferation/drug effects , PAX7 Transcription Factor/metabolism , Male , Mice, Inbred C57BL , Muscular Diseases/drug therapy , Muscular Diseases/pathology , Muscular Diseases/metabolism
12.
Nutrients ; 16(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38892515

ABSTRACT

Fructose is a commonly consumed monosaccharide implicated in developing several metabolic diseases. Previously, elevated branched-chain amino acids (BCAA) have been correlated with the severity of insulin resistance. Most recently, the effect of fructose consumption on the downregulation of BCAA catabolic enzymes was observed. Thus, this mechanistic study investigated the effects of physiologically attainable levels of fructose, both with and without concurrent insulin resistance, in a myotube model of skeletal muscle. METHODS: C2C12 mouse myoblasts were treated with fructose at a concentration of 100 µM (which approximates physiologically attainable concentrations in peripheral circulation) both with and without hyperinsulinemic-mediated insulin resistance. Gene expression was assessed by qRT-PCR, and protein expression was assessed by Western blot. Oxygen consumption rate and extracellular acidification rate were used to assess mitochondrial oxidative and glycolytic metabolism, respectively. Liquid chromatography-mass spectrometry was utilized to analyze leucine, isoleucine and valine concentration values. RESULTS: Fructose significantly reduced peak glycolytic and peak mitochondrial metabolism without altering related gene or protein expression. Similarly, no effect of fructose on BCAA catabolic enzymes was observed; however, fructose treatment resulted in elevated total extracellular BCAA in insulin-resistant cells. DISCUSSION: Collectively, these observations demonstrate that fructose at physiologically attainable levels does not appear to alter insulin sensitivity or BCAA catabolic potential in cultured myotubes. However, fructose may depress peak cell metabolism and BCAA utilization during insulin resistance.


Subject(s)
Amino Acids, Branched-Chain , Fructose , Insulin Resistance , Muscle Fibers, Skeletal , Animals , Fructose/pharmacology , Amino Acids, Branched-Chain/metabolism , Mice , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/drug effects , Cell Line , Mitochondria/metabolism , Mitochondria/drug effects , Glycolysis/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Oxygen Consumption/drug effects
13.
Int J Med Sci ; 21(8): 1461-1471, 2024.
Article in English | MEDLINE | ID: mdl-38903922

ABSTRACT

Dasatinib is one of the second-generation tyrosine kinase inhibitors used to treat chronic myeloid leukemia and has a broad target spectrum, including KIT, PDGFR, and SRC family kinases. Due to its broad drug spectrum, dasatinib has been reported at the basic research level to improve athletic performance by eliminating senescent cell removal and to have an effect on muscle diseases such as Duchenne muscular dystrophy, but its effect on myoblasts has not been investigated. In this study, we evaluated the effects of dasatinib on skeletal muscle both under normal conditions and in the regenerating state. Dasatinib suppressed the proliferation and promoted the fusion of C2C12 myoblasts. During muscle regeneration, dasatinib increased the gene expressions of myogenic-related genes (Myod, Myog, and Mymx), and caused abnormally thin muscle fibers on the CTX-induced muscle injury mouse model. From these results, dasatinib changes the closely regulated gene expression pattern of myogenic regulatory factors during muscle differentiation and disrupts normal muscle regeneration. Our data suggest that when using dasatinib, its effects on skeletal muscle should be considered, particularly at regenerating stages.


Subject(s)
Cell Differentiation , Dasatinib , Muscle Development , Muscle, Skeletal , Myoblasts , Regeneration , Dasatinib/pharmacology , Animals , Mice , Regeneration/drug effects , Cell Differentiation/drug effects , Muscle Development/drug effects , Muscle Development/genetics , Muscle, Skeletal/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/cytology , Cell Proliferation/drug effects , Humans , Cell Line , Protein Kinase Inhibitors/pharmacology
14.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928510

ABSTRACT

The decline in the function and mass of skeletal muscle during aging or other pathological conditions increases the incidence of aging-related secondary diseases, ultimately contributing to a decreased lifespan and quality of life. Much effort has been made to surmise the molecular mechanisms underlying muscle atrophy and develop tools for improving muscle function. Enhancing mitochondrial function is considered critical for increasing muscle function and health. This study is aimed at evaluating the effect of an aqueous extract of Gloiopeltis tenax (GTAE) on myogenesis and muscle atrophy caused by dexamethasone (DEX). The GTAE promoted myogenic differentiation, accompanied by an increase in peroxisome proliferator-activated receptor γ coactivator α (PGC-1α) expression and mitochondrial content in myoblast cell culture. In addition, the GTAE alleviated the DEX-mediated myotube atrophy that is attributable to the Akt-mediated inhibition of the Atrogin/MuRF1 pathway. Furthermore, an in vivo study using a DEX-induced muscle atrophy mouse model demonstrated the efficacy of GTAE in protecting muscles from atrophy and enhancing mitochondrial biogenesis and function, even under conditions of atrophy. Taken together, this study suggests that the GTAE shows propitious potential as a nutraceutical for enhancing muscle function and preventing muscle wasting.


