Satellite cell-specific deletion of Cipc alleviates myopathy in mdx mice

卫星 细胞生物学 杜氏肌营养不良 肌病 生物 mdx鼠标 细胞 化学 肌营养不良蛋白 遗传学 航空航天工程 工程类
作者
Zheng Jian,Jing Lou,Yanfang Li,Panting Qian,Wei He,Yingxue Hao,Ting Xue,Yangxin Li,Yao-Hua Song
出处
期刊:Cell Reports [Elsevier]
卷期号:39 (11): 110939-110939 被引量:2
标识
DOI:10.1016/j.celrep.2022.110939
摘要

•CIPC is up-regulated during myoblast differentiation•Specific deletion of Cipc in satellite cells alleviates myopathy in mdx mice•Cipc deficiency leads to activation of SP1 to trigger the transcription of Pax7•Loss of Cipc in satellite cells promotes muscle regeneration Skeletal muscle regeneration relies on satellite cells that can proliferate, differentiate, and form new myofibers upon injury. Emerging evidence suggests that misregulation of satellite cell fate and function influences the severity of Duchenne muscular dystrophy (DMD). The transcription factor Pax7 determines the myogenic identity and maintenance of the pool of satellite cells. The circadian clock regulates satellite cell proliferation and self-renewal. Here, we show that the CLOCK-interacting protein Circadian (CIPC) a negative-feedback regulator of the circadian clock, is up-regulated during myoblast differentiation. Specific deletion of Cipc in satellite cells alleviates myopathy, improves muscle function, and reduces fibrosis in mdx mice. Cipc deficiency leads to activation of the ERK1/2 and JNK1/2 signaling pathways, which activates the transcription factor SP1 to trigger the transcription of Pax7 and MyoD. Therefore, CIPC is a negative regulator of satellite cell function, and loss of Cipc in satellite cells promotes muscle regeneration. Skeletal muscle regeneration relies on satellite cells that can proliferate, differentiate, and form new myofibers upon injury. Emerging evidence suggests that misregulation of satellite cell fate and function influences the severity of Duchenne muscular dystrophy (DMD). The transcription factor Pax7 determines the myogenic identity and maintenance of the pool of satellite cells. The circadian clock regulates satellite cell proliferation and self-renewal. Here, we show that the CLOCK-interacting protein Circadian (CIPC) a negative-feedback regulator of the circadian clock, is up-regulated during myoblast differentiation. Specific deletion of Cipc in satellite cells alleviates myopathy, improves muscle function, and reduces fibrosis in mdx mice. Cipc deficiency leads to activation of the ERK1/2 and JNK1/2 signaling pathways, which activates the transcription factor SP1 to trigger the transcription of Pax7 and MyoD. Therefore, CIPC is a negative regulator of satellite cell function, and loss of Cipc in satellite cells promotes muscle regeneration. Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disorder caused by mutations of the gene encoding the protein dystrophin, which anchors the extracellular matrix to the cytoskeleton inside a myofiber. Many Duchenne mutations are caused by multiple exon deletions. Some of these deletions (or insertions) can result in frameshift mutations or nonsense mutations, leading to the production of truncated and non-functional dystrophin protein (Duan et al., 2021Duan D. Goemans N. Takeda S. Mercuri E. Aartsma-Rus A. Duchenne muscular dystrophy.Nat. Rev. Dis. Primers. 2021; 7: 13https://doi.org/10.1038/s41572-021-00248-3Crossref PubMed Scopus (100) Google Scholar; Verhaart and Aartsma-Rus, 2019Verhaart I.E.C. Aartsma-Rus A. Therapeutic developments for Duchenne muscular dystrophy.Nat. Rev. Neurol. 2019; 15: 373-386https://doi.org/10.1038/s41582-019-0203-3Crossref PubMed Scopus (166) Google Scholar). Dystrophin is critical for stabilizing the plasma membrane of a myofiber during muscle contraction. In the absence of dystrophin, muscle contraction causes myofiber necrosis, triggering subsequent inflammation and fibrosis, leading to progressive muscle weakness and wasting. There is no cure for DMD, but steroids may promote muscle recovery (Goemans, 2018Goemans N. How glucocorticoids change life in Duchenne muscular dystrophy.Lancet. 