Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.578
Filter
1.
Food Res Int ; 186: 114396, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729738

ABSTRACT

Cell culture meat is based on the scaled-up expansion of seed cells. The biological differences between seed cells from large yellow croakers in the two-dimensional (2D) and three-dimensional (3D) culture systems have not been explored. Here, satellite cells (SCs) from large yellow croakers (Larimichthys crocea) were grown on cell climbing slices, hydrogels, and microcarriers for five days to analyze the biological differences of SCs on different cell scaffolds. The results exhibited that SCs had different cell morphologies in 2D and 3D cultures. Cell adhesion receptors (Itgb1andsdc4) and adhesion spot markervclof the 3D cultures were markedly expressed. Furthermore, myogenic decision markers (Pax7andmyod) were significantly enhanced. However, the expression of myogenic differentiation marker (desmin) was significantly increased in the microcarrier group. Combined with the transcriptome data, this suggests that cell adhesion of SCs in 3D culture was related to the integrin signaling pathway. In contrast, the slight spontaneous differentiation of SCs on microcarriers was associated with rapid cell proliferation. This study is the first to report the biological differences between SCs in 2D and 3D cultures, providing new perspectives for the rapid expansion of cell culture meat-seeded cells and the development of customized scaffolds.


Subject(s)
Cell Adhesion , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Hydrogels , Satellite Cells, Skeletal Muscle , Tissue Scaffolds , Animals , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/cytology , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Cell Culture Techniques, Three Dimensional/methods , Cells, Cultured , Desmin/metabolism , PAX7 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , Muscle Development
2.
PLoS One ; 19(5): e0300850, 2024.
Article in English | MEDLINE | ID: mdl-38718005

ABSTRACT

Essential for muscle fiber formation and hypertrophy, muscle stem cells, also called satellite cells, reside beneath the basal lamina of the muscle fiber. Satellite cells have been commonly identified by the expression of the Paired box 7 (Pax7) due to its specificity and the availability of antibodies in tetrapods. In fish, the identification of satellite cells remains difficult due to the lack of specific antibodies in most species. Based on the development of a highly sensitive in situ hybridization (RNAScope®) for pax7, we showed that pax7+ cells were detected in the undifferentiated myogenic epithelium corresponding to the dermomyotome at day 14 post-fertilization in rainbow trout. Then, from day 24, pax7+ cells gradually migrated into the deep myotome and were localized along the muscle fibers and reach their niche in satellite position of the fibres after hatching. Our results showed that 18 days after muscle injury, a large number of pax7+ cells accumulated at the wound site compared to the uninjured area. During the in vitro differentiation of satellite cells, the percentage of pax7+ cells decreased from 44% to 18% on day 7, and some differentiated cells still expressed pax7. Taken together, these results show the dynamic expression of pax7 genes and the follow-up of these muscle stem cells during the different situations of muscle fiber formation in trout.


Subject(s)
Cell Differentiation , Oncorhynchus mykiss , PAX7 Transcription Factor , Regeneration , Satellite Cells, Skeletal Muscle , Animals , Oncorhynchus mykiss/metabolism , Oncorhynchus mykiss/genetics , PAX7 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/cytology , Muscle Development , Gene Expression Regulation, Developmental
3.
Genomics ; 116(3): 110851, 2024 May.
Article in English | MEDLINE | ID: mdl-38692440

ABSTRACT

Skeletal muscle satellite cells (SMSCs) play an important role in regulating muscle growth and regeneration. Chromatin accessibility allows physical interactions that synergistically regulate gene expression through enhancers, promoters, insulators, and chromatin binding factors. However, the chromatin accessibility altas and its regulatory role in ovine myoblast differentiation is still unclear. Therefore, ATAC-seq and RNA-seq analysis were performed on ovine SMSCs at the proliferation stage (SCG) and differentiation stage (SCD). 17,460 DARs (differential accessibility regions) and 3732 DEGs (differentially expressed genes) were identified. Based on joint analysis of ATAC-seq and RNA-seq, we revealed that PI3K-Akt, TGF-ß and other signaling pathways regulated SMSCs differentiation. We identified two novel candidate genes, FZD5 and MAP2K6, which may affect the proliferation and differentiation of SMSCs. Our data identify potential cis regulatory elements of ovine SMSCs. This study can provide a reference for exploring the mechanisms of the differentiation and regeneration of SMSCs in the future.


