Satellite cell

卫星小区
  • 文章类型: Journal Article
    骨骼肌在受伤时经历强大的再生,浸润的免疫细胞不仅在清除受损组织中起主要作用,而且还通过分泌的细胞因子调节成肌过程。趋化因子C-C基序配体8(Ccl8),已报道与Ccl2和Ccl7一起介导炎症反应以抑制肌肉再生。Ccl8也由肌肉细胞表达,但是肌肉细胞来源的Ccl8在肌生成中的作用尚未被报道。在这项研究中,我们发现,敲除Ccl8,而不是Ccl2或Ccl7,导致C2C12成肌细胞分化增加。对现有单细胞转录组数据集的分析显示,再生肌肉中的免疫细胞和肌肉干细胞(MuSC)都表达Ccl8,而MuSC的表达水平要低得多。CCl8表达的时间模式在MuSCs和巨噬细胞中不同。为了探索肌细胞来源的Ccl8在体内的功能,我们使用了一个小鼠系统,其中Cas9在Pax7+生肌祖细胞(MPCs)中表达,Ccl8基因编辑由AAV9递送的sgRNA诱导.Pax7MPCs中Ccl8的消耗导致年轻和中年小鼠氯化钡诱导的损伤后肌肉再生加速,和肌内施用重组Ccl8逆转了表型。当通过类似方法在Myf5+或MyoD+MPC中耗尽Ccl8时,也观察到加速再生。我们的结果表明,肌细胞来源的Ccl8在调节损伤诱导的肌肉再生过程中肌源性分化的启动中起着独特的作用。
    Skeletal muscles undergo robust regeneration upon injury, and infiltrating immune cells play a major role in not only clearing damaged tissues but also regulating the myogenic process through secreted cytokines. Chemokine C-C motif ligand 8 (Ccl8), along with Ccl2 and Ccl7, has been reported to mediate inflammatory responses to suppress muscle regeneration. Ccl8 is also expressed by muscle cells, but a role of the muscle cell-derived Ccl8 in myogenesis has not been reported. In this study, we found that knockdown of Ccl8, but not Ccl2 or Ccl7, led to increased differentiation of C2C12 myoblasts. Analysis of existing single-cell transcriptomic datasets revealed that both immune cells and muscle stem cells (MuSCs) in regenerating muscles express Ccl8, with the expression by MuSCs at a much lower level, and that the temporal patterns of Ccl8 expression were different in MuSCs and macrophages. To probe a function of muscle cell-derived Ccl8 in vivo, we utilized a mouse system in which Cas9 was expressed in Pax7+ myogenic progenitor cells (MPCs) and Ccl8 gene editing was induced by AAV9-delivered sgRNA. Depletion of Ccl8 in Pax7+ MPCs resulted in accelerated muscle regeneration after barium chloride-induced injury in both young and middle-aged mice, and intramuscular administration of a recombinant Ccl8 reversed the phenotype. Accelerated regeneration was also observed when Ccl8 was depleted in Myf5+ or MyoD+ MPCs by similar approaches. Our results suggest that muscle cell-derived Ccl8 plays a unique role in regulating the initiation of myogenic differentiation during injury-induced muscle 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
    骨骼肌是再生能力最高的组织之一,一个精细控制的过程,关键取决于肌肉干细胞。肌肉干细胞功能取决于内在信号通路和与其直接生态位的相互作用。受伤后,静止的肌肉干细胞被激活,增殖并融合形成新的肌纤维,涉及骨骼肌再生中多种细胞类型相互作用的过程。肌肉干细胞中的受体通过直接的细胞-细胞相互作用接收各自的信号,通过分泌因子或细胞-基质相互作用的信号传导,从而调节肌肉干细胞对外部刺激的反应。这里,我们讨论了肌肉干细胞如何与它们的直接生态位相互作用,重点是如何控制它们的静止,激活和自我更新,以及这些过程如何在年龄和疾病中改变。
    Skeletal muscle is one of the tissues with the highest ability to regenerate, a finely controlled process which is critically depending on muscle stem cells. Muscle stem cell functionality depends on intrinsic signaling pathways and interaction with their immediate niche. Upon injury quiescent muscle stem cells get activated, proliferate and fuse to form new myofibers, a process involving the interaction of multiple cell types in regenerating skeletal muscle. Receptors in muscle stem cells receive the respective signals through direct cell-cell interaction, signaling via secreted factors or cell-matrix interactions thereby regulating responses of muscle stem cells to external stimuli. Here, we discuss how muscle stem cells interact with their immediate niche focusing on how this controls their quiescence, activation and self-renewal and how these processes are altered in age and 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
