Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 680
Filtrar
1.
Curr Top Dev Biol ; 158: 1-14, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670701

RESUMEN

Embryonic skeletal muscle growth is contingent upon a population of somite derived satellite cells, however, the contribution of these cells to early postnatal skeletal muscle growth remains relatively high. As prepubertal postnatal development proceeds, the activity and contribution of satellite cells to skeletal muscle growth diminishes. Eventually, at around puberty, a population of satellite cells escapes terminal commitment, continues to express the paired box transcription factor Pax7, and reside in a quiescent state orbiting the myofiber periphery adjacent to the basal lamina. After adolescence, some satellite cell contributions to muscle maintenance and adaptation occur, however, their necessity is reduced relative to embryonic, early postnatal, and prepubertal growth.


Asunto(s)
Desarrollo de Músculos , Músculo Esquelético , Células Satélite del Músculo Esquelético , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Animales , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Humanos , Factor de Transcripción PAX7/metabolismo , Factor de Transcripción PAX7/genética , Diferenciación Celular
2.
Curr Top Dev Biol ; 158: 179-201, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670705

RESUMEN

The role of the cellular microenvironment has recently gained attention in the context of muscle health, adaption, and disease. Emerging evidence supports major roles for the extracellular matrix (ECM) in regeneration and the dynamic regulation of the satellite cell niche. Satellite cells normally reside in a quiescent state in healthy muscle, but upon muscle injury, they activate, proliferate, and fuse to the damaged fibers to restore muscle function and architecture. This chapter reviews the composition and mechanical properties of skeletal muscle ECM and the role of these factors in contributing to the satellite cell niche that impact muscle regeneration. In addition, the chapter details the effects of satellite cell-matrix interactions and provides evidence that there is bidirectional regulation affecting both the cellular and extracellular microenvironment within skeletal muscle. Lastly, emerging methods to investigate satellite cell-matrix interactions will be presented.


Asunto(s)
Microambiente Celular , Matriz Extracelular , Músculo Esquelético , Células Satélite del Músculo Esquelético , Humanos , Animales , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Células Satélite del Músculo Esquelético/metabolismo , Matriz Extracelular/metabolismo , Músculo Esquelético/fisiología , Músculo Esquelético/citología , Adaptación Fisiológica , Nicho de Células Madre/fisiología , Regeneración/fisiología , Enfermedades Musculares/patología , Enfermedades Musculares/fisiopatología , Células Madre/citología , Células Madre/fisiología
3.
Curr Top Dev Biol ; 158: 221-238, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670707

RESUMEN

The skeletal muscle is well known for its remarkable ability to regenerate after injuries. The regeneration is a complex and dynamic process that involves muscle stem cells (also called muscle satellite cells, MuSCs), fibro-adipogenic progenitors (FAPs), immune cells, and other muscle-resident cell populations. The MuSCs are the myogenic cell populaiton that contribute nuclei directly to the regenerated myofibers, while the other cell types collaboratively establish a microenvironment that facilitates myogenesis of MuSCs. The myogenic process includes activation, proliferation and differentiationof MuSCs, and subsequent fusion their descendent mononuclear myocytes into multinuclear myotubes. While the contributions of FAPs and immune cells to this microenvironment have been well studied, the influence of MuSCs on other cell types remains poorly understood. This review explores recent evidence supporting the potential role of MuSCs as immunomodulators during muscle regeneration, either through cytokine production or ligand-receptor interactions.


Asunto(s)
Músculo Esquelético , Regeneración , Regeneración/fisiología , Animales , Humanos , Músculo Esquelético/fisiología , Músculo Esquelético/citología , Desarrollo de Músculos , Células Madre/citología , Células Madre/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Diferenciación Celular , Factores Inmunológicos/farmacología , Factores Inmunológicos/metabolismo , Inmunomodulación
4.
Curr Top Dev Biol ; 158: 15-51, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670703

