- 无标题文档
查看论文信息

中文题名:

 转录因子Tgif1对成肌细胞增殖分化能力的影响及机理研究     

姓名:

 赵赶    

学号:

 2022105028    

保密级别:

 公开    

论文语种:

 chi    

学科代码:

 090501    

学科名称:

 农学 - 畜牧学 - 动物遗传育种与繁殖    

学生类型:

 硕士    

学位:

 农学硕士    

学校:

 南京农业大学    

院系:

 动物科技学院    

专业:

 动物遗传育种与繁殖    

研究方向:

 猪肌肉生长发育    

第一导师姓名:

 侯黎明    

第一导师单位:

 南京农业大学    

第二导师姓名:

 吴望军    

完成日期:

 2025-04-10    

答辩日期:

 2025-05-27    

外文题名:

 Impact and Mechanistic Study of Transcription Factor Tgif1 on Myoblast Proliferation and Differentiation    

中文关键词:

  ; 小鼠 ; Tgif1 ; 肌卫星细胞 ; C2C12成肌细胞 ; 肌肉生长发育    

外文关键词:

 pig ; mouse ; Tgif1 ; muscle satellite cells ; C2C12 ; muscle growth and development    

中文摘要:

肌肉重量和质量对养猪业经济效益至关重要。骨骼肌是猪肉的主要来源,鉴定骨骼肌生长发育的关键调控因子与调控机理能够为猪产肉性状的精准遗传改良提供理论依据与潜在靶点。哺乳动物骨骼肌来源于骨骼肌中胚层间充质干细胞 (Mesenchymal Stem Cells, MSCs),哺乳动物出生后,肌纤维数量不再增加,后续肌肉量的增加主要依赖肌纤维的肥大。当骨骼肌受损时,位于肌纤维肌浆膜和基底膜之间的肌卫星细胞 (Muscle Satellite Cells, MuSCs) 被激活,启动自我更新程序,在经历增殖、分化和融合后形成新的肌纤维,最终修复受损的肌肉组织。肌肉生长发育是一个多阶段的复杂过程,涉及细胞周期调控、肌肉特异性基因转录程序启动、细胞融合等多个关键生物学事件,这一复杂的调控过程是多种调控因子和信号通路相互作用的结果。TG结合因子1 (TG-interacting factor 1, Tgif1) 是三氨基酸环延伸家族的成员,其分子结构特征包含碱性螺旋环螺旋 (basic Helix-Loop-Helix, bHLH) 结构域。在鸽子与猪骨骼肌的研究中发现,Tgif1是影响骨骼肌发育和生长的候选基因。而课题组前期单细胞转录组测序分析发现,Tgif1在猪MuSCs中高表达。所以,我们推测Tgif1是一个影响肌肉生长发育的关键候选因子,但其在调控肌生成过程的具体分子机理仍有待研究。因此,本研究以猪MuSCs和小鼠稳定成肌细胞系 (C2C12) 为模型,研究Tgif1基因在肌生成过程中的作用和分子调控机理。

本研究首先通过公共数据库分析了Tgif1基因在猪不同组织和细胞中的表达模式;然后利用CRISPR/Cas9技术在猪MuSCs中沉默Tgif1基因,并利用免疫荧光 (Immunofluorescence, IF)、蛋白质印迹 (Western blot, WB)、荧光定量PCR (Real Time quantitative Polymerase Chain Reaction, RT-qPCR)、细胞计数试剂盒-8 (Cell Counting Kit-8, CCK-8) 和5’-乙炔基-2’-脱氧尿苷 (5-Ethynyl-2'-deoxyuridine, EdU) 标记细胞增殖检测法,系统评价了Tgif1基因沉默对猪MuSCs增殖与分化能力的影响;进一步利用RNA-seq技术分析了Tgif1基因沉默对猪MuSCs基因表达变化的影响,根据基因表达变化发现了一系列Tgif1基因的潜在下游靶基因。为了深入研究Tgif1基因在肌生成中的具体调控机理,我们以C2C12细胞为模型,利用多种分子生物学技术开展了分子机理研究。首先通过数据库分析和RT-qPCR检测了Tgif1基因在小鼠不同组织和细胞中的表达模式;随后,分别利用慢病毒载体系统和CRISPR/Cas9技术建立了Dox诱导过表达Tgif1基因和Tgif1基因敲除的稳定细胞系,利用IF、WB、RT-qPCR、CCK-8和EdU等方法检测了Tgif1基因对成肌细胞增殖与分化能力的影响;进一步,基于CUT&Tag和RNA-seq的联合分析,鉴定了Tgif1直接作用的下游靶基因,胆碱能受体烟碱α1亚单位 (Cholinergic receptor nicotinic alpha 1 subunit, Chrna1) 基因,利用siRNA干扰技术在成肌细胞中敲低Chrna1基因,并利用IF、WB、RT-qPCR和EdU等方法系统评价Chrna1基因对成肌细胞增殖与分化能力的影响;最后,利用WB和RT-qPCR检测了Tgif1基因在成肌细胞分化过程中的表达模式,并利用流式细胞仪分析了Tgif1基因对细胞周期的影响。本研究的主要结果如下:

1. Tgif1是猪MuSCs和小鼠成肌细胞分化的正调控因子

表达模式分析结果显示,Tgif1基因在猪和小鼠肌肉组织,猪MuSCs和小鼠成肌细胞中均高表达,提示Tgif1基因参与肌生成这一过程。在Tgif1基因沉默的猪MuSCs、Tgif1基因超表达和Tgif1基因敲除的小鼠成肌细胞系开展的细胞分化能力评价试验中发现,超表达Tgif1基因可以显著提升小鼠成肌细胞的分化能力;沉默或敲除Tgif1基因可以显著抑制猪MuSCs和小鼠成肌细胞的肌管生成。

2. CUT&Tag和RNA-seq联合分析鉴定Tgif1基因的下游靶基因

Tgif1基因沉默的猪MuSCs (sTG-KO) 与对照组 (sCrtl) 转录组分析结果显示,Chrna1、肌源性分化因子1 (Myoblast determination protein 1, Myod1)、早期生长反应基因1 (Early growth response 1, Egr1) 和双特异性磷酸酶1 (Dual specificity phosphatases 1, Dusp1) 等基因差异表达,京都基因与基因组百科全书 (Kyoto Encyclopedia of Genes and Genomes, KEGG) 和基因本体分析 (Gene Ontology Analysis, GO) 分析表明差异基因主要富集在cAMP信号通路、Pi3k-Akt信号通路和钙信号通路等肌肉分化相关过程中。

Tgif1基因敲除的小鼠成肌细胞 (mTG-KO) 与对照组 (mCrtl) 在分化阶段和增殖阶段的转录组分析结果显示,差异表达基因 (Differentially Expressed Genes, DEGs) 主要富集在钙信号通路、Rap1信号通路、MAPK信号通路、Pi3k-Akt信号通路等肌肉分化相关过程中。Tgif1抗体在小鼠成肌细胞中的CUT&Tag富集结果显示,Tgif1主要结合下游靶基因的转录起始位点 (Transcription Start Site, TSS) 区域,且靶基因主要富集在Wnt信号通路、mTor信号通路、AMPK信号通路、MAPK信号通路、Notch信号通路中。RNA-seq和CUT&Tag联合分析筛选到细胞周期蛋白依赖性激酶1 (Cyclin-dependent kinase 1,Cdk1)、信号转导和转录激活因子3 (Signal Transducer and Activator of Transcription 3, Stat3)、Dusp1、Egr1和Chrna1等潜在靶基因。其中,Chrna1基因是在增殖和分化阶段都被Tgif1直接结合其TSS区的靶基因,并且也是猪MuSCs中Tgif1基因沉默后的差异表达基因。而先前有研究报道,Chrna1基因参与肌肉分化的过程。因此,本研究将Chrna1基因作为Tgif1基因的下游关键靶基因进行下一步验证。

3. Tgif1-Chrna1基因轴促进小鼠成肌细胞分化的调控机理

在小鼠成肌细胞中的试验结果显示,Tgif1基因超表达后,ChIP富集到更多Chrna1基因TSS位点附近的序列。启动子活性分析试验表明,Chrna1基因启动子驱动的荧光素酶活性更高。细胞分化能力评价试验结果显示,Chrna1基因敲低显著抑制了小鼠成肌细胞的分化能力,表明了Tgif1可靶定Chrna1基因的TSS区域进而促进肌管生成。增殖能力评价试验结果显示,Tgif1和Chrna1基因对小鼠成肌细胞的增殖能力并没有显著影响。另外,基因表达模式检测发现,Tgif1和Chrna1基因在小鼠成肌细胞分化的早期阶段,尤其是肌管形成时具有相对高丰度的表达。细胞周期试验发现,在分化24 h时,超表达Tgif1基因可增强G1期细胞的积累,而敲除Tgif1基因会显著增加S期细胞,从而影响细胞周期阻滞。试验表明,敲除Tgif1基因可造成小鼠成肌细胞无法进入分化程序,从而降低成肌细胞分化能力。

