Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 255
Filtrar
1.
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
2.
Exp Cell Res ; 411(2): 112990, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34973262

RESUMEN

Human pluripotent stem cells (hPSCs) provide a human model for developmental myogenesis, disease modeling and development of therapeutics. Differentiation of hPSCs into muscle stem cells has the potential to provide a cell-based therapy for many skeletal muscle wasting diseases. This review describes the current state of hPSCs towards recapitulating human myogenesis ex vivo, considerations of stem cell and progenitor cell state as well as function for future use of hPSC-derived muscle cells in regenerative medicine.


Asunto(s)
Desarrollo de Músculos/fisiología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Diferenciación Celular/fisiología , Humanos , Modelos Biológicos , Desarrollo de Músculos/genética , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/fisiología , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología
3.
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
4.
Gene ; 802: 145869, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34352298

RESUMEN

Skeletal myoblasts are activated satellite cells capable of proliferation and differentiation. Studies on mammalian myoblast differentiation and myogenesis could be carried out in vitro thanks to the availability of mouse myoblast cell line C2C12. Lacking of muscle cell line hinders the studies of teleost fish myogenesis. Here, we established a continuous skeletal muscle cell line from juvenile rockfish (Sebastes schlegelii) muscle using explant method and subcultured more than 50 passages for over 150 days. Stable expression of myoblast-specific marker, MyoD (myoblast determination protein) and the potential of differentiation into multi-nucleated skeletal myotubes upon induction suggested the cell line were predominately composed of myoblasts. Transcriptome analysis revealed a total of 4375 genes differentially expressed at four time points after the switch to differentiation medium, which were mainly involved in proliferation and differentiation of myoblasts. KIF22 (kinesin family member 22) and POLA1 (DNA polymerase alpha 1) were identified as the key genes involved in fish myoblast proliferation whereas MYL3 (myosin light chain 3) and TNNT2 (troponin T2) were determined as the crucial genes responsible for differentiation. In all, the continuous myoblasts cultured in this study provided a cell platform for future studies on marine fish myoblast differentiation and myogenesis. The molecular process of myoblast differentiation revealed in this study will open a window into the understanding of indeterminate muscle growth of large teleost.


Asunto(s)
Técnicas de Cultivo de Célula , Línea Celular , Desarrollo de Músculos/genética , Mioblastos Esqueléticos/fisiología , Perciformes/anatomía & histología , Animales , Criopreservación , Transcriptoma
5.
Elife ; 102021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-34323217

RESUMEN

During aging and neuromuscular diseases, there is a progressive loss of skeletal muscle volume and function impacting mobility and quality of life. Muscle loss is often associated with denervation and a loss of resident muscle stem cells (satellite cells or MuSCs); however, the relationship between MuSCs and innervation has not been established. Herein, we administered severe neuromuscular trauma to a transgenic murine model that permits MuSC lineage tracing. We show that a subset of MuSCs specifically engraft in a position proximal to the neuromuscular junction (NMJ), the synapse between myofibers and motor neurons, in healthy young adult muscles. In aging and in a mouse model of neuromuscular degeneration (Cu/Zn superoxide dismutase knockout - Sod1-/-), this localized engraftment behavior was reduced. Genetic rescue of motor neurons in Sod1-/- mice reestablished integrity of the NMJ in a manner akin to young muscle and partially restored MuSC ability to engraft into positions proximal to the NMJ. Using single cell RNA-sequencing of MuSCs isolated from aged muscle, we demonstrate that a subset of MuSCs are molecularly distinguishable from MuSCs responding to myofiber injury and share similarity to synaptic myonuclei. Collectively, these data reveal unique features of MuSCs that respond to synaptic perturbations caused by aging and other stressors.