RESUMEN

Skeletal muscle is a highly represented tissue in mammals and is composed of fibers that are extremely adaptable and capable of regeneration. This characteristic of muscle fibers is made possible by a cell type called satellite cells. Adjacent to the fibers, satellite cells are found in a quiescent state and located between the muscle fibers membrane and the basal lamina. These cells are required for the growth and regeneration of skeletal muscle through myogenesis. This process is known to be tightly sequenced from the activation to the differentiation/fusion of myofibers. However, for the past fifteen years, researchers have been interested in examining satellite cell heterogeneity and have identified different subpopulations displaying distinct characteristics based on localization, quiescence state, stemness capacity, cell-cycle progression or gene expression. A small subset of satellite cells appears to represent multipotent long-term self-renewing muscle stem cells (MuSC). All these distinctions led us to the hypothesis that the characteristics of myogenesis might not be linear and therefore may be more permissive based on the evidence that satellite cells are a heterogeneous population. In this review, we discuss the different subpopulations that exist within the satellite cell pool to highlight the heterogeneity and to gain further understanding of the myogenesis progress. Finally, we discuss the long term self-renewing MuSC subpopulation that is capable of dividing asymmetrically and discuss the molecular mechanisms regulating MuSC polarization during health and disease.


Asunto(s)
Desarrollo de Músculos , Músculo Esquelético , Células Satélite del Músculo Esquelético , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Células Satélite del Músculo Esquelético/metabolismo , Animales , Humanos , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Diferenciación Celular , Regeneración/fisiología
5.
Curr Top Dev Biol ; 158: 407-431, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670714

RESUMEN

Skeletal muscle stem cells (MuSCs), also known as satellite cells, are essential for muscle growth and injury induced regeneration. In healthy adult muscle, MuSCs remain in a quiescent state located in a specialized niche beneath the basal lamina. Upon injury, these dormant MuSCs can quickly activate to re-enter the cell cycle and differentiate into new myofibers, while a subset undergoes self-renewal and returns to quiescence to restore the stem cell pool. The myogenic lineage progression is intricately controlled by complex intrinsic and extrinsic cues and coupled with dynamic transcriptional programs. In transcriptional regulation, enhancers are key regulatory elements controlling spatiotemporal gene expression through physical contacting promoters of target genes. The three-dimensional (3D) chromatin architecture is known to orchestrate the establishment of proper enhancer-promoter interactions throughout development and aging. However, studies dissecting the 3D organization of enhancers in MuSCs are just emerging. Here, we provide an overview of the general properties of enhancers and newly developed methods for assessing their activity. In particular, we summarize recent discoveries regarding the 3D rewiring of enhancers during MuSC specification, lineage progression as well as aging.


Asunto(s)
Elementos de Facilitación Genéticos , Animales , Humanos , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Desarrollo de Músculos/genética , Diferenciación Celular , Linaje de la Célula , Cromatina/metabolismo , Cromatina/genética , Regulación del Desarrollo de la Expresión Génica
6.
Curr Top Dev Biol ; 158: 307-339, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670711

RESUMEN

Recent research has highlighted an important role for the molecular circadian machinery in the regulation of tissue-specific function and stress responses. Indeed, disruption of circadian function, which is pervasive in modern society, is linked to accelerated aging, obesity, and type 2 diabetes. Furthermore, evidence supporting the importance of the circadian clock within both the mature muscle tissue and satellite cells to regulate the maintenance of muscle mass and repair capacity in response injury has recently emerged. Here, we review the discovery of circadian clocks within the satellite cell (a.k.a. adult muscle stem cell) and how they act to regulate metabolism, epigenetics, and myogenesis during both healthy and diseased states.


Asunto(s)
Ritmo Circadiano , Regeneración , Células Satélite del Músculo Esquelético , Células Satélite del Músculo Esquelético/fisiología , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Animales , Regeneración/fisiología , Humanos , Ritmo Circadiano/fisiología , Músculo Esquelético/fisiología , Desarrollo de Músculos , Relojes Circadianos/fisiología , Epigénesis Genética
7.
Curr Top Dev Biol ; 158: 375-406, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670713

RESUMEN

The proper functioning of skeletal muscles is essential throughout life. A crucial crosstalk between the environment and several cellular mechanisms allows striated muscles to perform successfully. Notably, the skeletal muscle tissue reacts to an injury producing a completely functioning tissue. The muscle's robust regenerative capacity relies on the fine coordination between muscle stem cells (MuSCs or "satellite cells") and their specific microenvironment that dictates stem cells' activation, differentiation, and self-renewal. Critical for the muscle stem cell pool is a fine regulation of chromatin organization and gene expression. Acquiring a lineage-specific 3D genome architecture constitutes a crucial modulator of muscle stem cell function during development, in the adult stage, in physiological and pathological conditions. The context-dependent relationship between genome structure, such as accessibility and chromatin compartmentalization, and their functional effects will be analysed considering the improved 3D epigenome knowledge, underlining the intimate liaison between environmental encounters and epigenetics.


