Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 716
Filtrar
1.
Anim Biotechnol ; 35(1): 2351973, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38753962

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Desarrollo de Músculos , Proteína MioD , Transducción de Señal , Tretinoina , Animales , Tretinoina/farmacología , Porcinos , Desarrollo de Músculos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína MioD/genética , Proteína MioD/metabolismo , Diferenciación Celular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Receptores de Ácido Retinoico/metabolismo , Receptores de Ácido Retinoico/genética , Proliferación Celular/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Células Cultivadas
2.
Mol Med Rep ; 30(1)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38785149

RESUMEN

Promotion of myoblast differentiation by activating mitochondrial biogenesis and protein synthesis signaling pathways provides a potential alternative strategy to balance energy and overcome muscle loss and muscle disorders. Saururus chinensis (Lour.) Baill. extract (SCE) has been used extensively as a traditional herbal medicine and has several physiological activities, including anti­asthmatic, anti­oxidant, anti­inflammatory, anti­atopic, anticancer and hepatoprotective properties. However, the effects and mechanisms of action of SCE on muscle differentiation have not yet been clarified. In the present study, it was investigated whether SCE affects skeletal muscle cell differentiation through the regulation of mitochondrial biogenesis and protein synthesis in murine C2C12 myoblasts. The XTT colorimetric assay was used to determine cell viability, and myosin heavy chain (MyHC) levels were determined using immunocytochemistry. SCE was applied to C2C12 myotube at different concentrations (1, 5, or 10 ng/ml) and times (1,3, or 5 days). Reverse transcription­quantitative PCR and western blotting were used to analyze the mRNA and protein expression change of factors related to differentiation, mitochondrial biogenesis and protein synthesis. Treatment of C2C12 cells with SCE at 1,5, and 10 ng/ml did not affect cell viability. SCE promoted C2C12 myotube formation and significantly increased MyHC expression in a concentration­ and time­dependent manner. SCE significantly increased the mRNA and protein expression of muscle differentiation­specific markers, such as MyHC, myogenic differentiation 1, myogenin, Myogenic Factor 5, and ß­catenin, mitochondrial biosynthesis­related factors, such as peroxisome proliferator­activated receptor­gamma coactivator­1α, nuclear respirator factor­1, AMP­activated protein kinase phosphorylation, and histone deacetylase 5 and AKT/mTOR signaling factors related to protein synthesis. SCE may prevent skeletal muscle dysfunction by enhancing myoblast differentiation through the promotion of mitochondrial biogenesis and protein synthesis.


Asunto(s)
Diferenciación Celular , Biogénesis de Organelos , Extractos Vegetales , Proteínas Proto-Oncogénicas c-akt , Saururaceae , Transducción de Señal , Serina-Treonina Quinasas TOR , Animales , Ratones , Diferenciación Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Extractos Vegetales/farmacología , Línea Celular , Saururaceae/química , Supervivencia Celular/efectos de los fármacos , Mioblastos/metabolismo , Mioblastos/efectos de los fármacos , Mioblastos/citología , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Desarrollo de Músculos/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/citología , Cadenas Pesadas de Miosina/metabolismo , Cadenas Pesadas de Miosina/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/citología
3.
Nutrients ; 16(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38732549

RESUMEN

Oleocanthal (OC) is a monophenol of extra-virgin olive oil (EVOO) endowed with antibiotic, cardioprotective and anticancer effects, among others, mainly in view of its antioxidant and anti-inflammatory properties. OC has been largely investigated in terms of its anticancer activity, in Alzheimer disease and in collagen-induced arthritis; however, the possibility that it can also affect muscle biology has been totally overlooked so far. This study is the first to describe that OC modulates alterations induced in C2C12 myotubes by stimuli known to induce muscle wasting in vivo, namely TNF-α, or in the medium conditioned by the C26 cachexia-inducing tumor (CM-C26). C2C12 myotubes were exposed to CM-C26 or TNF-α in the presence or absence of OC for 24 and 48 h and analyzed by immunofluorescence and Western blotting. In combination with TNF-α or CM-C26, OC was revealed to be able to restore both the myotube's original size and morphology and normal levels of both atrogin-1 and MuRF1. OC seems unable to impinge on the autophagic-lysosomal proteolytic system or protein synthesis. Modulations towards normal levels of the expression of molecules involved in myogenesis, such as Pax7, myogenin and MyHC, were also observed in the myotube cultures exposed to OC and TNF-α or CM-C26. In conclusion, the data presented here show that OC exerts a protective action in C2C12 myotubes exposed to TNF-α or CM-C26, with mechanisms likely involving the downregulation of ubiquitin-proteasome-dependent proteolysis and the partial relief of myogenic differentiation impairment.


