201
|
Dong W, Chen X, Wang M, Zheng Z, Zhang X, Xiao Q, Peng X. Mir-206 partially rescues myogenesis deficiency by inhibiting CUGBP1 accumulation in the cell models of myotonic dystrophy. Neurol Res 2018; 41:9-18. [PMID: 30281408 DOI: 10.1080/01616412.2018.1493963] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objectives: In this study, we aim to determine how CUG-expansion and the abundance of Celf1 regulates normal myocyte differentiation and reveal the role ofmiR-206 in myotonic dystrohy and explore a possible gene therapy vector. Methods: we set up CUG-expansion and Celf1 overexpression C2C12 cell models to imitate the myocyte differentiation defects of DM1, then transfected AdvmiR-206 into cell models, tested the level of myogenic markers MyoD, MyoG, Mef2c, Celf1 by RT-PCRand Western Blotting, detected myotube formation by myosin heavy chain immunostaining. Result: 3'-UTR CUG-expansion leads to myotube defects and impaired myoblasts differentiation. Overexpression of Celf1 inhibits myoblast differentiation and impairs differentiation. Knockdown of Celf1 partially rescues differentiation defects of myoblasts harboring CUG-expansion. miR-206 incompletely reverses myoblast differentiation inhibition induced by CUG-expansion and partially recuses myoblast differentiation defects induced by Celf1 overexpression. Conclusions: Ectopic miR-206 mimicking the endogenous temporal patterns specifically drives a myocyte program that boosts myoblast lineages, likely by promoting the expression of MyoD to rectify the myogenic deficiency by stimulating the accumulation of Celf1. Abbreviations: DMPK: (dystrophia myotonica protein kinase); 3'-UTR: (3'-untranslated region); MBNL1: (muscleblind-like [Drosophila]); DM1: (myotonic dystrophy type 1); GFP: (green fluorescent protein); RT-PCR: (quantitative reverse transcriptase-polymerase chain reaction); shRNA: (short hairpin RNA).
Collapse
Affiliation(s)
- Wei Dong
- a Department of Cardiology , The First Affiliated Hospital of Nanchang University , Nanchang , China.,b Hypertension Research Institute of Jiangxi , Nanchang , China
| | - Xuanying Chen
- a Department of Cardiology , The First Affiliated Hospital of Nanchang University , Nanchang , China.,b Hypertension Research Institute of Jiangxi , Nanchang , China
| | - Menghong Wang
- a Department of Cardiology , The First Affiliated Hospital of Nanchang University , Nanchang , China.,b Hypertension Research Institute of Jiangxi , Nanchang , China
| | - Zeqi Zheng
- a Department of Cardiology , The First Affiliated Hospital of Nanchang University , Nanchang , China.,b Hypertension Research Institute of Jiangxi , Nanchang , China
| | - Xing Zhang
- a Department of Cardiology , The First Affiliated Hospital of Nanchang University , Nanchang , China.,b Hypertension Research Institute of Jiangxi , Nanchang , China
| | - Qunlin Xiao
- a Department of Cardiology , The First Affiliated Hospital of Nanchang University , Nanchang , China.,b Hypertension Research Institute of Jiangxi , Nanchang , China
| | - Xiaoping Peng
- a Department of Cardiology , The First Affiliated Hospital of Nanchang University , Nanchang , China.,b Hypertension Research Institute of Jiangxi , Nanchang , China
| |
Collapse
|
202
|
Inflammation-associated miR-155 activates differentiation of muscular satellite cells. PLoS One 2018; 13:e0204860. [PMID: 30273359 PMCID: PMC6166968 DOI: 10.1371/journal.pone.0204860] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/14/2018] [Indexed: 11/25/2022] Open
Abstract
Tissue renewal and muscle regeneration largely rely on the proliferation and differentiation of muscle stem cells called muscular satellite cells (MuSCs). MuSCs are normally quiescent, but they are activated in response to various stimuli, such as inflammation. Activated MuSCs proliferate, migrate, differentiate, and fuse to form multinucleate myofibers. Meanwhile, inappropriate cues for MuSC activation induce premature differentiation and bring about stem cell loss. Recent studies revealed that stem cell regulation is disrupted in various aged tissues. We found that the expression of microRNA (miR)-155, which is an inflammation-associated miR, is upregulated in MuSCs of aged muscles, and this upregulation activates the differentiation process through suppression of C/ebpβ, which is an important molecule for maintaining MuSC self-renewal. We also found that Notch1 considerably repressed miR-155 expression, and loss of Notch1 induced miR-155 overexpression. Our findings suggest that miR-155 can act as an activator of muscular differentiation and might be responsible for accelerating aging-associated premature differentiation of MuSCs.
Collapse
|
203
|
Wang Y, Ma J, Qiu W, Zhang J, Feng S, Zhou X, Wang X, Jin L, Long K, Liu L, Xiao W, Tang Q, Zhu L, Jiang Y, Li X, Li M. Guanidinoacetic Acid Regulates Myogenic Differentiation and Muscle Growth Through miR-133a-3p and miR-1a-3p Co-mediated Akt/mTOR/S6K Signaling Pathway. Int J Mol Sci 2018; 19:ijms19092837. [PMID: 30235878 PMCID: PMC6163908 DOI: 10.3390/ijms19092837] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/14/2018] [Accepted: 09/17/2018] [Indexed: 12/12/2022] Open
Abstract
Guanidinoacetic acid (GAA), an amino acid derivative that is endogenous to animal tissues including muscle and nerve, has been reported to enhance muscular performance. MicroRNA (miRNA) is a post-transcriptional regulator that plays a key role in nutrient-mediated myogenesis. However, the effects of GAA on myogenic differentiation and skeletal muscle growth, and the potential regulatory mechanisms of miRNA in these processes have not been elucidated. In this study, we investigated the effects of GAA on proliferation, differentiation, and growth in C2C12 cells and mice. The results showed that GAA markedly inhibited the proliferation of myoblasts, along with the down-regulation of cyclin D1 (CCND1) and cyclin dependent kinase 4 (CDK4) mRNA expression, and the upregulation of cyclin dependent kinase inhibitor 1A (P21) mRNA expression. We also demonstrated that GAA treatment stimulated myogenic differentiation 1 (MyoD) and myogenin (MyoG) mRNA expression, resulting in an increase in the myotube fusion rate. Meanwhile, GAA supplementation promoted myotube growth through increase in total myosin heavy chain (MyHC) protein level, myotubes thickness and gastrocnemius muscle cross-sectional area. Furthermore, small RNA sequencing revealed that a total of eight miRNAs, including miR-133a-3p and miR-1a-3p cluster, showed differential expression after GAA supplementation. To further study the function of miR-133a-3p and miR-1a-3p in GAA-induced skeletal muscle growth, we transfected miR-133a-3p and miR-1a-3p mimics into myotube, which also induced muscle growth. Through bioinformatics and a dual-luciferase reporter system, the target genes of miR-133a-3p and miR-1a-3p were determined. These two miRNAs were shown to modulate the Akt/mTOR/S6K signaling pathway by restraining target gene expression. Taken together, these findings suggest that GAA supplementation can promote myoblast differentiation and skeletal muscle growth through miR-133a-3p- and miR-1a-3p-induced activation of the AKT/mTOR/S6K signaling pathway.