综上所述,Tgif1在骨骼肌中高度表达,是一个影响肌细胞分化的关键调节因子。Tgif1基因可增强G1期细胞的积累,促进细胞退出细胞周期,进而调控成肌细胞分化启动,最终促进成肌细胞分化。Chrna1基因是Tgif1基因的关键下游靶标,本研究阐明了Tgif1靶向Chrna1基因TSS区域促进肌细胞分化的作用机理。本研究不仅加深了人们对肌肉生长发育的理解,而且为肌肉相关疾病治疗提供了潜在的靶点。

外文摘要:

Muscle weight and quality are critical to the economic efficiency of pig farming. Skeletal muscle is the main source of pork, and the identification of key regulators and mechanisms of skeletal muscle growth and development can provide a theoretical basis and potential targets for precise genetic improvement of pork production traits. Mammalian skeletal muscle is derived from Mesenchymal Stem Cells (MSCs). After birth, the number of myofibers does not increase, and the subsequent increase in muscle mass depends on the hypertrophy of myofibers. When skeletal muscle is damaged, Muscle Satellite Cells (MuSCs) located between the plasma membrane and the basement membrane of myofibers are activated, initiating a self-renewal program that undergoes proliferation, differentiation, and fusion to form new muscle fibers, and ultimately repairing the damaged muscle tissue. Muscle growth and development is a multi-stage and complex process involving multiple key biological events such as cell cycle regulation, initiation of muscle-specific gene transcription programs, and cell fusion. This complex regulatory process is the result of the interaction of multiple regulatory factors and signaling pathways. TG-interacting factor 1 (Tgif1) is a member of a family of three amino acid loop extensions whose molecular structure is characterized by a basic Helix-Loop-Helix (bHLH) domain. Previous studies in pigeon and pig skeletal muscle revealed that Tgif1 is a candidate gene that affects skeletal muscle development and growth. And the group's previous single-cell transcriptome sequencing analysis found that Tgif1 was highly expressed in pig MuSCs. Therefore, we hypothesized that Tgif1 is a key candidate factor affecting muscle growth and development, but its specific molecular mechanism in myogenesis remains to be investigated. Therefore, in this study, we investigated the role and potential molecular regulatory mechanisms of Tgif1 gene in the myogenesis using pig MuSCs and mouse stable myoblast cell line (C2C12) as models.

In this study, we first analyzed the expression pattern of Tgif1 gene in different tissues and cells of pigs through public databases; then we silenced the Tgif1 gene in pig MuSCs using CRISPR/Cas9 technology, and systematically evaluated the Tgif1 gene silencing on the proliferation and differentiation ability of pig MuSCs using Immunofluorescence (IF), Western blot (WB), Real Time quantitative Polymerase Chain Reaction (RT-qPCR), Cell Counting Kit-8 (CCK-8) and 5'-ethynyl-2'-deoxyuridine (EdU)-labeled cell proliferation assay. We further analyzed the effects of Tgif1 gene silencing on gene expression changes in pig MuSCs using RNA-seq technology, and identified a series of potential downstream target genes of Tgif1 gene based on gene expression changes. In order to deeply investigate the specific regulatory mechanism of Tgif1 gene in myogenesis, we carried out molecular mechanistic studies using various molecular biology techniques with mouse stable myogenic cell line (C2C12) as a model. Firstly, we detected the expression pattern of Tgif1 gene in different tissues and cells of mice by database analysis and RT-qPCR; subsequently, Dox-induced overexpression of Tgif1 gene and Tgif1 gene knockout stable cell lines were established by using the lentiviral vector system and CRISPR/Cas9 technology, respectively. The effects of Tgif1 gene on the proliferation and differentiation ability of myoblasts were detected using IF, WB, RT-qPCR, CCK-8, and EdU. Further, based on the combined analysis of CUT&Tag and RNA-seq, the downstream target gene of Tgif1 direct action, cholinergic receptor nicotinic alpha 1 subunit (Chrna1), was identified. Knockdown of Chrna1 gene in C2C12 cells using siRNA interference and evaluation of the effect of Chrna1 gene on proliferation and differentiation ability using IF, WB, RT-qPCR, and EdU. Finally, the expression pattern of Tgif1 gene during differentiation of myoblasts was detected by WB and RT-qPCR, and the effect of Tgif1 gene on cell cycle was analyzed by flow cytometry. The main results of this study are as follows:

1. Tgif1 is a positive regulator of differentiation of pig MuSCs and mouse myoblasts

Expression pattern analysis showed that Tgif1 gene was highly expressed in both pig and mouse muscle tissues, pig MuSCs and mouse myoblasts, suggesting that Tgif1 gene is involved in the process of myogenesis. In the cell differentiation evaluation of Tgif1-silenced pig MuSCs, Tgif1 gene overexpression and Tgif1 gene knockout mouse myoblast cell lines, it was found that overexpression of Tgif1 gene significantly enhanced the differentiation ability of mouse myoblasts, and that silencing or knocking out of Tgif1 gene significantly inhibited myotubular myogenesis in both pig MuSCs and mouse myoblasts.

2. Combined CUT&Tag and RNA-seq analysis to identify downstream target genes of Tgif1 gene

Transcriptome analysis of Tgif1-silenced pig MuSCs (sTG-KO) and controls (sCrtl) showed that Chrna1, myogenic differentiation factor 1 (Myod1), early growth response 1 (Egr1), and dual specificity phosphatases 1 (Dusp1) were differentially expressed. The Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology Analysis (GO) analyses showed that the differentially expressed genes were mainly enriched in the cAMP signaling pathway, Pi3k-Akt signaling pathway, and calcium signaling pathway, which are related to muscle differentiation.

Transcriptome analysis of Tgif1-knockout C2C12 cells (mTG-KO) and control cells (mCrtl) at differentiation and proliferation stages showed that Differentially Expressed Genes (DEGs) were mainly enriched in the calcium signaling pathway, the Rap1 signaling pathway, MAPK signaling pathway, and the Pi3k−Akt signaling pathway. The CUT&Tag enrichment of Tgif1 antibody in mouse myoblasts showed that Tgif1 mainly binds to the Transcription Start Site (TSS) region of the downstream target genes, and the target genes were mainly enriched in Wnt signaling, mTor signaling, AMPK signaling, MAPK signaling, and Notch signaling pathways. RNA-seq and CUT&Tag combined analysis screened for Cyclin-dependent kinase 1 (Cdk1), Dusp1, Egr1, Signal Transducer and Activator of Transcription 3 (Stat3) and Chrna1 are potential target genes. Among them, the Chrna1 geneis a target gene that is directly bound to its TSS region by Tgif1 at both the proliferation and differentiation stages, and is also a DEG after silencing Tgif1 gene in pig MuSCs. Therefore, the Chrna1 gene was next validated as a key target gene downstream of Tgif1 in this study.

3. Mechanisms by which the Tgif1-Chrna1 gene axis promotes differentiation of mouse myoblasts

Validation assays in mouse myoblasts showed that ChIP was enriched to more sequences near the TSS site of the Chrna1 gene after Tgif1 gene overexpression. Promoter activity analysis assay showed higher luciferase activity driven by Chrna1 gene promoter. The results of the cell differentiation ability evaluation assay showed that Chrna1 knockdown significantly inhibited the differentiation ability of mouse myoblasts, suggesting that Tgif1 can target the TSS region of the Chrna1 gene and thus promote myotube generation. The results of proliferative capacity evaluation assay showed that Tgif1 and Chrna1 genes did not significantly affect the proliferative capacity of mouse myoblasts. In addition, gene expression patterns revealed that Tgif1 and Chrna1 genes were expressed in high relative abundance at the early stage of myoblast differentiation, especially during myotube formation. Cell cycle assay revealed that overexpression of Tgif1 gene enhanced the accumulation of G1-phase cells at 24 h of differentiation, whereas knockout of Tgif1 gene significantly increased the S-phase cells, which affected the cell cycle block and might result in the inability of mouse myoblasts to enter the differentiation program, thus reducing the differentiation capacity of myoblasts.

In summary, Tgif1 is highly expressed in skeletal muscle and is a key regulator affecting myoblast differentiation. Tgif1 gene enhances the accumulation of G1-phase cells and promotes cell exit from the cell cycle, which in turn regulates the initiation of myoblast differentiation and ultimately promotes myoblast differentiation. Chrna1 gene is a key downstream target of Tgif1 gene, and this study elucidated the reciprocal mechanism by which Tgif1 targets the TSS region of Chrna1 gene to promote myoblast differentiation. This study not only deepens the understanding of muscle growth and development, but also provides potential targets for the treatment of muscle-related diseases.

参考文献:

[1] A., J., Whitmarshr., et al. 1996. Transcription factor AP-1 regulation by mitogen-activated protein kinase signal transduction pathways. Journal of Molecular Medicine [J].

[2] Aikawa K, Nishikimi N, Sakurai T, et al. 2001. SA channel mediates superoxide production in HUVECs. Life Sci [J], 69: 1717-1724.

[3] Akpan I, Goncalves M D, Dhir R, et al. 2009. The effects of a soluble activin type IIB receptor on obesity and insulin sensitivity. Int J Obes [J], 33: 1265-1273.