Asunto(s)
Catecoles , Monoterpenos Ciclopentánicos , Fibras Musculares Esqueléticas , Proteínas Musculares , Atrofia Muscular , Factor de Necrosis Tumoral alfa , Animales , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Ratones , Factor de Necrosis Tumoral alfa/metabolismo , Atrofia Muscular/prevención & control , Atrofia Muscular/metabolismo , Proteínas Musculares/metabolismo , Monoterpenos Ciclopentánicos/farmacología , Catecoles/farmacología , Línea Celular , Proteínas Ligasas SKP Cullina F-box/metabolismo , Proteínas Ligasas SKP Cullina F-box/genética , Desarrollo de Músculos/efectos de los fármacos , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/metabolismo , Autofagia/efectos de los fármacos , Fenoles/farmacología , Caquexia/prevención & control , Medios de Cultivo Condicionados/farmacología , Aldehídos
4.
ACS Appl Bio Mater ; 7(5): 3258-3270, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38593039

RESUMEN

Reliable in vitro models closely resembling native tissue are urgently needed for disease modeling and drug screening applications. Recently, conductive biomaterials have received increasing attention in the development of in vitro models as they permit exogenous electrical signals to guide cells toward a desired cellular response. Interestingly, they have demonstrated that they promote cellular proliferation and adhesion even without external electrical stimulation. This paper describes the development of a conductive, fully synthetic hydrogel based on hybrids of the peptide-modified polyisocyanide (PIC-RGD) and the relatively conductive poly(aniline-co-N-(4-sulfophenyl)aniline) (PASA) and its suitability as the in vitro matrix. We demonstrate that incorporating PASA enhances the PIC-RGD hydrogel's electroactive nature without significantly altering the fibrous architecture and nonlinear mechanics of the PIC-RGD network. The biocompatibility of our model was assessed through phenotyping cultured human foreskin fibroblasts (HFF) and murine C2C12 myoblasts. Immunofluorescence analysis revealed that PIC-PASA hydrogels inhibit the fibrotic behavior of HFFs while promoting myogenesis in C2C12 cells without electrical stimulation. The composite PIC-PASA hydrogel can actively change the cell fate of different cell types, providing an attractive tool to improve skin and muscle repair.


Asunto(s)
Materiales Biocompatibles , Hidrogeles , Ensayo de Materiales , Hidrogeles/química , Hidrogeles/farmacología , Humanos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/síntesis química , Animales , Ratones , Desarrollo de Músculos/efectos de los fármacos , Fibrosis/tratamiento farmacológico , Tamaño de la Partícula , Fibroblastos/efectos de los fármacos , Línea Celular , Estructura Molecular , Proliferación Celular/efectos de los fármacos , Conductividad Eléctrica
5.
J Biosci Bioeng ; 137(6): 480-486, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38604883

RESUMEN

Functional tissue-engineered artificial skeletal muscle tissue has great potential for pharmacological and academic applications. This study demonstrates an in vitro tissue engineering system to construct functional artificial skeletal muscle tissues using self-organization and signal inhibitors. To induce efficient self-organization, we optimized the substrate stiffness and extracellular matrix (ECM) coatings. We modified the tissue morphology to be ring-shaped under optimized self-organization conditions. A bone morphogenetic protein (BMP) inhibitor was added to improve overall myogenic differentiation. This supplementation enhanced the myogenic differentiation ratio and myotube hypertrophy in two-dimensional cell cultures. Finally, we found that myotube hypertrophy was enhanced by a combination of self-organization with ring-shaped tissue and a BMP inhibitor. BMP inhibitor treatment significantly improved myogenic marker expression and contractile force generation in the self-organized tissue. These observations indicated that this procedure may provide a novel and functional artificial skeletal muscle for pharmacological studies.


Asunto(s)
Proteínas Morfogenéticas Óseas , Diferenciación Celular , Desarrollo de Músculos , Fibras Musculares Esqueléticas , Músculo Esquelético , Transducción de Señal , Ingeniería de Tejidos , Diferenciación Celular/efectos de los fármacos , Animales , Ingeniería de Tejidos/métodos , Ratones , Proteínas Morfogenéticas Óseas/metabolismo , Transducción de Señal/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/efectos de los fármacos , Desarrollo de Músculos/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/citología , Línea Celular , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Andamios del Tejido/química
6.
Int J Biol Macromol ; 268(Pt 2): 131547, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38641281