Collapse
Affiliation(s)
- Yujie Wang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Jideng Ma
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Wanling Qiu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Jinwei Zhang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Siyuan Feng
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Xiankun Zhou
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Xun Wang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Long Jin
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Keren Long
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Lingyan Liu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Weihang Xiao
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Qianzi Tang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Li Zhu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Yanzhi Jiang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Xuewei Li
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Mingzhou Li
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| |
Collapse
|
204
|
MiR‐499 regulates myoblast proliferation and differentiation by targeting transforming growth factor β receptor 1. J Cell Physiol 2018; 234:2523-2536. [DOI: 10.1002/jcp.26903] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 06/12/2018] [Indexed: 12/21/2022]
|
205
|
Pietraszek-Gremplewicz K, Kozakowska M, Bronisz-Budzynska I, Ciesla M, Mucha O, Podkalicka P, Madej M, Glowniak U, Szade K, Stepniewski J, Jez M, Andrysiak K, Bukowska-Strakova K, Kaminska A, Kostera-Pruszczyk A, Jozkowicz A, Loboda A, Dulak J. Heme Oxygenase-1 Influences Satellite Cells and Progression of Duchenne Muscular Dystrophy in Mice. Antioxid Redox Signal 2018; 29:128-148. [PMID: 29669436 DOI: 10.1089/ars.2017.7435] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS Muscle damage in Duchenne muscular dystrophy (DMD) caused by the lack of dystrophin is strongly linked to inflammation. Heme oxygenase-1 (HO-1; Hmox1) is an anti-inflammatory and cytoprotective enzyme affecting myoblast differentiation by inhibiting myomiRs. The role of HO-1 has not been so far well addressed in DMD. RESULTS In dystrophin-deficient mdx mice, expression of Hmox1 in limb skeletal muscles and diaphragm is higher than in wild-type animals, being consistently elevated from 8 up to 52 weeks, both in myofibers and inflammatory leukocytes. Accordingly, HO-1 expression is induced in muscles of DMD patients. Pharmacological inhibition of HO-1 activity or genetic ablation of Hmox1 aggravates muscle damage and inflammation in mdx mice. Double knockout animals (Hmox1-/-mdx) demonstrate impaired exercise capacity in comparison with mdx mice. Interestingly, in contrast to the effect observed in muscle fibers, in dystrophin-deficient muscle satellite cells (SCs) expression of Hmox1 is decreased, while MyoD, myogenin, and miR-206 are upregulated compared with wild-type counterparts. Mdx SCs demonstrate disturbed and enhanced differentiation, which is further intensified by Hmox1 deficiency. RNA sequencing revealed downregulation of Atf3, MafK, Foxo1, and Klf2 transcription factors, known to activate Hmox1 expression, as well as attenuation of nitric oxide-mediated cGMP-dependent signaling in mdx SCs. Accordingly, treatment with NO-donor induces Hmox1 expression and inhibits differentiation. Finally, differentiation of mdx SCs was normalized by CO, a product of HO-1 activity. Innovation and Conclusions: HO-1 is induced in DMD, and HO-1 inhibition aggravates DMD pathology. Therefore, HO-1 can be considered a therapeutic target to alleviate this disease. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Katarzyna Pietraszek-Gremplewicz
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Magdalena Kozakowska
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Iwona Bronisz-Budzynska
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Maciej Ciesla
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Olga Mucha
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Paulina Podkalicka
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Magdalena Madej
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Urszula Glowniak
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Krzysztof Szade
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Jacek Stepniewski
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Mateusz Jez
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Kalina Andrysiak
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Karolina Bukowska-Strakova
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland .,2 Department of Clinical Immunology and Transplantology, Institute of Paediatrics, Medical College, Jagiellonian University , Krakow, Poland
| | - Anna Kaminska
- 3 Department of Neurology, Medical University of Warsaw , Warsaw, Poland
| | | | - Alicja Jozkowicz
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Agnieszka Loboda
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| | - Jozef Dulak
- 1 Faculty of Biochemistry, Biophysics and Biotechnology, Department of Medical Biotechnology, Jagiellonian University , Krakow, Poland
| |
Collapse
|
206
|
Abreu P. Bioenergetics mechanisms regulating muscle stem cell self-renewal commitment and function. Biomed Pharmacother 2018; 103:463-472. [DOI: 10.1016/j.biopha.2018.04.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 11/25/2022] Open
|
207
|
Scully D, Naseem KM, Matsakas A. Platelet biology in regenerative medicine of skeletal muscle. Acta Physiol (Oxf) 2018; 223:e13071. [PMID: 29633517 DOI: 10.1111/apha.13071] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/07/2018] [Accepted: 04/01/2018] [Indexed: 12/13/2022]
Abstract
Platelet-based applications such as platelet-rich plasma (PRP) and platelet releasate have gained unprecedented attention in regenerative medicine across a variety of tissues as of late. The rationale behind utilizing PRP originates in the delivery of key cytokines and growth factors from α-granules to the targeted area, which in turn act as cell cycle regulators and promote the healing process across a variety of tissues. The aim of the present review is to assimilate current experimental evidence on the role of platelets as biomaterials in tissue regeneration, particularly in skeletal muscle, by integrating findings from human, animal and cell studies. This review is composed of 3 parts: firstly, we review key aspects of platelet biology that precede the preparation and use of platelet-related applications for tissue regeneration. Secondly, we critically discuss relevant evidence on platelet-mediated regeneration in skeletal muscle focusing on findings from (i) clinical trials, (ii) experimental animal studies and (iii) cell culture studies; and thirdly, we discuss the application of platelets in the regeneration of several other tissues including tendon, bone, liver, vessels and nerve. Finally, we review key technical variations in platelet preparation that may account for the large discrepancy in outcomes from different studies. This review provides an up-to-date reference tool for biomedical and clinical scientists involved in platelet-mediated tissue regenerative applications.