Asunto(s)
Envejecimiento , Músculo Esquelético/lesiones , Mioblastos Esqueléticos/fisiología , Unión Neuromuscular/fisiología , Superóxido Dismutasa-1/deficiencia , Animales , Femenino , Masculino , Ratones Noqueados
6.
Cell Transplant ; 30: 9636897211009559, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33880968

RESUMEN

Colorectal anastomotic leakage is one of the most feared and fatal complications of colorectal surgery. To date, no external coating material that can prevent anastomotic leakage has been developed. As myoblasts possess anti-inflammatory capacity and improve wound healing, we developed a multilayered human skeletal muscle myoblast (HSMM) sheet by periodic exposure to supraphysiological hydrostatic pressure during repeated cell seeding. We assessed whether the application of an HSMM sheet can promote the healing process after colonic anastomosis. Partial colectomy and insufficient suturing were employed to create a high-risk colo-colonic anastomosis model in 60 nude rats. Rats were divided into a control group (n = 30) and an HSMM sheet group (n = 30). Macroscopic findings, anastomotic bursting pressure, and histology at the colonic anastomotic site were evaluated on postoperative day (POD) 3, 5, 7, 14, and 28. The application of an HSMM sheet significantly suppressed abscess formation at the anastomotic site compared to the control group on POD3 and 5. The anastomotic bursting pressure in the HSMM sheet group was higher than that in the control group on POD3 and 5. Inflammatory cell infiltration in the HSMM sheet group was significantly suppressed compared to that in the control group throughout the time course. Collagen deposition in the HSMM sheet group on POD3 was significantly abundant compared to that in the control group. Regeneration of the mucosa at the colonic anastomotic site was promoted in the HSMM sheet group compared to that in the control group on POD14 and 28. Immunohistochemical analysis demonstrated that surviving cells in the HSMM sheet gradually decreased with postoperative time and none were detected on POD14. These results suggest that the application of a multilayered HSMM sheet may prevent postoperative colonic anastomotic leakage.


Asunto(s)
Anastomosis Quirúrgica/métodos , Colon/cirugía , Mioblastos Esqueléticos/fisiología , Animales , Colon/patología , Humanos , Masculino , Ratones Desnudos , Ratas
7.
Crit Rev Biochem Mol Biol ; 56(3): 284-300, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33823731

RESUMEN

Muscle stem cells (MuSCs) also called satellite cells are the building blocks of skeletal muscle, the largest tissue in the human body which is formed primarily of myofibers. While MuSCs are the principal cells that directly contribute to the formation of the muscle fibers, their ability to do so depends on critical interactions with a vast array of nonmyogenic cells within their niche environment. Therefore, understanding the nature of communication between MuSCs and their niche is of key importance to understand how the skeletal muscle is maintained and regenerated after injury. MuSCs are rare and therefore difficult to study in vivo within the context of their niche environment. The advent of single-cell technologies, such as switching mechanism at 5' end of the RNA template (SMART) and tagmentation based technologies using hyperactive transposase, afford the unprecedented opportunity to perform whole transcriptome and epigenome studies on rare cells within their niche environment. In this review, we will delve into how single-cell technologies can be applied to the study of MuSCs and muscle-resident niche cells and the impact this can have on our understanding of MuSC biology and skeletal muscle regeneration.


Asunto(s)
Epigenoma , Estudio de Asociación del Genoma Completo , Mioblastos Esqueléticos/fisiología , Regeneración , Análisis de la Célula Individual , Nicho de Células Madre , Transcriptoma , Animales , Humanos
8.
Int J Mol Sci ; 21(20)2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096920

RESUMEN

The Duchenne muscular dystrophy (DMD) gene has a complex expression pattern regulated by multiple tissue-specific promoters and by alternative splicing (AS) of the resulting transcripts. Here, we used an RNAi-based approach coupled with DMD-targeted RNA-seq to identify RNA-binding proteins (RBPs) that regulate splicing of its skeletal muscle isoform (Dp427m) in a human muscular cell line. A total of 16 RBPs comprising the major regulators of muscle-specific splicing events were tested. We show that distinct combinations of RBPs maintain the correct inclusion in the Dp427m of exons that undergo spatio-temporal AS in other dystrophin isoforms. In particular, our findings revealed the complex networks of RBPs contributing to the splicing of the two short DMD exons 71 and 78, the inclusion of exon 78 in the adult Dp427m isoform being crucial for muscle function. Among the RBPs tested, QKI and DDX5/DDX17 proteins are important determinants of DMD exon inclusion. This is the first large-scale study to determine which RBP proteins act on the physiological splicing of the DMD gene. Our data shed light on molecular mechanisms contributing to the expression of the different dystrophin isoforms, which could be influenced by a change in the function or expression level of the identified RBPs.