Subject(s)
Cell Differentiation , Muscle Development , Satellite Cells, Skeletal Muscle , Animals , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/cytology , Sheep/genetics , Muscle Development/genetics , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , RNA-Seq , Signal Transduction , Cells, Cultured , Chromatin Immunoprecipitation Sequencing , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Cell Proliferation
4.
Proc Natl Acad Sci U S A ; 121(21): e2317495121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38753506

ABSTRACT

Myogenic regeneration relies on the proliferation and differentiation of satellite cells. TECRL (trans-2,3-enoyl-CoA reductase like) is an endoplasmic reticulum protein only expressed in cardiac and skeletal muscle. However, its role in myogenesis remains unknown. We show that TECRL expression is increased in response to injury. Satellite cell-specific deletion of TECRL enhances muscle repair by increasing the expression of EGR2 through the activation of the ERK1/2 signaling pathway, which in turn promotes the expression of PAX7. We further show that TECRL deletion led to the upregulation of the histone acetyltransferase general control nonderepressible 5, which enhances the transcription of EGR2 through acetylation. Importantly, we showed that AAV9-mediated TECRL silencing improved muscle repair in mice. These findings shed light on myogenic regeneration and muscle repair.


Subject(s)
Early Growth Response Protein 2 , Muscle Development , Muscle, Skeletal , Regeneration , Animals , Mice , Muscle, Skeletal/metabolism , Early Growth Response Protein 2/metabolism , Early Growth Response Protein 2/genetics , Muscle Development/genetics , Regeneration/genetics , Up-Regulation , Satellite Cells, Skeletal Muscle/metabolism , PAX7 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , MAP Kinase Signaling System , Mice, Knockout , Cell Differentiation
5.
Rheumatol Int ; 44(6): 1077-1087, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38581449

ABSTRACT

Sporadic inclusion body myositis (sIBM) is a subgroup of idiopathic inflammatory myopathies characterised by progressive muscle weakness and skeletal muscle inflammation. Quantitative data on the myofibre morphology in sIBM remains scarce. Further, no previous study has examined fibre type association of satellite cells (SC), myonuclei number, macrophages, capillaries, and myonuclear domain (MD) in sIBM patients. Muscle biopsies from sIBM patients (n = 18) obtained previously (NCT02317094) were included in the analysis for fibre type-specific myofibre cross-sectional area (mCSA), SCs, myonuclei and macrophages, myonuclear domain, and capillarisation. mCSA (p < 0.001), peripheral myonuclei (p < 0.001) and MD (p = 0.005) were higher in association with type 1 (slow-twitch) than type 2 (fast-twitch) fibres. Conversely, quiescent SCs (p < 0.001), centrally placed myonuclei (p = 0.03), M1 macrophages (p < 0.002), M2 macrophages (p = 0.013) and capillaries (p < 0.001) were higher at type 2 fibres compared to type 1 fibres. In contrast, proliferating (Pax7+/Ki67+) SCs (p = 0.68) were similarly associated with each fibre type. Type 2 myofibres of late-phase sIBM patients showed marked signs of muscle atrophy (i.e. reduced mCSA) accompanied by higher numbers of associated quiescent SCs, centrally placed myonuclei, macrophages and capillaries compared to type 1 fibres. In contrast, type 1 fibres were suffering from pathological enlargement with larger MDs as well as fewer nuclei and capillaries per area when compared with type 2 fibres. More research is needed to examine to which extent different therapeutic interventions including targeted exercise might alleviate these fibre type-specific characteristics and countermeasure their consequences in impaired functional performance.