Collapse
Affiliation(s)
- D. Scully
- Molecular Physiology Laboratory; Centre for Atherothrombotic & Metabolic Disease; Hull York Medical School; University of Hull; Hull UK
| | - K. M. Naseem
- Leeds Institute of Cardiovascular and Metabolic Medicine; University of Leeds; Leeds UK
| | - A. Matsakas
- Molecular Physiology Laboratory; Centre for Atherothrombotic & Metabolic Disease; Hull York Medical School; University of Hull; Hull UK
| |
Collapse
|
208
|
mir-127-3p inhibits the proliferation of myocytes by targeting KMT5a. Biochem Biophys Res Commun 2018; 503:970-976. [PMID: 29932923 DOI: 10.1016/j.bbrc.2018.06.104] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 06/19/2018] [Indexed: 01/06/2023]
Abstract
MicroRNAs are a class of highly conserved ∼20 nucleotides non-coding RNAs that post-transcriptionally regulate gene expression. Many miRNAs were studied in the development of skeletal muscle, such as miR-1, miR-206, and miR-133. In our previous study, miR-127-3p was found highly expressed in porcine fetal skeletal muscle, whereas the detailed functions of miR-127-3p in muscle development is still unclear. In this study, we detected that miR-127-3p also highly expressed in skeletal muscle, cardiac muscle of adult mice and proliferative C2C12 cell lines. Overexpression of miR-127-3p almost has no effects on differentiation of C2C12 cell lines. However, miR-127-3p significantly inhibited the cell proliferation of C2C12 cells. Moreover, we identified KMT5a as a target gene that was down-regulated in both mRNA and protein level when miR-127-3p mimics were introduced. Furthermore, KMT5a overexpression in miR-127-3p treated cells rescued the influence of miR-127-3p on C2C12 proliferation. In brief, our data reveals that miR-127-3p regulates the proliferation of myocytes through KMT5a.
Collapse
|
209
|
Bulaklak K, Xiao B, Qiao C, Li J, Patel T, Jin Q, Li J, Xiao X. MicroRNA-206 Downregulation Improves Therapeutic Gene Expression and Motor Function in mdx Mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:283-293. [PMID: 30195767 PMCID: PMC6011022 DOI: 10.1016/j.omtn.2018.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 12/17/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscle-wasting disorder caused by a mutation in the dystrophin gene. Numerous gene therapies have been developed to replace or repair the defective dystrophin gene; however, these treatments cannot restore the full-length protein or completely resolve dystrophic symptoms. Secondary pathological mechanisms, such as functional ischemia and fibrosis, are thought to exacerbate the primary defect and cause the profound muscle degeneration found in dystrophic muscle. Surrogate therapies utilizing alternative therapeutic genes, or “booster genes,” such as VEGFA and utrophin, seek to address these secondary mechanisms and have shown impressive benefit in mdx mice. A skeletal muscle-specific microRNA, miR-206, is particularly overexpressed in dystrophic muscle and inhibits the expression of known booster genes. Thus, we aimed to determine if miR-206 contributes to dystrophic pathology by repressing beneficial gene expression. Here, we show that AAV-mediated expression of a miR-206 decoy target effectively downregulated miR-206 expression and increased endogenous therapeutic gene expression in mature mdx muscle. Furthermore, treatment significantly improved motor function and dystrophic pathology in mdx mice. In summary, we have identified a contributing factor to the dystrophic phenotype and characterized a novel therapeutic avenue for DMD.
Collapse
Affiliation(s)
- Karen Bulaklak
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bin Xiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chunping Qiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jianbin Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tejash Patel
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Quan Jin
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Juan Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiao Xiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
210
|
Effects of microRNAs on skeletal muscle development. Gene 2018; 668:107-113. [PMID: 29775754 DOI: 10.1016/j.gene.2018.05.039] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/04/2018] [Accepted: 05/13/2018] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are small (about 22 nucleotides) noncoding RNAs, which were highly conserved among mammals. They have ushered in a new era in molecular biology over twenty years. They can negatively regulate gene expression at the posttranscriptional level through the principle of complementary base pairing with the 3' untranslated region (UTR) of their target mRNAs and induce their degradation. They involve in tissue morphogenesis, cellular processes like apoptosis, and major signaling pathways. Previous studies have promoted our understanding that miRNAs play an important role in myogenesis and have a big impact on muscle mass, muscle fiber type and muscle diseases. Many researchers have provided evidence of the involvement of muscle-specific and enriched miRNAs in the individual stages of skeletal muscle development as well as of their significant influence on muscle metabolism during quiescence, proliferation, differentiation and regeneration. Here, we focus on the microRNAs that related to the development of skeletal muscle. For example, some microRNAs are upregulated in differentiated skeletal muscle and can promote differentiation, like, miR-1, miR-24, miR-26a, miR-181 and miR-206. However, some microRNAs highly expressed in proliferating myoblasts, downregulated in differentiated and could inhibit differentiation, like MiR-221 and miR-222. Some others not only promote skeletal muscle proliferation, but also promote differentiation, like miR-214. Studying the miRNAs' regulatory mechanisms in skeletal development will help us know more about the knowledge of miRNAs in muscle developmental biology and make us learn more about involved signal pathway.
Collapse
|
211
|
Forcina L, Miano C, Musarò A. The physiopathologic interplay between stem cells and tissue niche in muscle regeneration and the role of IL-6 on muscle homeostasis and diseases. Cytokine Growth Factor Rev 2018; 41:1-9. [PMID: 29778303 DOI: 10.1016/j.cytogfr.2018.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022]
Abstract
Skeletal muscle is a complex, dynamic tissue characterized by an elevated plasticity. Although the adult muscle is mainly composed of multinucleated fibers with post mitotic nuclei, it retains a remarkable ability to regenerate in response to traumatic events. The regenerative potential of the adult skeletal muscle relies in the activity of satellite cells, mononucleated cells residing within the muscle in intimate association with myofibers. Satellite cells normally remain quiescent in their sublaminar position, sporadically entering the cell cycle to guarantee an efficient cellular turnover, by fusing with pre-existing myofibers, and to maintain the stem cell pool. However, after muscle injury satellite cells undergo an extensive increase of their activity in response to environmental stimuli, thereby participating to the regeneration of a functional muscle tissue. Nevertheless, regeneration is affected in several pathologic conditions and by a wide range of environmental signals that are highly variable, not only through time, but also depending on the physiological or pathological conditions of the musculature. Among these factors, the interleukin-6 (IL-6) plays a critical physiopathologic role on muscle homeostasis and diseases. The basis of muscle regeneration and the impact of IL-6 on the physiopathology of skeletal muscle will be discussed.
Collapse
Affiliation(s)
- Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy
| | - Carmen Miano
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy.