Asunto(s)
Distrofina/genética , Exones , Proteínas con Motivos de Reconocimiento de ARN/genética , Adulto , Empalme Alternativo , Línea Celular , Regulación de la Expresión Génica , Humanos , Intrones , Mioblastos Esqueléticos/fisiología , Interferencia de ARN
9.
Math Biosci ; 327: 108424, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32681914

RESUMEN

This study investigates the effect that upper body vibration has on the recovery rate of the biceps muscle. A mathematical model that accounts for vibration is developed by adapting three vibration terms into the Stephenson and Kojourahov skeletal muscle regeneration mathematical model. The first term accounts for the increase in the influx rate of type 1 macrophages (P1). These cells are part of the body's immune response to muscle damage. They control the proliferation rate of satellite cells (S) and phagocytize dead myofiber cells. The second term accounts for the rate of the phenotype change of P1 to type 2 macrophages (P2). P2 are used to support S differentiation and prevent apoptosis of myoblasts (Mb). The final term accounts for the fusion rate of Mb. Mb fuse with each other to create myotubes which align to create myofibers. The addition of these three terms decreases the overall skeletal muscle regeneration time by 47%. The model is validated on the macroscopic scale by subjecting test participants to a muscle damage and recovery protocol involving vibration therapy.


Asunto(s)
Modelos Biológicos , Músculo Esquelético/fisiología , Regeneración/fisiología , Vibración/uso terapéutico , Adulto , Simulación por Computador , Músculos Isquiosurales/citología , Músculos Isquiosurales/lesiones , Músculos Isquiosurales/fisiología , Humanos , Macrófagos/inmunología , Masculino , Conceptos Matemáticos , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Mioblastos Esqueléticos/fisiología , Regeneración/inmunología , Células Satélite del Músculo Esquelético/fisiología , Adulto Joven
10.
FASEB J ; 34(9): 12367-12378, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32686868

RESUMEN

HMGB2, a DNA-binding protein, highly expresses during embryogenesis and plays an important role in development of some organs and tissues. However, it remains to be further investigated weather HMGB2 influences muscle development. In this work, we identified HMGB2 as an essential factor in myogenesis. Compared to wild type (WT) mice, body weights of systemic hmgb2 homozygous knockout (hmgb2-/- ) mice especially males were reduced. Diameter and cross-section area of tibialis anterior (TA) muscle fibers as well as expression of Myogenin and MyHC were all decreased in hmgb2-/- mice. CTX injury model revealed that HMGB2 was required for satellite cell proliferation and muscle regeneration. Moreover, HMGB2 interacted with S6K1 and regulated the kinase activity of S6K1 during cell proliferation. Knockdown and inactivation of S6K1 in C2C12 cells both resulted in impaired proliferation and differentiation. Furthermore, expression of cyclin D1 and Myf5 were both decreased when HMGB2 or S6K1 were knocked down and kinase activity of S6K1 was inhibited. These results indicate that HMGB2 is required for skeletal muscle development and regeneration, and HMGB2 maintains proliferation of myoblasts through regulating kinase activity of S6K1.


Asunto(s)
Proteína HMGB2/fisiología , Desarrollo de Músculos/fisiología , Músculo Esquelético/fisiología , Proteínas Quinasas S6 Ribosómicas 90-kDa/fisiología , Animales , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/fisiología , Regeneración , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología
11.
FASEB J ; 34(6): 7687-7702, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32277852

RESUMEN

miR-206, miR-1a-1, and miR-1a-2 are induced during differentiation of skeletal myoblasts and promote myogenesis in vitro. miR-206 is required for skeletal muscle regeneration in vivo. Although this miRNA family is hypothesized to play an essential role in differentiation, a triple knock-out (tKO) of the three genes has not been done to test this hypothesis. We report that tKO C2C12 myoblasts generated using CRISPR/Cas9 method differentiate despite the expected derepression of the miRNA targets. Surprisingly, their mitochondrial function is diminished. tKO mice demonstrate partial embryonic lethality, most likely due to the role of miR-1a in cardiac muscle differentiation. Two tKO mice survive and grow normally to adulthood with smaller myofiber diameter, diminished physical performance, and an increase in PAX7 positive satellite cells. Thus, unlike other miRNAs important in other differentiation pathways, the miR-206 family is not absolutely essential for myogenesis and is instead a modulator of optimal differentiation of skeletal myoblasts.