2018; 391: 406-407https://doi.org/10.1016/s0140-6736(17)32405-4Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar; Quattrocelli et al., 2017Quattrocelli M. Barefield D.Y. Warner J.L. Vo A.H. Hadhazy M. Earley J.U. Demonbreun A.R. McNally E.M. Intermittent glucocorticoid steroid dosing enhances muscle repair without eliciting muscle atrophy.J. Clin. Invest. 2017; 127: 2418-2432https://doi.org/10.1172/jci91445Crossref PubMed Scopus (0) Google Scholar). Several studies have attempted to restore functional expression of the dystrophin protein through genetic intervention (Amoasii et al., 2018Amoasii L. Hildyard J.C.W. Li H. Sanchez-Ortiz E. Mireault A. Caballero D. Harron R. Stathopoulou T.R. Massey C. Shelton J.M. et al.Gene editing restores dystrophin expression in a canine model of Duchenne muscular dystrophy.Science. 2018; 362: 86-91https://doi.org/10.1126/science.aau1549Crossref PubMed Scopus (271) Google Scholar; Chemello et al., 2020Chemello F. Bassel-Duby R. Olson E.N. Correction of muscular dystrophies by CRISPR gene editing.J. Clin. Invest. 2020; 130: 2766-2776https://doi.org/10.1172/jci136873Crossref PubMed Scopus (0) Google Scholar; Cirak et al., 2011Cirak S. Arechavala-Gomeza V. Guglieri M. Feng L. Torelli S. Anthony K. Abbs S. Garralda M.E. Bourke J. Wells D.J. et al.Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study.Lancet. 2011; 378: 595-605https://doi.org/10.1016/s0140-6736(11)60756-3Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar; Doudna, 2020Doudna J.A. The promise and challenge of therapeutic genome editing.Nature. 2020; 578: 229-236https://doi.org/10.1038/s41586-020-1978-5Crossref PubMed Scopus (257) Google Scholar; Nelson et al., 2017Nelson C.E. Robinson-Hamm J.N. Gersbach C.A. Genome engineering: a new approach to gene therapy for neuromuscular disorders.Nat. Rev. Neurol. 2017; 13: 647-661https://doi.org/10.1038/nrneurol.2017.126Crossref PubMed Scopus (45) Google Scholar). Although the results are promising, these therapies are still in the early stages of study in humans. Alternative treatments that block signaling pathways that contribute to disease progression are also emerging. Deletion of periostin can reduce muscle fibrosis in the δ-sarcoglycan null (Sgcd(−/−)) mouse model of muscular dystrophy (Lorts et al., 2012Lorts A. Schwanekamp J.A. Baudino T.A. McNally E.M. Molkentin J.D. Deletion of periostin reduces muscular dystrophy and fibrosis in mice by modulating the transforming growth factor-beta pathway.Proc. Natl. Acad. Sci. U S A. 2012; 109: 10978-10983https://doi.org/10.1073/pnas.1204708109Crossref PubMed Scopus (83) Google Scholar). Pharmacological inhibition of the BET proteins ameliorated muscle function by reducing oxidative stress (Segatto et al., 2020Segatto M. Szokoll R. Fittipaldi R. Bottino C. Nevi L. Mamchaoui K. Filippakopoulos P. Caretti G. BETs inhibition attenuates oxidative stress and preserves muscle integrity in Duchenne muscular dystrophy.Nat. Commun. 2020; 11: 6108https://doi.org/10.1038/s41467-020-19839-xCrossref PubMed Scopus (15) Google Scholar). Muscle-specific deletion of SLK enhances myogenic differentiation through up-regulation of myogenin in the mdx model (Pryce et al., 2021Pryce B.R. Labreche C. Hamoudi D. Abou-Hamad J. Al-Zahrani K.N. Hodgins J.J. Boulanger-Piette A. Bosse S. Balog-Alvarez C. Frenette J. et al.Muscle-specific deletion of SLK/Stk2 enhances p38 activity and myogenesis in mdx mice.Biochim. Biophys. Acta Mol. Cell Res. 2021; 1868: 118917https://doi.org/10.1016/j.bbamcr.2020.118917Crossref PubMed Scopus (3) Google Scholar). Recent studies revealed stem cells as a new avenue of a potential treatment for DMD (Blau and Daley, 2019Blau H.M. Daley G.Q. Stem cells in the treatment of disease.New Engl. J. Med. 2019; 380: 1748-1760https://doi.org/10.1056/nejmra1716145Crossref