Subject(s)
Dexamethasone , Muscle Development , Muscular Atrophy , Plant Extracts , Animals , Muscular Atrophy/chemically induced , Muscular Atrophy/metabolism , Muscular Atrophy/drug therapy , Muscular Atrophy/pathology , Dexamethasone/adverse effects , Dexamethasone/pharmacology , Muscle Development/drug effects , Mice , Plant Extracts/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Cell Differentiation/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Cell Line , Muscle Proteins/metabolism , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Mice, Inbred C57BL , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Rhodophyta
15.
Tissue Cell ; 89: 102423, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38875923

ABSTRACT

Skeletal muscle function is highly dependent on the energy supply provided by mitochondria. Besides ATP production, mitochondria have several other roles, such as calcium storage, heat production, cell death signaling, autophagy regulation and redox state modulation. Mitochondrial function is crucial for skeletal muscle fiber formation. Disorders that affect mitochondria have a major impact in muscle development and function. Here we studied the role of mitochondria during chick skeletal myogenesis. We analyzed the intracellular distribution of mitochondria in myoblasts, fibroblasts and myotubes using Mitotracker labeling. Mitochondrial respiration was investigated in chick muscle cells. Our results show that (i) myoblasts and myotubes have more mitochondria than muscle fibroblasts; (ii) mitochondria are organized in long lines within the whole cytoplasm and around the nuclei of myotubes, while in myoblasts they are dispersed in the cytoplasm; (iii) the area of mitochondria in myotubes increases during myogenesis, while in myoblasts and fibroblasts there is a slight decrease; (iv) mitochondrial length increases in the three cell types (myoblasts, fibroblasts and myotubes) during myogenesis; (v) the distance of mitochondria to the nucleus increases in myoblasts and myotubes during myogenesis; (vi) Rotenone inhibits muscle fiber formation, while FCCP increases the size of myotubes; (vii) N-acetyl cysteine (NAC), an inhibitor of ROS formation, rescues the effects of Rotenone on muscle fiber size; and (viii) Rotenone induces the production of ROS in chick myogenic cells. The collection of our results suggests a role of ROS signaling in mitochondrial function during chick myogenesis.


Subject(s)
Muscle Development , Muscle Fibers, Skeletal , Myoblasts , Reactive Oxygen Species , Rotenone , Animals , Reactive Oxygen Species/metabolism , Muscle Development/drug effects , Chick Embryo , Rotenone/pharmacology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/cytology , Myoblasts/metabolism , Myoblasts/drug effects , Myoblasts/cytology , Fibroblasts/metabolism , Fibroblasts/drug effects , Mitochondria/metabolism , Mitochondria/drug effects
16.
Mol Nutr Food Res ; 68(14): e2300685, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38860356

ABSTRACT

SCOPE: Kaempferol (KMP), a bioactive flavonoid compound found in fruits and vegetables, contributes to human health in many ways but little is known about its relationship with muscle mass. The effect of KMP on C2C12 myoblast differentiation and the mechanisms that might underlie that effect are studied. METHODS AND RESULTS: This study finds that KMP (1, 10 µM) increases the migration and differentiation of C2C12 myoblasts in vitro. Studying the possible mechanism underlying its effect on migration, the study finds that KMP activates Integrin Subunit Beta 1 (ITGB1) in C2C12 myoblasts, increasing p-FAK (Tyr398) and its downstream cell division cycle 42 (CDC42), a protein previously associated with cell migration. Regarding differentiation, KMP upregulates the expression of myosin heavy chain (MHC) and activates IGF1/AKT/mTOR/P70S6K. Interestingly, pretreatment with an AKT inhibitor (LY294002) and siRNA knockdown of IGF1R leads to a decrease in cell differentiation, suggesting that IGF1/AKT activation is required for KMP to induce C2C12 myoblast differentiation. CONCLUSION: Together, the findings suggest that KMP enhances the migration and differentiation of C2C12 myoblasts through the ITG1B/FAK/paxillin and IGF1R/AKT/mTOR pathways. Thus, KMP supplementation might potentially be used to prevent or delay age-related loss of muscle mass and help maintain muscle health.