RESUMEN

Eicosapentaenoic acid regulates glucose uptake in skeletal muscle and significantly affects whole-body energy metabolism. However, the underlying molecular mechanism remains unclear. Here we report that eicosapentaenoic acid activates phosphoglycerate mutase 2, which mediates the conversion of 2-phosphoglycerate into 3-phosphoglycerate. This enzyme plays a pivotal role in glycerol degradation, thereby facilitating the proliferation and differentiation of satellite cells in skeletal muscle. Interestingly, phosphoglycerate mutase 2 inhibits mitochondrial metabolism, promoting the formation of fast-type muscle fibers. Treatment with eicosapentaenoic acid and phosphoglycerate mutase 2 knockdown induced opposite transcriptomic changes, most of which were enriched in the PI3K-AKT signaling pathway. Phosphoglycerate mutase 2 activated the PI3K-AKT signaling pathway, which inhibited the phosphorylation of FOXO1, and, in turn, inhibited mitochondrial function and promoted the formation of fast-type muscle fibers. Our results suggest that eicosapentaenoic acid promotes skeletal muscle growth and regulates glucose metabolism by targeting phosphoglycerate mutase 2 and activating the PI3K/AKT signaling pathway.


Asunto(s)
Ácido Eicosapentaenoico , Músculo Esquelético , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Músculo Esquelético/metabolismo , Músculo Esquelético/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Ácido Eicosapentaenoico/farmacología , Ratones , Fosfoglicerato Mutasa/metabolismo , Fosfoglicerato Mutasa/genética , Desarrollo de Músculos/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL
7.
Food Funct ; 15(8): 4389-4398, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38563085

RESUMEN

ß-Hydroxy-ß-methylbutyrate (HMB) is a breakdown product of leucine, which promotes muscle growth. Although some studies indicate that HMB activates AKT and mTOR, others show activation of the downstream effectors, P70S6K and S6, independent of mTOR. Our aim was to study the metabolic effect of HMB around the circadian clock in order to determine more accurately the signaling pathway involved. C2C12 myotubes were treated with HMB and clock, metabolic and myogenic markers were measured around the clock. HMB-treated C2C12 myotubes showed no activation of AKT and mTOR, but did show activation of P70S6K and S6. Activation of P70S6K and S6 was also found when myotubes were treated with HMB combined with metformin, an indirect mTOR inhibitor, or rapamycin, a direct mTOR inhibitor. The activation of the P70S6K and S6 independent of AKT and mTOR, was accompanied by increased activation of phospholipase D2 (PLD). In addition, HMB led to high amplitude and advanced circadian rhythms. In conclusion, HMB induces myogenesis in C2C12 by activating P70S6K and S6 via PLD2, rather than AKT and mTOR, leading to high amplitude advanced rhythms.


Asunto(s)
Ritmo Circadiano , Fibras Musculares Esqueléticas , Fosfolipasa D , Valeratos , Valeratos/farmacología , Animales , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Ratones , Fosfolipasa D/metabolismo , Ritmo Circadiano/efectos de los fármacos , Línea Celular , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Desarrollo de Músculos/efectos de los fármacos
8.
Arch Toxicol ; 98(6): 1645-1658, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38546836

RESUMEN

A number of environmental toxicants are noted for their activity that leads to declined motor function. However, the role of muscle as a proximal toxicity target organ for environmental agents has received considerably less attention than the toxicity targets in the nervous system. Nonetheless, the effects of conventional neurotoxicants on processes of myogenesis and muscle maintenance are beginning to resolve a concerted role of muscle as a susceptible toxicity target. A large body of evidence from epidemiological, animal, and in vitro studies has established that methylmercury (MeHg) is a potent developmental toxicant, with the nervous system being a preferred target. Despite its well-recognized status as a neurotoxicant, there is accumulating evidence that MeHg also targets muscle and neuromuscular development as well as contributes to the etiology of motor defects with prenatal MeHg exposure. Here, we summarize evidence for targets of MeHg in the morphogenesis and maintenance of skeletal muscle that reveal effects on MeHg distribution, myogenesis, myotube formation, myotendinous junction formation, neuromuscular junction formation, and satellite cell-mediated muscle repair. We briefly recapitulate the molecular and cellular mechanisms of skeletal muscle development and highlight the pragmatic role of alternative model organisms, Drosophila and zebrafish, in delineating the molecular underpinnings of muscle development and MeHg-mediated myotoxicity. Finally, we discuss how toxicity targets in muscle development may inform the developmental origins of health and disease theory to explain the etiology of environmentally induced adult motor deficits and accelerated decline in muscle fitness with aging.