[4] Alameddine H S, Dehaupas M, Fardeau M 1989. Regeneration of skeletal muscle fibers from autologous satellite cells multiplied in vitro. An experimental model for testing cultured cell myogenicity. Muscle Nerve [J], 12: 544-555.

[5] álamo D D, Rouault H, Schweisguth F O 2011. Mechanism and significance of cis-inhibition in Notch signalling. Current Biology Cb [J], 21: R40-R47.

[6] Alan K, Milton C B, Abigail P, et al. 2010. A soluble activin receptor type IIb prevents the effects of androgen deprivation on body composition and bone health. Endocrinology [J]: 4289-4300.

[7] Alter J, Rozentzweig D, Bengal E 2008. Inhibition of myoblast differentiation by tumor necrosis factor alpha is mediated by c-Jun N-terminal kinase 1 and leukemia inhibitory factor. J Biol Chem [J], 283: 23224-23234.

[8] Arnold S J, Robertson E J 2009. Making a commitment: cell lineage allocation and axis patterning in the early mouse embryo. Nat Rev Mol Cell Biol [J], 10: 91-103.

[9] Atilla, Bier, Serena, et al. 2017. ChIP-Seq analysis identifies p27(Kip1)-target genes involved in cell adhesion and cell signalling in mouse embryonic fibroblasts. PLoS ONE [J], 12.

[10] Baraldo M, Geremia A, Pirazzini M, et al. 2020. Skeletal muscle mTORC1 regulates neuromuscular junction stability. J Cachexia Sarcopenia Muscle [J], 11: 208-225.

[11] Bartholin L, Melhuish T A, Powers S E, et al. 2008. Maternal Tgif is required for vascularization of the embryonic placenta. Developmental Biology [J], 319: 285-297.

[12] Bartkova J, Lukas J, Strauss M, et al. 1998. Cyclin D3: requirement for G1/S transition and high abundance in quiescent tissues suggest a dual role in proliferation and differentiation. Oncogene [J], 17: 1027-1037.

[13] Bean C, Salamon M, Raffaello A, et al. 2005. The Ankrd2, Cdkn1c and Calcyclin Genes are Under the Control of MyoD During Myogenic Differentiation. Journal of Molecular Biology [J], 349: 349-366.

[14] Benavides Damm T, Egli M 2014. Calcium's Role in Mechanotransduction during Muscle Development. Cellular Physiology & Biochemistry International Journal of Experimental Cellular Physiology Biochemistry & Pharmacology [J], 33: 249-272.

[15] Bentzinger C F, Wang Y X, Rudnicki M A 2012. Building muscle: molecular regulation of myogenesis. Cold Spring Harb Perspect Biol [J], 4.

[16] Berridge M J, Bootman M D, Roderick H L 2003. Calcium signalling: dynamics, homeostasis and remodelling. Nat Rev Mol Cell Biol [J], 4: 517-529.

[17] Birchmeier C, Brohmann H 2000. Genes that control the development of migrating muscle precursor cells. Curr Opin Cell Biol [J], 12: 725-730.

[18] Bjornson C R R, Cheung T H, Liu L, et al. 2012. Notch Signaling Is Necessary to Maintain Quiescence in Adult Muscle Stem Cells. Stem Cells [J].

[19] Blokzijl A, Dahlqvist C, Reissmann E, et al. 2003. Cross-talk between the Notch and TGF-β signaling pathways mediated by interaction of the Notch intracellular domain with Smad3. Journal of Cell Biology [J], 163: 723-728.

[20] Blumer S, Fang L, Chen W C, et al. 2021. IPF-Fibroblast Erk1/2 Activity Is Independent from microRNA Cluster 17-92 but Can Be Inhibited by Treprostinil through DUSP1. Cells [J], 10: 2836-.

[21] Bober E, Franz T, Arnold H H, et al. 1994. Pax-3 is required for the development of limb muscles: a possible role for the migration of dermomyotomal muscle progenitor cells. Development [J], 120: 603-612.

[22] Bogdanovich S, Krag T O B, Barton E R, et al. 2002. Functional improvement of dystrophic muscle by myostatin blockade. Nature [J], 420: 418-421.

[23] Bolamperti S, Saito H, Heerdmann S, et al. 2024. Tgif1-deficiency impairs cytoskeletal architecture in osteoblasts by activating PAK3 signaling. eLife [J].

[24] Bopp D, Burri M, Baumgartner S, et al. 1986. Conservation of a large protein domain in the segmentation gene paired and in functionally related genes of Drosophila. Cell [J], 47: 1033-1040.

[25] Borello, U. 2006. The Wnt/beta-catenin pathway regulates Gli-mediated Myf5 expression during somitogenesis. Development [J], 133: 3723.

[26] Borriello A, Caldarelli I, Bencivenga D, et al. 2011. p57(Kip2) and cancer: time for a critical appraisal. Mol Cancer Res [J], 9: 1269-1284.

[27] Borycki A G, Brunk B, Tajbakhsh S, et al. 1999a. Sonic hedgehog controls epaxial muscle determination through Myf5 activation. Development [J], 126: 4053-4063.

[28] Borycki A G, Li J, Jin F, et al. 1999b. Pax3 functions in cell survival and in pax7 regulation. Development [J], 126: 1665-1674.

[29] Brack A S, Conboy I M, Conboy M J, et al. 2008. A Temporal Switch from Notch to Wnt Signaling in Muscle Stem Cells Is Necessary for Normal Adult Myogenesis - ScienceDirect. Cell Stem Cell [J], 2: 50-59.

[30] Braun T, Arnold H H 1995. Inactivation of Myf-6 and Myf-5 genes in mice leads to alterations in skeletal muscle development. Embo Journal [J], 14: 1176-1186.

[31] Brendan, Egan, And, et al. 2013. Exercise Metabolism and the Molecular Regulation of Skeletal Muscle Adaptation. Cell Metabolism [J].

[32] Brennan T J, Edmondson D G, Li L, et al. 1991. Transforming growth factor beta represses the actions of myogenin through a mechanism independent of DNA binding. Proc Natl Acad Sci U S A [J], 88: 3822-3826.

[33] Brett J O, Arjona M, Ikeda M, et al. 2020. Exercise rejuvenates quiescent skeletal muscle stem cells in old mice through restoration of Cyclin D1. Nat Metab [J], 2: 307-317.

[34] Brunelli S, Relaix F, Baesso S, et al. 2007. Beta catenin-independent activation of MyoD in presomitic mesoderm requires PKC and depends on Pax3 transcriptional activity. Developmental Biology [J], 304: 604-614.

[35] Buas M F, Kabak S, Kadesch T 2010. The Notch Effector Hey1 Associates with Myogenic Target Genes to Repress Myogenesis. Journal of Biological Chemistry [J], 285.

[36] Burglin T R 1997. Analysis of TALE superclass homeobox genes (MEIS, PBC, KNOX, Iroquois, TGIF) reveals a novel domain conserved between plants and animals. Nucleic Acids Res [J], 25: 4173-4180.

[37] Cai S, Hu B, Wang X, et al. 2023. Integrative single-cell RNA-seq and ATAC-seq analysis of myogenic differentiation in pig. BMC biology [J], 21: 19.

[38] Carling D 2017. AMPK signalling in health and disease. Curr Opin Cell Biol [J], 45: 31-37.

[39] Carlson M E, Hsu M, Conboy I M 2008. Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature [J], 454: 528.

[40] Chakkalakal J V, Christensen J, Xiang W, et al. 2014. Early forming label-retaining muscle stem cells require p27kip1 for maintenance of the primitive state. Development [J], 141: 1649-1659.

[41] Chal J, Pourquie O 2017. Making muscle: skeletal myogenesis in vivo and in vitro. Development [J], 144: 2104-2122.

[42] Chang Y H, Tseng Y H, Wang J M, et al. 2024. TG-interacting factor 1 regulates mitotic clonal expansion during adipocyte differentiation. BBA - Molecular and Cell Biology of Lipids [J], 1869.

[43] Chen, Alice, E., et al. 2005. Protein kinase A signalling via CREB controls myogenesis induced by Wnt proteins. Nature [J].

[44] Chen, Jian-Fu, Tao, et al. 2010. microRNA-1 and microRNA-206 regulate skeletal muscle satellite cell proliferation and differentiation by repressing Pax7. Journal of Cell Biology [J].

[45] Cheung T H, Rando T A 2013. Molecular regulation of stem cell quiescence. Nature Reviews Molecular Cell Biology [J].

[46] Chinzei N, Hayashi S, Ueha T, et al. 2015. P21 Deficiency Delays Regeneration of Skeletal Muscular Tissue. PLoS ONE [J], 10.

[47] Choi Y J, Li X, Hydbring P, et al. 2012. The requirement for cyclin D function in tumor maintenance. Cancer Cell [J], 22: 438-451.

[48] Christ B, Ordahl C P 1995. Early stages of chick somite development. Anat Embryol (Berl) [J], 191: 381-396.

[49] Chu C Y, Lim R W 2000. Involvement of p27~(kip1) and cyclin D3 in the regulation of cdk2 activity during skeletal muscle differentiation. Biochimica Et Biophysica Acta [J], 1497: 175-185.

[50] Cobrinik, David 2005. Pocket proteins and cell cycle control. Oncogene [J], 24: 2796-2809.