Asunto(s)
Cromatina , Cromatina/metabolismo , Cromatina/genética , Animales , Humanos , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Diferenciación Celular , Células Madre/citología , Células Madre/metabolismo , Epigénesis Genética , Desarrollo de Músculos , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/fisiología
8.
Results Probl Cell Differ ; 72: 105-118, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38509254

RESUMEN

The classic conception of tissue regeneration assumed the existence of tissue-proper regeneration stem cells that are set aside during normal tissue development and reserved as stem cells for regeneration. However, modern studies using cell tracing and other approaches have ruled out the presence of regeneration-proper stem cells in most cases in vertebrate tissue regeneration. The only experimentally validated regeneration-dedicated reserve cells are the satellite cells in skeletal muscle (e.g., Michele 2022) (see Sect. 5.2.3 ). Here, we will first discuss examples of large-scale tissue regeneration, liver regeneration in mammals, and lens and limb regeneration in newts. Then, attempts to widen the tissue regeneration capacity in mammals with exogenous transcription factor genes will be reviewed.


Asunto(s)
Músculo Esquelético , Células Satélite del Músculo Esquelético , Animales , Músculo Esquelético/fisiología , Células Satélite del Músculo Esquelético/fisiología , Células Madre , Regeneración , Mamíferos , Diferenciación Celular/genética
9.
Cells ; 12(23)2023 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-38067102

RESUMEN

Satellite cells (SCs) are adult muscle stem cells that are mobilized when muscle homeostasis is perturbed. Here we show that RhoA in SCs is indispensable to have correct muscle regeneration and hypertrophy. In particular, the absence of RhoA in SCs prevents a correct SC fusion both to other RhoA-deleted SCs (regeneration context) and to growing control myofibers (hypertrophy context). We demonstrated that RhoA is dispensable for SCs proliferation and differentiation; however, RhoA-deleted SCs have an inefficient movement even if their cytoskeleton assembly is not altered. Proliferative myoblast and differentiated myotubes without RhoA display a decreased expression of Chordin, suggesting a crosstalk between these genes for myoblast fusion regulation. These findings demonstrate the importance of RhoA in SC fusion regulation and its requirement to achieve an efficient skeletal muscle homeostasis restoration.


Asunto(s)
Fusión Celular , Fibras Musculares Esqueléticas , Células Satélite del Músculo Esquelético , Proteína de Unión al GTP rhoA , Humanos , Comunicación Celular , Hipertrofia/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/fisiología
10.
Elife ; 122023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37963071

RESUMEN

In vitro culture systems that structurally model human myogenesis and promote PAX7+ myogenic progenitor maturation have not been established. Here we report that human skeletal muscle organoids can be differentiated from induced pluripotent stem cell lines to contain paraxial mesoderm and neuromesodermal progenitors and develop into organized structures reassembling neural plate border and dermomyotome. Culture conditions instigate neural lineage arrest and promote fetal hypaxial myogenesis toward limb axial anatomical identity, with generation of sustainable uncommitted PAX7 myogenic progenitors and fibroadipogenic (PDGFRa+) progenitor populations equivalent to those from the second trimester of human gestation. Single-cell comparison to human fetal and adult myogenic progenitor /satellite cells reveals distinct molecular signatures for non-dividing myogenic progenitors in activated (CD44High/CD98+/MYOD1+) and dormant (PAX7High/FBN1High/SPRY1High) states. Our approach provides a robust 3D in vitro developmental system for investigating muscle tissue morphogenesis and homeostasis.