| |
Collapse
|
212
|
Guilbaud M, Gentil C, Peccate C, Gargaun E, Holtzmann I, Gruszczynski C, Falcone S, Mamchaoui K, Ben Yaou R, Leturcq F, Jeanson-Leh L, Piétri-Rouxel F. miR-708-5p and miR-34c-5p are involved in nNOS regulation in dystrophic context. Skelet Muscle 2018; 8:15. [PMID: 29703249 PMCID: PMC5924477 DOI: 10.1186/s13395-018-0161-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/04/2018] [Indexed: 12/30/2022] Open
Abstract
Background Duchenne (DMD) and Becker (BMD) muscular dystrophies are caused by mutations in the DMD gene coding for dystrophin, a protein being part of a large sarcolemmal protein scaffold that includes the neuronal nitric oxide synthase (nNOS). The nNOS was shown to play critical roles in a variety of muscle functions and alterations of its expression and location in dystrophic muscle fiber leads to an increase of the muscle fatigability. We previously revealed a decrease of nNOS expression in BMD patients all presenting a deletion of exons 45 to 55 in the DMD gene (BMDd45-55), impacting the nNOS binding site of dystrophin. Since several studies showed deregulation of microRNAs (miRNAs) in dystrophinopathies, we focused on miRNAs that could target nNOS in dystrophic context. Methods By a screening of 617 miRNAs in BMDd45-55 muscular biopsies using TLDA and an in silico study to determine which one could target nNOS, we selected four miRNAs. In order to select those that targeted a sequence of 3′UTR of NOS1, we performed luciferase gene reporter assay in HEK393T cells. Finally, expression of candidate miRNAs was modulated in control and DMD human myoblasts (DMDd45-52) to study their ability to target nNOS. Results TLDA assay and the in silico study allowed us to select four miRNAs overexpressed in muscle biopsies of BMDd45-55 compared to controls. Among them, only the overexpression of miR-31, miR-708, and miR-34c led to a decrease of luciferase activity in an NOS1-3′UTR-luciferase assay, confirming their interaction with the NOS1-3′UTR. The effect of these three miRNAs was investigated on control and DMDd45-52 myoblasts. First, we showed a decrease of nNOS expression when miR-708 or miR-34c were overexpressed in control myoblasts. We then confirmed that DMDd45-52 cells displayed an endogenous increased of miR-31, miR-708, and miR-34c and a decreased of nNOS expression, the same characteristics observed in BMDd45-55 biopsies. In DMDd45-52 cells, we demonstrated that the inhibition of miR-708 and miR-34c increased nNOS expression, confirming that both miRNAs can modulate nNOS expression in human myoblasts. Conclusion These results strongly suggest that miR-708 and miR-34c, overexpressed in dystrophic context, are new actors involved in the regulation of nNOS expression in dystrophic muscle. Electronic supplementary material The online version of this article (10.1186/s13395-018-0161-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marine Guilbaud
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Christel Gentil
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Cécile Peccate
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Elena Gargaun
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Isabelle Holtzmann
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Carole Gruszczynski
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Sestina Falcone
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Kamel Mamchaoui
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Rabah Ben Yaou
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France.,AP-HP, Centre de Référence Maladies Neuromusculaire Nord, Est, Ile-de-France, G.H. Pitié-Salpêtrière, F-75013, Paris, France
| | - France Leturcq
- Laboratoire de Génétique et Biologie Moléculaire, Hôpital Cochin, Paris, France
| | | | - France Piétri-Rouxel
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France.
| |
Collapse
|
213
|
Chodkowska KA, Ciecierska A, Majchrzak K, Ostaszewski P, Sadkowski T. Effect of β-hydroxy-β-methylbutyrate on miRNA expression in differentiating equine satellite cells exposed to hydrogen peroxide. GENES AND NUTRITION 2018; 13:10. [PMID: 29662554 PMCID: PMC5892041 DOI: 10.1186/s12263-018-0598-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 03/14/2018] [Indexed: 12/25/2022]
Abstract
Background Skeletal muscle injury activates satellite cells to initiate processes of proliferation, differentiation, and hypertrophy in order to regenerate muscle fibers. The number of microRNAs and their target genes are engaged in satellite cell activation. β-Hydroxy-β-methylbutyrate (HMB) is known to prevent exercise-induced muscle damage. The purpose of this study was to evaluate the effect of HMB on miRNA and relevant target gene expression in differentiating equine satellite cells exposed to H2O2. We hypothesized that HMB may regulate satellite cell activity, proliferation, and differentiation, hence attenuate the pathological processes induced during an in vitro model of H2O2-related injury by changing the expression of miRNAs. Methods Equine satellite cells (ESC) were isolated from the samples of skeletal muscle collected from young horses. ESC were treated with HMB (24 h) and then exposed to H2O2 (1 h). For the microRNA and gene expression assessment microarrays, technique was used. Identified miRNAs and genes were validated using real-time qPCR. Cell viability, oxidative stress, and cell damage were measured using colorimetric method and flow cytometry. Results Analysis of miRNA and gene profile in differentiating ESC pre-incubated with HMB and then exposed to H2O2 revealed difference in the expression of 27 miRNAs and 4740 genes, of which 344 were potential target genes for identified miRNAs. Special attention was focused on differentially expressed miRNAs and their target genes involved in processes related to skeletal muscle injury. Western blot analysis showed protein protection in HMB-pre-treated group compared to control. The viability test confirmed that HMB enhanced cell survival after the hydrogen peroxide exposition. Conclusions Our results suggest that ESC pre-incubated with HMB and exposed to H2O2 could affect expression on miRNA levels responsible for skeletal muscle development, cell proliferation and differentiation, and activation of tissue repair after injury. Enrichment analyses for targeted genes revealed that a large group of genes was associated with the regulation of signaling pathways crucial for muscle tissue development, protein metabolism, muscle injury, and regeneration, as well as with oxidative stress response.
Collapse
Affiliation(s)
- Karolina A Chodkowska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Anna Ciecierska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Kinga Majchrzak
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Piotr Ostaszewski
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Tomasz Sadkowski
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| |
Collapse
|
214
|
Gan M, Du J, Shen L, Yang D, Jiang A, Li Q, Jiang Y, Tang G, Li M, Wang J, Li X, Zhang S, Zhu L. miR-152 regulates the proliferation and differentiation of C2C12 myoblasts by targeting E2F3. In Vitro Cell Dev Biol Anim 2018; 54:304-310. [PMID: 29508126 DOI: 10.1007/s11626-017-0219-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 11/27/2017] [Indexed: 01/19/2023]
Abstract
The development of skeletal muscle is a complex process involving the proliferation, differentiation, apoptosis, and changing of muscle fiber types in myoblasts. Many reports have described the involvement of microRNAs in the myogenesis of myoblasts. In this study, we found that the expression of miR-152 was gradually down-regulated during myoblast proliferation, but gradually up-regulated during the differentiation of myoblasts. Transfection with miR-152 mimics restrained cell proliferation and decreased the expression levels of cyclin E, CDK4, and cyclin D1, but promoted myotube formation and significantly increased the mRNA expression levels of MyHC, MyoD, MRF4, and MyoG in C2C12 myoblasts. However, treatment with miR-152 inhibitors promoted cell proliferation and restrained differentiation. Moreover, over-expression of miR-152 significantly decreased E2F3 production in C2C12 myoblasts. A luciferase assay confirmed that miR-152 could bind to the 3' UTR of E2F3. In conclusion, this study showed that miR-152 inhibited proliferation and promoted myoblast differentiation by targeting E2F3.