    股四头肌在膝骨关节炎(OA)的进展和症状表现中起着关键作用,特别是疼痛。这项研究调查了肌肉增强和支持疗法(MEST)的治疗效果,一种最近开发的用于肌内插入cog聚二恶烷酮细丝的装置,在股四头肌修复中缓解OA疼痛。通过单碘乙酸盐注射在SpragueDawley大鼠中诱导膝关节OA。在OA或幼稚大鼠股四头肌中进行MEST或假治疗。使用爪退缩阈值和负重来评估疼痛。使用生物标志物评估股四头肌损伤和通过MEST恢复,组织形态学,肌肉质量,收缩力和后肢扭矩。卫星细胞和巨噬细胞活化,以及他们的活化剂,也进行了评估。在MEST治疗后1周和3周比较数据(M-W1和M-W3)。MEST治疗OA大鼠引起的肌肉损伤,血清天冬氨酸转移酶和肌酐激酶水平升高,和M-W1处的局部β-肌动蛋白变化。这种损伤引发了促炎巨噬细胞和卫星细胞活化,伴随着白细胞介素-6和胰岛素样生长因子-1水平升高。然而,通过M-W3,这些过程逐渐转向炎症消退和肌肉恢复。这在抗炎巨噬细胞表型中可见,持续的卫星细胞活化和损伤标志物回归基线。在M-W3时,股四头肌从萎缩中恢复的质量和力量与显著减轻OA疼痛相关。这项研究表明,MEST诱导的轻微肌肉损伤会触发巨噬细胞和卫星细胞激活,导致OA大鼠股四头肌萎缩恢复和疼痛缓解。
    Quadriceps muscles play a pivotal role in knee osteoarthritis (OA) progression and symptom manifestation, particularly pain. This research investigates the therapeutic effectiveness of muscle enhancement and support therapy (MEST), a recently developed device intended for intramuscular insertion of cog polydioxanone filaments, in quadriceps restoration to alleviate OA pain. Knee OA was induced in Sprague Dawley rats via monoiodoacetate injections. MEST or sham treatment was performed in OA or Naive rat quadriceps. Pain was assessed using paw withdrawal threshold and weight bearing. Quadriceps injury and recovery via MEST were evaluated using biomarkers, tissue morphology, muscle mass, contractile force and hindlimb torque. Satellite cell and macrophage activation, along with their activators, were also assessed. Data were compared at 1- and 3-weeks post-MEST treatment (M-W1 and M-W3). MEST treatment in OA rats caused muscle injury, indicated by elevated serum aspartate transferase and creatinine kinase levels, and local β-actin changes at M-W1. This injury triggered pro-inflammatory macrophage and satellite cell activation, accompanied by heightened interleukin-6 and insulin-like growth factor-1 levels. However, by M-W3, these processes gradually shifted toward inflammation resolution and muscle restoration. This was seen in anti-inflammatory macrophage phenotypes, sustained satellite cell activation and injury markers regressing to baseline. Quadriceps recovery in mass and strength from atrophy correlated with substantial OA pain reduction at M-W3. This study suggests that MEST-induced minor muscle injury triggers macrophage and satellite cell activation, leading to recovery of atrophied quadriceps and pain relief in OA rats.
    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
    内皮和骨骼肌谱系产生于常见的胚胎祖细胞。尽管他们共同的发展起源,成人内皮细胞(ECs)和肌肉干细胞(MuSCs)(卫星细胞)被认为具有不同的基因特征和信号通路.在这里,我们通过揭示成年MuSC行为如何受到EC转录本子集表达的影响来改变这种范式。我们使用了几种计算分析,包括单细胞RNAseq,以显示MuSC在小鼠中表达低水平的经典EC标记。我们证明了MuSC的存活受一个这样的原型内皮信号通路(VEGFA-FLT1)的调节。使用药理学和遗传功能增益和丧失研究,我们确定FLT1-AKT1轴是VEGFA介导的MuSC存活调节的关键效应子.一起,我们的数据支持VEGFA-FLT1-AKT1途径促进肌肉再生过程中的MuSC存活,并强调了选择转录物的次要表达如何足以影响细胞行为。
    Endothelial and skeletal muscle lineages arise from common embryonic progenitors. Despite their shared developmental origin, adult endothelial cells (ECs) and muscle stem cells (MuSCs; satellite cells) have been thought to possess distinct gene signatures and signaling pathways. Here, we shift this paradigm by uncovering how adult MuSC behavior is affected by the expression of a subset of EC transcripts. We used several computational analyses including single-cell RNA-seq (scRNA-seq) to show that MuSCs express low levels of canonical EC markers in mice. We demonstrate that MuSC survival is regulated by one such prototypic endothelial signaling pathway (VEGFA-FLT1). Using pharmacological and genetic gain- and loss-of-function studies, we identify the FLT1-AKT1 axis as the key effector underlying VEGFA-mediated regulation of MuSC survival. All together, our data support that the VEGFA-FLT1-AKT1 pathway promotes MuSC survival during muscle regeneration, and highlights how the minor expression of select transcripts is sufficient for affecting cell behavior.