[51] Collins C A, Olsen I, Zammit P S, et al. 2005. Stem Cell Function, Self-Renewal, and Behavioral Heterogeneity of Cells from the Adult Muscle Satellite Cell Niche. CELL -CAMBRIDGE MA- [J].

[52] Corton J M, Gillespie J G, Hawley S A, et al. 1995. 5-aminoimidazole-4-carboxamide ribonucleoside. A specific method for activating AMP-activated protein kinase in intact cells? Eur J Biochem [J], 229: 558-565.

[53] Croce J C, Mcclay D R 2008. Evolution of the Wnt Pathways. methods in molecular biology [J].

[54] Cuenda, A. 1999. Stress-activated Protein Kinase-2/p38 and a Rapamycin-sensitive Pathway Are Required for C2C12 Myogenesis. Journal of Biological Chemistry [J], 274: 4341-4346.

[55] Curtis D H, Zalin R J 1981. Regulation of muscle differentiation: stimulation of myoblast fusion in vitro by catecholamines. Science [J], 214: 1355-1357.

[56] De A 2011. Wnt/Ca2+ signaling pathway: a brief overview. Acta Biochimica Et Biophysica Sinica [J], 43: 745.

[57] De Luca G, Ferretti R, Bruschi M, et al. 2013. Cyclin D3 critically regulates the balance between self-renewal and differentiation in skeletal muscle stem cells. Stem Cells [J], 31: 2478-2491.

[58] Delaney K, Kasprzycka P, Ciemerych M A, et al. 2017. The role of TGF‐β1 during skeletal muscle regeneration. Cell Biology International [J].

[59] Denetclaw W F, Jr., Christ B, Ordahl C P 1997. Location and growth of epaxial myotome precursor cells. Development [J], 124: 1601-1610.

[60] Deng C 1995. Mice lacking p21CIP1/WAF1 undergo normal development, but are defective in G1 checkpoint control. Cell [J], 82: 675-684.

[61] Desjardins C A, Naya F J 2016. The Function of the MEF2 Family of Transcription Factors in Cardiac Development, Cardiogenomics, and Direct Reprogramming. Journal of Cardiovascular Development and Disease [J], 3.

[62] Ding H, Lin Y, Zhang T, et al. 2021. Transcriptome Analysis of Differentially Expressed mRNA Related to Pigeon Muscle Development. Animals : an open access journal from MDPI [J], 11: 2311.

[63] Ding X, He Z, Zhou K, et al. 2010. Essential role of TRPC6 channels in G2/M phase transition and development of human glioma. J Natl Cancer Inst [J], 102: 1052-1068.

[64] Dong Y, Zhu C, Liu X, et al. 2024. [Effect of CircCCND1 on the Malignant Biological Behaviors of H446 Lung Cancer Cells by Regulating the MiR-340-5p/TGIF1 Axis]. Zhongguo Fei Ai Za Zhi [J], 27: 161-169.

[65] Dumont N A, Bentzinger C F, Sincennes M C, et al. 2015. Satellite Cells and Skeletal Muscle Regeneration. Compr Physiol [J], 5: 1027-1059.

[66] Dumont N A, Rudnicki M A 2017. Characterizing Satellite Cells and Myogenic Progenitors During Skeletal Muscle Regeneration.

[67] Egerman, Marc A, Glass, et al. 2019. Signaling pathways controlling skeletal muscle mass.

[68] Epstein J A, Shapiro D N, Cheng J, et al. 1996. Pax3 modulates expression of the c-Met receptor during limb muscle development. Proc Natl Acad Sci U S A [J], 93: 4213-4218.

[69] Fantl V, Stamp G, Andrews A, et al. 1995. Mice lacking cyclin D1 are small and show defects in eye and mammary gland development. Genes Dev [J], 9: 2364-2372.

[70] Ferey J L, Brault J J, Smith C A, et al. 2014. Constitutive activation of CaMKKalpha signaling is sufficient but not necessary for mTORC1 activation and growth in mouse skeletal muscle. Am J Physiol Endocrinol Metab [J], 307: E686-694.

[71] Florini J R, Ewton D Z, Coolican S A 1996. Growth Hormone and the Insulin-Like Growth Factor System in Myogenesis*. Endocrine Reviews [J]: 5.

[72] Fujii N, Hayashi T, Hirshman M F, et al. 2000. Exercise induces isoform-specific increase in 5'AMP-activated protein kinase activity in human skeletal muscle. Biochem Biophys Res Commun [J], 273: 1150-1155.

[73] Fukada S I, Yamaguchi M, Kokubo H, et al. 2011. Hesr1 and Hesr3 are essential to generate undifferentiated quiescent satellite cells and to maintain satellite cell numbers. Development [J], 138: 4609.

[74] Galceran, J. 2004. LEF1-mediated regulation of Delta-like1 links Wnt and Notch signaling in somitogenesis. Genes & Development [J], 18: 2718.

[75] Galli L M, Willert K, Nusse R, et al. 2004. A proliferative role for Wnt-3a in chick somites. Developmental Biology [J], 269: 489-504.

[76] Ge X, Mcfarlane C, Vajjala A, et al. 2011. Smad3 signaling is required for satellite cell function and myogenic differentiation of myoblasts. Cell Research [J], 21: 1591-1604.

[77] Gioftsidi S, Relaix F, Mourikis P 2022. The Notch signaling network in muscle stem cells during development, homeostasis, and disease. Skeletal muscle [J], 12: 9.

[78] Girardi F, Grand F L 2018. Wnt Signaling in Skeletal Muscle Development and Regeneration. Progress in molecular biology and translational ence [J].

[79] Girardi F, Taleb A, Ebrahimi M, et al. 2021. TGFβ signaling curbs cell fusion and muscle regeneration. Nature Communications [J], 12.

[80] Girgenrath S, Song K, Whittemore L A 2005. Loss of myostatin expression alters fiber-type distribution and expression of myosin heavy chain isoforms in slow- and fast-type skeletal muscle. Muscle & Nerve [J], 31: 34-40.

[81] Glass, David J 2010. Signaling pathways perturbing muscle mass. Curr Opin Clin Nutr Metab Care [J], 13: 225-229.

[82] Gopinath S D, Webb A E, Brunet A, et al. 2014. FOXO3 Promotes Quiescence in Adult Muscle Stem Cells during the Process of Self-Renewal. Stem Cell Reports [J], 2: 414-426.

[83] Goulding M D, Lumsden A, Paquette A J 1994. Regulation of Pax-3 expression in the dermomyotome and its role in muscle development. Development [J], 120: 957.

[84] Grand F L, Jones A E, Seale V, et al. 2009. Wnt7a Activates the Planar Cell Polarity Pathway to Drive the Symmetric Expansion of Satellite Stem Cells. Cell Stem Cell [J], 4: 535-547.

[85] Griffin C A, Kafadar K A, Pavlath G K 2009. MOR23 Promotes Muscle Regeneration and Regulates Cell Adhesion and Migration. Developmental Cell [J], 17: 649-661.

[86] Grifone R, Demignon J, Houbron C, et al. 2005. Six1 and Six4 homeoproteins are required for Pax3 and Mrf expression during myogenesis in the mouse embryo. Development [J], 132: 2235-2249.

[87] Gros J M, Scaal M, Marcelle C 2004. A two-step mechanism for myotome formation in chick. Developmental Cell [J], 6: 875-882.

[88] Guo K, Wang J, Andrés V, et al. 1995. MyoD-induced expression of p21 inhibits cyclin-dependent kinase activity upon myocyte terminal differentiation. Molecular & Cellular Biology [J].

[89] Gurung R, Parnaik V K 2011. Cyclin D3 promotes myogenic differentiation and Pax7 transcription. Journal of Cellular Biochemistry [J], 113: 209-219.

[90] Halevy O, Novitch B, Spicer D, et al. 1995. Correlation of terminal cell cycle arrest of skeletal muscle with induction of p21 by MyoD. Science [J], 267: 1018-1021.

[91] Ham D J, Borsch A, Lin S, et al. 2020. The neuromuscular junction is a focal point of mTORC1 signaling in sarcopenia. Nat Commun [J], 11: 4510.

[92] Hamrick M W, Pennington C, Webb C N, et al. 2006. Resistance to body fat gain in 'double-muscled' mice fed a high-fat diet. International Journal of Obesity [J], 30: 868.

[93] Hardie D G, Ross F A, Hawley S A 2012. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol [J], 13: 251-262.

[94] Hawke T J, Meeson A P, Jiang N, et al. 2003. p21 is essential for normal myogenic progenitor cell function in regenerating skeletal muscle. American Journal of Physiology Cell Physiology [J], 285: C1019.

[95] He F, Wu Z, Wang Y, et al. 2022. Downregulation of tripartite motif protein 11 attenuates cardiomyocyte apoptosis after ischemia/reperfusion injury via DUSP1-JNK1/2. Cell Biol Int [J], 46: 148-157.

[96] He M, Li Y, Tang Q, et al. 2018. Genome-Wide Chromatin Structure Changes During Adipogenesis and Myogenesis. International journal of biological sciences [J], 14: 1571-1585.