PubMed Scopus (0) Google Scholar; Feige et al., 2018Feige P. Brun C.E. Ritso M. Rudnicki M.A. Orienting muscle stem cells for regeneration in homeostasis, aging, and disease.Cell Stem Cell. 2018; 23: 653-664https://doi.org/10.1016/j.stem.2018.10.006Abstract Full Text Full Text PDF PubMed Scopus (100) Google Scholar). DMD has also been considered a stem cell disease, because dystrophin deficiency is related to abnormalities in satellite cell polarity, asymmetric division, and function, which contribute to the development of the DMD phenotype (Almada and Wagers, 2016Almada A.E. Wagers A.J. Molecular circuitry of stem cell fate in skeletal muscle regeneration, ageing and disease.Nat. Rev. Mol. Cell Biol. 2016; 17: 267-279https://doi.org/10.1038/nrm.2016.7Crossref PubMed Scopus (158) Google Scholar; Chang et al., 2016Chang N.C. Chevalier F.P. Rudnicki M.A. Satellite cells in muscular dystrophy - lost in polarity.Trends Mol. Med. 2016; 22: 479-496https://doi.org/10.1016/j.molmed.2016.04.002Abstract Full Text Full Text PDF PubMed Scopus (85) Google Scholar; Matre et al., 2019Matre P.R. Mu X. Wu J. Danila D. Hall M.A. Kolonin M.G. Darabi R. Huard J. CRISPR/Cas9-Based dystrophin restoration reveals a novel role for dystrophin in bioenergetics and stress resistance of muscle progenitors.Stem Cell. 2019; 37: 1615-1628https://doi.org/10.1002/stem.3094Crossref PubMed Scopus (15) Google Scholar; Xie et al., 2016Xie X. Tsai S.Y. Tsai M.J. COUP-TFII regulates satellite cell function and muscular dystrophy.J. Clin. Invest. 2016; 126: 3929-3941https://doi.org/10.1172/jci87414Crossref PubMed Scopus (0) Google Scholar). Satellite cells are muscle stem cells that remain quiescent in resting muscles and become activated in response to injury to proliferate, differentiate, and fuse with either existing myofibers or with each other to form new myofibers to repair the injured muscle (Akhmedov and Berdeaux, 2013Akhmedov D. Berdeaux R. The effects of obesity on skeletal muscle regeneration.Front. Physiol. 2013; 4: 371https://doi.org/10.3389/fphys.2013.00371Crossref PubMed Scopus (137) Google Scholar; Dinulovic et al., 2016Dinulovic I. Furrer R. Beer M. Ferry A. Cardel B. Handschin C. Muscle PGC-1α modulates satellite cell number and proliferation by remodeling the stem cell niche.Skelet. Muscle. 2016; 6: 39https://doi.org/10.1186/s13395-016-0111-9Crossref PubMed Scopus (16) Google Scholar; Gurevich et al., 2016Gurevich D.B. Nguyen P.D. Siegel A.L. Ehrlich O.V. Sonntag C. Phan J.M.N. Berger S. Ratnayake D. Hersey L. Berger J. et al.Asymmetric division of clonal muscle stem cells coordinates muscle regeneration in vivo.Science. 2016; 353: aad9969https://doi.org/10.1126/science.aad9969Crossref PubMed Scopus (85) Google Scholar; Moyer and Wagner, 2011Moyer A.L. Wagner K.R. Regeneration versus fibrosis in skeletal muscle.Curr. Opin. Rheumatol. 2011; 23: 568-573https://doi.org/10.1097/bor.0b013e32834bac92Crossref PubMed Scopus (0) Google Scholar; Musaro, 2020Musaro A. Muscle homeostasis and regeneration: from molecular mechanisms to therapeutic opportunities.Cells. 2020; 9: 2033https://doi.org/10.3390/cells9092033Crossref Scopus (5) Google Scholar; Olguin and Olwin, 2004Olguin H.C. Olwin B.B. Pax-7 up-regulation inhibits myogenesis and cell cycle progression in satellite cells: a potential mechanism for self-renewal.Dev. Biol. 2004; 275: 375-388https://doi.org/10.1016/j.ydbio.2004.08.015Crossref PubMed Scopus (372) Google Scholar; Zammit et al., 2004Zammit P.S. Golding J.P. Nagata Y. Hudon V. Partridge T.A. Beauchamp J.R. Muscle satellite cells adopt divergent fates.J. Cell Biol. 2004; 166: 347-357https://doi.org/10.1083/jcb.200312007Crossref PubMed Scopus (665) Google Scholar). The paired box protein 7 (Pax7) is a transcription factor that plays an essential role in satellite cell specification, survival, proliferation, and self-renewal (Egerman et al., 2015Egerman M.A. Cadena S.M. Gilbert J.A. Meyer A. Nelson H.N. Swalley S.E. Mallozzi C. Jacobi C. Jennings L.L. Clay I. et al.GDF11 increases with age and inhibits skeletal muscle regeneration.Cell Metab. 