    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
    由各种各样的细胞组成,骨骼肌是人体损伤后具有显著再生能力的组织之一。再生过程中的关键参与者之一是肌肉卫星细胞(MuSC),骨骼肌干细胞群,因为它是新肌纤维的来源。在稳态期间维持MuSC静止涉及MuSC与成年骨骼肌中相应小生境中的其他细胞之间的复杂相互作用。受伤后,MuSCs被激活进入细胞周期进行细胞增殖并分化为肌管,然后是成熟的肌纤维再生肌肉。尽管经过几十年的研究,MuSC维持和激活的确切机制仍然难以捉摸。分析MuSCs的传统方法,包括细胞培养,动物模型,和基因表达分析,提供对MuSC生物学的一些见解,但缺乏复制3维(3-D)体内肌肉环境和全面捕获动态过程的能力。成像技术的最新进展,包括共焦,活体内,和多光子显微镜,为观察和表征动态MuSC形态和行为提供了有希望的途径。本章旨在回顾3-D和实时成像方法,这些方法有助于发现对MuSC行为的见解,形态变化,肌肉小生境内的相互作用,和在静止到激活(Q-A)过渡期间的内部信号通路。整合先进的成像模式和计算工具为研究骨骼肌再生和肌肉退行性疾病(例如肌肉减少症和杜氏肌营养不良症(DMD))中的复杂生物学过程提供了新的途径。
    Composed of a diverse variety of cells, the skeletal muscle is one of the body\'s tissues with the remarkable ability to regenerate after injury. One of the key players in the regeneration process is the muscle satellite cell (MuSC), a stem cell population for skeletal muscle, as it is the source of new myofibers. Maintaining MuSC quiescence during homeostasis involves complex interactions between MuSCs and other cells in their corresponding niche in adult skeletal muscle. After the injury, MuSCs are activated to enter the cell cycle for cell proliferation and differentiate into myotubes, followed by mature myofibers to regenerate muscle. Despite decades of research, the exact mechanisms underlying MuSC maintenance and activation remain elusive. Traditional methods of analyzing MuSCs, including cell cultures, animal models, and gene expression analyses, provide some insight into MuSC biology but lack the ability to replicate the 3-dimensional (3-D) in vivo muscle environment and capture dynamic processes comprehensively. Recent advancements in imaging technology, including confocal, intra-vital, and multi-photon microscopies, provide promising avenues for dynamic MuSC morphology and behavior to be observed and characterized. This chapter aims to review 3-D and live-imaging methods that have contributed to uncovering insights into MuSC behavior, morphology changes, interactions within the muscle niche, and internal signaling pathways during the quiescence to activation (Q-A) transition. Integrating advanced imaging modalities and computational tools provides a new avenue for studying complex biological processes in skeletal muscle regeneration and muscle degenerative diseases such as sarcopenia and Duchenne muscular dystrophy (DMD).
    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
    生肌再生依赖于卫星细胞的增殖和分化。TECRL(反式-2,3-烯酰-CoA还原酶样)是仅在心肌和骨骼肌中表达的内质网蛋白。然而,其在肌生成中的作用尚不清楚。我们显示TECRL表达响应于损伤而增加。TECRL的卫星细胞特异性缺失通过激活ERK1/2信号通路增加EGR2的表达来增强肌肉修复,进而促进PAX7的表达。我们进一步表明,TECRL缺失导致组蛋白乙酰转移酶一般控制不可去抑制的5上调,从而通过乙酰化增强EGR2的转录。重要的是,我们发现AAV9介导的TECRL沉默能改善小鼠的肌肉修复。这些发现揭示了肌源性再生和肌肉修复。
    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.