Subject(s)
Cell Differentiation , Cell Movement , Integrin beta1 , Kaempferols , Myoblasts , Paxillin , Proto-Oncogene Proteins c-akt , Receptor, IGF Type 1 , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Kaempferols/pharmacology , TOR Serine-Threonine Kinases/metabolism , Mice , Proto-Oncogene Proteins c-akt/metabolism , Cell Differentiation/drug effects , Signal Transduction/drug effects , Cell Movement/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Integrin beta1/metabolism , Paxillin/metabolism , Cell Line , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/genetics , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics
17.
Ecotoxicol Environ Saf ; 281: 116607, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908055

ABSTRACT

Deoxynivalenol (DON), commonly known as vomitoxin, is a mycotoxin produced by fungi and is frequently found as a contaminant in various cereal-based food worldwide. While the harmful effects of DON have been extensively studied in different tissues, its specific impact on the proliferation of skeletal muscle cells remains unclear. In this study, we utilized murine C2C12 myoblasts as a model to explore the influence of DON on their proliferation. Our observations indicated that DON exhibits dose-dependent toxicity, significantly inhibiting the proliferation of C2C12 cells. Through the application of RNA-seq analysis combined with gene set enrichment analysis, we identified a noteworthy downregulation of genes linked to the extracellular matrix (ECM) and condensed chromosome. Concurrently with the reduced expression of ECM genes, immunostaining analysis revealed notable changes in the distribution of fibronectin, a vital ECM component, condensing into clusters and punctate formations. Remarkably, the exposure to DON induced the formation of multipolar spindles, leading to the disruption of the normal cell cycle. This, in turn, activated the p53-p21 signaling pathway and ultimately resulted in apoptosis. These findings contribute significant insights into the mechanisms through which DON induces toxicity within skeletal muscle cells.


Subject(s)
Apoptosis , Myoblasts , Trichothecenes , Animals , Trichothecenes/toxicity , Apoptosis/drug effects , Mice , Myoblasts/drug effects , Cell Line , Mitosis/drug effects , Cell Proliferation/drug effects , Signal Transduction/drug effects , Extracellular Matrix/drug effects
18.
SLAS Discov ; 29(5): 100171, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38917882

ABSTRACT

DNA-encoded small molecule library technology has recently emerged as a new paradigm for identifying ligands against drug targets. To date, it has been used to identify ligands against targets that are soluble or overexpressed on cell surfaces. Here, we report applying cell-based selection methods to profile surfaces of mouse C2C12 myoblasts and myotube cells in an unbiased, target agnostic manner. A panel of on-DNA compounds were identified and confirmed for cell binding selectivity. We optimized the cell selection protocol and employed a novel data analysis method to identify cell selective ligands against a panel of human B and T lymphocytes. We discuss the generality of using this workflow for DNA encoded small molecule library selection and data analysis against different cell types, and the feasibility of applying this method to profile cell surfaces for biomarker and target identification.


Subject(s)
Small Molecule Libraries , Humans , Animals , Mice , Small Molecule Libraries/pharmacology , Ligands , Cell Line , Myoblasts/drug effects , Myoblasts/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Drug Discovery/methods , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Membrane/metabolism , Cell Membrane/drug effects
19.
Phytomedicine ; 130: 155718, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38795694