2015; 22: 164-174https://doi.org/10.1016/j.cmet.2015.05.010Abstract Full Text Full Text PDF PubMed Scopus (367) Google Scholar; Olguin and Olwin, 2004Olguin H.C. Olwin B.B. Pax-7 up-regulation inhibits myogenesis and cell cycle progression in satellite cells: a potential mechanism for self-renewal.Dev. Biol. 2004; 275: 375-388https://doi.org/10.1016/j.ydbio.2004.08.015Crossref PubMed Scopus (372) Google Scholar; Relaix et al., 2004Relaix F. Rocancourt D. Mansouri A. Buckingham M. Divergent functions of murine Pax3 and Pax7 in limb muscle development.Genes Dev. 2004; 18: 1088-1105https://doi.org/10.1101/gad.301004Crossref PubMed Scopus (234) Google Scholar, Relaix et al., 2005Relaix F. Rocancourt D. Mansouri A. Buckingham M. A Pax3/Pax7-dependent population of skeletal muscle progenitor cells.Nature. 2005; 435: 948-953https://doi.org/10.1038/nature03594Crossref PubMed Scopus (792) Google Scholar; Seale et al., 2000Seale P. Sabourin L.A. Girgis-Gabardo A. Mansouri A. Gruss P. Rudnicki M.A. Pax7 is required for the specification of myogenic satellite cells.Cell. 2000; 102: 777-786https://doi.org/10.1016/s0092-8674(00)00066-0Abstract Full Text Full Text PDF PubMed Google Scholar). Pax7-null mice showed a marked reduction in myonuclear numbers and myofiber diameters due to a progressive loss of satellite cells (Kuang et al., 2006Kuang S. Charge S.B. Seale P. Huh M. Rudnicki M.A. Distinct roles for Pax7 and Pax3 in adult regenerative myogenesis.J. Cell Biol. 2006; 172: 103-113https://doi.org/10.1083/jcb.200508001Crossref PubMed Scopus (342) Google Scholar; Oustanina et al., 2004Oustanina S. Hause G. Braun T. Pax7 directs postnatal renewal and propagation of myogenic satellite cells but not their specification.EMBO J. 2004; 23: 3430-3439https://doi.org/10.1038/sj.emboj.7600346Crossref PubMed Scopus (380) Google Scholar; Relaix et al., 2006Relaix F. Montarras D. Zaffran S. Gayraud-Morel B. Rocancourt D. Tajbakhsh S. Mansouri A. Cumano A. Buckingham M. Pax3 and Pax7 have distinct and overlapping functions in adult muscle progenitor cells.J. Cell Biol. 2006; 172: 91-102https://doi.org/10.1083/jcb.200508044Crossref PubMed Scopus (495) Google Scholar; Seale et al., 2000Seale P. Sabourin L.A. Girgis-Gabardo A. Mansouri A. Gruss P. Rudnicki M.A. Pax7 is required for the specification of myogenic satellite cells.Cell. 2000; 102: 777-786https://doi.org/10.1016/s0092-8674(00)00066-0Abstract Full Text Full Text PDF PubMed Google Scholar). Conditional deletion of Pax7 in adult muscles resulted in the loss of satellite cells and impaired muscle regeneration (Gunther et al., 2013Gunther S. Kim J. Kostin S. Lepper C. Fan C.M. Braun T. Myf5-positive satellite cells contribute to Pax7-dependent long-term maintenance of adult muscle stem cells.Cell Stem Cell. 2013; 13: 590-601https://doi.org/10.1016/j.stem.2013.07.016Abstract Full Text Full Text PDF PubMed Scopus (168) Google Scholar; Lepper et al., 2011Lepper C. Partridge T.A. Fan C.M. An absolute requirement for Pax7-positive satellite cells in acute injury-induced skeletal muscle regeneration.Development. 2011; 138: 3639-3646https://doi.org/10.1242/dev.067595Crossref PubMed Scopus (674) Google Scholar; Murphy et al., 2011Murphy M.M. Lawson J.A. Mathew S.J. Hutcheson D.A. Kardon G. Satellite cells, connective tissue fibroblasts and their interactions are crucial for muscle regeneration.Development. 2011; 138: 3625-3637https://doi.org/10.1242/dev.064162Crossref PubMed Scopus (721) Google Scholar; Sambasivan et al., 2011Sambasivan R. Yao R. Kissenpfennig A. Van Wittenberghe L. Paldi A. Gayraud-Morel B. Guenou H. Malissen B. Tajbakhsh S. Galy A. Pax7-expressing satellite cells are indispensable for adult skeletal muscle regeneration.Development. 