Subject(s)
Myositis, Inclusion Body , Regeneration , Humans , Myositis, Inclusion Body/pathology , Myositis, Inclusion Body/physiopathology , Male , Female , Aged , Middle Aged , Muscle Fibers, Skeletal/pathology , Macrophages/pathology , Inflammation/pathology , Biomarkers/analysis , Muscle, Skeletal/pathology , Satellite Cells, Skeletal Muscle/pathology , Biopsy , Muscle Fibers, Slow-Twitch/pathology , Muscle Fibers, Fast-Twitch/pathology
6.
Elife ; 122024 Apr 25.
Article in English | MEDLINE | ID: mdl-38661532

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disorder characterized by progressive weakness of almost all skeletal muscles, whereas extraocular muscles (EOMs) are comparatively spared. While hindlimb and diaphragm muscles of end-stage SOD1G93A (G93A) mice (a familial ALS mouse model) exhibit severe denervation and depletion of Pax7+satellite cells (SCs), we found that the pool of SCs and the integrity of neuromuscular junctions (NMJs) are maintained in EOMs. In cell sorting profiles, SCs derived from hindlimb and diaphragm muscles of G93A mice exhibit denervation-related activation, whereas SCs from EOMs of G93A mice display spontaneous (non-denervation-related) activation, similar to SCs from wild-type mice. Specifically, cultured EOM SCs contain more abundant transcripts of axon guidance molecules, including Cxcl12, along with more sustainable renewability than the diaphragm and hindlimb counterparts under differentiation pressure. In neuromuscular co-culture assays, AAV-delivery of Cxcl12 to G93A-hindlimb SC-derived myotubes enhances motor neuron axon extension and innervation, recapitulating the innervation capacity of EOM SC-derived myotubes. G93A mice fed with sodium butyrate (NaBu) supplementation exhibited less NMJ loss in hindlimb and diaphragm muscles. Additionally, SCs derived from G93A hindlimb and diaphragm muscles displayed elevated expression of Cxcl12 and improved renewability following NaBu treatment in vitro. Thus, the NaBu-induced transcriptomic changes resembling the patterns of EOM SCs may contribute to the beneficial effects observed in G93A mice. More broadly, the distinct transcriptomic profile of EOM SCs may offer novel therapeutic targets to slow progressive neuromuscular functional decay in ALS and provide possible 'response biomarkers' in pre-clinical and clinical studies.


Subject(s)
Amyotrophic Lateral Sclerosis , Disease Models, Animal , Neuromuscular Junction , Satellite Cells, Skeletal Muscle , Transcriptome , Animals , Neuromuscular Junction/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Mice , Satellite Cells, Skeletal Muscle/metabolism , Mice, Transgenic , Oculomotor Muscles/innervation , Oculomotor Muscles/metabolism
7.
Cells ; 13(8)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38667318

ABSTRACT

Muscle satellite cells (MuSCs) are crucial for muscle development and regeneration. The primary pig MuSCs (pMuSCs) is an ideal in vitro cell model for studying the pig's muscle development and differentiation. However, the long-term in vitro culture of pMuSCs results in the gradual loss of their stemness, thereby limiting their application. To address this conundrum and maintain the normal function of pMuSCs during in vitro passaging, we generated an immortalized pMuSCs (SV40 T-pMuSCs) by stably expressing SV40 T-antigen (SV40 T) using a lentiviral-based vector system. The SV40 T-pMuSCs can be stably sub-cultured for over 40 generations in vitro. An evaluation of SV40 T-pMuSCs was conducted through immunofluorescence staining, quantitative real-time PCR, EdU assay, and SA-ß-gal activity. Their proliferation capacity was similar to that of primary pMuSCs at passage 1, and while their differentiation potential was slightly decreased. SiRNA-mediated interference of SV40 T-antigen expression restored the differentiation capability of SV40 T-pMuSCs. Taken together, our results provide a valuable tool for studying pig skeletal muscle development and differentiation.