Asunto(s)
MicroARNs/genética , Mitocondrias/genética , Desarrollo de Músculos/genética , Músculo Esquelético/fisiología , Mioblastos Esqueléticos/fisiología , Animales , Sistemas CRISPR-Cas/genética , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/genética , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Enfermedades Musculares/genética
12.
Nat Commun ; 11(1): 1025, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32094341

RESUMEN

A bioengineered skeletal muscle construct that mimics structural and functional characteristics of native skeletal muscle is a promising therapeutic option to treat extensive muscle defect injuries. We previously showed that bioprinted human skeletal muscle constructs were able to form multi-layered bundles with aligned myofibers. In this study, we investigate the effects of neural cell integration into the bioprinted skeletal muscle construct to accelerate functional muscle regeneration in vivo. Neural input into this bioprinted skeletal muscle construct shows the improvement of myofiber formation, long-term survival, and neuromuscular junction formation in vitro. More importantly, the bioprinted constructs with neural cell integration facilitate rapid innervation and mature into organized muscle tissue that restores normal muscle weight and function in a rodent model of muscle defect injury. These results suggest that the 3D bioprinted human neural-skeletal muscle constructs can be rapidly integrated with the host neural network, resulting in accelerated muscle function restoration.


Asunto(s)
Bioimpresión/métodos , Regeneración Tisular Dirigida/métodos , Enfermedades Musculares/terapia , Mioblastos Esqueléticos/fisiología , Neuronas/fisiología , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/uso terapéutico , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Estudios de Factibilidad , Humanos , Hidrogeles/química , Masculino , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Músculo Esquelético/fisiología , Enfermedades Musculares/fisiopatología , Red Nerviosa/fisiología , Unión Neuromuscular/citología , Unión Neuromuscular/fisiología , Impresión Tridimensional , Ratas , Factores de Tiempo
13.
J Mol Endocrinol ; 64(3): 195-208, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31940280

RESUMEN

The biologically active metabolite of vitamin D, 1,25-dihydroxyvitamin D3 (VD3), exerts its tissue-specific actions through binding to its intracellular vitamin D receptor (VDR) which functions as a heterodimer with retinoid X receptor (RXR) to recognize vitamin D response elements (VDRE) and activate target genes. Upregulation of VDR in murine skeletal muscle cells occurs concomitantly with transcriptional regulation of key myogenic factors upon VD3 administration, reinforcing the notion that VD3 exerts beneficial effects on muscle. Herein we elucidated the regulatory role of VD3/VDR axis on the expression of dystrobrevin alpha (DTNA), a member of dystrophin-associated protein complex (DAPC). In C2C12 cells, Dtna and VDR gene and protein expression were upregulated by 1-50 nM of VD3 during all stages of myogenic differentiation. In the dystrophic-derived H2K-mdx52 cells, upregulation of DTNA by VD3 occurred upon co-transfection of VDR and RXR expression vectors. Silencing of MyoD1, an E-box binding myogenic transcription factor, did not alter the VD3-mediated Dtna induction, but Vdr silencing abolished this effect. We also demonstrated that VD3 administration enhanced the muscle-specific Dtna promoter activity in presence of VDR/RXR only. Through site-directed mutagenesis and chromatin immunoprecipitation assays, we have validated a VDRE site in Dtna promoter in myogenic cells. We have thus proved that the positive regulation of Dtna by VD3 observed during in vitro murine myogenic differentiation is VDR mediated and specific. The current study reveals a novel mechanism of VDR-mediated regulation for Dtna, which may be positively explored in treatments aiming to stabilize the DAPC in musculoskeletal diseases.