[97] Hindi S M, Sato S, Xiong G, et al. 2018. TAK1 regulates skeletal muscle mass and mitochondrial function. JCI Insight [J], 3.

[98] Hirsinger E, Duprez D, Jouve C, et al. 1997. Noggin acts downstream of Wnt and Sonic Hedgehog to antagonize BMP4 in avian somite patterning. Development [J], 124: 4605-4614.

[99] Hirsinger E, Malapert P, Dubrulle J, et al. 2001. Notch signalling acts in postmitotic avian myogenic cells to control MyoD activation. Development [J], 128: 107-116.

[100] Hlaing M, Shen X, Dazin P, et al. 2002. The Hypertrophic Response in C2C12 Myoblasts Recruits the G1 Cell Cycle Machinery. Journal of Biological Chemistry [J], 277: 23794.

[101] Holland J D, Klaus A, Garratt A N, et al. 2013. Wnt signaling in stem and cancer stem cells. Curr Opin Cell Biol [J], 25: 254-264.

[102] Hoogaars W M H, Jaspers R T 2018. Past, Present, and Future Perspective of Targeting Myostatin and Related Signaling Pathways to Counteract Muscle Atrophy.

[103] Horie T, Ono K, Kinoshita M, et al. 2008. TG-interacting factor is required for the differentiation of preadipocytes. Journal of Lipid Research [J], 49: 1224.

[104] Hou L, Wang Y, Liu Y, et al. 2019. Paf1C regulates RNA polymerase II progression by modulating elongation rate. Proc Natl Acad Sci U S A [J], 116: 14583-14592.

[105] Hughes A, Kleine-Albers J, Helfrich M H, et al. 2012. A Class III Semaphorin (Sema3e) Inhibits Mouse Osteoblast Migration and Decreases Osteoclast Formation In Vitro. Calcified Tissue International [J], 90: 151-162.

[106] Hulin J A, Nguyen T D T, Cui S, et al. 2016. Barx2 and Pax7 Regulate Axin2 Expression in Myoblasts by Interaction with β-Catenin and Chromatin Remodelling. Stem Cells [J], 34.

[107] Inoh H, Ishiguro N, Sawazaki S, et al. 2002. Uni-axial cyclic stretch induces the activation of transcription factor nuclear factor kappaB in human fibroblast cells. FASEB J [J], 16: 405-407.

[108] Inoki K, Guan K L 2006. Complexity of the TOR signaling network. Trends in Cell Biology [J], 16: 206-212.

[109] Inoki K, Li Y, Zhu T, et al. 2002. TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nature Cell Biology [J], 4: 648-657.

[110] Inoki K, Ouyang H, Zhu T, et al. 2006. TSC2 Integrates Wnt and Energy Signals via a Coordinated Phosphorylation by AMPK and GSK3 to Regulate Cell Growth - ScienceDirect.

[111] Jensen P B, Pedersen L, Krishna S, et al. 2010. A Wnt oscillator model for somitogenesis. Biophys J [J], 98: 943-950.

[112] Jensen T E, Rose A J, Jorgensen S B, et al. 2007. Possible CaMKK-dependent regulation of AMPK phosphorylation and glucose uptake at the onset of mild tetanic skeletal muscle contraction. Am J Physiol Endocrinol Metab [J], 292: E1308-1317.

[113] Jessica S, Eusebio P, Pura M O-C 2016. Regulation of Muscle Stem Cell Functions: A Focus on the p38 MAPK Signaling Pathway. Frontiers in Cell and Developmental Biology [J], 4.

[114] Joulia D, Bernardi H, Garandel V, et al. 2003. Mechanisms involved in the inhibition of myoblast proliferation and differentiation by myostatin. Experimental Cell Research [J], 286: 263-275.

[115] Ju Q, Jiang M, Huang W, et al. 2022. CtBP2 interacts with TGIF to promote the progression of esophageal squamous cell cancer through the Wnt/beta‑catenin pathway. Oncol Rep [J], 47.

[116] Kahl C R, Means A R 2003. Regulation of cell cycle progression by calcium/calmodulin-dependent pathways. Endocr Rev [J], 24: 719-736.

[117] Kassar-Duchossoy L, Gayraud-Morel B, Gomes D, et al. 2004. Mrf4 determines skeletal muscle identity in Myf5:Myod double-mutant mice. Nature [J], 431: 466-471.

[118] Kawakami K, Sato S, Ozaki H, et al. 2000. Six family genes-structure and function as transcription factors and their roles in development. Bioessays [J], 22: 616-626.

[119] Keren A, Tamir Y, Bengal E 2006. The p38 MAPK signaling pathway: A major regulator of skeletal muscle development. Molecular and Cellular Endocrinology [J], 252: 224-230.

[120] Khilji S, Hamed M, Chen J, et al. 2020. Dissecting myogenin-mediated retinoid X receptor signaling in myogenic differentiation. Communications Biology [J], 3: 315.

[121] Kitamoto T, Hanaoka K 2011. Notch3 Null Mutation in Mice Causes Muscle Hyperplasia by Repetitive Muscle Regeneration. Stem Cells [J], 28: 2205-2216.

[122] Kollias H D, Mcdermott J C 2008. Transforming growth factor-beta and myostatin signaling in skeletal muscle. Journal of applied physiology [J]: 104.

[123] Kong L, Yu Y, Guan H, et al. 2021. TGIF1 plays a carcinogenic role in esophageal squamous cell carcinoma through the Wnt/beta‑catenin and Akt/mTOR signaling pathways. Int J Mol Med [J], 47.

[124] Kuroda K, Tani S, Tamura K, et al. 1999. Delta-induced Notch Signaling Mediated by RBP-J Inhibits MyoD Expression and Myogenesis. Journal of Biological Chemistry [J], 274: 7238.

[125] Labbe E, Lock L, Letamendia A, et al. 2007. Transcriptional cooperation between the transforming growth factor-beta and Wnt pathways in mammary and intestinal tumorigenesis. Cancer Research [J], 67: 75-84.

[126] Lahmann I, Brohl D, Zyrianova T, et al. 2019. Oscillations of MyoD and Hes1 proteins regulate the maintenance of activated muscle stem cells. Genes Dev [J], 33: 524-535.

[127] Le May M, Mach H, Lacroix N, et al. 2011. Contribution of Retinoid X Receptor Signaling to the Specification of Skeletal Muscle Lineage. Journal of Biological Chemistry [J], 286: 26806-26812.

[128] Le Peuch C J, Ferraz C, Walsh M P, et al. 1979. Calcium and cyclic nucleotide dependent regulatory mechanisms during development of chick embryo skeletal muscle. Biochemistry [J], 18: 5267-5273.

[129] Lebrasseur N K, Schelhorn T M, Bernardo B L, et al. 2009. Myostatin inhibition enhances the effects of exercise on performance and metabolic outcomes in aged mice. J Gerontol A Biol Med [J]: 940-948.

[130] Lepper C, Fan C M 2010. Inducible lineage tracing of Pax7-descendant cells reveals embryonic origin of adult satellite cells. Genesis [J], 48: 424-436.

[131] Li J G, Cheng X, Huang Y X, et al. 2021. Wnt7a promotes muscle regeneration in branchiomeric orbicularis oris muscle. International journal of clinical and experimental pathology [J], 14: 693-704.

[132] Li L, Heller-Harrison R, Czech M, et al. 1992. Cyclic AMP-dependent protein kinase inhibits the activity of myogenic helix-loop-helix proteins. Molecular and Cellular Biology [J], 12: 4478-4485.

[133] Lima J E D, Blavet C, Bonnin M A, et al. 2022. TMEM8C-mediated fusion is regionalized and regulated by NOTCH signalling during foetal myogenesis. Development [J], 149: -.

[134] Lipina C, Kendall H, Mcpherron A C, et al. 2010. Mechanisms involved in the enhancement of mammalian target of rapamycin signalling and hypertrophy in skeletal muscle of myostatin-deficient mice - ScienceDirect. Febs Letters [J], 584: 2403-2408.

[135] Liu N, Nelson B R, Bezprozvannaya S, et al. 2014. Requirement of MEF2A, C, and D for skeletal muscle regeneration. Proceedings of the National Academy of Sciences of the United States of America [J], 111.

[136] Liu Q C, Zha X H, Faralli H, et al. 2012. Comparative expression profiling identifies differential roles for Myogenin and p38alpha MAPK signaling in myogenesis. J Mol Cell Biol [J], 4: 386-397.

[137] Liu X, Wang Y, Wang Y, et al. 2024. Effect of myristic acid supplementation on triglyceride synthesis and related genes in the pectoral muscles of broiler chickens. Poult Sci [J], 103: 104038.

[138] Lluis F, Perdiguero E, Nebreda A R, et al. 2006. Regulation of skeletal muscle gene expression by p38 MAP kinases. Trends Cell Biol [J], 16: 36-44.

[139] Ma Y, Jiang X D, Zhang D W, et al. 2023. Molecular characterization and effects of the TGIF1 gene on proliferation and steroidogenesis in yak (Bos grunniens) granulosa cells. Theriogenology [J], 211: 224-231.

[140] Macdonald B T, Tamai K, He X 2009. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell [J], 17: 9-26.