Humans contains around 650 skeletal muscles which allow the body to move around and maintain its posture. Skeletal muscles are made up of individual cells that bundle together into highly organized structures. If this group of muscles fail to develop correctly in the embryo and/or fetus, this can lead to muscular disorders that can make it painful and difficult to move. One way to better understand how skeletal muscles are formed, and how this process can go wrong, is to grow them in the laboratory. This can be achieved using induced pluripotent stem cells (iPSCs), human adult cells that have been 'reprogrammed' to behave like cells in the embryo that can develop in to almost any cell in the body. The iPSCs can then be converted into specific cell types in the laboratory, including the cells that make up skeletal muscle. Here, Mavrommatis et al. created a protocol for developing iPSCs into three-dimensional organoids which resemble how cells of the skeletal muscle look and arrange themselves in the fetus. To form the skeletal muscle organoid, Mavrommatis et al. treated iPSCs that were growing in a three-dimensional environment with various factors that are found early on in development. This caused the iPSCs to organize themselves in to embryonic and fetal structures that will eventually give rise to the parts of the body that contain skeletal muscle, such as the limbs. Within the organoid were cells that produced Pax7, a protein commonly found in myogenic progenitors that specifically mature into skeletal muscle cells in the fetus. Pax 7 is also present in 'satellite cells' that help to regrow damaged skeletal muscle in adults. Indeed, Mavrommatis et al. found that the myogenic progenitors produced by the organoid were able to regenerate muscle when transplanted in to adult mice. These findings suggest that this organoid protocol can generate cells that will give rise to skeletal muscle. In the future, these lab-grown progenitors could potentially be created from cells isolated from patients and used to repair muscle injuries. The organoid model could also provide new insights in to how skeletal muscles develop in the fetus, and how genetic mutations linked with muscular disorders disrupt this process.


Asunto(s)
Músculo Esquelético , Células Satélite del Músculo Esquelético , Humanos , Músculo Esquelético/metabolismo , Diferenciación Celular , Feto/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Desarrollo de Músculos/fisiología , Factor de Transcripción PAX7/metabolismo
11.
Nat Cell Biol ; 25(12): 1758-1773, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37919520

RESUMEN

Skeletal muscle stem and progenitor cells including those derived from human pluripotent stem cells (hPSCs) offer an avenue towards personalized therapies and readily fuse to form human-mouse myofibres in vivo. However, skeletal muscle progenitor cells (SMPCs) inefficiently colonize chimeric stem cell niches and instead associate with human myofibres resembling foetal niches. We hypothesized competition with mouse satellite cells (SCs) prevented SMPC engraftment into the SC niche and thus generated an SC ablation mouse compatible with human engraftment. Single-nucleus RNA sequencing of SC-ablated mice identified the absence of a transient myofibre subtype during regeneration expressing Actc1. Similarly, ACTC1+ human myofibres supporting PAX7+ SMPCs increased in SC-ablated mice, and after re-injury we found SMPCs could now repopulate into chimeric niches. To demonstrate ACTC1+ myofibres are essential to supporting PAX7 SMPCs, we generated caspase-inducible ACTC1 depletion human pluripotent stem cells, and upon SMPC engraftment we found a 90% reduction in ACTC1+ myofibres and a 100-fold decrease in PAX7 cell numbers compared with non-induced controls. We used spatial RNA sequencing to identify key factors driving emerging human niche formation between ACTC1+ myofibres and PAX7+ SMPCs in vivo. This revealed that transient regenerating human myofibres are essential for emerging niche formation in vivo to support PAX7 SMPCs.


Asunto(s)
Músculo Esquelético , Factor de Transcripción PAX7 , Regeneración , Células Satélite del Músculo Esquelético , Animales , Humanos , Ratones , Músculo Esquelético/fisiología , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Células Madre Pluripotentes , Células Satélite del Músculo Esquelético/fisiología
12.
Dev Biol ; 504: 86-97, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37758009

RESUMEN

Human satellite cells (HuSCs) have been deemed to be the potential cure to treat muscular atrophy diseases such as Duchenne muscular dystrophy. However, the clinical trials of HuSCs were restricted to the inadequacy of donors because of that freshly isolated HuSCs quickly lost the Pax7 expression and myogenesis capacity in vivo after a few days of culture. Here we found that oleanic acid, a kind of triterpenoid endowed with diverse biological functions with treatment potential, could efficiently promote HuSCs proliferation. The HuSCs cultured in the medium supplement with oleanic acid could maintain a high expression level of Pax7 and retain the ability to differentiate into myotubes as well as facilitate muscle regeneration in injured muscles of recipient mice. We further revealed that Tenascin-C acts as the core mechanism to activate the EGFR signaling pathway followed by HuSCs proliferation. Taken together, our data provide an efficient method to expand functional HuSCs and a novel mechanism that controls HuSCs proliferation, which sheds light on the HuSCs-based therapy to treat muscle diseases.