2011; 138: 3647-3656https://doi.org/10.1242/dev.073601Crossref PubMed Scopus (1) Google Scholar; von Maltzahn et al., 2013von Maltzahn J. Jones A.E. Parks R.J. Rudnicki M.A. Pax7 is critical for the normal function of satellite cells in adult skeletal muscle.Proc. Natl. Acad. Sci. U S A. 2013; 110: 16474-16479https://doi.org/10.1073/pnas.1307680110Crossref PubMed Scopus (319) Google Scholar). Accumulating evidence suggests that the circadian clock regulates satellite cell function and skeletal muscle development. The core clock genes BMAL1 and CLOCK form a CLOCK-BMAL1 complex to induce the expression of CRY and PER, which in turn form a complex repressing the expression of CLOCK and BMAL1. MYOD1 has been identified as a clock amplifier that transcriptionally regulates Bmal1 expression (Hodge et al., 2019Hodge B.A. Zhang X. Gutierrez-Monreal M.A. Cao Y. Hammers D.W. Yao Z. Wolff C.A. Du P. Kemler D. Judge A.R. Esser K.A. MYOD1 functions as a clock amplifier as well as a critical co-factor for downstream circadian gene expression in muscle.Elife. 2019; 8https://doi.org/10.7554/elife.43017Crossref PubMed Google Scholar). The circadian clock is also reinforced by two additional negative-feedback regulators, including the nuclear receptors Rev-erbα and β (Solt et al., 2012Solt L.A. Wang Y. Banerjee S. Hughes T. Kojetin D.J. Lundasen T. Shin Y. Liu J. Cameron M.D. Noel R. et al.Regulation of circadian behaviour and metabolism by synthetic REV-ERB agonists.Nature. 2012; 485: 62-68https://doi.org/10.1038/nature11030Crossref PubMed Scopus (473) Google Scholar; Yin et al., 2006Yin L. Wang J. Klein P.S. Lazar M.A. Nuclear receptor rev-erbα is a critical lithium-sensitive component of the circadian clock.Science. 2006; 311: 1002-1005https://doi.org/10.1126/science.1121613Crossref PubMed Scopus (441) Google Scholar) and CLOCK-interacting protein, Circadian (CIPC) (Zhao et al., 2007Zhao W.N. Malinin N. Yang F.C. Staknis D. Gekakis N. Maier B. Reischl S. Kramer A. Weitz C.J. CIPC is a mammalian circadian clock protein without invertebrate homologues.Nat. Cell Biol. 2007; 9: 268-275https://doi.org/10.1038/ncb1539Crossref PubMed Scopus (61) Google Scholar). Bmal1−/− mice display reduced muscle mass and body weight, which is rescued by muscle-specific expression of Bmal1 (Kondratov et al., 2006Kondratov R.V. Kondratova A.A. Gorbacheva V.Y. Vykhovanets O.V. Antoch M.P. Early aging and age-related pathologies in mice deficient in BMAL1, the core component of the circadian clock.Genes Dev. 2006; 20: 1868-1873https://doi.org/10.1101/gad.1432206Crossref PubMed Scopus (767) Google Scholar; McDearmon et al., 2006McDearmon E.L. Patel K.N. Ko C.H. Walisser J.A. Schook A.C. Chong J.L. Wilsbacher L.D. Song E.J. Hong H.K. Bradfield C.A. Takahashi J.S. Dissecting the functions of the mammalian clock protein BMAL1 by tissue-specific rescue in mice.Science. 2006; 314: 1304-1308https://doi.org/10.1126/science.1132430Crossref PubMed Scopus (238) Google Scholar). Genetic loss of function of Bmal1 aggravated muscle damage in the dystrophic mouse model (mdx) (Gao et al., 2020Gao H. Xiong X. Lin Y. Chatterjee S. Ma K. The clock regulator Bmal1 protects against muscular dystrophy.Exp. Cell Res. 2020; 397: 112348https://doi.org/10.1016/j.yexcr.2020.112348Crossref PubMed Scopus (6) Google Scholar). CLOCK mutant mice display a 1.8-fold reduction in mean daily locomotor activity relative to the wild-type mice (Pastore and Hood, 2013Pastore S. Hood D.A. Endurance training ameliorates the metabolic and performance characteristics of circadian Clock mutant mice.J. Appl. Physiol. 2013; 114: 1076-1084https://doi.org/10.1152/japplphysiol.01505.2012Crossref PubMed Scopus (39) Google Scholar). Loss of Per2 affects forced locomotor performance in mice without altering muscle contractility (Bae et al., 2006Bae K. Lee K. Seo Y. Lee H. Kim D. Choi I. Differential effects of two period genes on the physiology and proteomic profiles of mouse anterior tibialis muscles.Mol. Cells. 