[141] Machado L, Geara P, Camps J, et al. 2021. Tissue damage induces a conserved stress response that initiates quiescent muscle stem cell activation. Cell Stem Cell [J].

[142] Macoritto M, Nguyen-Yamamoto L, Huang D C, et al. 2008. Phosphorylation of the human retinoid X receptor alpha at serine 260 impairs coactivator(s) recruitment and induces hormone resistance to multiple ligands. Journal of Biological Chemistry [J], 283: 4943-4956.

[143] Mal, A. 2000. P21 and Retinoblastoma Protein Control the Absence of DNA Replication in Terminally Differentiated Muscle Cells. The Journal of Cell Biology [J], 149: 281-292.

[144] Malumbres M 2014. Cyclin-dependent kinases. Genome Biol [J], 15: 122.

[145] Massagué, Joan 2012. TGFβ signalling in context. Nature Reviews Molecular Cell Biology [J], 13: 616-630.

[146] Massague J, Cheifetz S, Endo T, et al. 1986. Type beta transforming growth factor is an inhibitor of myogenic differentiation. Proc Natl Acad Sci U S A [J], 83: 8206-8210.

[147] Massague J, Seoane J, Wotton D 2005. Smad transcription factors. Genes Dev [J], 19: 2783-2810.

[148] Mccroskery, S. 2003. Myostatin negatively regulates satellite cell activation and self-renewal. Journal of Cell Biology [J], 162: 1135-1147.

[149] Mckinnell I W, Ishibashi J, Le Grand F, et al. 2008. Pax7 activates myogenic genes by recruitment of a histone methyltransferase complex. Nature Cell Biology [J], 10: 77-84.

[150] Mcpherron, Alexandra, C., et al. 1997. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature [J].

[151] Mcpherron A C, Lawler A M, Lee S J Regulation of skeletal muscle mass in mice by a new TGF-p superfamily member. Nature [J].

[152] Mcpherron A C, Lee S J 2002. Suppression of body fat accumulation in myostatin-deficient mice. Journal of Clinical Investigation [J], 109: 595-601.

[153] Melendez J, Sieiro D, Salgado D, et al. 2021. TGFβ signalling acts as a molecular brake of myoblast fusion. Nature Communications [J], 12.

[154] Meng L, Lu Y, Zhang C, et al. 2023. NPRC deletion attenuates cardiac fibrosis in diabetic mice by activating PKA/PKG and inhibiting TGF-β1/Smad pathways. Science Advances [J], 9.

[155] Meriane M, Roux P, Primig M, et al. 2000. Critical activities of Rac1 and Cdc42Hs in skeletal myogenesis: antagonistic effects of JNK and p38 pathways. Mol Biol Cell [J], 11: 2513-2528.

[156] Messina, G. 2005. p27Kip1 Acts Downstream of N-Cadherin-mediated Cell Adhesion to Promote Myogenesis beyond Cell Cycle RegulationD⃞. Molecular Biology of the Cell [J], 16: 1469-1480.

[157] Miao W, Ma Z, Tang Z, et al. 2021. Integrative ATAC-seq and RNA-seq Analysis of the Longissimus Muscle of Luchuan and Duroc Pigs. Front Nutr [J], 8: 742672.

[158] Min C, Chuncheng L, Meng W, et al. 2017. Clenbuterol Induces Cell Cycle Arrest in C2C12 Myoblasts by Delaying p27 Degradation through β-arrestin 2 Signaling. International journal of biological sciences [J], 13: 1341-1350.

[159] Minetti G C, Feige J N, Rosenstiel A, et al. 2011. Gαi2 Signaling Promotes Skeletal Muscle Hypertrophy, Myoblast Differentiation, and Muscle Regeneration. Science Signaling [J], 4: ra80.

[160] Mohamed T M A, Ang Y S, Radzinsky E, et al. 2018. Regulation of Cell Cycle to Stimulate Adult Cardiomyocyte Proliferation and Cardiac Regeneration. Cell [J]: S0092867418301582.

[161] Molkentin J D, Olson E N 1996. Combinatorial control of muscle development by basic helix-loop-helix and MADS-box transcription factors. Proc Natl Acad Sci U S A [J], 93: 9366-9373.

[162] Morissette M R, Cook S A, Buranasombati C, et al. 2009. Myostatin inhibits IGF-I-induced myotube hypertrophy through Akt. Am J Physiol Cell Physiol [J], 297: C1124-1132.

[163] Murphy L O, Smith S, Chen R H, et al. 2002. Molecular interpretation of ERK signal duration by immediate early gene products. Nature Cell Biology [J]: 556-564.

[164] Nabeshima Y, Hanaoka K, Hayasaka M, et al. 1993. Myogenin gene disruption results in perinatal lethality because of severe muscle defect. Nature [J], 364: 532-535.

[165] Noguchi Y T, Nakamura M, Hino N, et al. 2019. Cell-autonomous and redundant roles of Hey1 and HeyL in muscle stem cells: HeyL requires Hes1 to bind diverse DNA sites. Development (Cambridge, England) [J], 146.

[166] Olguin H C, Olwin B B 2004. Pax-7 up-regulation inhibits myogenesis and cell cycle progression in satellite cells: a potential mechanism for self-renewal. Developmental Biology [J], 275: 375-388.

[167] Olguin H C, Yang Z, Tapscott S J, et al. 2007. Reciprocal inhibition between Pax7 and muscle regulatory factors modulates myogenic cell fate determination. J Cell Biol [J], 177: 769-779.

[168] Ornatsky O I, Andreucci J J, Mcdermott J C 1997. A Dominant-Negative Form of Transcription Factor MEF2 Inhibits Myogenesis. Journal of Biological Chemistry [J], 272: 33271-33278.

[169] Otto A, Schmidt C, Patel K 2006. Pax3 and Pax7 expression and regulation in the avian embryo. Anatomy & Embryology [J], 211: 293-310.

[170] Parajuli P, Singh P, Wang Z, et al. 2019. TGIF1 functions as a tumor suppressor in pancreatic ductal adenocarcinoma. The EMBO Journal [J], 38: e101067.

[171] Parisi A, Lacour F, Giordani L, et al. 2015. APC is required for muscle stem cell proliferation and skeletal muscle tissue repair. Journal of Cell Biology [J], 210: 717-726.

[172] Parker S B, Eichele G, Zhang P, et al. 1995. p53-independent expression of p21$^{cip1}$ in muscle and other terminally. Science [J], 267: 1024-1024.

[173] Patrick S, Bryan B, Wenli Y, et al. 2019. PRDM16 controls a brown fat/skeletal muscle switch. Nature [J], 2008年454卷7207期: 961-967页.

[174] Perdiguero E, Ruiz-Bonilla V, Serrano A L, et al. 2007. Genetic deficiency of p38alpha reveals its critical role in myoblast cell cycle exit: the p38alpha-JNK connection. Cell Cycle [J], 6: 1298-1303.

[175] Pollock R, Treisman R 1992. Human SRF-related proteins: DNA-binding properties and potential regulatory targets. Genes & Development [J], 5: 2327.

[176] Potthoff M J, Arnold M A, Mcanally J, et al. 2007. Regulation of skeletal muscle sarcomere integrity and postnatal muscle function by Mef2c. Molecular and Cellular Biology [J]: 27.

[177] Potthoff M J, Olson E N 2007. MEF2: a central regulator of diverse developmental programs. Development [J], 134: 4131-4140.

[178] Pourquie O, Fan C M, Coltey M, et al. 1996. Lateral and axial signals involved in avian somite patterning: a role for BMP4. Cell [J], 84: 461-471.

[179] Puri P L, Avantaggiati M L, Burgio V L, et al. 1995. Reactive oxygen intermediates (ROIs) are involved in the intracellular transduction of angiotensin II signal in C2C12 cells. Ann N Y Acad Sci [J], 752: 394-405.

[180] Rawls A, Valdez M R, Zhang W, et al. 1998. Overlapping functions of the myogenic bHLH genes MRF4 and MyoD revealed in double mutant mice. Development [J], 125: 2349-2358.

[181] Relaix F, Rocancourt D, Mansouri A, et al. 2005. A Pax3/Pax7-dependent population of skeletal muscle progenitor cells. Nature [J], 435: 948-953.

[182] Reynaud E G, Pelpel K, Guillier M, et al. 1999. p57Kip2 Stabilizes the MyoD Protein by Inhibiting Cyclin E-Cdk2 Kinase Activity in Growing Myoblasts. Molecular & Cellular Biology [J], 19: 7621-7629.

[183] Rodgers B D, Ward C W 2022. Myostatin/Activin Receptor Ligands in Muscle and the Development Status of Attenuating Drugs. Endocr Rev [J], 43: 329-365.

[184] Rodgers B D, Wiedeback B D, Hoversten K E, et al. 2014a. Myostatin Stimulates, Not Inihibits, C2C12 Myoblast Proliferation. Endocrinology [J]: 670-675.

[185] Rodgers J T, King K Y, Brett J O, et al. 2014b. mTORC1 controls the adaptive transition of quiescent stem cells from G0 to G(Alert). Nature [J], 510: 393-396.