Collapse
Affiliation(s)
- Mailin Gan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jingjing Du
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Linyuan Shen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Dongli Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Anan Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Qiang Li
- Sichuan Province General Station of Animal Husbandry, Chengdu, 611130, China
| | - Yanzhi Jiang
- College of Life and Science, Sichuan Agricultural University, Chengdu, 611130, China
| | - Guoqing Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Mingzhou Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jinyong Wang
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
| | - Xuewei Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Shunhua Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Li Zhu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
215
|
Inhibition of the JNK/MAPK signaling pathway by myogenesis-associated miRNAs is required for skeletal muscle development. Cell Death Differ 2018; 25:1581-1597. [PMID: 29449644 PMCID: PMC6143622 DOI: 10.1038/s41418-018-0063-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 12/24/2017] [Accepted: 01/04/2018] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle differentiation is controlled by multiple cell signaling pathways, however, the JNK/MAPK signaling pathway dominating this process has not been fully elucidated. Here, we report that the JNK/MAPK pathway was significantly downregulated in the late stages of myogenesis, and in contrast to P38/MAPK pathway, it negatively regulated skeletal muscle differentiation. Based on the PAR-CLIP-seq analysis, we identified six elevated miRNAs (miR-1a-3p, miR-133a-3p, miR-133b-3p, miR-206-3p, miR-128-3p, miR-351-5p), namely myogenesis-associated miRNAs (mamiRs), negatively controlled the JNK/MAPK pathway by repressing multiple factors for the phosphorylation of the JNK/MAPK pathway, including MEKK1, MEKK2, MKK7, and c-Jun but not JNK protein itself, and as a result, expression of transcriptional factor MyoD and mamiRs were further promoted. Our study revealed a novel double-negative feedback regulatory pattern of cell-specific miRNAs by targeting phosphorylation kinase signaling cascade responsible for skeletal muscle development.
Collapse
|
216
|
Zhao Y, Cao F, Yu X, Chen C, Meng J, Zhong R, Zhang Y, Zhu D. Linc-RAM is required for FGF2 function in regulating myogenic cell differentiation. RNA Biol 2018; 15:404-412. [PMID: 29364044 DOI: 10.1080/15476286.2018.1431494] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Myogenic differentiation of skeletal muscle stem cells, also known satellite cells, is tightly orchestrated by extrinsic and intrinsic regulators. Basic fibroblast growth factor (FGF2) is well documented to be implicated in satellite cell self-renewal and differentiation by repressing MyoD. We recently identified a MyoD-regulated and skeletal muscle-specifically expressed long non-coding RNA Linc-RAM which enhances myogenic differentiation by facilitating MyoD/Baf60c/Brg1 complex assembly. Herein, we investigated the transcriptional regulation and intracellular signaling pathway in mediating Linc-RAM gene expression during muscle cell differentiation. Firstly, we demonstrate Linc-RAM is negatively regulated by FGF2 via Ras/Raf/Mek/Erk signaling pathway in muscle cells. Overexpression of MyoD significantly attenuates repression of Linc-RAM promoter activities in C2C12 cells treated with FGF2. Knockout of MyoD abolishes FGF2-mediated repression of Linc-RAM gene transcription in satellite cells sorted from skeletal muscle of MyoD-/-;Pax7-nGFP mice, suggesting inhibition of MyoD is required for FGF2-mediated expression of Linc-RAM. For the functional significance, we show that overexpression of Linc-RAM rescues FGF2-induced inhibition of C2C12 cell differentiation, indicating inhibition of Linc-RAM is required for FGF2-mediated suppression of myogenic differentiation. Consistently, we are able to further corroborate the requirement of Linc-RAM inhibition for FGF2-modulated repression of myogenic differentiation by using an ex vivo cultured single fiber system and satellite cells sorted from Linc-RAM-/-;Pax7-nGFP knockout mice. Collectively, the present study not only reveals the intracellular signaling in FGF2-mediated Linc-RAM gene expression but also demonstrate the functional significance of Linc-RAM in FGF2-mediated muscle cell differentiation.
Collapse
Affiliation(s)
- Yixia Zhao
- a The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College , 5 Dong Dan San Tiao, Beijing , China
| | - Fengqi Cao
- a The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College , 5 Dong Dan San Tiao, Beijing , China
| | - Xiaohua Yu
- a The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College , 5 Dong Dan San Tiao, Beijing , China
| | - Chuyan Chen
- b Peking Union Medical College Hospital , Shuaifuyuan No.1, Beijing , China
| | - Jiao Meng
- a The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College , 5 Dong Dan San Tiao, Beijing , China
| | - Ran Zhong
- a The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College , 5 Dong Dan San Tiao, Beijing , China
| | - Yong Zhang
- a The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College , 5 Dong Dan San Tiao, Beijing , China
| | - Dahai Zhu
- a The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College , 5 Dong Dan San Tiao, Beijing , China
| |
Collapse
|
217
|
Vertii A, Kaufman PD, Hehnly H, Doxsey S. New dimensions of asymmetric division in vertebrates. Cytoskeleton (Hoboken) 2018; 75:87-102. [DOI: 10.1002/cm.21434] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/20/2017] [Accepted: 01/16/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Anastassiia Vertii
- Department of MolecularCell and Cancer Biology University of Massachusetts Medical SchoolWorcester Massachusetts
| | - Paul D. Kaufman
- Department of MolecularCell and Cancer Biology University of Massachusetts Medical SchoolWorcester Massachusetts
| | - Heidi Hehnly
- Department of Cell and Developmental BiologySUNY Upstate Medical UniversitySyracuse New York13210
| | - Stephen Doxsey
- Program in Molecular Medicine University of Massachusetts Medical SchoolWorcester Massachusetts
| |
Collapse
|
218
|
Bem J, Grabowska I, Daniszewski M, Zawada D, Czerwinska AM, Bugajski L, Piwocka K, Fogtman A, Ciemerych MA. Transient MicroRNA Expression Enhances Myogenic Potential of Mouse Embryonic Stem Cells. Stem Cells 2018; 36:655-670. [PMID: 29314416 DOI: 10.1002/stem.2772] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 12/07/2017] [Accepted: 12/27/2017] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) are known regulators of various cellular processes, including pluripotency and differentiation of embryonic stem cells (ESCs). We analyzed differentiation of two ESC lines-D3 and B8, and observed significant differences in the expression of miRNAs and genes involved in pluripotency and differentiation. We also examined if transient miRNA overexpression could serve as a sufficient impulse modulating differentiation of mouse ESCs. ESCs were transfected with miRNA Mimics and differentiated in embryoid bodies and embryoid body outgrowths. miRNAs involved in differentiation of mesodermal lineages, such as miR145 and miR181, as well as miRNAs regulating myogenesis (MyomiRs)-miR1, miR133a, miR133b, and miR206 were tested. Using such approach, we proved that transient overexpression of molecules selected by us modulated differentiation of mouse ESCs. Increase in miR145 levels upregulated Pax3, Pax7, Myod1, Myog, and MyHC2, while miR181 triggered the expression of such crucial myogenic factors as Myf5 and MyHC2. As a result, the ability of ESCs to initiate myogenic differentiation and form myotubes was enhanced. Premature expression of MyomiRs had, however, an adverse effect on myogenic differentiation of ESCs. Stem Cells 2018;36:655-670.