Subject(s)
Antigens, Polyomavirus Transforming , Cell Differentiation , Satellite Cells, Skeletal Muscle , Animals , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Swine , Antigens, Polyomavirus Transforming/metabolism , Antigens, Polyomavirus Transforming/genetics , Cell Proliferation , Muscle Development , Antigens, Viral, Tumor/metabolism , Antigens, Viral, Tumor/genetics , Simian virus 40/genetics
8.
Biomolecules ; 14(4)2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38672432

ABSTRACT

Sarcopenia has a complex pathophysiology that encompasses metabolic dysregulation and muscle ultrastructural changes. Among the drivers of intracellular and ultrastructural changes of muscle fibers in sarcopenia, mitochondria and their quality control pathways play relevant roles. Mononucleated muscle stem cells/satellite cells (MSCs) have been attributed a critical role in muscle repair after an injury. The involvement of mitochondria in supporting MSC-directed muscle repair is unclear. There is evidence that a reduction in mitochondrial biogenesis blunts muscle repair, thus indicating that the delivery of functional mitochondria to injured muscles can be harnessed to limit muscle fibrosis and enhance restoration of muscle function. Injection of autologous respiration-competent mitochondria from uninjured sites to damaged tissue has been shown to reduce infarct size and enhance cell survival in preclinical models of ischemia-reperfusion. Furthermore, the incorporation of donor mitochondria into MSCs enhances lung and cardiac tissue repair. This strategy has also been tested for regeneration purposes in traumatic muscle injuries. Indeed, the systemic delivery of mitochondria promotes muscle regeneration and restores muscle mass and function while reducing fibrosis during recovery after an injury. In this review, we discuss the contribution of altered MSC function to sarcopenia and illustrate the prospect of harnessing mitochondrial delivery and restoration of MSCs as a therapeutic strategy against age-related sarcopenia.


Subject(s)
Sarcopenia , Satellite Cells, Skeletal Muscle , Signal Transduction , Sarcopenia/metabolism , Sarcopenia/therapy , Sarcopenia/pathology , Humans , Satellite Cells, Skeletal Muscle/metabolism , Animals , Mitochondria/metabolism , Aging/metabolism , Regeneration , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology
9.
Cell Rep ; 43(4): 114052, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38573860

ABSTRACT

Skeletal muscles exert remarkable regenerative or adaptive capacities in response to injuries or mechanical loads. However, the cellular networks underlying muscle adaptation are poorly understood compared to those underlying muscle regeneration. We employed single-cell RNA sequencing to investigate the gene expression patterns and cellular networks activated in overloaded muscles and compared these results with those observed in regenerating muscles. The cellular composition of the 4-day overloaded muscle, when macrophage infiltration peaked, closely resembled that of the 10-day regenerating muscle. In addition to the mesenchymal progenitor-muscle satellite cell (MuSC) axis, interactome analyses or targeted depletion experiments revealed communications between mesenchymal progenitors-macrophages and macrophages-MuSCs. Furthermore, granulin, a macrophage-derived factor, inhibited MuSC differentiation, and Granulin-knockout mice exhibited blunted muscle hypertrophy due to the premature differentiation of overloaded MuSCs. These findings reveal the critical role of granulin through the relayed communications of mesenchymal progenitors, macrophages, and MuSCs in facilitating efficient muscle hypertrophy.