Asunto(s)
Células Satélite del Músculo Esquelético , Tenascina , Animales , Humanos , Ratones , Diferenciación Celular , Proliferación Celular , Receptores ErbB/metabolismo , Músculo Esquelético/fisiología , Células Satélite del Músculo Esquelético/fisiología , Células Madre , Tenascina/metabolismo
13.
J Appl Physiol (1985) ; 135(6): 1403-1414, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37705447

RESUMEN

Changes in skeletal muscle are an important aspect of overall health. The collection of human muscle to study cellular and molecular processes for research requires a needle biopsy procedure which, in itself, can induce changes in the tissue. To investigate the effect of repeat tissue sampling, we collected skeletal muscle biopsy samples from vastus lateralis separated by 7 days. Cellular infiltrate, central nucleation, enlarged extracellular matrix, and rounding of muscle fibers were used as indices to define muscle damage, and we found that 16/26 samples (61.5%) revealed at least two of these symptoms in the secondary biopsy. The presence of damage influenced outcome measures usually obtained in human biopsies. Damaged muscle showed an increase in the number of small fibers even though average fiber and fiber type-specific cross-sectional area (CSA) were not different. This included higher numbers of embryonic myosin heavy chain-positive fibers (P = 0.001) as well as elevated satellite cell number (P = 0.02) in the damaged areas and higher variability in satellite cell count in the total area (P = 0.04). Collagen content was higher in damaged (P = 0.0003) as well as nondamaged areas (P = 0.05) of the muscle sections of the damaged compared with the nondamaged group. Myofibrillar protein and ribonucleic acid (RNA) fractional synthesis rates were not significantly different between the damaged compared with the nondamaged group. Results indicate that common outcomes as well as outcome variability in human muscle tissue are affected by previous biopsies. Therefore, the extent of potential damage should be assessed when performing repeated biopsies.NEW & NOTEWORTHY Indices of damage can be found in repeated biopsy samples of nonintervened control legs. Variables, directly and not directly related to muscle damage or regeneration, were compromised in second biopsy. There is a need to determine potential damage within muscle tissue when repeated muscle sampling is part of the study design. Muscle biopsy sampling may be a source of increased heterogeneity in human muscle data.


Asunto(s)
Músculo Esquelético , Células Satélite del Músculo Esquelético , Humanos , Biopsia , Músculo Esquelético/metabolismo , Fibras Musculares Esqueléticas/fisiología , Células Satélite del Músculo Esquelético/fisiología , Músculo Cuádriceps
14.
Int J Mol Sci ; 24(7)2023 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-37047747

RESUMEN

Myogenic differentiation is a complex biological process that is regulated by multiple factors, among which long noncoding RNAs (lncRNAs) play an essential role. However, in-depth studies on the regulatory mechanisms of long noncoding RNAs (lncRNAs) in myogenic differentiation are limited. In this study, we characterized the role of the novel lncRNA TCONS_00323213, which is upregulated during porcine skeletal muscle satellite cell (PSC) differentiation in myogenesis. We found that TCONS_00323213 affected the proliferation and differentiation of PSC in vitro. We performed quantitative polymerase chain reaction (qPCR), 5-ethynyl-20-deoxyuridine (EdU), western blotting, immunofluorescence staining, pull-down assays, and cleavage under targets and tagmentation (CUT and Tag) assays to clarify the effects and action mechanisms of TCONS_00323213. LncRNA TCONS_00323213 inhibited myoblast proliferation based on analyses of cell survival rates during PSC proliferation. Functional analyses revealed that TCONS_00323213 promotes cell differentiation and enhances myogenin (MyoG), myosin heavy chain (MyHC), and myocyte enhancer factor 2 (MEF2C) during myoblast differentiation. As determined by pull-down and RNA immunoprecipitation (RIP) assays, the lncRNA TCONS_00323213 interacted with PBX/Knotted Homeobox 2 (PKNOX2). CUT and Tag assays showed that PKNOX2 was significantly enriched on the MyoG promoter after lncRNA TCONS_00323213 knockdown. Our findings demonstrate that the interaction between lncRNA TCONS_00323213 and PKNOX2 relieves the inhibitory effect of PKNOX2 on the MyoG promoter, increases its expression, and promotes PSC differentiation. This novel role of lncRNA TCONS_00323213 sheds light on the molecular mechanisms by which lncRNAs regulate porcine myogenesis.


Asunto(s)
Desarrollo de Músculos , ARN Largo no Codificante , Células Satélite del Músculo Esquelético , ARN Largo no Codificante/genética , ARN Largo no Codificante/fisiología , Desarrollo de Músculos/genética , Diferenciación Celular/genética , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Animales , Porcinos , Miogenina/genética , Miogenina/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Técnicas de Silenciamiento del Gen
15.
Methods Mol Biol ; 2640: 227-248, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36995599

RESUMEN

Muscle regeneration models have revealed mechanisms of inflammation, wound clearance, and stem cell-directed repair of damage, thereby informing therapy. Whereas studies of muscle repair are most advanced in rodents, the zebrafish is emerging as an additional model organism with genetic and optical advantages. Various muscle wounding protocols (both chemical and physical) have been published. Here we describe simple, cheap, precise, adaptable, and effective wounding protocols and analysis methods for two stages of a larval zebrafish skeletal muscle regeneration model. We show examples of how muscle damage, ingression of muscle stem cells, immune cells, and regeneration of fibers can be monitored over an extended timecourse in individual larvae. Such analyses have the potential to greatly enhance understanding, by reducing the need to average regeneration responses across individuals subjected to an unavoidably variable wound stimulus.


Asunto(s)
Células Satélite del Músculo Esquelético , Pez Cebra , Animales , Pez Cebra/fisiología , Fibras Musculares Esqueléticas , Células Madre , Células Satélite del Músculo Esquelético/fisiología , Proliferación Celular , Músculo Esquelético/fisiología
16.
Methods Mol Biol ; 2640: 463-477, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36995614

RESUMEN

Skeletal muscle is a highly ordered tissue composed of a complex network of a diverse variety of cells. The dynamic spatial and temporal interaction between these cells during homeostasis and during times of injury gives the skeletal muscle its regenerative capacity. In order to properly understand the process of regeneration, a three-dimensional (3-D) imaging process must be conducted. While there have been several protocols studying 3-D imaging, it has primarily been focused on the nervous system. This protocol aims to outline the workflow for rendering a 3-D image of the skeletal muscle using spatial data from confocal microscope images. This protocol uses the ImageJ, Ilastik, and Imaris software for 3-D rendering and computational image analysis as both are relatively easy to use and have powerful segmentation capabilities.


Asunto(s)
Imagenología Tridimensional , Células Satélite del Músculo Esquelético , Células Satélite del Músculo Esquelético/fisiología , Músculo Esquelético/fisiología , Procesamiento de Imagen Asistido por Computador , Desarrollo de Músculos/fisiología , Diferenciación Celular
17.
J Appl Physiol (1985) ; 134(3): 753-765, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36794689

RESUMEN

We have previously shown that maximal over-the-counter doses of ibuprofen, compared with low doses of acetylsalicylic acid, reduce muscle hypertrophy in young individuals after 8 wk of resistance training. Because the mechanism behind this effect has not been fully elucidated, we here investigated skeletal muscle molecular responses and myofiber adaptations in response to acute and chronic resistance training with concomitant drug intake. Thirty-one young (aged 18-35 yr) healthy men (n = 17) and women (n = 14) were randomized to receive either ibuprofen (IBU; 1,200 mg daily; n = 15) or acetylsalicylic acid (ASA; 75 mg daily; n = 16) while undergoing 8 wk of knee extension training. Muscle biopsies from the vastus lateralis were obtained before, at week 4 after an acute exercise session, and after 8 wk of resistance training and analyzed for mRNA markers and mTOR signaling, as well as quantification of total RNA content (marker of ribosome biogenesis) and immunohistochemical analysis of muscle fiber size, satellite cell content, myonuclear accretion, and capillarization. There were only two treatment × time interaction in selected molecular markers after acute exercise (atrogin-1 and MuRF1 mRNA), but several exercise effects. Muscle fiber size, satellite cell and myonuclear accretion, and capillarization were not affected by chronic training or drug intake. RNA content increased comparably (∼14%) in both groups. Collectively, these data suggest that established acute and chronic hypertrophy regulators (including mTOR signaling, ribosome biogenesis, satellite cell content, myonuclear accretion, and angiogenesis) were not differentially affected between groups and therefore do not explain the deleterious effects of ibuprofen on muscle hypertrophy in young adults.NEW & NOTEWORTHY Here we show that mTOR signaling, fiber size, ribosome biogenesis, satellite cell content, myonuclear accretion, and angiogenesis were not differentially affected between groups undergoing 8 wk of resistance training with concomitant anti-inflammatory medication (ibuprofen versus low-dose aspirin). Atrogin-1 and MuRF-1 mRNA were more downregulated after acute exercise in the low-dose aspirin group than in the ibuprofen group. Taken together it appears that these established hypertrophy regulators do not explain the previously reported deleterious effects of high doses of ibuprofen on muscle hypertrophy in young adults.