[186] Rosalind, J., Zalin, et al. 1975. Changes in cyclic AMP, adenylate cyclase and protein kinase levels during the development of embryonic chick skeletal muscle. Experimental Cell Research [J].

[187] Ross S E, Hemati N, Longo K A, et al. 2000. Inhibition of Adipogenesis by Wnt Signaling. Science [J], 289: 950.

[188] Rudnicki M A, Schnegelsberg P N, Stead R H, et al. 1993. MyoD or Myf-5 is required for the formation of skeletal muscle. Cell [J], 75: 1351-1359.

[189] Rudolf A, Schirwis E, Giordani L, et al. 2016. β-Catenin Activation in Muscle Progenitor Cells Regulates Tissue Repair. Cell Reports [J], 15.

[190] Ruiz-Bonilla V, Perdiguero E, Gresh L, et al. 2008. Efficient adult skeletal muscle regeneration in mice deficient in p38beta, p38gamma and p38delta MAP kinases. Cell Cycle [J], 7: 2208-2214.

[191] Ryall J G, Schertzer J D, Alabakis T M, et al. 2008. Intramuscular beta2-agonist administration enhances early regeneration and functional repair in rat skeletal muscle after myotoxic injury. J Appl Physiol (1985) [J], 105: 165-172.

[192] Saab R, Bills J L, Miceli A P, et al. 2006. Pharmacologic inhibition of cyclin-dependent kinase 4/6 activity arrests proliferation in myoblasts and rhabdomyosarcoma-derived cells. Mol Cancer Ther [J], 5: 1299-1308.

[193] Saito H, Gasser A, Bolamperti S, et al. 2019. TG-interacting factor 1 (Tgif1)-deficiency attenuates bone remodeling and blunts the anabolic response to parathyroid hormone. Nat Commun [J], 10: 1354.

[194] Sakamoto K, Mccarthy A, Smith D, et al. 2005. Deficiency of LKB1 in skeletal muscle prevents AMPK activation and glucose uptake during contraction. EMBO J [J], 24: 1810-1820.

[195] Sander V D H, Dyson N J 2008. Conserved functions of the pRB and E2F families. Nat Rev Mol Cell Biol [J], 9: 713-724.

[196] Sartorelli V, Caretti G 2005. Mechanisms underlying the transcriptional regulation of skeletal myogenesis. Curr Opin Genet Dev [J], 15: 528-535.

[197] Schabort E J, Merwe M V D, Loos B, et al. 2009. TGF-β's delay skeletal muscle progenitor cell differentiation in an isoform-independent manner. Experimental Cell Research [J], 315: 373-384.

[198] Schmidt C, Stoeckelhuber M, Mckinnell I, et al. 2004. Wnt 6 regulates the epithelialisation process of the segmental plate mesoderm leading to somite formation. Developmental Biology [J], 271: 198-209.

[199] Schmidt M, Schüler S C, Hüttner S S, et al. 2019. Adult stem cells at work: regenerating skeletal muscle. Cellular and Molecular Life Sciences [J], 76: 2559-2570.

[200] Schuster-Gossler K, Cordes R, Gossler A 2007. Premature myogenic differentiation and depletion of progenitor cells cause severe muscle hypotrophy in Delta1 mutants. Proc Natl Acad Sci U S A [J], 104: 537-542.

[201] Segales J, Perdiguero E, Munoz-Canoves P 2015. Epigenetic control of adult skeletal muscle stem cell functions. FEBS J [J], 282: 1571-1588.

[202] Seigneurin-Venin S, Parrish E, Marty I, et al. 1996. Involvement of the dihydropyridine receptor and internal Ca2+ stores in myoblast fusion. Exp Cell Res [J], 223: 301-307.

[203] Serra C, Palacios D, Mozzetta C, et al. 2007. Functional interdependence at the chromatin level between the MKK6/p38 and IGF1/PI3K/AKT pathways during muscle differentiation. Molecular Cell [J], 28: 200-213.

[204] Shainberg A, Yagil G, Yaffe D 1969. Control of myogenesis in vitro by Ca 2 + concentration in nutritional medium. Exp Cell Res [J], 58: 163-167.

[205] Shan T, Zhang P, Liang X, et al. 2014. Lkb1 is indispensable for skeletal muscle development, regeneration, and satellite cell homeostasis. Stem Cells [J], 32: 2893-2907.

[206] Shaulian E, Karin M 2001. AP-1 in cell proliferation and survival. Oncogene [J], 20: 2390-2400.

[207] Shaw T, Barr F G, Ren A 2024. The PAX Genes: Roles in Development, Cancer, and Other Diseases. Cancers [J], 16.

[208] Shi L, Zha H, Pan Z, et al. 2023. DUSP1 protects against ischemic acute kidney injury through stabilizing mtDNA via interaction with JNK. Cell Death & Disease [J], 14.

[209] Shin, Fujimaki, Daiki, et al. 2018. Notch1 and Notch2 Coordinately Regulate Stem Cell Function in the Quiescent and Activated States of Muscle Satellite Cells. Stem Cells [J].

[210] Shin K S, Park J Y, Ha D B, et al. 1996. Involvement of K(Ca) channels and stretch-activated channels in calcium influx, triggering membrane fusion of chick embryonic myoblasts. Dev Biol [J], 175: 14-23.

[211] Sicinski P, Donaher J L, Parker S B, et al. 1995. Cyclin D1 provides a link between development and oncogenesis in the retina and breast. Cell [J], 82: 621-630.

[212] Singh K, Dilworth F J 2013. Differential modulation of cell cycle progression distinguishes members of the myogenic regulatory factor family of transcription factors. FEBS J [J], 280: 3991-4003.

[213] Snyder C M, Rice A L, Estrella N L, et al. 2013. MEF2A regulates the Gtl2-Dio3 microRNA mega-cluster to modulate WNT signaling in skeletal muscle regeneration. Development (Cambridge, England) [J], 140: 31.

[214] Soleimani V D, Punch V G, Kawabe Y I, et al. 2012. Transcriptional Dominance of Pax7 in Adult Myogenesis Is Due to High-Affinity Recognition of Homeodomain Motifs. Developmental Cell [J], 22.

[215] Souilhol C, Cormier S, Tanigaki K, et al. 2006. RBP-Jκ-Dependent Notch Signaling Is Dispensable for Mouse Early Embryonic Development. Molecular & Cellular Biology [J], 26: 4769.

[216] Stewart R, Flechner L, Montminy M, et al. 2011. CREB Is Activated by Muscle Injury and Promotes Muscle Regeneration. PLoS ONE [J], 6: e24714.

[217] Steyn P J, Dzobo K, Smith R I, et al. 2019. Interleukin-6 Induces Myogenic Differentiation via JAK2-STAT3 Signaling in Mouse C2C12 Myoblast Cell Line and Primary Human Myoblasts. International Journal of Molecular Sciences [J], 20.

[218] Stürken C, Mbus V, Milde-Langosch K, et al. 2021. TGFB-Induced Factor Homeobox 1 (TGIF) Expression in Breast Cancer. BMC Cancer [J], 21.

[219] Szade K 2020. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris.

[220] Tajbakhsh S 2009. Skeletal muscle stem cells in developmental versus regenerative myogenesis. J Intern Med [J], 266: 372-389.

[221] Tajbakhsh S, Borello U, Vivarelli E, et al. 1998. Differential activation of Myf5 and MyoD by different Wnts in explants of mouse paraxial mesoderm and the later activation of myogenesis in the absence of Myf5. Development [J], 125: 4155-4162.

[222] Tajbakhsh S, Rocancourt D, Buckingham M 1996. Muscle progenitor cells failing to respond to positional cues adopt non-myogenic fates in myf-5 null mice. Nature [J], 384: 266-270.

[223] Tajeddine N, Gailly P 2012. TRPC1 protein channel is major regulator of epidermal growth factor receptor signaling. J Biol Chem [J], 287: 16146-16157.

[224] Taskinen P, Ruskoaho H 1996. Stretch-induced increase in atrial natriuretic peptide secretion is blocked by thapsigargin. Eur J Pharmacol [J], 308: 295-300.

[225] Taylor W E, Bhasin S, Artaza J, et al. 2001. Myostatin inhibits cell proliferation and protein synthesis in C2C12 muscle cells. Am J Physiol Endocrinol Metab [J], 280: E221-228.

[226] Teng J, Gao Y, Zeng H, et al. 2024. A compendium of genetic regulatory effects across pig tissues. Nature Genetics [J], 56: 34.

[227] Thomas M, Langley B, Berry C, et al. 2000. Myostatin, a Negative Regulator of Muscle Growth, Functions by Inhibiting Myoblast Proliferation. Journal of Biological Chemistry [J], 275: 40235-40243.

[228] Thomson D 2018. The Role of AMPK in the Regulation of Skeletal Muscle Size, Hypertrophy, and Regeneration. International Journal of Molecular Sciences [J], 19.

[229] Tintignac L A, Sirri V, Leibovitch M P, et al. 2004. Mutant MyoD lacking Cdc2 phosphorylation sites delays M-phase entry. Mol Cell Biol [J], 24: 1809-1821.