    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
    木乳肌病导致快速生长的重体重肉型肉鸡的乳腺肌纤维坏死和纤维化。需要成肌卫星细胞来修复和再生受损的肌纤维。利用全基因组关联,先前已经报道了受木胸影响的候选基因。这些基因对卫星细胞增殖的影响,分化,卫星细胞合成的脂质是未知的。使用从商业Ross708肉鸡的胸大肌分离的卫星细胞和随机繁殖的鸡(RBch)系。通过沉默干扰RNA降低钙蛋白1(CNN1)和PHD和环指结构域1(PHRF1)的表达,以确定它们对卫星细胞介导的增殖的影响,分化,和脂质积累。CNN1和PHRF1影响两个系中的卫星细胞活性和脂质积累。通过敲低这两个基因的表达,Ross708和RBch系的增殖减少,当增殖开始时基因表达降低时,分化受到品系和处理相互作用的影响。在分化过程中,随着CNN1和PHRF1表达的降低,脂质积累减少。CNN1和PHRF1以前在骨骼肌中都没有报道,需要进一步的研究来确定它们对卫星细胞介导的生长和胸大肌(乳房)再生的影响。
    The Wooden Breast myopathy results in the necrosis and fibrosis of breast muscle fibers in fast-growing heavy weight meat-type broiler chickens. Myogenic satellite cells are required to repair and regenerate the damaged muscle fibers. Using Genome Wide Association, candidate genes affected with Wooden Breast have been previously reported. The effect of these genes on satellite cell proliferation, differentiation, and the synthesis of lipids by satellite cells is unknown. Satellite cells isolated from the pectoralis major muscle from commercial Ross 708 broilers and a Randombred chicken (RBch) line were used. Expression of calponin 1 (CNN1) and PHD and ring fingers domains 1 (PHRF1) were knocked down by silent interfering RNA to determine their effect on satellite cell-mediated proliferation, differentiation, and lipid accumulation. CNN1 and PHRF1 affected satellite cell activity and lipid accumulation in both lines. Proliferation was reduced in the Ross 708 and RBch lines by knocking down the expression of both genes, and differentiation was affected with a line and treatment interaction when gene expression was reduced at the beginning of proliferation. During differentiation lipid accumulation was decreased with knocking down the expression of CNN1 and PHRF1. Both CNN1 and PHRF1 have not been reported previously in skeletal muscle and further research is required to determine their effect on satellite cell-mediated growth and regeneration of the pectoralis major (breast) muscle.
    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
    肌萎缩侧索硬化症(ALS)是一种致命的神经肌肉疾病,其特征是几乎所有骨骼肌的进行性无力,而眼外肌(EOM)相对较少。尽管晚期SOD1G93A(G93A)小鼠(家族性ALS小鼠模型)的后肢和diaphragm肌表现出严重的神经支配和Pax7卫星细胞(SC)的消耗,我们发现SCs池和神经肌肉接头(NMJs)的完整性在EOMs中得以维持。来自G93A小鼠后肢和diaphragm肌的SC表现出神经相关的激活,而来自G93A小鼠EOM的SCs表现出自发(非去神经相关)激活,与野生型小鼠的SC相似。具体来说,培养的EOMSCs含有更丰富的轴突导向分子转录本,包括Cxcl12,以及在分化压力下比隔膜和后肢对应物更可持续的可再生性。在神经肌肉共培养试验中,AAV将Cxcl12递送至G93A后肢SC衍生的肌管可增强运动神经元轴突延伸和神经支配,概述EOMSC衍生的肌管的神经支配能力。补充丁酸钠(NaBu)的G93A小鼠后肢和diaphragm肌的NMJ损失较少。此外,源自G93A后肢和diaphragm肌的SC在体外NaBu治疗后显示出Cxcl12的表达升高和改善的可再生性。因此,类似于EOMSCs模式的NaBu诱导的转录组变化可能有助于在G93A小鼠中观察到的有益效应.更广泛地说,EOMSCs独特的转录组特征可能为减缓ALS中进行性神经肌肉功能衰退提供新的治疗靶点,并在临床前和临床研究中提供可能的“应答生物标志物”.
    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.