Asunto(s)
Exposición a Riesgos Ambientales , Contaminantes Ambientales , Compuestos de Metilmercurio , Desarrollo de Músculos , Músculo Esquelético , Compuestos de Metilmercurio/toxicidad , Animales , Desarrollo de Músculos/efectos de los fármacos , Humanos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Contaminantes Ambientales/toxicidad , Exposición a Riesgos Ambientales/efectos adversos , Unión Neuromuscular/efectos de los fármacos
9.
Radiat Res ; 201(5): 429-439, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38253061

RESUMEN

The current geopolitical context has brought the radiological nuclear risk to the forefront of concerns. High-dose localized radiation exposure leads to the development of a musculocutaneous radiation syndrome affecting the skin and subcutaneous muscles. Despite the implementation of a gold standard treatment based on an invasive surgical procedure coupled with autologous cell therapy, a muscular defect frequently persists. Targeting the modulation of the Hedgehog (Hh) signaling pathway appears to be a promising therapeutic approach. Activation of this pathway enhances cell survival and promotes proliferation after irradiation, while inhibition by Cyclopamine facilitates differentiation. In this study, we compared the effects of three antagonists of Hh, Cyclopamine (CA), Vismodegib (VDG) and Sonidegib (SDG) on differentiation. A stable cell line of murine myoblasts, C2C12, was exposed to X-ray radiation (5 Gy) and treated with CA, VDG or SDG. Analysis of proliferation, survival (apoptosis), morphology, myogenesis genes expression and proteins production were performed. According to the results, VDG does not have a significant impact on C2C12 cells. SDG increases the expression/production of differentiation markers to a similar extent as CA, while morphologically, SDG proves to be more effective than CA. To conclude, SDG can be used in the same way as CA but already has a marketing authorization with an indication against basal cell cancers, facilitating their use in vivo. This proof of concept demonstrates that SDG represents a promising alternative to CA to promotes differentiation of murine myoblasts. Future studies on isolated and cultured satellite cells and in vivo will test this proof of concept.


Asunto(s)
Proteínas Hedgehog , Músculo Esquelético , Regeneración , Transducción de Señal , Animales , Ratones , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/antagonistas & inhibidores , Músculo Esquelético/efectos de la radiación , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/citología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Línea Celular , Regeneración/efectos de los fármacos , Regeneración/efectos de la radiación , Piridinas/farmacología , Alcaloides de Veratrum/farmacología , Anilidas/farmacología , Compuestos de Bifenilo/farmacología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Desarrollo de Músculos/efectos de los fármacos , Desarrollo de Músculos/efectos de la radiación
10.
Mol Biotechnol ; 66(5): 948-959, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38198052

RESUMEN

Obestatin is derived from the same gene as that of ghrelin and their functions were perceived to be antagonistic. Recent developments have shown that although they are known to have contradictory functions, effect of obestatin on skeletal muscle regeneration is similar to that of ghrelin. Obestatin works through a receptor called GPR39, a ghrelin and motilin family receptor and transduces signals in skeletal muscle similar to that of ghrelin. Not only there is a similarity in the receptor family, but also obestatin targets similar proteins and transcription factors as that of ghrelin (for example, FoxO family members) for salvaging skeletal muscle atrophy. Moreover, like ghrelin, obestatin also works by inducing the transcription of Pax7 which is required for muscle stem cell mobilisation. Hence, there are quite some evidences which points to the fact that obestatin can be purposed as a peptide intervention to prevent skeletal muscle wasting and induce myogenesis. This review elaborates these aspects of obestatin which can be further exploited and addressed to bring obestatin as a clinical intervention towards preventing skeletal muscle atrophy and sarcopenia.


Asunto(s)
Ghrelina , Músculo Esquelético , Atrofia Muscular , Regeneración , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/efectos de los fármacos , Regeneración/efectos de los fármacos , Animales , Atrofia Muscular/metabolismo , Atrofia Muscular/prevención & control , Atrofia Muscular/tratamiento farmacológico , Ghrelina/farmacología , Ghrelina/metabolismo , Ghrelina/uso terapéutico , Transducción de Señal/efectos de los fármacos , Desarrollo de Músculos/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Factor de Transcripción PAX7/metabolismo , Factor de Transcripción PAX7/genética
11.
Aging Cell ; 22(3): e13764, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36625257

RESUMEN

Cellular senescence leads to the depletion of myogenic progenitors and decreased regenerative capacity. We show that the small molecule 2,6-disubstituted purine, reversine, can improve some well-known hallmarks of cellular aging in senescent myoblast cells. Reversine reactivated autophagy and insulin signaling pathway via upregulation of Adenosine Monophosphate-activated protein kinase (AMPK) and Akt2, restoring insulin sensitivity and glucose uptake in senescent cells. Reversine also restored the loss of connectivity of glycolysis to the TCA cycle, thus restoring dysfunctional mitochondria and the impaired myogenic differentiation potential of senescent myoblasts. Altogether, our data suggest that cellular senescence can be reversed by treatment with a single small molecule without employing genetic reprogramming technologies.