Collapse
Affiliation(s)
- Joanna Bem
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Maciej Daniszewski
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Dorota Zawada
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Areta M Czerwinska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Lukasz Bugajski
- Laboratory of Cytometry, Nencki Institute of Experimental Biology
| | | | - Anna Fogtman
- Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Maria A Ciemerych
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| |
Collapse
|
219
|
|
220
|
Rovira M, Arrey G, Planas JV. Exercise-Induced Hypertrophic and Oxidative Signaling Pathways and Myokine Expression in Fast Muscle of Adult Zebrafish. Front Physiol 2017; 8:1063. [PMID: 29326600 PMCID: PMC5741866 DOI: 10.3389/fphys.2017.01063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/04/2017] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle is a plastic tissue that undergoes cellular and metabolic adaptations under conditions of increased contractile activity such as exercise. Using adult zebrafish as an exercise model, we previously demonstrated that swimming training stimulates hypertrophy and vascularization of fast muscle fibers, consistent with the known muscle growth-promoting effects of exercise and with the resulting increased aerobic capacity of this tissue. Here we investigated the potential involvement of factors and signaling mechanisms that could be responsible for exercise-induced fast muscle remodeling in adult zebrafish. By subjecting zebrafish to swimming-induced exercise, we observed an increase in the activity of mammalian target of rapamycin (mTOR) and Mef2 protein levels in fast muscle. We also observed an increase in the protein levels of the mitotic marker phosphorylated histone H3 that correlated with an increase in the protein expression levels of Pax7, a satellite-like cell marker. Furthermore, the activity of AMP-activated protein kinase (AMPK) was also increased by exercise, in parallel with an increase in the mRNA expression levels of pgc1α and also of pparda, a β-oxidation marker. Changes in the mRNA expression levels of slow and fast myosin markers further supported the notion of an exercise-induced aerobic phenotype in zebrafish fast muscle. The mRNA expression levels of il6, il6r, apln, aplnra and aplnrb, sparc, decorin and igf1, myokines known in mammals to be produced in response to exercise and to signal through mTOR/AMPK pathways, among others, were increased in fast muscle of exercised zebrafish. These results support the notion that exercise increases skeletal muscle growth and myogenesis in adult zebrafish through the coordinated activation of the mTOR-MEF2 and AMPK-PGC1α signaling pathways. These results, coupled with altered expression of markers for oxidative metabolism and fast-to-slow fiber-type switch, also suggest improved aerobic capacity as a result of swimming-induced exercise. Finally, the induction of myokine expression by swimming-induced exercise support the hypothesis that these myokines may have been produced and secreted by the exercised zebrafish muscle and acted on fast muscle cells to promote metabolic remodeling. These results support the use of zebrafish as a suitable model for studies on muscle remodeling in vertebrates, including humans.
Collapse
Affiliation(s)
- Mireia Rovira
- Departament de Biologia Cel·lular, Facultat de Biologia, Fisiologia i Immunologia, Universitat de Barcelona, Barcelona, Spain
| | - Gerard Arrey
- Departament de Biologia Cel·lular, Facultat de Biologia, Fisiologia i Immunologia, Universitat de Barcelona, Barcelona, Spain
| | - Josep V Planas
- Departament de Biologia Cel·lular, Facultat de Biologia, Fisiologia i Immunologia, Universitat de Barcelona, Barcelona, Spain.,Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
221
|
Yalvac ME, Amornvit J, Braganza C, Chen L, Hussain SRA, Shontz KM, Montgomery CL, Flanigan KM, Lewis S, Sahenk Z. Impaired regeneration in calpain-3 null muscle is associated with perturbations in mTORC1 signaling and defective mitochondrial biogenesis. Skelet Muscle 2017; 7:27. [PMID: 29241457 PMCID: PMC5731057 DOI: 10.1186/s13395-017-0146-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/06/2017] [Indexed: 12/25/2022] Open
Abstract
Background Previous studies in patients with limb-girdle muscular dystrophy type 2A (LGMD2A) have suggested that calpain-3 (CAPN3) mutations result in aberrant regeneration in muscle. Methods To gain insight into pathogenesis of aberrant muscle regeneration in LGMD2A, we used a paradigm of cardiotoxin (CTX)-induced cycles of muscle necrosis and regeneration in the CAPN3-KO mice to simulate the early features of the dystrophic process in LGMD2A. The temporal evolution of the regeneration process was followed by assessing the oxidative state, size, and the number of metabolic fiber types at 4 and 12 weeks after last CTX injection. Muscles isolated at these time points were further investigated for the key regulators of the pathways involved in various cellular processes such as protein synthesis, cellular energy status, metabolism, and cell stress to include Akt/mTORC1 signaling, mitochondrial biogenesis, and AMPK signaling. TGF-β and microRNA (miR-1, miR-206, miR-133a) regulation were also assessed. Additional studies included in vitro assays for quantifying fusion index of myoblasts from CAPN3-KO mice and development of an in vivo gene therapy paradigm for restoration of impaired regeneration using the adeno-associated virus vector carrying CAPN3 gene in the muscle. Results At 4 and 12 weeks after last CTX injection, we found impaired regeneration in CAPN3-KO muscle characterized by excessive numbers of small lobulated fibers belonging to oxidative metabolic type (slow twitch) and increased connective tissue. TGF-β transcription levels in the regenerating CAPN3-KO muscles were significantly increased along with microRNA dysregulation compared to wild type (WT), and the attenuated radial growth of muscle fibers was accompanied by perturbed Akt/mTORC1 signaling, uncoupled from protein synthesis, through activation of AMPK pathway, thought to be triggered by energy shortage in the CAPN3-KO muscle. This was associated with failure to increase mitochondria content, PGC-1α, and ATP5D transcripts in the regenerating CAPN3-KO muscles compared to WT. In vitro studies showed defective myotube fusion in CAPN3-KO myoblast cultures. Replacement of CAPN3 by gene therapy in vivo increased the fiber size and decreased the number of small oxidative fibers. Conclusion Our findings provide insights into understanding of the impaired radial growth phase of regeneration in calpainopathy. Electronic supplementary material The online version of this article (10.1186/s13395-017-0146-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mehmet E Yalvac
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Jakkrit Amornvit
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Current Address: King Chulalongkorn Memorial Hospital and Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Cilwyn Braganza
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Lei Chen
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Syed-Rehan A Hussain
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Kimberly M Shontz
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Chrystal L Montgomery
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Kevin M Flanigan
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics and Neurology, Nationwide Children's Hospital and The Ohio State University, Columbus, USA
| | - Sarah Lewis
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Zarife Sahenk
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA. .,Department of Pediatrics and Neurology, Nationwide Children's Hospital and The Ohio State University, Columbus, USA. .,Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH, USA. .,Neuromuscular Pathology, Nationwide Children's Hospital, 700 Children's Drive Rm WA 3024, Columbus, USA.