Asunto(s)
Proteínas Asociadas a la Distrofina/genética , Músculos/metabolismo , Neuropéptidos/genética , Receptores de Calcitriol/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculos/efectos de los fármacos , Músculos/fisiología , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/fisiología , Activación Transcripcional/efectos de los fármacos , Vitamina D/análogos & derivados , Vitamina D/farmacología , Elemento de Respuesta a la Vitamina D/efectos de los fármacos , Elemento de Respuesta a la Vitamina D/genética
14.
Cell Mol Life Sci ; 77(8): 1551-1569, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31642939

RESUMEN

Skeletal muscle plays essential roles in motor function, energy, and glucose metabolism. Skeletal muscle formation occurs through a process called myogenesis, in which a crucial step is the fusion of mononucleated myoblasts to form multinucleated myofibers. The myoblast/myocyte fusion is triggered and coordinated in a muscle-specific way that is essential for muscle development and post-natal muscle regeneration. Many molecules and proteins have been found and demonstrated to have the capacity to regulate the fusion of myoblast/myocytes. Interestingly, two newly discovered muscle-specific membrane proteins, Myomaker and Myomixer (also called Myomerger and Minion), have been identified as fusogenic regulators in vertebrates. Both Myomaker and Myomixer-Myomerger-Minion have the capacity to directly control the myogenic fusion process. Here, we review and discuss the latest studies related to these two proteins, including the discovery, structure, expression pattern, functions, and regulation of Myomaker and Myomixer-Myomerger-Minion. We also emphasize and discuss the interaction between Myomaker and Myomixer-Myomerger-Minion, as well as their cooperative regulatory roles in cell-cell fusion. Moreover, we highlight the areas for exploration of Myomaker and Myomixer-Myomerger-Minion in future studies and consider their potential application to control cell fusion for cell-therapy purposes.


Asunto(s)
Proteínas de la Membrana/metabolismo , Desarrollo de Músculos , Proteínas Musculares/metabolismo , Músculo Esquelético/fisiología , Regeneración , Secuencia de Aminoácidos , Animales , Fusión Celular , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Proteínas Musculares/análisis , Proteínas Musculares/genética , Mioblastos Esqueléticos/fisiología , Alineación de Secuencia
15.
J Exp Biol ; 223(Pt 2)2020 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-31871118

RESUMEN

Muscle fibres are classified as fast, intermediate and slow. In vitro myoblast cell culture model from fast muscle is a very useful tool to study muscle growth and development; however, similar models for slow muscle do not exist. Owing to the compartmentalization of fish muscle fibres, we have developed a slow myoblast cell culture for rainbow trout (Oncorhynchus mykiss). Slow and fast muscle-derived myoblasts have similar morphology, but with differential expression of slow muscle markers such as slow myhc, sox6 and pgc-1α We also characterized the mir-133 and mir-499 microRNA families in trout slow and fast myoblasts as a case study during myogenesis and in response to electrostimulation. Three mir-133 (a-1a, a-1b and a-2) and four mir-499 (aa, ab, ba and bb) paralogues were identified for rainbow trout and named base on their phylogenetic relationship to zebrafish and Atlantic salmon orthologues. Omy-mir-499ab and omy-mir-499bb had 0.6 and 0.5-fold higher expression in slow myoblasts compared with fast myoblasts, whereas mir-133 duplicates had similar levels in both phenotypes and little variation during development. Slow myoblasts also showed increased expression for omy-mir-499b paralogues in response to chronic electrostimulation (7-fold increase for omy-mir-499ba and 2.5-fold increase for omy-mir-499bb). The higher expression of mir-499 paralogues in slow myoblasts suggests a role in phenotype determination, while the lack of significant differences of mir-133 copies during culture development might indicate a different role in fish compared with mammals. We have also found signs of sub-functionalization of mir-499 paralogues after electrostimulation, with omy-mir-499b copies more responsive to electrical signals.


Asunto(s)
MicroARNs/metabolismo , Mioblastos Esqueléticos/fisiología , Oncorhynchus mykiss , Animales , Técnicas de Cultivo de Célula/métodos , Desarrollo de Músculos , Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Lenta/metabolismo , Mioblastos Esqueléticos/metabolismo
17.
Exp Cell Res ; 381(1): 121-128, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31082374