Asunto(s)
Autofagia , Senescencia Celular , Morfolinas , Desarrollo de Músculos , Mioblastos Esqueléticos , Inhibidores de Proteínas Quinasas , Purinas , Senescencia Celular/efectos de los fármacos , Morfolinas/farmacología , Purinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Humanos , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/fisiología , Autofagia/efectos de los fármacos , Insulina/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Glucólisis/efectos de los fármacos , Ciclo del Ácido Cítrico/efectos de los fármacos , Resistencia a la Insulina , Células Cultivadas , Desarrollo de Músculos/efectos de los fármacos
12.
Food Funct ; 13(24): 12721-12732, 2022 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-36408829

RESUMEN

Cyanocobalamin (CNCbl, the compound name of Vitamin B12) is the only mineral vitamin that is essential for growth and development and cannot be produced by animals. Some studies have found that CNCbl can promote the proliferation and migration of C2C12 cells, but the mechanism by which it affects muscle development is still unknown. In this study, we elucidated the effect of CNCbl on muscle development and studied its underlying mechanism. CNCbl could promote the differentiation of C2C12 cells and upregulate Acvr1, p-Smad2 and p-Smad3 in the TGF-ß signaling pathway in vitro. CD320 (the receptor in cell surface for binding with CNCbl transporter transcobalamin II) inhibition could reduce the uptake of CNCbl and significantly downregulate the expression of differentiation marker proteins MyoG and MYH2. Furthermore, the levels of p-Smad2 and p-Smad3 were also reduced with the inhibition of CD320, even though CNCbl was added to the C2C12 culture medium. In addition, the injection of CNCbl could accelerate the process of mouse muscle injury repair, enlarge the diameter of newly formed myofibers and upregulate the expression of MYH2, PAX7, CD320, Acvr1, p-Smad2 and p-Smad3 in vivo. These results suggest that CNCbl can promote muscle development and may play its role by regulating the expression of Acvr1, p-Smad2 and p-Smad3 related to the TGF-ß signaling pathway.


Asunto(s)
Desarrollo de Músculos , Factor de Crecimiento Transformador beta , Vitamina B 12 , Animales , Ratones , Diferenciación Celular , Desarrollo de Músculos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Vitamina B 12/farmacología , Línea Celular
13.
Molecules ; 27(13)2022 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-35807547

RESUMEN

Myostatin (MSTN), a negative regulator of muscle mass, is reported to be increased in conditions linked with muscle atrophy, sarcopenia, and other muscle-related diseases. Most pharmacologic approaches that treat muscle disorders are ineffective, emphasizing the emergence of MSTN inhibition. In this study, we used computational screening to uncover natural small bioactive inhibitors from the Traditional Chinese Medicine database (~38,000 compounds) for the MSTN protein. Potential ligands were screened, based on binding affinity (150), physicochemical (53) and ADMET properties (17). We found two hits (ZINC85592908 and ZINC85511481) with high binding affinity and specificity, and their binding patterns with MSTN protein. In addition, molecular dynamic simulations were run on each complex to better understand the interaction mechanism of MSTN with the control (curcumin) and the hit compounds (ZINC85592908 and ZINC85511481). We determined that the hits bind to the active pocket site (Helix region) and trigger conformational changes in the MSTN protein. Since the stability of the ZINC85592908 compound was greater than the MSTN control, we believe that ZINC85592908 has therapeutic potential against the MSTN protein and may hinder downstream singling by inhibiting the MSTN protein and increasing myogenesis in the skeletal muscle tissues.


Asunto(s)
Medicina Tradicional China , Enfermedades Musculares/tratamiento farmacológico , Miostatina/antagonistas & inhibidores , Simulación por Computador , Evaluación Preclínica de Medicamentos , Simulación de Dinámica Molecular , Desarrollo de Músculos/efectos de los fármacos , Enfermedades Musculares/fisiopatología , Unión Proteica
14.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-35163382