| |
Collapse
|
222
|
Qiang J, Bao JW, Li HX, Chen DJ, He J, Tao YF, Xu P. miR-1338-5p Modulates Growth Hormone Secretion and Glucose Utilization by Regulating ghitm in Genetically Improved Farmed Tilapia (GIFT, Oreochromis niloticus). Front Physiol 2017; 8:998. [PMID: 29270127 PMCID: PMC5723647 DOI: 10.3389/fphys.2017.00998] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/20/2017] [Indexed: 11/13/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenous, non-coding small RNA molecules about 22 nt in length, which could regulate the expressions of target genes and participate in growth and development of organisms. Genetically improved farmed tilapia (GIFT, Oreochromis niloticus) is an important economic freshwater species in China and the growth performance is one of the main breeding indicators. Growth hormone inducible transmembrane protein (ghitm) plays an important role in growth and development of both mammals and invertebrates; however, little studies have been reported on fish. Our previous experiments indicated that miR-1338-5p expression may be negatively correlated with ghitm expression. In this study, we firstly used qRT-PCR and northern blot to verify the expression of miR-1338-5p and ghitm, and determined the binding site of miR-1338-5p in the ghitm 3'-untranslated region (UTR) by luciferase reporter assay. Secondly, juveniles GIFT injected with miR-1338-5p antagomir were used to analyze the regulatory function of the miR-1338-5p-ghitm pair in vivo. The results showed that the ghitm 3'-UTR was complementary to the 5' 2-8-nt site of miR-1338-5p. Inhibition of miR-1338-5p promoted ghitm expression in the pituitary and liver of GIFT. ghitm could interfere in the growth hormone (Gh)-growth hormone receptor (Ghr)-insulin-like growth factor (Igf) signaling pathway by competing with the ghr1 for combination with Gh, and then reduce the growth of GIFT. Moreover, the reduction of Gh in serum may regulate insulin secretion and result in the increasing sugar and fat storage in serum and liver. Our results suggest that miR-1338-5p participates in the growth and development of GIFT through the regulation of ghitm, which provides theoretical support for the study of the fish growth mechanism.
Collapse
Affiliation(s)
- Jun Qiang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Jing Wen Bao
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Hong Xia Li
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - De Ju Chen
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Jie He
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Yi Fan Tao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Pao Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| |
Collapse
|
223
|
MicroRNA-95 promotes myogenic differentiation by down-regulation of aminoacyl-tRNA synthase complex-interacting multifunctional protein 2. Oncotarget 2017; 8:111356-111368. [PMID: 29340059 PMCID: PMC5762327 DOI: 10.18632/oncotarget.22796] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/16/2017] [Indexed: 12/15/2022] Open
Abstract
MicroRNA-95 (miR-95) is well known for its ability to promote the proliferation of a variety of cancer cells, but its function in skeletal muscle development has not been reported so far. Our laboratory has recently generated genetically engineered Meishan pigs containing a loss-of-function myostatin (MSTN) mutant (MSTN-/-). These MSTN-/- pigs grow and develop normally but show clear double muscle phenotype as observed in Belgian cattle. We observed that the expression of miR-95 was up-regulated in the longissimus dorsi from MSTN-/- Meishan pigs at day 65 during embryo development. In this study, we investigated the role of miR-95 in the myogenic differentiation using a murine myoblast cell line C2C12. Our results revealed that miR-95 may play a very important role in regulating the expression of myogenic differentiation marker genes myosin heavy chain (MHC) and myogenin. By use of bioinformatical analysis and luciferase reporter gene assay, aminoacyl-tRNA synthase complex-interacting multifunctional protein 2 (AIMP2) gene was identified as a miR-95 target gene involved in myogenic differentiation. Our results indicated that higher miR-95 expression level leads to lower level of AIMP2 protein expression. When the endogenous expression of AIMP2 is inhibited by siRNA, the expression levels of myogenic differentiation marker genes MHC and myogenin increased, implying that AIMP2 negatively regulates myogenic differentiation. Taken together, it is likely that miR-95 promotes myogenic differentiation in C2C12 myoblasts and may play a positive functional role in skeletal muscle development by down regulating the expression of AIMP2 at protein level.
Collapse
|
224
|
Freire PP, Cury SS, de Oliveira G, Fernandez GJ, Moraes LN, da Silva Duran BO, Ferreira JH, Fuziwara CS, Kimura ET, Dal-Pai-Silva M, Carvalho RF. Osteoglycin inhibition by microRNA miR-155 impairs myogenesis. PLoS One 2017; 12:e0188464. [PMID: 29161332 PMCID: PMC5697837 DOI: 10.1371/journal.pone.0188464] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 11/07/2017] [Indexed: 01/22/2023] Open
Abstract
Skeletal myogenesis is a regulated process in which mononucleated cells, the myoblasts, undergo proliferation and differentiation. Upon differentiation, the cells align with each other, and subsequently fuse to form terminally differentiated multinucleated myotubes. Previous reports have identified the protein osteoglycin (Ogn) as an important component of the skeletal muscle secretome, which is expressed differentially during muscle development. However, the posttranscriptional regulation of Ogn by microRNAs during myogenesis is unknown. Bioinformatic analysis showed that miR-155 potentially targeted the Ogn transcript at the 3´-untranslated region (3´ UTR). In this study, we tested the hypothesis that miR-155 inhibits the expression of the Ogn to regulate skeletal myogenesis. C2C12 myoblast cells were cultured and miR-155 overexpression or Ogn knockdown was induced by transfection with miR-155 mimic, siRNA-Ogn, and negative controls with lipofectamine for 15 hours. Near confluence (80–90%), myoblasts were induced to differentiate myotubes in a differentiation medium. Luciferase assay was used to confirm the interaction between miR-155 and Ogn 3’UTR. RT-qPCR and Western blot analyses were used to confirm that the differential expression of miR-155 correlates with the differential expression of myogenic molecular markers (Myh2, MyoD, and MyoG) and inhibits Ogn protein and gene expression in myoblasts and myotubes. Myoblast migration and proliferation were assessed using Wound Healing and MTT assays. Our results show that miR-155 interacts with the 3’UTR Ogn region and decrease the levels of Ogn in myotubes. The overexpression of miR-155 increased MyoG expression, decreased myoblasts wound closure rate, and decreased Myh2 expression in myotubes. Moreover, Ogn knockdown reduced the expression levels of MyoD, MyoG, and Myh2 in myotubes. These results reveal a novel pathway in which miR-155 inhibits Ogn expression to regulate proliferation and differentiation of C2C12 myoblast cells.