[230] Trendelenburg A U, Meyer A, Rohner D, et al. 2009. Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. Am J Physiol Cell Physiol [J], 296: C1258-1270.

[231] Tsai C C, Saffitz J E, Billadello J J 1997. Expression of the Gs protein alpha-subunit disrupts the normal program of differentiation in cultured murine myogenic cells. Journal of Clinical Investigation [J], 99: 67-76.

[232] Vainshtein A, Sandri M 2020. Signaling Pathways That Control Muscle Mass. Int J Mol Sci [J], 21.

[233] Vavvas D, Apazidis A, Saha A K, et al. 1997. Contraction-induced changes in acetyl-CoA carboxylase and 5'-AMP-activated kinase in skeletal muscle. J Biol Chem [J], 272: 13255-13261.

[234] Vertino, A. M 2005. Wnt10b Deficiency Promotes Coexpression of Myogenic and Adipogenic Programs in Myoblasts. Molecular Biology of the Cell [J], 16: 2039-2048.

[235] Von Maltzahn J, Bentzinger C F, Rudnicki M A 2011. Wnt7a-Fzd7 signalling directly activates the Akt/mTOR anabolic growth pathway in skeletal muscle. Nature Cell Biology [J], 14: 186.

[236] Von Maltzahn J, Jones A E, Parks R J, et al. 2013. Pax7 is critical for the normal function of satellite cells in adult skeletal muscle. Proceedings of the National Academy of Sciences of the United States of America [J], 110: 16474-16479.

[237] Wahrmann J P, Luzzati D, Winand R 1973. Changes in adenyl cyclase specific activity during differentiation on an established myogenic cell line. Biochemical & Biophysical Research Communications [J], 52: 576-581.

[238] Wang B, Ma Q, Wang X, et al. 2022. TGIF1 overexpression promotes glioma progression and worsens patient prognosis. Cancer Med [J], 11: 5113-5128.

[239] Wang G, Zhu H, Situ C, et al. 2018. p110α of PI3K is necessary and sufficient for quiescence exit in adult muscle satellite cells. The EMBO Journal [J], 37.

[240] Wang H, Xu Q, Xiao F, et al. 2008. Involvement of the p38 mitogen-activated protein kinase alpha, beta, and gamma isoforms in myogenic differentiation. Mol Biol Cell [J], 19: 1519-1528.

[241] Wang J G, Miyazu M, Matsushita E, et al. 2001. Uniaxial cyclic stretch induces focal adhesion kinase (FAK) tyrosine phosphorylation followed by mitogen-activated protein kinase (MAPK) activation. Biochem Biophys Res Commun [J], 288: 356-361.

[242] Wang J L, Qi Z, Li Y H, et al. 2017. TGFβ induced factor homeobox 1 promotes colorectal cancer development through activating Wnt/β-catenin signaling. Oncotarget [J], 8.

[243] Wang Y X, Liu S Y, Yan Y Q, et al. 2019. SPARCL1 promotes C2C12 cell differentiation via BMP7-mediated BMP/TGF-β cell signaling pathway. Cell Death & Disease [J], 10.

[244] Whittemore L A, Song K, Li X, et al. 2003. Inhibition of myostatin in adult mice increases skeletal muscle mass and strength. Biochemical & Biophysical Research Communications [J], 300: 965-971.

[245] Wilson E M, Rotwein P 2006. Control of MyoD function during initiation of muscle differentiation by an autocrine signaling pathway activated by insulin-like growth factor-II. J Biol Chem [J], 281: 29962-29971.

[246] Wilson E M, Rotwein P 2007. Selective Control of Skeletal Muscle Differentiation by Akt1. Journal of Biological Chemistry [J], 282: 5106.

[247] Wu J, Yue B, Lan X, et al. 2019. MiR‐499 regulates myoblast proliferation and differentiation by targeting transforming growth factor β receptor 1. Journal of Cellular Physiology [J], 234.

[248] Wu Z, Woodring P J, Bhakta K S, et al. 2000. p38 and Extracellular Signal-Regulated Kinases Regulate the Myogenic Program at Multiple Steps. Molecular & Cellular Biology [J], 20: 3951.

[249] Xiang X, Zhao J, Xu G, et al. 2011. mTOR and the differentiation of mesenchymal stem cells. 生物化学与生物物理学报:英文版 [J], 43: 10.

[250] Xiao W, Jiang N, Ji Z, et al. 2024. Single-Cell RNA Sequencing Reveals the Cellular Landscape of Longissimus Dorsi in a Newborn Suhuai Pig. International Journal of Molecular Sciences [J], 25.

[251] Xie B, Wang S, Jiang N, et al. 2019. Cyclin B1/CDK1-regulated mitochondrial bioenergetics in cell cycle progression and tumor resistance. Cancer letters [J], 443: 56-66.

[252] Xiong Z, Wang M, Wu J, et al. 2023. Tceal7 Regulates Skeletal Muscle Development through Its Interaction with Cdk1. Int J Mol Sci [J], 24.

[253] Yablonka-Reuveni Z, Rivera A J 1997. Proliferative Dynamics and the Role of FGF2 During Myogenesis of Rat Satellite Cells on Isolated Fibers. Basic Appl Myol [J], 7: 189-202.

[254] Yang Y, Zhang Y, Lin Z, et al. 2022. Silencing of histone deacetylase 3 suppresses the development of esophageal squamous cell carcinoma through regulation of miR-494-mediated TGIF1. Cancer cell international [J], 22: 191.

[255] Yin H, Price F, Rudnicki M A 2013. Satellite cells and the muscle stem cell niche. Physiol Rev [J], 93: 23-67.

[256] Yun K, Wold B 1996. Skeletal muscle determination and differentiation: story of a core regulatory network and its context. Curr Opin Cell Biol [J], 8: 877-889.

[257] Zalc A, Hayashi S, Aurade F, et al. 2014. Antagonistic regulation of p57kip2 by Hes/Hey downstream of Notch signaling and muscle regulatory factors regulates skeletal muscle growth arrest. Development [J], 141: 2780-2790.

[258] Zalin R J, Leaver R 1975. The effect of a transient increase in intracellular cyclic AMP upon muscle cell fusion. Febs Letters [J], 53.

[259] Zavadil J, Cermak L, Sotonieves N, et al. 2004. Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. Embo Journal [J], 23: 1155-1165.

[260] Zhang J, Zhang F, Fan J, et al. 2021a. TGIF1 Knockdown Inhibits the Proliferation and Invasion of Gastric Cancer via AKT Signaling Pathway. Cancer Management and Research [J], Volume 13: 2603-2612.

[261] Zhang J M, Wei Q, Zhao X, et al. 1999a. Coupling of the cell cycle and myogenesis through the cyclin D1-dependent interaction of MyoD with cdk4. EMBO J [J], 18: 926-933.

[262] Zhang J M, Zhao X, Wei Q, et al. 1999b. Direct inhibition of G(1) cdk kinase activity by MyoD promotes myoblast cell cycle withdrawal and terminal differentiation. EMBO J [J], 18: 6983-6993.

[263] Zhang S, Chen X, Hu Y, et al. 2016. All-trans retinoic acid modulates Wnt3A-induced osteogenic differentiation of mesenchymal stem cells via activating the PI3K/AKT/GSK3β signalling pathway. Molecular & Cellular Endocrinology [J], 422: 243-253.

[264] Zhang W, Behringer R R, Olson E N 1995. Inactivation of the myogenic bHLH gene MRF4 results in up-regulation of myogenin and rib anomalies. Genes & Development [J], 9: 1388-1399.

[265] Zhang Y, Lahmann I, Baum K, et al. 2021b. Author Correction: Oscillations of Delta-like1 regulate the balance between differentiation and maintenance of muscle stem cells. Nat Commun [J], 12: 1766.

[266] Zhu G H, Huang J, Bi Y, et al. 2009. Activation of RXR and RAR signaling promotes myogenic differentiation of myoblastic C2C12 cells. Differentiation [J], 78: 195-204.

[267] Zhu Q, Liang Z, Cai S, et al. 2023. E3 ligase Deltex2 accelerates myoblast proliferation and inhibits myoblast differentiation by targeting Pax7 and MyoD,respectively. 生物化学与生物物理学报:英文版 [J], 55: 12.

[268] 曹贺然 2019. TGIF1对奶山羊卵巢颗粒细胞E2/P4分泌及凋亡作用的研究 [M]. 西北农林科技大学.

[269] 代立志 2023. Egr1在小鼠成肌细胞增殖过程中的作用机制 [M]. 东北农业大学.

[270] 侯黎明, 倪梦茹, 赵清波, 等. 一种猪永生化肌卫星细胞及其构建方法 [M].

[271] 季铮渝, 倪梦茹, 张兆博, 等. 2023. 苏淮猪背最长肌FAPs细胞体外成脂能力及其基因表达模式的研究. 畜牧兽医学报 [J], 54: 4126-4142.

[272] 刘雪云 2015. 阻血诱导大鼠骨骼肌肥大的基因组学分析研究(免疫组化部分) [M]. 浙江中医药大学.

中图分类号:

 S81    

开放日期:

 2025-06-12    

无标题文档

   建议浏览器: 谷歌 火狐 360请用极速模式,双核浏览器请用极速模式