RESUMEN

Transient receptor potential melastatin-4 (TRPM4) is activated by an increase in intracellular Ca2+ concentration and is expressed on smooth muscle cells (SMCs). It is implicated in the myogenic constriction of cerebral arteries. We hypothesized that TRPM4 has a general role in intracellular Ca2+ signal amplification in a wide range of blood vessels. TRPM4 function was tested with the TRPM4 antagonist 9-phenanthrol and the TRPM4 activator A23187 on the cardiovascular responses of the rat, in vivo and in isolated basilar, mesenteric, and skeletal muscle arteries. TRPM4 inhibition by 9-phenanthrol resulted in hypotension and a decreased heart rate in the rat. TRPM4 inhibition completely antagonized myogenic tone development and norepinephrine-evoked vasoconstriction, and depolarization (high extracellular KCl concentration) evoked vasoconstriction in a wide range of peripheral arteries. Vasorelaxation caused by TRPM4 inhibition was accompanied by a significant decrease in intracellular Ca2+ concentration, suggesting an inhibition of Ca2+ signal amplification. Immunohistochemistry confirmed TRPM4 expression in the smooth muscle cells of the peripheral arteries. Finally, TRPM4 activation by the Ca2+ ionophore A23187 was competitively inhibited by 9-phenanthrol. In summary, TRPM4 was identified as an essential Ca2+-amplifying channel in peripheral arteries, contributing to both myogenic tone and agonist responses. These results suggest an important role for TRPM4 in the circulation. The modulation of TRPM4 activity may be a therapeutic target for hypertension. Furthermore, the Ca2+ ionophore A23187 was identified as the first high-affinity (nanomolar) direct activator of TRPM4, acting on the 9-phenanthrol binding site.


Asunto(s)
Señalización del Calcio , Canales Catiónicos TRPM/metabolismo , Vasoconstricción , Administración Intravenosa , Animales , Arterias/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Calcimicina/farmacología , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Frecuencia Cardíaca/efectos de los fármacos , Ionóforos/farmacología , Masculino , Desarrollo de Músculos/efectos de los fármacos , Músculo Esquelético/irrigación sanguínea , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Norepinefrina/farmacología , Fenantrenos/administración & dosificación , Fenantrenos/farmacología , Cloruro de Potasio/farmacología , Ratas Wistar , Canales Catiónicos TRPM/agonistas , Vasoconstricción/efectos de los fármacos
15.
Biochem Biophys Res Commun ; 592: 87-92, 2022 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-35033871

RESUMEN

We screened pre-approved drugs for the survival of the Hu5/KD3 human myogenic progenitors. We found that meclozine, an anti-histamine drug that has long been used for motion sickness, promoted the proliferation and survival of Hu5/KD3 cells. Meclozine increased expression of MyoD, but reduced expression of myosin heavy chain and suppressed myotube formation. Withdrawal of meclozine, however, resumed the ability of Hu5/KD3 cells to differentiate into myotubes. We examined the effects of meclozine on mdx mouse carrying a nonsense mutation in the dystrophin gene and modeling for Duchenne muscular dystrophy. Intragastric administration of meclozine in mdx mouse increased the body weight, the muscle mass in the lower limbs, the cross-sectional area of the paravertebral muscle, and improved exercise performances. Previous reports show that inhibition of phosphorylation of ERK1/2 improves muscle functions in mouse models for Emery-Dreifuss muscular dystrophy and cancer cachexia, as well as in mdx mice. We and others previously showed that meclozine blocks the phosphorylation of ERK1/2 in cultured cells. We currently showed that meclozine decreased phosphorylation of ERK1/2 in muscles in mdx mice but not in wild-type mice. This was likely to be one of the underlying mechanisms of the effects of meclozine on mdx mice.


Asunto(s)
Meclizina/farmacología , Fuerza Muscular/fisiología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Masculino , Meclizina/uso terapéutico , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Actividad Motora/efectos de los fármacos , Desarrollo de Músculos/efectos de los fármacos , Fuerza Muscular/efectos de los fármacos , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Fosforilación/efectos de los fármacos
16.
Cells ; 10(12)2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34943981

RESUMEN

Satellite cells (SC) are a population of muscle resident stem cells that are responsible for postnatal muscle growth and repair. With investigation into the genomic regulation of SC fate, the role of the epigenome in governing SC myogenesis is becoming clearer. Histone deacetylase (HDAC) inhibitors have been demonstrated to be effective at enhancing the myogenic program of SC, but their role in altering the epigenetic landscape of SC remains undetermined. Our objective was to determine how an HDAC inhibitor, butyrate, promotes myogenic differentiation. SC from tributyrin treated neonatal piglets showed a decrease relative to SC from control animals in the expression of enhance of zeste homologue-2 (EZH2), a chromatin modifier, ex vivo. Chromatin Immunoprecipitation-Sequencing (ChIP-Seq) analysis of SC isolated from tributyrin treated pigs showed a global reduction of the tri-methylation of lysine 27 of histone H3 (H3K27me3) repressive chromatin mark. To determine if reductions in EZH2 was the primary mechanism through which butyrate affects SC behavior, SC were transfected with siRNA targeting EZH2, treated with 0.5 mM butyrate, or both. Treatment with butyrate reduced paired-box-7 (Pax7) and myogenic differentiation-1 (MyoD) gene expression, while siRNA caused reductions in EZH2 had no effect on their expression. EZH2 depletion did result in an increase in differentiating SC, but not in myotube hypertrophy. These results indicate that while EZH2 reduction may force myogenic differentiation, butyrate may operate through a parallel mechanism to enhance the myogenic program.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína MioD/genética , Factor de Transcripción PAX7/genética , Células Satélite del Músculo Esquelético/efectos de los fármacos , Triglicéridos/farmacología , Animales , Butiratos/química , Butiratos/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Ensamble y Desensamble de Cromatina/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Hipertrofia/genética , Hipertrofia/patología , Desarrollo de Músculos/efectos de los fármacos , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , Profármacos/química , Profármacos/farmacología , ARN Interferente Pequeño/farmacología , Células Satélite del Músculo Esquelético/metabolismo , Porcinos
17.
Front Endocrinol (Lausanne) ; 12: 785242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34917036

RESUMEN

Intrauterine growth restricted (IUGR) fetuses are born with lower skeletal muscle mass, fewer proliferating myoblasts, and fewer myofibers compared to normally growing fetuses. Plasma concentrations of insulin, a myogenic growth factor, are lower in IUGR fetuses. We hypothesized that a two-week insulin infusion at 75% gestation would increase myoblast proliferation and fiber number in IUGR fetal sheep. Catheterized control fetuses received saline (CON-S, n=6), and the IUGR fetuses received either saline (IUGR-S, n=7) or insulin (IUGR-I, 0.014 ± 0.001 units/kg/hr, n=11) for 14 days. Fetal arterial blood gases and plasma amino acid levels were measured. Fetal skeletal muscles (biceps femoris, BF; and flexor digitorum superficialis, FDS) and pancreases were collected at necropsy (126 ± 2 dGA) for immunochemistry analysis, real-time qPCR, or flow cytometry. Insulin concentrations in IUGR-I and IUGR-S were lower vs. CON-S (P ≤ 0.05, group). Fetal arterial PaO2, O2 content, and glucose concentrations were lower in IUGR-I vs. CON-S (P ≤ 0.01) throughout the infusion period. IGF-1 concentrations tended to be higher in IUGR-I vs. IUGR-S (P=0.06), but both were lower vs. CON-S (P ≤ 0.0001, group). More myoblasts were in S/G2 cell cycle stage in IUGR-I vs. both IUGR-S and CON-S (145% and 113%, respectively, P ≤ 0.01). IUGR-I FDS muscle weighed 40% less and had 40% lower fiber number vs. CON-S (P ≤ 0.05) but were not different from IUGR-S. Myonuclear number per fiber and the mRNA expression levels of muscle regulatory factors were not different between groups. While the pancreatic ß-cell mass was lower in both IUGR-I and IUGR-S compared to CON-S, the IUGR groups were not different from each other indicating that feedback inhibition by endogenous insulin did not reduce ß-cell mass. A two-week insulin infusion at 75% gestation promoted myoblast proliferation in the IUGR fetus but did not increase fiber or myonuclear number. Myoblasts in the IUGR fetus retain the capacity to proliferate in response to mitogenic stimuli, but intrinsic defects in the fetal myoblast by 75% gestation may limit the capacity to restore fiber number.


Asunto(s)
Desarrollo Fetal/efectos de los fármacos , Retardo del Crecimiento Fetal/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Insulina/administración & dosificación , Fibras Musculares Esqueléticas/efectos de los fármacos , Mioblastos Esqueléticos/efectos de los fármacos , Animales , Esquema de Medicación , Femenino , Desarrollo Fetal/fisiología , Retardo del Crecimiento Fetal/patología , Infusiones Intravenosas , Desarrollo de Músculos/efectos de los fármacos , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Mioblastos Esqueléticos/patología , Mioblastos Esqueléticos/fisiología , Embarazo , Ovinos
18.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34830317

RESUMEN

Clostridium sporogenes (C. sporogenes), as a potential probiotic, metabolizes tryptophan and produces an anti-inflammatory metabolite, indole-3-propionic acid (IPA). Herein, we studied the effects of C. sporogenes and its bioactive metabolite, IPA, on skeletal muscle development and chronic inflammation in mice. In the in vivo study, the muscle tissues and serum samples of mice with C. sporogenes supplementation were used to analyze the effects of C. sporogenes on muscle metabolism; the IPA content was determined by metabonomics and ELISA. In an in vitro study, C2C12 cells were exposed to lipopolysaccharide (LPS) alone or LPS + IPA to verify the effect of IPA on muscle cell inflammation by transcriptome, and the involved mechanism was revealed by different functional assays. We observed that C. sporogenes colonization significantly increased the body weight and muscle weight gain, as well as the myogenic regulatory factors' (MRFs) expression. In addition, C. sporogenes significantly improved host IPA content and decreased pro-inflammatory cytokine levels in the muscle tissue of mice. Subsequently, we confirmed that IPA promoted C2C12 cells' proliferation by activating MRF signaling. IPA also effectively protected against LPS-induced C2C12 cells inflammation by activating Pregnane X Receptor and restoring the inhibited miR-26a-2-3p expression. miR-26a-2-3p serves as a novel muscle inflammation regulatory factor that could directly bind to the 3'-UTR of IL-1ß, a key initiator factor in inflammation. The results suggested that C. sporogenes with its functional metabolite IPA not only helps muscle growth development, but also protects against inflammation, partly by the IPA/ miR-26a-2-3p /IL-1ß cascade.


Asunto(s)
Clostridium/metabolismo , Indoles/metabolismo , Interleucina-1beta/genética , MicroARNs/genética , Células Musculares/efectos de los fármacos , Receptor X de Pregnano/genética , Propionatos/metabolismo , Regiones no Traducidas 3' , Animales , Línea Celular , Microbioma Gastrointestinal/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Indoles/farmacología , Inflamación/prevención & control , Interleucina-1beta/metabolismo , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Células Musculares/citología , Células Musculares/metabolismo , Desarrollo de Músculos/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Receptor X de Pregnano/metabolismo , Probióticos/metabolismo , Propionatos/farmacología , Transducción de Señal , Transcriptoma , Triptófano/metabolismo
19.
Cells ; 10(11)2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34831292

RESUMEN

Muscle stem cells (MuSCs) isolated ex vivo are essential original cells to produce cultured meat. Currently, one of the main obstacles for cultured meat production derives from the limited capacity of large-scale amplification of MuSCs, especially under high-density culture condition. Here, we show that at higher cell densities, proliferation and differentiation capacities of porcine MuSCs are impaired. We investigate the roles of Hippo-YAP signaling, which is important regulators in response to cell contact inhibition. Interestingly, abundant but not functional YAP proteins are accumulated in MuSCs seeded at high density. When treated with lysophosphatidic acid (LPA), the activator of YAP, porcine MuSCs exhibit increased proliferation and elevated differentiation potential compared with control cells. Moreover, constitutively active YAP with deactivated phosphorylation sites, but not intact YAP, promotes cell proliferation and stemness maintenance of MuSCs. Together, we reveal a potential molecular target that enables massive MuSCs expansion for large-scale cultured meat production under high-density condition.


Asunto(s)
Mioblastos/citología , Mioblastos/metabolismo , Proteínas Señalizadoras YAP/metabolismo , Secuencia de Aminoácidos , Animales , Recuento de Células , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Secuencia Conservada , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Lisofosfolípidos/farmacología , Desarrollo de Músculos/efectos de los fármacos , Desarrollo de Músculos/genética , Fosforilación , Porcinos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Proteínas Señalizadoras YAP/química
20.
Molecules ; 26(21)2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34770984

RESUMEN

Sarcopenia is an aging associated disorder involving skeletal muscle atrophy and a reduction in muscle strength, and there are no pharmaceutical interventions available thus far. Moreover, conditions such as hyperglycaemia are known to further intensify muscle degradation. Therefore, novel strategies to attenuate skeletal muscle loss are essential to enhance muscle function and thereby improve the quality of life in diabetic individuals. In this study, we have investigated the efficiency of a potato peptide hydrolysate PPH902 for its cytoprotective effects in skeletal muscle cells. PPH902 treatment in C2C12 cells showed the dose-dependent activation of the Akt/mTOR signalling pathway that is involved in skeletal myogenesis. According to Western blotting analysis, PPH902 induced the phosphorylation of Akt, mTOR proteins and induced the myogenic differentiation of C2C12 myoblasts in a differentiation medium. The phosphorylation myogenic transcription factor Foxo3A was also found to be increased in the cells treated with PPH902. In addition, treatment with PPH902 ameliorated the high glucose induced reduction in cell viability in a dose-dependent manner. Moreover, the number of myotubes in a differentiation medium reduced upon high glucose challenge, but treatment with PPH902 increased the number of differentiated myotubes. Further, the phosphorylations of AMPK and mitochondrial-related transcription factors such as PGC-1α were suppressed upon high glucose challenge but PPH902 treatment restored the protein levels. We demonstrate, for the first time, that a specific potato peptide has a therapeutic effect against sarcopenia. In addition, PPH902 improved the myogenic differentiation and their mitochondrial biogenesis and further improved myogenic protein and inhibited muscle protein degradation in C2C12 cells challenged under a high glucose condition.


Asunto(s)
Proteína Forkhead Box O3/biosíntesis , Glucosa/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Proteína Forkhead Box O3/química , Ratones , Desarrollo de Músculos/efectos de los fármacos , Hidrolisados de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...