Subject(s)
Cell Differentiation , Hypertrophy , Macrophages , Mesenchymal Stem Cells , Mice, Knockout , Satellite Cells, Skeletal Muscle , Animals , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Granulins , Cell Communication , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Male , Regeneration
10.
FASEB J ; 38(8): e23621, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38651653

ABSTRACT

Denervated myofibers and senescent cells are hallmarks of skeletal muscle aging. However, sparse research has examined how resistance training affects these outcomes. We investigated the effects of unilateral leg extensor resistance training (2 days/week for 8 weeks) on denervated myofibers, senescent cells, and associated protein markers in apparently healthy middle-aged participants (MA, 55 ± 8 years old, 17 females, 9 males). We obtained dual-leg vastus lateralis (VL) muscle cross-sectional area (mCSA), VL biopsies, and strength assessments before and after training. Fiber cross-sectional area (fCSA), satellite cells (Pax7+), denervated myofibers (NCAM+), senescent cells (p16+ or p21+), proteins associated with denervation and senescence, and senescence-associated secretory phenotype (SASP) proteins were analyzed from biopsy specimens. Leg extensor peak torque increased after training (p < .001), while VL mCSA trended upward (interaction p = .082). No significant changes were observed for Type I/II fCSAs, NCAM+ myofibers, or senescent (p16+ or p21+) cells, albeit satellite cells increased after training (p = .037). While >90% satellite cells were not p16+ or p21+, most p16+ and p21+ cells were Pax7+ (>90% on average). Training altered 13 out of 46 proteins related to muscle-nerve communication (all upregulated, p < .05) and 10 out of 19 proteins related to cellular senescence (9 upregulated, p < .05). Only 1 out of 17 SASP protein increased with training (IGFBP-3, p = .031). In conclusion, resistance training upregulates proteins associated with muscle-nerve communication in MA participants but does not alter NCAM+ myofibers. Moreover, while training increased senescence-related proteins, this coincided with an increase in satellite cells but not alterations in senescent cell content or SASP proteins. These latter findings suggest shorter term resistance training is an unlikely inducer of cellular senescence in apparently healthy middle-aged participants. However, similar study designs are needed in older and diseased populations before definitive conclusions can be drawn.


Subject(s)
Cellular Senescence , Resistance Training , Humans , Resistance Training/methods , Male , Female , Middle Aged , Cellular Senescence/physiology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology , Biomarkers/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , PAX7 Transcription Factor/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Adult , Quadriceps Muscle/metabolism , Quadriceps Muscle/innervation
11.
Food Funct ; 15(8): 4575-4585, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38587267

ABSTRACT

Previous studies have shown that vitamin C (VC), an essential vitamin for the human body, can promote the differentiation of muscle satellite cells (MuSCs) in vitro and play an important role in skeletal muscle post-injury regeneration. However, the molecular mechanism of VC regulating MuSC proliferation has not been elucidated. In this study, the role of VC in promoting MuSC proliferation and its molecular mechanism were explored using cell molecular biology and animal experiments. The results showed that VC accelerates the progress of skeletal muscle post-injury regeneration by promoting MuSC proliferation in vivo. VC can also promote skeletal muscle regeneration in the case of atrophy. Using the C2C12 myoblast murine cell line, we observed that VC also stimulated cell proliferation. In addition, after an in vitro study establishing the occurrence of a physical interaction between VC and Pax7, we observed that VC also upregulated the total and nuclear Pax7 protein levels. This mechanism increased the expression of Myf5 (Myogenic Factor 5), a Pax7 target gene. This study establishes a theoretical foundation for understanding the regulatory mechanisms underlying VC-mediated MuSC proliferation and skeletal muscle regeneration. Moreover, it develops the application of VC in animal muscle nutritional supplements and treatment of skeletal muscle-related diseases.


Subject(s)
Ascorbic Acid , Cell Proliferation , Muscle, Skeletal , Myoblasts , PAX7 Transcription Factor , Regeneration , Animals , Male , Mice , Ascorbic Acid/pharmacology , Cell Line , Cell Proliferation/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factor 5/genetics , PAX7 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , Regeneration/drug effects , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/drug effects
12.
Int J Mol Sci ; 25(8)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38673893

ABSTRACT

During embryogenesis, basic fibroblast growth factor (bFGF) is released from neural tube and myotome to promote myogenic fate in the somite, and is routinely used for the culture of adult skeletal muscle (SKM) stem cells (MuSC, called satellite cells). However, the mechanism employed by bFGF to promote SKM lineage and MuSC proliferation has not been analyzed in detail. Furthermore, the question of if the post-translational modification (PTM) of bFGF is important to its stemness-promoting effect has not been answered. In this study, GST-bFGF was expressed and purified from E.coli, which lacks the PTM system in eukaryotes. We found that both GST-bFGF and commercially available bFGF activated the Akt-Erk pathway and had strong cell proliferation effect on C2C12 myoblasts and MuSC. GST-bFGF reversibly compromised the myogenesis of C2C12 myoblasts and MuSC, and it increased the expression of Myf5, Pax3/7, and Cyclin D1 but strongly repressed that of MyoD, suggesting the maintenance of myogenic stemness amid repressed MyoD expression. The proliferation effect of GST-bFGF was conserved in C2C12 over-expressed with MyoD (C2C12-tTA-MyoD), implying its independence of the down-regulation of MyoD. In addition, the repressive effect of GST-bFGF on myogenic differentiation was almost totally rescued by the over-expression of MyoD. Together, these evidences suggest that (1) GST-bFGF and bFGF have similar effects on myogenic cell proliferation and differentiation, and (2) GST-bFGF can promote MuSC stemness and proliferation by differentially regulating MRFs and Pax3/7, (3) MyoD repression by GST-bFGF is reversible and independent of the proliferation effect, and (4) GST-bFGF can be a good substitute for bFGF in sustaining MuSC stemness and proliferation.


Subject(s)
Cell Proliferation , Fibroblast Growth Factor 2 , Muscle Development , MyoD Protein , Myoblasts , Muscle Development/genetics , Animals , Mice , MyoD Protein/metabolism , MyoD Protein/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/genetics , Myoblasts/metabolism , Myoblasts/cytology , Cell Line , PAX7 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , PAX3 Transcription Factor/metabolism , PAX3 Transcription Factor/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factor 5/genetics , Cyclin D1/metabolism , Cyclin D1/genetics , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/cytology , Cell Differentiation , Proto-Oncogene Proteins c-akt/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology
13.
Int J Mol Sci ; 25(7)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38612477

ABSTRACT

Cell division cycle 23 (CDC23) is a component of the tetratricopeptide repeat (TPR) subunit in the anaphase-promoting complex or cyclosome (APC/C) complex, which participates in the regulation of mitosis in eukaryotes. However, the regulatory model and mechanism by which the CDC23 gene regulates muscle production in pigs are largely unknown. In this study, we investigated the expression of CDC23 in pigs, and the results indicated that CDC23 is widely expressed in various tissues and organs. In vitro cell experiments have demonstrated that CDC23 promotes the proliferation of myoblasts, as well as significantly positively regulating the differentiation of skeletal muscle satellite cells. In addition, Gene Set Enrichment Analysis (GSEA) revealed a significant downregulation of the cell cycle pathway during the differentiation process of skeletal muscle satellite cells. The protein-protein interaction (PPI) network showed a high degree of interaction between genes related to the cell cycle pathway and CDC23. Subsequently, in differentiated myocytes induced after overexpression of CDC23, the level of CDC23 exhibited a significant negative correlation with the expression of key factors in the cell cycle pathway, suggesting that CDC23 may be involved in the inhibition of the cell cycle signaling pathway in order to promote the differentiation process. In summary, we preliminarily determined the function of CDC23 with the aim of providing new insights into molecular regulation during porcine skeletal muscle development.


Subject(s)
Muscle, Skeletal , Satellite Cells, Skeletal Muscle , Animals , Anaphase-Promoting Complex-Cyclosome , Muscle Cells , Swine
14.
Sci Rep ; 14(1): 9668, 2024 04 26.
Article in English | MEDLINE | ID: mdl-38671006

ABSTRACT

Massive rotator cuff (RC) tendon tears are associated with progressive fibro-adipogenesis and muscle atrophy that altogether cause shoulder muscle wasting. Platelet derived growth factor ß (PDGFRß) lineage cells, that co-express PDGFRα have previously been shown to directly contribute to scar formation and fat accumulation in a mouse model of irreversible tendon and nerve transection (TTDN). Conversely, PDGFRß+ lineage cells have also been  shown to be myogenic in cultures and in other models of skeletal muscle injury. We therefore hypothesized that PDGFRß demarcates two distinct RC residing subpopulations, fibro-adipogenic and myogenic, and aimed to elucidate the identity of the PDGFRß myogenic precursors and evaluate their contribution, if any, to RC myo-regeneration. Lineage tracing revealed increasing contribution of PDGFRß+ myo-progenitors to the formation of GFP+ myofibers, which were the most abundant myofiber type in regenerated muscle at 2 weeks post-TTDN. Muscle regeneration preceded muscle atrophy and both advanced from the lateral site of tendon transection to the farthest medial region. GFP+/PDGFRß+Sca-1-lin-CXCR4+Integrin-ß1+ marked a novel subset of satellite cells with confirmed myogenic properties. Further studies are warranted to identify the existence of PDGFRß+ satellite cells in human and other mouse muscles and to define their myo-regenerative potential following acute and chronic muscle injury.


Subject(s)
Cell Lineage , Receptor, Platelet-Derived Growth Factor beta , Regeneration , Rotator Cuff Injuries , Satellite Cells, Skeletal Muscle , Animals , Receptor, Platelet-Derived Growth Factor beta/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Mice , Rotator Cuff Injuries/pathology , Rotator Cuff Injuries/metabolism , Muscle Development , Disease Models, Animal , Rotator Cuff/pathology , Rotator Cuff/metabolism , Male
15.
Am J Physiol Cell Physiol ; 326(4): C1193-C1202, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38581669

ABSTRACT

Satellite cells (SCs) and fibroadipogenic progenitors (FAPs) are progenitor populations found in muscle that form new myofibers postinjury. Muscle development, regeneration, and tissue-engineering experiments require robust progenitor populations, yet their isolation and expansion are difficult given their scarcity in muscle, limited muscle biopsy sizes in humans, and lack of methodological detail in the literature. Here, we investigated whether a dispase and collagenase type 1 and 2 cocktail could allow dual isolation of SCs and FAPs, enabling significantly increased yield from human skeletal muscle. Postdissociation, we found that single cells could be sorted into CD56 + CD31-CD45- (SC) and CD56-CD31-CD45- (FAP) cell populations, expanded in culture, and characterized for lineage-specific marker expression and differentiation capacity; we obtained ∼10% SCs and ∼40% FAPs, with yields twofold better than what is reported in current literature. SCs were PAX7+ and retained CD56 expression and myogenic fusion potential after multiple passages, expanding up to 1012 cells. Conversely, FAPs expressed CD140a and differentiated into either fibroblasts or adipocytes upon induction. This study demonstrates robust isolation of both SCs and FAPs from the same muscle sample with SC recovery more than two times higher than previously reported, which could enable translational studies for muscle injuries.NEW & NOTEWORTHY We demonstrated that a dispase/collagenase cocktail allows for simultaneous isolation of SCs and FAPs with 2× higher SC yield compared with other studies. We provide a thorough characterization of SC and FAP in vitro expansion that other studies have not reported. Following our dissociation, SCs and FAPs were able to expand by up to 1012 cells before reaching senescence and maintained differentiation capacity in vitro demonstrating their efficacy for clinical translation for muscle injury.