Asunto(s)
Entrenamiento de Fuerza , Células Satélite del Músculo Esquelético , Masculino , Humanos , Adulto Joven , Femenino , Ibuprofeno/uso terapéutico , Ibuprofeno/farmacología , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/fisiología , Hipertrofia , Aspirina/farmacología , ARN , ARN Mensajero , Serina-Treonina Quinasas TOR , Células Satélite del Músculo Esquelético/fisiología
18.
Life Sci Alliance ; 6(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36446523

RESUMEN

Muscle satellite cells (MuSCs), myogenic stem cells in skeletal muscles, play an essential role in muscle regeneration. After skeletal muscle injury, quiescent MuSCs are activated to enter the cell cycle and proliferate, thereby initiating regeneration; however, the mechanisms that ensure successful MuSC division, including chromosome segregation, remain unclear. Here, we show that PIEZO1, a calcium ion (Ca2+)-permeable cation channel activated by membrane tension, mediates spontaneous Ca2+ influx to control the regenerative function of MuSCs. Our genetic engineering approach in mice revealed that PIEZO1 is functionally expressed in MuSCs and that Piezo1 deletion in these cells delays myofibre regeneration after injury. These results are, at least in part, due to a mitotic defect in MuSCs. Mechanistically, this phenotype is caused by impaired PIEZO1-Rho signalling during myogenesis. Thus, we provide the first concrete evidence that PIEZO1, a bona fide mechanosensitive ion channel, promotes proliferation and regenerative functions of MuSCs through precise control of cell division.


Asunto(s)
Canales Iónicos , Regeneración , Células Satélite del Músculo Esquelético , Animales , Ratones , Segregación Cromosómica/genética , Segregación Cromosómica/fisiología , Canales Iónicos/genética , Canales Iónicos/fisiología , Músculo Esquelético/fisiología , Mioblastos/fisiología , Transducción de Señal , Células Satélite del Músculo Esquelético/fisiología , Regeneración/genética , Regeneración/fisiología
19.
Sports Med ; 53(2): 457-480, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36266373

RESUMEN

BACKGROUND: Skeletal muscle has extraordinary regenerative capabilities against challenge, mainly owing to its resident muscle stem cells, commonly identified by Pax7+, which expediently donate nuclei to the regenerating multinucleated myofibers. This local reserve of stem cells in damaged muscle tissues is replenished by undifferentiated bone marrow stem cells (CD34+) permeating into the surrounding vascular system. OBJECTIVE: The purpose of the study was to provide a quantitative estimate for the changes in Pax7+ muscle stem cells (satellite cells) in humans following an acute bout of exercise until 96 h, in temporal relation to circulating CD34+ bone marrow stem cells. A subgroup analysis of age was also performed. METHODS: Four databases (Web of Science, PubMed, Scopus, and BASE) were used for the literature search until February 2022. Pax7+ cells in human skeletal muscle were the primary outcome. Circulating CD34+ cells were the secondary outcome. The standardized mean difference (SMD) was calculated using a random-effects meta-analysis. Subgroup analyses were conducted to examine the influence of age, training status, type of exercise, and follow-up time after exercise. RESULTS: The final search identified 20 studies for Pax7+ cells comprising a total of 370 participants between the average age of 21 and 74 years and 26 studies for circulating CD34+ bone marrow stem cells comprising 494 participants between the average age of 21 and 67 years. Only one study assessed Pax7+ cells immediately after aerobic exercise and showed a 32% reduction in exercising muscle followed by a fast repletion to pre-exercise level within 3 h. A large effect on increasing Pax7+ cell content in skeletal muscles was observed 24 h after resistance exercise (SMD = 0.89, p < 0.001). Pax7+ cells increased to ~ 50% above pre-exercise level 24-72 h after resistance exercise. For a subgroup analysis of age, a large effect (SMD = 0.81, p < 0.001) was observed on increasing Pax7+ cells in exercised muscle among adults aged > 50 years, whereas adults at younger age presented a medium effect (SMD = 0.64, p < 0.001). Both resistance exercise and aerobic exercise showed a medium overall effect in increasing circulating CD34+ cells (SMD = 0.53, p < 0.001), which declined quickly to the pre-exercise baseline level after exercise within 6 h. CONCLUSIONS: An immediate depletion of Pax7+ cells in exercising skeletal muscle concurrent with a transient release of CD34+ cells suggest a replenishment of the local stem cell reserve from bone marrow. A protracted Pax7+ cell expansion in the muscle can be observed during 24-72 h after resistance exercise. This result provides a scientific basis for exercise recommendations on weekly cycles allowing for adequate recovery time. Exercise-induced Pax7+ cell expansion in muscle remains significant at higher age, despite a lower stem cell reserve after age 50 years. More studies are required to confirm whether Pax7+ cell increment can occur after aerobic exercise. CLINICAL TRIAL REGISTRATION: Registered at the International Prospective Register of Systematic Reviews (PROSPERO) [identification code CRD42021265457].


Asunto(s)
Músculo Esquelético , Células Satélite del Músculo Esquelético , Adulto , Humanos , Adulto Joven , Persona de Mediana Edad , Anciano , Músculo Esquelético/fisiología , Ejercicio Físico , Células Satélite del Músculo Esquelético/fisiología , Factor de Transcripción PAX7/metabolismo
20.
Acta Physiol (Oxf) ; 237(1): e13889, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36164969

RESUMEN

AIM: It has been suggested that the proliferation and early differentiation of myoblasts are impaired in Marfan syndrome (MFS) mice during muscle regeneration. However, the underlying cellular and molecular mechanisms remain poorly understood. Here, we investigated muscle regeneration in MFS mouse models by analyzing the influence of the fibrotic niche on satellite cell function. METHODS: In vivo, ex vivo, and in vitro experiments were performed. In addition, we evaluated the effect of the pharmacological inhibition of fibrosis using Ang-(1-7) on regenerating skeletal muscles of MFS mice. RESULTS: The skeletal muscle of MFS mice shows an increased accumulation of collagen fibers (81.2%), number of fibroblasts (157.1%), and Smad2/3 signaling (110.5%), as well as an aberrant number of fibro-adipogenic progenitor cells in response to injury compared with wild-type mice. There was an increased number of proinflammatory and anti-inflammatory macrophages (3.6- and 3.1-fold, respectively) in regenerating muscles of wild-type mice, but not in the regenerating muscles of MFS mice. Our data show that proliferation and differentiation of satellite cells are altered (p ≤ 0.05) in MFS mice. Myoblast transplantation assay revealed that the regenerating muscles from MFS mice have reduced satellite cell self-renewal capacity (74.7%). In addition, we found that treatment with Ang-(1-7) reduces fibrosis (71.6%) and ameliorates satellite cell dysfunction (p ≤ 0.05) and muscle contractile function (p ≤ 0.05) in MFS mice. CONCLUSION: The fibrotic niche, caused by Fbn1 mutations, reduces the myogenic potential of satellite cells, affecting structural and functional muscle regeneration. In addition, the fibrosis inhibitor Ang-(1-7) partially counteracts satellite cell abnormalities and restores myofiber size and contractile force in regenerating muscles.


Asunto(s)
Síndrome de Marfan , Células Satélite del Músculo Esquelético , Ratones , Animales , Síndrome de Marfan/patología , Músculo Esquelético/fisiología , Células Satélite del Músculo Esquelético/fisiología , Diferenciación Celular , Modelos Animales de Enfermedad , Regeneración/fisiología , Fibrosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...