RESUMEN

An in vitro system of electrical stimulation was used to explore whether an innovative "noisy" stimulation protocol derived from human electromyographic recordings (EMGstim) could promote muscle regeneration. EMGstim was delivered to cultured mouse myofibers isolated from Flexor Digitorum Brevis, preserving their satellite cells. In response to EMGstim, immunostaining for the myogenic regulatory factor myogenin, revealed an increased percentage of elongated myogenin-positive cells surrounding the myofibers. Conditioned medium collected from EMGstim-treated cell cultures, promoted satellite cells differentiation in unstimulated myofiber cell cultures, suggesting that extracellular soluble factors could mediate the process. Interestingly, the myogenic effect of EMGstim was mimicked by exogenously applied ATP (0.1 µM), reduced by the ATP diphosphohydrolase apyrase and prevented by blocking endogenous ATP release with carbenoxolone. In conclusion, our results show that "noisy" electrical stimulations favor muscle progenitor cell differentiation most likely via the release of endogenous ATP from contracting myofibres. Our data also suggest that "noisy" stimulation protocols could be potentially more efficient than regular stimulations to promote in vivo muscle regeneration after traumatic injury or in neuropathological diseases.


Asunto(s)
Adenosina Trifosfato/metabolismo , Fibras Musculares Esqueléticas/fisiología , Regeneración , Animales , Estimulación Eléctrica , Electromiografía , Masculino , Ratones , Ratones Endogámicos C57BL , Desarrollo de Músculos , Mioblastos Esqueléticos/fisiología , Miogenina/metabolismo , Factor de Transcripción PAX7/metabolismo
18.
Ann Biomed Eng ; 47(7): 1596-1610, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30963383

RESUMEN

A number of significant muscle diseases, such as cachexia, sarcopenia, systemic chronic inflammation, along with inflammatory myopathies share TNF-α-dominated inflammation in their pathogenesis. In addition, inflammatory episodes may increase susceptibility to drug toxicity. To assess the effect of TNF-α-induced inflammation on drug responses, we engineered 3D, human skeletal myobundles, chronically exposed them to TNF-α during maturation, and measured the combined response of TNF-α and the chemotherapeutic doxorubicin on muscle function. First, the myobundle inflammatory environment was characterized by assessing the effects of TNF-α on 2D human skeletal muscle cultures and 3D human myobundles. High doses of TNF-α inhibited maturation in human 2D cultures and maturation and function in 3D myobundles. Then, a tetanus force dose-response curve was constructed to characterize doxorubicin's effects on function alone. The combination of TNF-α and 10 nM doxorubicin exhibited a synergistic effect on both twitch and tetanus force production. Overall, the results demonstrated that inflammation of a 3D, human skeletal muscle inflammatory system alters the response to doxorubicin.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Músculo Esquelético/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Línea Celular , Humanos , Ratones , Modelos Biológicos , Músculo Esquelético/fisiología , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/fisiología , Ingeniería de Tejidos
19.
Biochimie ; 162: 55-65, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30959082

RESUMEN

The homeoprotein Msx1 plays a critical role in embryonic patterning, particularly to maintain myogenic progenitor cell fate. However, the mechanisms underlying Msx1-mediated inhibition of myogenesis remain largely unknown. Here, we show that Msx1 cooperates with the protein kinase C-related kinase 1 (Pkn1), a member of the protein kinase C-related kinase family, to prevent the terminal differentiation of myogenic precursor cells. In mouse C2C12 cells, Pkn1 knockout partly impaired Msx1-mediated inhibition of myogenic differentiation, indicating a role for Pkn1 in this process. Furthermore, we found that Pkn1 was required for Msx1 enrichment at the promoter of Myf5, a myogenic regulatory gene. In Pkn1 knockout cells, this reduced Msx1 enrichment at the Myf5 promoter coincided with attenuated repression of Myf5 transcription. Together with our observation that Msx1 and Pkn1 were associated in a protein complex, these findings strongly suggest that Msx1 cooperates with Pkn1 to down-regulate Myf5 and, therefore, prevent the differentiation of myogenic precursor cells. Collectively, our data provide key insights into the mechanisms underlying Msx1 function in the prevention of myogenic differentiation.


Asunto(s)
Factor de Transcripción MSX1/metabolismo , Desarrollo de Músculos , Mioblastos Esqueléticos/fisiología , Factor 5 Regulador Miogénico/metabolismo , Proteína Quinasa C/metabolismo , Animales , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Ratones , Mioblastos Esqueléticos/citología , Factor 5 Regulador Miogénico/genética , Regiones Promotoras Genéticas , Proteína Quinasa C/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA