Skeletal muscle stem cells

  • 文章类型: Journal Article
    成体骨骼肌干细胞(MuSC)是肌纤维的再生前体,在肌纤维生长中也有重要作用。适应,和通过融合到肌纤维来维持-这个过程被称为“肌核增生”。\"由于在再生过程中专注于MuSC功能,肌纤维仍然是影响MuSC命运的MuSC生态位的一个很大程度上被忽视的组成部分。这里,我们描述了一种使用基于乙炔基-2'-脱氧尿苷(EdU)的MuSC后代示踪直接测量体外和体内肌核增生率的方法。该方法支持解剖影响肌核增生的MuSC内在和肌纤维衍生因子,作为支持肌纤维稳态和可塑性的MuSC的替代命运。
    Adult skeletal muscle stem cells (MuSC) are the regenerative precursors of myofibers and also have an important role in myofiber growth, adaptation, and maintenance by fusing to the myofibers-a process referred to as \"myonuclear accretion.\" Due to a focus on MuSC function during regeneration, myofibers remain a largely overlooked component of the MuSC niche influencing MuSC fate. Here, we describe a method to directly measure the rate of myonuclear accretion in vitro and in vivo using ethynyl-2\'-deoxyuridine (EdU)-based tracing of MuSC progeny. This method supports the dissection of MuSC intrinsic and myofiber-derived factors influencing myonuclear accretion as an alternative fate of MuSCs supporting myofiber homeostasis and plasticity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    进行性肌肉无力和变性是杜氏肌营养不良症(DMD)的特征,一个致命的,影响1/5,000男孩的x连锁神经肌肉疾病。肌养蛋白蛋白的丢失导致复发性肌肉变性,进行性纤维化,慢性炎症,和骨骼肌干细胞的功能障碍,称为卫星细胞。不幸的是,目前没有治疗DMD的方法。在这个迷你评论中,我们讨论了营养不良肌肉中的卫星细胞是如何功能受损的,以及这如何导致DMD病理学,以及恢复内源性卫星细胞功能的巨大潜力,作为治疗这种使人衰弱和致命的疾病的可行治疗策略。
    Progressive muscle weakness and degeneration characterize Duchenne muscular dystrophy (DMD), a lethal, x-linked neuromuscular disorder that affects 1 in 5,000 boys. Loss of dystrophin protein leads to recurrent muscle degeneration, progressive fibrosis, chronic inflammation, and dysfunction of skeletal muscle resident stem cells, called satellite cells. Unfortunately, there is currently no cure for DMD. In this mini review, we discuss how satellite cells in dystrophic muscle are functionally impaired, and how this contributes to the DMD pathology, and the tremendous potential of restoring endogenous satellite cell function as a viable treatment strategy to treat this debilitating and fatal disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    干细胞具有分化成特化细胞类型的独特能力。这些专门的细胞类型可用于再生医学目的,例如细胞治疗。肌细胞,也被称为骨骼肌干细胞(MuSCs),在成长中发挥重要作用,修复,和骨骼肌组织的再生。然而,尽管它具有治疗潜力,成功的差异化,扩散,由于多种因素,MuSC的扩增过程仍然是一个重大挑战。例如,通过使用机械力积极复制MuSCs微环境(称为生态位),可以极大地影响MuSCs的生长和分化。然而,机械生物学在MuSC生长中的分子作用,扩散,和再生医学的分化仍然知之甚少。在本次审查中,我们全面总结,比较,并批判性地分析不同的机械线索如何塑造干细胞生长,扩散,分化,以及它们在疾病发展中的潜在作用(图。1).从干细胞的机械生物学中获得的见解也将有助于如何使用MuSC将这些应用用于再生目的。
    Stem cells possess the unique ability to differentiate into specialized cell types. These specialized cell types can be used for regenerative medicine purposes such as cell therapy. Myosatellite cells, also known as skeletal muscle stem cells (MuSCs), play important roles in the growth, repair, and regeneration of skeletal muscle tissues. However, despite its therapeutic potential, the successful differentiation, proliferation, and expansion processes of MuSCs remain a significant challenge due to a variety of factors. For example, the growth and differentiation of MuSCs can be greatly influenced by actively replicating the MuSCs microenvironment (known as the niche) using mechanical forces. However, the molecular role of mechanobiology in MuSC growth, proliferation, and differentiation for regenerative medicine is still poorly understood. In this present review, we comprehensively summarize, compare, and critically analyze how different mechanical cues shape stem cell growth, proliferation, differentiation, and their potential role in disease development (Fig. 1). The insights developed from the mechanobiology of stem cells will also contribute to how these applications can be used for regenerative purposes using MuSCs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Skeletal muscles have an extraordinary capacity to regenerate themselves when injured. Skeletal muscle stem cells, called satellite cells, play a central role in muscle regeneration via three major steps: activation, proliferation, and differentiation. These steps are affected by multiple types of cells, such as immune cells, fibro-adipogenic progenitor cells, and vascular endothelial cells. The widespread use of single-cell sequencing technologies has enabled the identification of novel cell subpopulations associated with muscle regeneration and their regulatory mechanisms. This review summarizes the dynamism of the cellular community that controls and promotes muscle regeneration, with a particular focus on skeletal muscle stem cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    骨骼肌干细胞,被称为卫星细胞(SC),在正常的成人肢体肌肉中处于静止状态。损伤刺激SC增殖,分化,和融合再生肌肉结构。在咽部肌肉中,这对吞咽食物和液体至关重要,SCs在没有损伤的情况下增殖和融合。尚不清楚哪些因素导致咽部SCs的基础活性增加。这里,我们确定了生态位因素如何影响咽部和肢体SCs的状态。在体内,咽部SCs的子集呈现激活的SCs的特征,包括大细胞大小和增加的线粒体含量。在这项研究中,我们发现咽肌含有高水平的活性肝细胞生长因子(HGF),已知在小鼠和人类中激活SCs。我们发现纤维脂肪原细胞(FAP)是提供HGF的主要细胞类型,因此负责咽肌中SC的基础增殖。最后,我们证实了FAP对于咽肌功能和维持的关键作用.这项研究为解释咽部肌肉的独特SC活动提供了新的见解。
    Skeletal muscle stem cells, known as satellite cells (SCs), are quiescent in normal adult limb muscles. Injury stimulates SC proliferation, differentiation, and fusion to regenerate muscle structure. In pharyngeal muscles, which are critical for swallowing foods and liquids, SCs proliferate and fuse in the absence of injury. It is unknown what factors drive increased basal activity of pharyngeal SCs. Here, we determined how niche factors influence the status of pharyngeal versus limb SCs. In vivo, a subset of pharyngeal SCs present features of activated SCs, including large cell size and increased mitochondrial content. In this study, we discovered that the pharyngeal muscle contains high levels of active hepatocyte growth factor (HGF), which is known to activate SCs in mice and humans. We found that fibroadipogenic progenitors (FAPs) are the major cell type providing HGF and are thus responsible for basal proliferation of SCs in pharyngeal muscles. Lastly, we confirmed the critical role of FAPs for pharyngeal muscle function and maintenance. This study gives new insights to explain the distinctive SC activity of pharyngeal muscles.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    人多能干细胞(hPSC)衍生的肌源性祖细胞(MPC)移植是多种退行性肌肉疾病的有希望的治疗方法。这里,使用MPC特异性荧光报告系统(PAX7::GFP),我们证明,hPSC衍生的MPC可以促进局部损伤后小鼠和缺乏肌营养不良蛋白(mdx)的小鼠的肌纤维再生。我们还证明了PAX7::GFPMPCs的子集在再生肌纤维的基底层内移植,采用静止状态,并有助于再损伤和mdx小鼠模型的再生。PAX7::GFPMPC的这个子集经历成熟过程并重塑其分子特征以类似于体内植入后的晚期胎儿MPC/成人卫星细胞的分子特征。这些体内成熟的PAX7::GFPMPC保留了细胞自主再生能力,并且可以在次级受体小鼠的小生境中重新繁殖,为未来基于hPSC的肌肉疾病细胞治疗提供原理证明。
    Human pluripotent stem cell (hPSC)-derived myogenic progenitor cell (MPC) transplantation is a promising therapeutic approach for a variety of degenerative muscle disorders. Here, using an MPC-specific fluorescent reporter system (PAX7::GFP), we demonstrate that hPSC-derived MPCs can contribute to the regeneration of myofibers in mice following local injury and in mice deficient of dystrophin (mdx). We also demonstrate that a subset of PAX7::GFP MPCs engraft within the basal lamina of regenerated myofibers, adopt a quiescent state, and contribute to regeneration upon reinjury and in mdx mouse models. This subset of PAX7::GFP MPCs undergo a maturation process and remodel their molecular characteristics to resemble those of late-stage fetal MPCs/adult satellite cells following in vivo engraftment. These in-vivo-matured PAX7::GFP MPCs retain a cell-autonomous ability to regenerate and can repopulate in the niche of secondary recipient mice, providing a proof of principle for future hPSC-based cell therapy for muscle disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    骨骼肌干细胞(SMSCs)对其生长至关重要,维护,和肌肉的修复;新出现的证据表明Toll样受体4(TLR4)可以潜在地调节肌肉再生。在目前的研究中,体外和体内实验探讨TLR4与富含亮氨酸的胶质瘤灭活因子1(LGI1)的相关性及其对SMSCs增殖和成骨潜能的影响。为了研究TLR4和LGI1在SMSCs中的调控机制,获得的细胞用脂多糖(LPS,用作不同时间点不同浓度的TLR4的激活剂)以及针对LGI1的siRNA。随后,为了测量SMSCs的增殖和分化潜能,进行了一系列检测,这涉及到相关因素的检测,细胞活性,和球体形成能力。LPS治疗后,观察到TLR4表达增加和LGI1表达减少。因此,我们还发现Erk信号通路失活,细胞增殖和成骨能力下降,相关因子如细胞周期蛋白B1和runt相关转录因子2的下调。此外,SMSCs的细胞活性和成球性能也下降。这些结果也在盲肠结扎和穿孔诱导的脓毒症大鼠模型中得到了验证。总之,本研究揭示了SMSCs中的调节机制,通过TLR4降低LGI1表达,从而阻碍细胞增殖和成骨,强调TLR4是针对许多与SMSCs相关的疾病的潜在治疗靶标。
    Skeletal muscle stem cells (SMSCs) are vital to the growth, maintenance, and repair of the muscles; emerging evidence has indicated that Toll-like receptor 4 (TLR4) can potentially regulate muscle regeneration. In present study, in vitro and in vivo experiments were performed to explore the correlation of TLR4 with leucine-rich glioma-inactivated 1 (LGI1) as well as their effects on the proliferation and osteogenesis potential of SMSCs. In order to examine the regulatory mechanisms of TLR4 and LGI1 in SMSCs, the obtained cells were treated with lipopolysaccharide (LPS, used as an activator of TLR4) of different concentration at different time points as well as the siRNA against LGI1. Subsequently, a series of detection was undertaken in order to measure the proliferation and differentiation potential of SMSCs, which involved detection of the related factors, cell activity, and the sphere-forming capability. Following LPS treatment, the increased TLR4 expression and reduced LGI1 expression were observed. Consequently, we also discovered that Erk signaling pathway was inactivated and cell proliferation and osteogenesis capabilities declined, presented by the downregulation of related factors such as cyclin B1 and runt-related transcription factor 2. Moreover, the cell activity and sphere-formation performance of SMSCs were also declined. These results were also validated in rats with cecal ligation and perforation-induced rat models with sepsis. In conclusion, the present study reveals a regulatory mechanism in SMSCs whereby LGI1 expression is reduced by TLR4, thus impeding cell proliferation and osteogenesis, highlighting TLR4 as a potential therapeutic target against many diseases related to SMSCs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    骨质疏松症是一种代谢性疾病,骨质疏松性骨折(OPF)是其最严重的并发症之一。常忽略骨折周围肌肉对OPF愈合的影响。我们旨在阐明骨骼肌卫星细胞(SMSCs)在β-catenin促进OPF愈合中的作用。为了提高我们对SMSC的理解,让我们探索它作为治疗靶点的潜力。
    骨骼肌是从对照非OPF或OPF患者中获得的,用于原代SMSCs培养(n=3,33%的女性,平均年龄60±15.52)。测量SMSCs的表达。在体内,3个月大的雌性C57BL/6小鼠接受OVX手术。三个月后,再次制作左胫骨骨折模型.对照组和治疗组(n=24,每组,female).治疗组用激动剂(蛇床子素)治疗。在7、14和28三个时间点检测肌肉中的SMSCs和骨折愈合(n=8、8、8,女性)。为了进一步澄清科学假设,我们创新性地使用Pax7-CreERT2/+;β-cateninfx/fx转基因小鼠(每组n=12,male).敲除SMSC中的β-catenin观察SMSCs的增殖和成骨分化,OPF治疗来自3月龄产仔阴性β-cateninfx/fx转基因小鼠的SMSCs的体外原代细胞。腺病毒CRE转染后,观察到SMSC的成肌和成骨分化。
    我们发现人SMSCs降低OPF患者的增殖和成骨分化(-38.63%,P<0.05)。通过动物实验,发现β-catenin的激活促进骨折部位SMSC的增殖和成骨分化,从而加速骨折部位的愈合(189.47%,P<0.05)。为了证明这个观点,在体内Pax7-CreERT2/+;β-cateninfx/fx转基因小鼠实验,我们创新性地发现,敲除SMSC中的β-catenin会导致骨量和骨微结构的减少,并伴有骨折延迟愈合(-35.04%,P<0.001)。同时,通过体外培养实验,发现它们的肌源性(-66.89%,P<0.01)和成骨分化(-16.5%,P<0.05)能力下降。
    这些结果为SMSCs直接促进OPF愈合提供了第一个实际证据,具有重要的临床意义,因为它可能有助于治疗OPFs的延迟愈合和不愈合。和动员自体干细胞疗法在骨科应用中的应用。
    Osteoporosis is a metabolic disease, and osteoporotic fracture (OPF) is one of its most serious complications. It is often ignored that the influence of the muscles surrounding the fracture on the healing of OPF. We aimed to clarify the role of skeletal muscle satellite cells (SMSCs) in promoting OPF healing by β-catenin, to improve our understanding of SMSCs, and let us explore its potential as a therapeutic target.
    Skeletal muscles were obtained from control non-OPF or OPF patients for primary SMSCs culture (n = 3, 33% females, mean age 60 ± 15.52). Expression of SMSCs was measured. In vivo, 3-month-old female C57BL/6 mice underwent OVX surgery. Three months later, the left tibia fracture model was again performed. The control and the treatment group (n = 24, per group, female). The treatment group was treated with an agonist (osthole). Detection of SMSCs in muscles and fracture healing at 7, 14, and 28 three time points (n = 8, 8, 8, female). To further clarify the scientific hypothesis, we innovatively used Pax7-CreERT2/+ ;β-cateninfx/fx transgenic mice (n = 12, per group, male). Knock out β-catenin in SMSC to observe the proliferation and osteogenic differentiation of SMSCs, and OPF healing. In vitro primary cells of SMSCs from 3-month-old litter-negative β-cateninfx/fx transgenic mice. After adenovirus-CRE transfection, the myogenic and osteogenic differentiation of SMSC was observed.
    We find that human SMSCs reduced proliferation and osteogenic differentiation in patients with OPF (-38.63%, P < 0.05). And through animal experiments, it was found that activation of β-catenin promoted the proliferation and osteogenic differentiation of SMSC at the fracture site, thereby accelerating the healing of the fracture site (189.47%, P < 0.05). To prove this point of view, in the in vivo Pax7-CreERT2/+ ;β-cateninfx/fx transgenic mouse experiment, we innovatively found that knocking out β-catenin in SMSC will cause a decrease in bone mass and bone microstructure, and accompanied by delayed fracture healing (-35.04%, P < 0.001). At the same time, through in vitro SMSC culture experiments, it was found that their myogenic (-66.89%, P < 0.01) and osteogenic differentiation (-16.5%, P < 0.05) ability decreased.
    These results provide the first practical evidence for a direct contribution of SMSCs to promote the healing of OPF with important clinical implications as it may help in the treatment of delayed healing and non-union of OPFs, and mobilization of autologous stem cell therapy in orthopaedic applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    骨骼肌干细胞,即,卫星细胞(SC),是损伤或慢性退行性肌病后骨骼肌再生的新肌核的重要来源。在老化过程中,SC数和再生容量都减少。然而,控制初始SC池大小的分子调节剂是未知的。我们证明成纤维细胞生长因子6(FGF6)对于SC池缩放至关重要。缺乏FGF6的小鼠具有减少的出生后早期起源的SCs和受损的再生。相比之下,在出生后早期增加FGF6足以进行SC扩张。一起,这些数据支持FGF6对于在关键的出生后时期调节SC数量以建立静止的成体肌肉干细胞池是必要且足够的.我们的工作强调了产后发育作为通过生长因子信号传导扩展体干细胞群的时间窗口,这可能与设计新的生物医学策略以增强组织再生有关。
    Skeletal muscle stem cells, i.e., satellite cells (SCs), are the essential source of new myonuclei for skeletal muscle regeneration following injury or chronic degenerative myopathies. Both SC number and regenerative capacity diminish during aging. However, molecular regulators that govern sizing of the initial SC pool are unknown. We demonstrate that fibroblast growth factor 6 (FGF6) is critical for SC pool scaling. Mice lacking FGF6 have reduced SCs of early postnatal origin and impaired regeneration. By contrast, increasing FGF6 during the early postnatal period is sufficient for SC expansion. Together, these data support that FGF6 is necessary and sufficient to modulate SC numbers during a critical postnatal period to establish the quiescent adult muscle stem cell pool. Our work highlights postnatal development as a time window receptive for scaling a somatic stem cell population via growth factor signaling, which might be relevant for designing new biomedical strategies to enhance tissue regeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    鉴定与肌肉发育相关的基因对于提高猪的肉产量和质量至关重要。胰岛素降解酶(IDE),巯基锌-金属内肽酶,已知可以调节小鼠和大鼠成肌细胞的成肌过程,而其在猪中的肌源性作用仍然难以捉摸。因此,本研究旨在探讨IDE对猪骨骼肌干细胞(PSMSCs)增殖和凋亡的影响及其分子机制。我们发现IDE在猪组织中广泛表达,包括肾脏,肺,脾,脾肝脏,心,和骨骼肌。然后,探讨IDE对PSMSCs增殖和凋亡的影响,我们对细胞进行siRNA介导的IDE表达敲低,促进细胞增殖,减少细胞凋亡。作为肌生成过程中的关键转录因子之一,MYOD,其表达也随着IDE敲低而降低。为了进一步阐明潜在的分子机制,进行RNA测序。在被IDE击倒后扰乱的成绩单中,被称为肌肉生长的负调节因子的下调基因肌肉生长抑制素(MSTN)吸引了我们的兴趣。的确,MSTN击倒导致与IDE击倒类似的结果,随着细胞周期相关基因的上调,MYOD以及凋亡相关基因的下调,增强细胞增殖。一起来看,我们的研究结果表明,IDE通过MSTN/MYOD通路调节PSMSCs的增殖和凋亡。因此,我们招募了猪骨骼肌发育调节因子基因家族的IDE,并建议将IDE作为基因的一个例子进行优先排序,以提高猪肉产量。
    Identifying the genes relevant for muscle development is pivotal to improve meat production and quality in pigs. Insulin-degrading enzyme (IDE), a thiol zinc-metalloendopeptidase, has been known to regulate the myogenic process of mouse and rat myoblast cell lines, while its myogenic role in pigs remained elusive. Therefore, the current study aimed to identify the effects of IDE on the proliferation and apoptosis of porcine skeletal muscle stem cells (PSMSCs) and underlying molecular mechanism. We found that IDE was widely expressed in porcine tissues, including kidney, lung, spleen, liver, heart, and skeletal muscle. Then, to explore the effects of IDE on the proliferation and apoptosis of PSMSCs, we subjected the cells to siRNA-mediated knockdown of IDE expression, which resulted in promoted cell proliferation and reduced apoptosis. As one of key transcription factors in myogenesis, MYOD, its expression was also decreased with IDE knockdown. To further elucidate the underlying molecular mechanism, RNA sequencing was performed. Among transcripts perturbed by the IDE knockdown after, a downregulated gene myostatin (MSTN) which is known as a negative regulator for muscle growth attracted our interest. Indeed, MSTN knockdown led to similar results as those of the IDE knockdown, with upregulation of cell cycle-related genes, downregulation of MYOD as well as apoptosis-related genes, and enhanced cell proliferation. Taken together, our findings suggest that IDE regulates the proliferation and apoptosis of PSMSCs via MSTN/MYOD pathway. Thus, we recruit IDE to the gene family of regulators for porcine skeletal muscle development and propose IDE as an example of gene to prioritize in order to improve pork production.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号