2006; 22: 275-284PubMed Google Scholar). Rev-erbα deficiency results in lower exercise capacities (Woldt et al., 2013Woldt E. Sebti Y. Solt L.A. Duhem C. Lancel S. Eeckhoute J. Hesselink M.K.C. Paquet C. Delhaye S. Shin Y. et al.Rev-erb-alpha modulates skeletal muscle oxidative capacity by regulating mitochondrial biogenesis and autophagy.Nat. Med. 2013; 19: 1039-1046https://doi.org/10.1038/nm.3213Crossref PubMed Scopus (271) Google Scholar). However, the role of CIPC in satellite cell function and skeletal muscle regeneration remains unknown. CIPC is a CLOCK-interacting protein identified through two-hybrid assays (Zhao et al., 2007Zhao W.N. Malinin N. Yang F.C. Staknis D. Gekakis N. Maier B. Reischl S. Kramer A. Weitz C.J. CIPC is a mammalian circadian clock protein without invertebrate homologues.Nat. Cell Biol. 2007; 9: 268-275https://doi.org/10.1038/ncb1539Crossref PubMed Scopus (61) Google Scholar). CIPC inhibits BMAL1-CLOCK transcriptional activity by binding to CLOCK, but not BMAL1. Cipc mRNA and protein expression exhibits circadian oscillations in a way similar to that of other negative-feedback clock genes, such as Pers and Crys. Like Pers and Crys, Cipc also contains an E-box element, suggesting that Cipc is a transcriptional target of CLOCK-BMAL1. Knockdown of endogenous Cipc shortens circadian-period length (Zhao et al., 2007Zhao W.N. Malinin N. Yang F.C. Staknis D. Gekakis N. Maier B. Reischl S. Kramer A. Weitz C.J. CIPC is a mammalian circadian clock protein without invertebrate homologues.Nat. Cell Biol. 2007; 9: 268-275https://doi.org/10.1038/ncb1539Crossref PubMed Scopus (61) Google Scholar). In addition to its role as a negative regulator of the circadian clock, CIPC also has other biological functions. Overexpression of Cipc in HEK293 cells inhibits cell proliferation through the inactivation of ERK (Matsunaga et al., 2016Matsunaga R. Nishino T. Yokoyama A. Nakashima A. Kikkawa U. Konishi H. Versatile function of the circadian protein CIPC as a regulator of Erk activation.Biochem. Biophys. Res. Commun. 2016; 469: 377-383https://doi.org/10.1016/j.bbrc.2015.11.117Crossref PubMed Scopus (2) Google Scholar). This study demonstrated that myopathy in mdx mice can be alleviated by deleting Cipc in satellite cells. Using a satellite-cell-specific Cipc knockout approach, we demonstrate that CIPC inhibits Pax7 expression and regenerative myogenesis. Our results show that deletion of Cipc in satellite cells activates ERK1/2 and JNK1/2 signaling pathways, leading to phosphorylation of the transcription factor SP1, which augments the gene expression of Pax7 and MyoD. To determine the role of CIPC in satellite cell function, we generated satellite-cell-specific Cipc knockout (Cipcscko) mice by crossing Cipcflox/flox mice with Pax7-Cre mice (Figure S1A), and the genotype was identified by PCR (Figure S1B). The efficiency of Cipc deletion in satellite cells was determined by qRT-PCR (Figure S1C). We then isolated primary myoblasts from 8-week-old wild-type (WT) mice and Cipcflox/flox and Cipcscko mice and investigated whether myoblasts of adult mice express CIPC by immunostaining and western blot. To verify the purity of primary myoblasts, we performed Pax7 staining, and the results showed that most cells were Pax7+ (Figures S2A and S2B). Confocal microscopy of myoblasts revealed co-localization of CIPC and Pax7 in primary myoblasts, and CIPC was expressed in the cytoplasm of myoblasts in WT and Cipcflox/flox mice but was not expressed in myoblasts from Cipcscko mice (Figure 1A ). Western blot results showed that CIPC was expressed in myoblasts of Cipcflox/flox mice, but not in myoblasts of Cipcscko mice (Figures 1B and 1C). Myoblasts undergo differentiation when cultured in the differentiation medium (DM) containing 2% horse serum. The differentiation is characterized by gradual reduction of Pax7 and increased myogenic markers, such as MyoD, myogenin, myosin heavy chain (MyHC) (Olguin and Olwin, 2004Olguin H.C. Olwin B.B. Pax-7 up-regulation inhibits myogenesis and cell cycle progression in satellite cells: a potential mechanism for self-renewal.Dev. Biol. 2004; 275: 375-388https://doi.org/10.1016/j.ydbio.2004.08.015Crossref PubMed Scopus (372) Google Scholar). To find out the relationship between CIPC and myogenic markers, we performed western blot using the cell lysates of WT myoblasts. We analyzed the levels of CIPC, Pax7, MyoD, and MyHC at different time points after switching the cells to DM. The results showed that CIPC, MyoD, and MyHC levels were increased during differentiation, whereas the level of Pax7 was decreased (Figures 1D and 1E). Immunostaining revealed that CIPC is expressed in the differentiated myotubes (Figure S2C). qRT-PCR also confirmed the inverse relationship between Pax7 and Cipc expression during differentiation (Figures S2D and S2E). To determine whether CIPC negatively regulates Pax7, we overexpressed Cipc in myoblasts and analyzed the expression of Pax7 by qRT-PCR. The results showed that Pax7 expression was reduced when Cipc was overexpressed (Figures S2F and S2G). Collectively, the results suggest that CIPC is negatively correlated with Pax7 expression. To assess the impact of Cipc deletion on muscle regeneration, BaCl2 solution was locally injected into the tibial anterior (TA) muscle of adult Cipcflox/flox and Cipcscko mice. The TA muscle was isolated at different time points after injury. Muscle weight was decreased in Cipcflox/flox after injury at day 5, while that of injured Cipcscko mice did not change significantly compared with uninjured mice (Figures S1D and S1E). H&E staining was performed to analyze muscle morphology and the numbers of centronucleated fibers (CNFs) myofibers. The number of newly formed CNFs and cross-sectional area (CSA) of CNFs was higher in TA muscle of Cipcscko mice than that of littermate Cipcflox/flox mice after BaCl2 injection (Figures 1F–1I). In Cipcscko mice, reconstitution of muscle structure was completed at day 10 post-injury, where muscle sections of the Cipcflox/flox mice or the Pax7-Cre mice still showed areas of regeneration (Figures 1F and S1F). These data indicate that the deletion of Cipc enhances muscle regeneration. The expression of the embryonic isoform of MyHC (eMyHC) is a hallmark of muscle regeneration (Addicks et al., 2019Addicks G.C. Brun C.E. Sincennes M.C. Saber J. Porter C.J. Francis Stewart A. Ernst P. Rudnicki M.A. MLL1 is required for PAX7 expression and satellite cell self-renewal in mice.Nat. Commun. 2019; 10: 4256https://doi.org/10.1038/s41467-019-12086-9Crossref PubMed Scopus (17) Google Scholar; Liu et al., 2020Liu Z. Zhang X. Lei H. Lam N. Carter S. Yockey O. Xu M. Mendoza A. Hernandez E.R. Wei J.S. et al.CASZ1 induces skeletal muscle and rhabdomyosarcoma differentiation through a feed-forward loop with MYOD and MYOG.Nat. Commun. 2020; 11: 911https://doi.org/10.1038/s41467-020-14684-4Crossref PubMed Scopus (13) Google Scholar; Schiaffino et al., 2015Schiaffino S. Rossi A.C. Smerdu V. Leinwand L.A. Reggiani C. Developmental myosins: expression patterns and functional significance.Skelet. Muscle. 2015; 5: 22https://doi.org/10.1186/s13395-015-0046-6Crossref PubMed Scopus (210) Google Scholar). Immunofluorescence staining showed that the number of eMyHC+ fibers was increased in injured muscles from Cipcscko mice compared with Cipcflox/flox mice up to 5 days post-injury. Then the number of eMyHC+ fibers gradually decreased as muscle regeneration neared completion (Figures S3A, S3B, and S3G). The CSA of eMyHC+ fibers followed a similar pattern (Figures S3A, S3C, and S3G). qRT-PCR confirmed that eMyHC mRNA (Myh3) expression in Cipcscko muscles was higher than that of Cipcflox/flox muscles (Figure S3D). Western blot analysis showed that the expression of eMyHC protein peaked on day 5 after injury in Cipcscko muscles. However, eMyHC protein did not reach the