ABSTRACT

BACKGROUND: Senile osteoporosis (SOP) is an age-related systemic metabolic bone disorder. Previous studies have proved that Zhuang-Gu-Fang (ZGF) modulates myokines, stimulates osteogenic differentiation, and mitigates osteoporosis. OBJECTIVE: To elucidate the mechanism by which ZGF promotes osteogenic differentiation via myoblast and myoblast exosomal microRNAs (miRNAs) and investigate its potential implications in senile osteoporosis. METHODS: Characterization of ZGF and ZGF serum using UHPLC-MS/MS. An alkaline phosphatase (ALP) activity assay and staining techniques were employed to corroborate the impacts of ZGF on the osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) via myoblasts. Subsequently, exosomes derived from myoblasts were isolated through ultracentrifugation. The effects of ZGF on the BMSCs' osteogenic differentiation were substantiated through ALP activity, alizarin red staining, and a quantitative real-time polymerase reaction system (qRT-PCR). Selected miRNAs were identified via high-throughput sequencing and subjected to differential expression analysis, and subsequently validated through qRT-PCR. The senescence-accelerated (SAMP6) mice were selected as the SOP models. qRT-PCR analyses were further conducted to confirm the expression levels of these selected miRNAs in the muscle and bone tissues of the SAMP6 mice, and the protein expression of osteogenesis-related transcription factors OCN and Osterix in its bone tissue was evaluated by immunofluorescence staining analysis (IF). RESULTS: ZGF may enhance the osteogenic differentiation of BMSCs through myoblasts and myoblast-derived exosomes. High-throughput sequencing, differential expression analysis, and subsequent qRT-PCR validation identified four miRNAs that stood out due to their significant differential expression: miR-5100, miR-142a-3p, miR-126a-3p, miR-450b-5p and miR-669a-5p. Moreover, the mice experiment corroborated these findings, which revealed that ZGF not only up-regulated the expression of miR-5100, miR-450b-5p and miR-126a-3p in muscle and bone tissues but also concurrently down-regulated the expression of miR-669a-5p in these tissues. IF staining analysis indicated that ZGF can significantly increase the protein expression of the osteogenic transcription factors OCN and Osterix in the bone tissue of mice with SOP. CONCLUSIONS: ZGF can promote osteogenic differentiation of osteoblasts, regulate bone metabolism, and thereby delay the process of SOP. Perhaps, its mechanism is to upregulate myoblast-derived exosomes miR-5100, miR-126a-3p, and miR-450b-5p or downregulate miR-669a-5p. This study reports for the first time that myoblast exosomes miR-669a-5p and miR-450b-5p are novel targets for the regulation of osteoblastic differentiation and the treatment of SOP.


Subject(s)
Cell Differentiation , Exosomes , Mesenchymal Stem Cells , MicroRNAs , Myoblasts , Osteoblasts , Osteogenesis , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Cell Differentiation/drug effects , Exosomes/metabolism , Osteogenesis/drug effects , Mice , Osteoblasts/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Drugs, Chinese Herbal/pharmacology , Osteoporosis , Male
20.
Clin Sci (Lond) ; 138(12): 711-723, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38804865

ABSTRACT

Myopenia is a condition marked by progressive decline of muscle mass and strength and is associated with aging or obesity. It poses the risk of falling, with potential bone fractures, thereby also increasing the burden on family and society. Skeletal muscle wasting is characterized by a reduced number of myoblasts, impaired muscle regeneration and increased muscle atrophy markers (Atrogin-1, MuRF-1). Endothelin-1 (ET-1) is a potent vasoconstrictor peptide. Increased circulating levels of ET-1 is noted with aging and is associated with muscular fibrosis and decline of strength. However, the regulatory mechanism controlling its effect on myogenesis and atrophy remains unknown. In the present study, the effects of ET-1 on myoblast proliferation, differentiation and development were investigated in C2C12 cells and in ET-1-infused mice. The results show that ET-1, acting via ETB receptors, reduced insulin-stimulated cell proliferation, and also reduced MyoD, MyoG and MyHC expression in the differentiation processes of C2C12 myoblasts. ET-1 inhibited myoblast differentiation through ETB receptors and the p38 mitogen-activated protein kinase (MAPK)-dependent pathway. Additionally, ET-1 decreased MyHC expression in differentiated myotubes. Inhibition of proteasome activity by MG132 ameliorated the ET-1-stimulated protein degradation in differentiated C2C12 myotubes. Furthermore, chronic ET-1 infusion caused skeletal muscle atrophy and impaired exercise performance in mice. In conclusion, ET-1 inhibits insulin-induced cell proliferation, impairs myogenesis and induces muscle atrophy via ETB receptors and the p38 MAPK-dependent pathway.


Subject(s)
Cell Differentiation , Cell Proliferation , Endothelin-1 , Muscle Development , Muscle, Skeletal , p38 Mitogen-Activated Protein Kinases , Animals , Muscle Development/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Endothelin-1/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/drug effects , Cell Proliferation/drug effects , Cell Line , Mice , Male , Mice, Inbred C57BL , Myoblasts/metabolism , Myoblasts/drug effects , Signal Transduction , MAP Kinase Signaling System , Muscular Atrophy/metabolism , Muscular Atrophy/pathology
SELECTION OF CITATIONS
SEARCH DETAIL