Subject(s)
Endopeptidases , Muscle, Skeletal , Satellite Cells, Skeletal Muscle , Humans , Muscle, Skeletal/metabolism , Cell Differentiation/physiology , Satellite Cells, Skeletal Muscle/metabolism , Fibroblasts/metabolism
16.
Development ; 151(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38573009

ABSTRACT

Adhesion of muscle stem cells to their niche provides stable anchorage, and biochemical and biomechanical signals required for quiescence. In their work, Robert Krauss and colleagues reveal the role of catenin/cadherin-based adhesion interactions in maintaining niche localization. To find out more about their work, we spoke to the first author, Margaret Hung, and the corresponding author, Robert Krauss, Professor in Cell, Developmental and Regenerative Biology at the Icahn School of Medicine, Mount Sinai.


Subject(s)
Cadherins , Satellite Cells, Skeletal Muscle , Humans , Cell Differentiation , Cell Division
17.
Curr Top Dev Biol ; 158: 1-14, 2024.
Article in English | MEDLINE | ID: mdl-38670701

ABSTRACT

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.


Subject(s)
Muscle Development , Muscle, Skeletal , Satellite Cells, Skeletal Muscle , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/physiology , Animals , Muscle, Skeletal/growth & development , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Humans , PAX7 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , Cell Differentiation
18.
Curr Top Dev Biol ; 158: 123-150, 2024.
Article in English | MEDLINE | ID: mdl-38670702

ABSTRACT

Preserving the potency of stem cells in adult tissues is very demanding and relies on the concerted action of various cellular and non-cellular elements in a precise stoichiometry. This balanced microenvironment is found in specific anatomical "pockets" within the tissue, known as the stem cell niche. In this review, we explore the interplay between stem cells and their niches, with a primary focus on skeletal muscle stem cells and the extracellular matrix (ECM). Quiescent muscle stem cells, known as satellite cells are active producers of a diverse array of ECM molecules, encompassing major constituents like collagens, laminins, and integrins, some of which are explored in this review. The conventional perception of ECM as merely a structural scaffold is evolving. Collagens can directly interact as ligands with receptors on satellite cells, while other ECM proteins have the capacity to sequester growth factors and regulate their release, especially relevant during satellite cell turnover in homeostasis or activation upon injury. Additionally, we explore an evolutionary perspective on the ECM across a range of multicellular organisms and discuss a model wherein satellite cells are self-sustained by generating their own niche. Considering the prevalence of ECM proteins in the connective tissue of various organs it is not surprising that mutations in ECM genes have pathological implications, including in muscle, where they can lead to myopathies. However, the particular role of certain disease-related ECM proteins in stem cell maintenance highlights the potential contribution of stem cell deregulation to the progression of these disorders.


Subject(s)
Extracellular Matrix , Satellite Cells, Skeletal Muscle , Stem Cell Niche , Humans , Extracellular Matrix/metabolism , Animals , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/cytology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics
19.
Curr Top Dev Biol ; 158: 179-201, 2024.
Article in English | MEDLINE | ID: mdl-38670705

ABSTRACT

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.


Subject(s)
Cellular Microenvironment , Extracellular Matrix , Muscle, Skeletal , Satellite Cells, Skeletal Muscle , Humans , Animals , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/physiology , Satellite Cells, Skeletal Muscle/metabolism , Extracellular Matrix/metabolism , Muscle, Skeletal/physiology , Muscle, Skeletal/cytology , Adaptation, Physiological , Stem Cell Niche/physiology , Regeneration/physiology , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Stem Cells/cytology , Stem Cells/physiology
20.
Curr Top Dev Biol ; 158: 221-238, 2024.
Article in English | MEDLINE | ID: mdl-38670707

ABSTRACT

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.


Subject(s)
Muscle, Skeletal , Regeneration , Regeneration/physiology , Animals , Humans , Muscle, Skeletal/physiology , Muscle, Skeletal/cytology , Muscle Development , Stem Cells/cytology , Stem Cells/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/physiology , Cell Differentiation , Immunologic Factors/pharmacology , Immunologic Factors/metabolism , Immunomodulation
SELECTION OF CITATIONS
SEARCH DETAIL
...