peak until day 7 in Cipcflox/flox muscles (Figures S3E and S3F). These findings suggest that deletion of Cipc in satellite cells promotes myogenic regeneration. The increased expression of eMyHC in the injured muscle of Cipcscko mice suggests that Cipc deletion led to an increased number of Pax7+ satellite cells, which subsequently differentiated and formed new myofibers. To determine whether CIPC affects satellite cell proliferation and quiescence, we performed immunofluorescence staining on muscle sections from uninjured or injured Cipcscko and Cipcflox/flox mice using antibodies against Pax7 and laminin (Figure 2A ). The number of Pax7+ cells was increased 2-fold in the uninjured muscles of Cipcscko mice compared with that of Cipcflox/flox mice (Figures 2A and 2B), and this increase was even more evident on day 5 post-injury (4-fold) (Figures 2A and 2C). By the end of the regeneration process at day 21 after injury, satellite cells in the TA muscle have returned to a quiescent state, and the number of Pax7+ cells in TA muscle of Cipcscko is still higher than that of Cipcflox/flox mice (Figures 2A and 2D). These results indicate that loss of Cipc increases the number of quiescent satellite cells in vivo and promotes their proliferation upon injury. To further investigate whether loss of Cipc affects the differentiation of satellite cells, we performed immunofluorescence staining on muscle sections from BaCl2-injured Cipcflox/flox mice and Cipcscko mice using antibodies against myogenin (Myog) and laminin (Figure 2E). On day 5 post-injury, Cipcscko mice exhibited a 3-fold increase in the numbers of myogenin-expressing cells compared with Cipcflox/flox mice (Figures 2E and 2F). The increased number of Myog+ cells could be a consequence of the increased number of satellite cells. Alternatively, it could suggest that loss of Cipc promotes differentiation. BaCl2 solution was locally injected into the TA muscles of adult WT and mdx mice to assess the expression level of Cipc during myogenesis. The mRNA levels of Cipc and Pax7 in TA muscle of WT and mdx mice after 0, 3, 5, 10, 15, and 21 days of injury were determined with qRT-PCR. Although the expression pattern of Pax7 is similar in the injured TA muscle of mdx mice, its expression level is significantly lower than WT mice at day 3 post-injury (Figure S4A). The mRNA levels of Cipc decreased at day 3 post-injury in TA muscles of WT mice compared with day 0. Then its expression level gradually increased and reached
最长约 10秒,即可获得该文献文件

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
张家小猫完成签到,获得积分20
刚刚
1秒前
胖一达完成签到 ,获得积分10
1秒前
2秒前
6秒前
王敬顺应助无念采纳,获得10
8秒前
wishe完成签到,获得积分10
9秒前
11秒前
11秒前
Lucas应助白白熊采纳,获得10
14秒前
哈拉斯完成签到,获得积分10
16秒前
无花果应助觉得就到家采纳,获得10
16秒前
17秒前
flxlei发布了新的文献求助200
18秒前
18秒前
18秒前
斯文败类应助调皮乐荷采纳,获得10
18秒前
fighting完成签到 ,获得积分10
18秒前
mmz完成签到,获得积分10
22秒前
动听的语堂完成签到,获得积分10
23秒前
pokemeow发布了新的文献求助10
24秒前
supcond发布了新的文献求助50
25秒前
充电宝应助加湿器采纳,获得20
27秒前
毛蛋爱吃汉堡包完成签到,获得积分10
28秒前
29秒前
舍得完成签到,获得积分10
29秒前
苏尔琳诺完成签到,获得积分10
30秒前
香蕉觅云应助moffy采纳,获得30
30秒前
38秒前
qphys完成签到,获得积分10
40秒前
41秒前
lizhiqian2024发布了新的文献求助10
43秒前
44秒前
moffy发布了新的文献求助30
45秒前
七羽完成签到 ,获得积分10
47秒前
ee发布了新的文献求助10
48秒前
凤梨完成签到,获得积分10
49秒前
很菜的研究生完成签到,获得积分10
49秒前
49秒前
刘丽忠发布了新的文献求助10
49秒前
高分求助中
One Man Talking: Selected Essays of Shao Xunmei, 1929–1939 1000
A Chronicle of Small Beer: The Memoirs of Nan Green 1000
From Rural China to the Ivy League: Reminiscences of Transformations in Modern Chinese History 900
Eric Dunning and the Sociology of Sport 850
QMS18Ed2 | process management. 2nd ed 800
Operative Techniques in Pediatric Orthopaedic Surgery 510
The Making of Détente: Eastern Europe and Western Europe in the Cold War, 1965-75 500
热门求助领域 (近24小时)
化学 医学 材料科学 生物 工程类 有机化学 生物化学 物理 内科学 纳米技术 计算机科学 化学工程 复合材料 基因 遗传学 物理化学 催化作用 免疫学 细胞生物学 电极
热门帖子
关注 科研通微信公众号,转发送积分 2914170
求助须知:如何正确求助?哪些是违规求助? 2551633
关于积分的说明 6904209
捐赠科研通 2214191
什么是DOI,文献DOI怎么找? 1176721
版权声明 588293
科研通“疑难数据库(出版商)”最低求助积分说明 576221