    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
    肌肉再生,代表一个基本的体内平衡过程,主要依赖于常驻卫星细胞的肌源性进展,它由多种物理和营养因素调节。这里,我们研究了肌源性分化相关因子和途径如何响应快速和慢速肌肉中的第一个限制性氨基酸赖氨酸(Lys),和他们的卫星细胞(SC),猪。30只体重相似的28日龄断奶仔猪接受三种饮食方案:对照组(d0-28:1.31%Lys,n=12),Lys缺乏组(d0-28:0.83%Lys,n=12),和Lys救援组(d0-14:0.83%Lys;d15-28:1.31%Lys,n=6)。对第15天和第29天的猪进行选择性屠宰以进行肌肉参数评估。还选择了从快速(半膜)和慢速(半腱肌)肌肉分离的卫星细胞来研究分化能力的变化。我们发现Lys缺乏通过对肌源性调节因子和Wnt/Ca2途径的独特操纵显着阻碍了快速和慢速肌肉的肌肉发育。在SC模型中,Lys缺乏抑制Wnt/Ca2+途径和肌球蛋白重链,Myogenin,和肌源性调节因子4在慢肌SCs中,但在快肌SCs中刺激它们。当获得足够的Lys时,快肌来源的SCsWnt/Ca2+途径(蛋白激酶C,钙调磷酸酶,钙/钙调蛋白依赖性蛋白激酶II,和活化T细胞核因子1)被抑制,同时刺激其对应物的Wnt/Ca2途径以进一步促进肌源性分化。Lys可能以相反的趋势通过Wnt/Ca2途径操纵猪慢肌纤维和快肌纤维的分化。
    Muscle regeneration, representing an essential homeostatic process, relies mainly on the myogenic progress of resident satellite cells, and it is modulated by multiple physical and nutritional factors. Here, we investigated how myogenic differentiation-related factors and pathways respond to the first limiting amino acid lysine (Lys) in the fast and slow muscles, and their satellite cells (SCs), of swine. Thirty 28-day-old weaned piglets with similar body weights were subjected to three diet regimens: control group (d 0-28: 1.31% Lys, n = 12), Lys-deficient group (d 0-28: 0.83% Lys, n = 12), and Lys rescue group (d 0-14: 0.83% Lys; d 15-28: 1.31% Lys, n = 6). Pigs on d 15 and 29 were selectively slaughtered for muscular parameters evaluation. Satellite cells isolated from fast (semimembranosus) and slow (semitendinosus) muscles were also selected to investigate differentiation ability variations. We found Lys deficiency significantly hindered muscle development in both fast and slow muscles via the distinct manipulation of myogenic regulatory factors and the Wnt/Ca2+ pathway. In the SC model, Lys deficiency suppressed the Wnt/Ca2+ pathways and myosin heavy chain, myogenin, and myogenic regulatory factor 4 in slow muscle SCs but stimulated them in fast muscle SCs. When sufficient Lys was attained, the fast muscle-derived SCs Wnt/Ca2+ pathway (protein kinase C, calcineurin, calcium/calmodulin-dependent protein kinase II, and nuclear factor of activated T cells 1) was repressed, while the Wnt/Ca2+ pathway of its counterpart was stimulated to further the myogenic differentiation. Lys potentially manipulates the differentiation of porcine slow and fast muscle myofibers via the Wnt/Ca2+ pathway in opposite trends.
    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
    短期热量限制(CR)的一个有趣的效果是某些干细胞群体的扩增,包括肌肉干细胞(卫星细胞),这有助于受伤后加速再生程序。这里,我们利用MetRSL274G(MetRS)转基因小鼠鉴定肝脏分泌型纤溶酶原作为短期CR期间调节卫星细胞扩增的候选物.循环纤溶酶原的敲除可防止短期CR期间的卫星细胞扩增。此外,纤溶酶原受体KT(Plg-RKT)的丢失也足以阻止CR相关的卫星细胞扩增,与纤溶酶原通过纤溶酶原受体Plg-RKT/ERK激酶促进卫星细胞增殖的直接信号传导一致。重要的是,我们能够在CALERIE试验的人类参与者中复制许多这些发现.我们的结果表明,CR增强纤溶酶原的肝脏蛋白分泌,通过Plg-RKT直接向肌肉卫星细胞发出信号,以促进CR期间的增殖和随后的肌肉弹性。
    An intriguing effect of short-term caloric restriction (CR) is the expansion of certain stem cell populations, including muscle stem cells (satellite cells), which facilitate an accelerated regenerative program after injury. Here, we utilized the MetRSL274G (MetRS) transgenic mouse to identify liver-secreted plasminogen as a candidate for regulating satellite cell expansion during short-term CR. Knockdown of circulating plasminogen prevents satellite cell expansion during short-term CR. Furthermore, loss of the plasminogen receptor KT (Plg-RKT) is also sufficient to prevent CR-related satellite cell expansion, consistent with direct signaling of plasminogen through the plasminogen receptor Plg-RKT/ERK kinase to promote proliferation of satellite cells. Importantly, we are able to replicate many of these findings in human participants from the CALERIE trial. Our results demonstrate that CR enhances liver protein secretion of plasminogen, which signals directly to the muscle satellite cell through Plg-RKT to promote proliferation and subsequent muscle resilience during CR.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号