Collapse
Affiliation(s)
- Paula Paccielli Freire
- Department of Morphology, Institute of Biosciences, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Sarah Santiloni Cury
- Department of Morphology, Institute of Biosciences, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Grasieli de Oliveira
- Department of Morphology, Institute of Biosciences, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Geysson Javier Fernandez
- Department of Morphology, Institute of Biosciences, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Leonardo Nazario Moraes
- Department of Morphology, Institute of Biosciences, São Paulo State University, Botucatu, São Paulo, Brazil
| | | | - Juarez Henrique Ferreira
- Department of Morphology, Institute of Biosciences, São Paulo State University, Botucatu, São Paulo, Brazil
| | - César Seigi Fuziwara
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Edna Teruko Kimura
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Maeli Dal-Pai-Silva
- Department of Morphology, Institute of Biosciences, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Robson Francisco Carvalho
- Department of Morphology, Institute of Biosciences, São Paulo State University, Botucatu, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
225
|
Lack of Heme Oxygenase-1 Induces Inflammatory Reaction and Proliferation of Muscle Satellite Cells after Cardiotoxin-Induced Skeletal Muscle Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:491-506. [PMID: 29169990 DOI: 10.1016/j.ajpath.2017.10.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 10/12/2017] [Accepted: 10/23/2017] [Indexed: 12/29/2022]
Abstract
Heme oxygenase-1 (HO-1, Hmox1) regulates viability, proliferation, and differentiation of many cell types; hence, it may affect regeneration of injured skeletal muscle. Here, we injected cardiotoxin into gastrocnemius muscle of Hmox1+/+ and Hmox1-/- animals and analyzed cellular response after muscle injury, focusing on muscle satellite cells (SCs), inflammatory reaction, fibrosis, and formation of new blood vessels. HO-1 is strongly induced after muscle injury, being expressed mostly in the infiltrating leukocytes (CD45+ cells), including macrophages (F4/80+ cells). Lack of HO-1 augments skeletal muscle injury, evidenced by increased creatinine kinase and lactate dehydrogenase, as well as expression of monocyte chemoattractant protein-1, IL-6, IL-1β, and insulin-like growth factor-1. This, together with disturbed proportion of M1/M2 macrophages, accompanied by enhanced formation of arterioles, may be responsible for shift of Hmox1-/- myofiber size distribution toward larger one. Importantly, HO-1-deficient SCs are prone to activation and have higher proliferation on injury. This effect can be partially mimicked by stimulation of Hmox1+/+ SCs with monocyte chemoattractant protein-1, IL-6, IL-1β, and is associated with increased MyoD expression, suggesting that Hmox1-/- SCs are shifted toward more differentiated myogenic population. However, multiple rounds of degeneration/regeneration in conditions of HO-1 deficiency may lead to exhaustion of SC pool, and the number of SCs is decreased in old Hmox1-/- mice. In summary, HO-1 modulates muscle repair mechanisms preventing its uncontrolled acceleration.
Collapse
|
226
|
Mok GF, Lozano-Velasco E, Münsterberg A. microRNAs in skeletal muscle development. Semin Cell Dev Biol 2017; 72:67-76. [PMID: 29102719 DOI: 10.1016/j.semcdb.2017.10.032] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/24/2017] [Accepted: 10/27/2017] [Indexed: 12/21/2022]
Abstract
A fundamental process during both embryo development and stem cell differentiation is the control of cell lineage determination. In developing skeletal muscle, many of the diffusible signaling molecules, transcription factors and more recently non-coding RNAs that contribute to this process have been identified. This has facilitated advances in our understanding of the molecular mechanisms underlying the control of cell fate choice. Here we will review the role of non-coding RNAs, in particular microRNAs (miRNAs), in embryonic muscle development and differentiation, and in satellite cells of adult muscle, which are essential for muscle growth and regeneration. Some of these short post-transcriptional regulators of gene expression are restricted to skeletal muscle, but their expression can also be more widespread. In addition, we discuss a few examples of long non-coding RNAs, which are numerous but much less well understood.
Collapse
Affiliation(s)
- Gi Fay Mok
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Estefania Lozano-Velasco
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Andrea Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| |
Collapse
|
227
|
Segersvärd H, Lakkisto P, Hänninen M, Forsten H, Siren J, Immonen K, Kosonen R, Sarparanta M, Laine M, Tikkanen I. Carbon monoxide releasing molecule improves structural and functional cardiac recovery after myocardial injury. Eur J Pharmacol 2017; 818:57-66. [PMID: 29055786 DOI: 10.1016/j.ejphar.2017.10.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 10/15/2017] [Accepted: 10/17/2017] [Indexed: 01/17/2023]
Abstract
Carbon monoxide (CO), produced by heme oxygenase-1 (HO-1), is an endogenous paracrine factor involved in the regulation of cardiovascular structure and function. We studied the effects of a synthetic CO releasing molecule (CORM-3) on cardiac recovery and myocardial microRNA expression after myocardial infarction (MI). Male Wistar rats with MI (n = 75) or sham-operated controls (n = 75) were treated from day 4 to day 14 after MI either with synthetic CORM-3 or with inactive iCORM and killed 2, 4 or 8 weeks post-MI. Infarct size, vascular and capillary densities, the amount of cardiomyocytes in the infarct area, and cardiomyocyte proliferation and apoptosis were determined. PCR was used for microRNA and mRNA quantification, western blotting to evaluate protein expression and echocardiography to assess cardiac structure and function. CORM-3 treatment increased vascular density (P< 0.05 vs. iCORM) and the proportion of cardiomyocytes (P< 0.05 vs. iCORM) in the infarct area. Ejection fraction improved (P< 0.05) and left ventricular volumes decreased (P< 0.05) in CORM-3 treated MI groups compared to iCORM treatment. CORM-3 treatment decreased the amount of proliferating Ki67 positive cardiomyocytes in the infarct/border area at week 2 after MI compared to iCORM treatment, whereas the amount of apoptotic cardiomyocytes did not differ between CORM-3 and iCORM groups. Compared to iCORM treatment, CORM-3 decreased expression on miR-206 in the remote area at week 2 after MI. The CO releasing molecule CORM-3 improved structural and functional cardiac recovery after MI. Modulation of HO-1-CO axis may prove novel drug targets to facilitate cardiac recovery after myocardial injury.
Collapse
Affiliation(s)
- Heli Segersvärd
- Minerva Institute for Medical Research, Biomedicum 2U Helsinki, Finland.
| | - Päivi Lakkisto
- Minerva Institute for Medical Research, Biomedicum 2U Helsinki, Finland; Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko Hänninen
- Minerva Institute for Medical Research, Biomedicum 2U Helsinki, Finland
| | - Hanna Forsten
- Minerva Institute for Medical Research, Biomedicum 2U Helsinki, Finland
| | - Juuso Siren
- Minerva Institute for Medical Research, Biomedicum 2U Helsinki, Finland
| | - Katariina Immonen
- Minerva Institute for Medical Research, Biomedicum 2U Helsinki, Finland
| | - Riikka Kosonen
- Minerva Institute for Medical Research, Biomedicum 2U Helsinki, Finland
| | | | - Mika Laine
- Minerva Institute for Medical Research, Biomedicum 2U Helsinki, Finland; Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ilkka Tikkanen
- Minerva Institute for Medical Research, Biomedicum 2U Helsinki, Finland; Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
228
|
Coenen-Stass AML, Wood MJA, Roberts TC. Biomarker Potential of Extracellular miRNAs in Duchenne Muscular Dystrophy. Trends Mol Med 2017; 23:989-1001. [PMID: 28988850 DOI: 10.1016/j.molmed.2017.09.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 12/12/2022]
Abstract
miRNAs are small, noncoding RNAs that not only regulate gene expression within cells, but might also constitute promising extracellular biomarkers for a variety of pathologies, including the progressive muscle-wasting disorder Duchenne Muscular Dystrophy (DMD). A set of muscle-enriched miRNAs, the myomiRs (miR-1, miR-133, and miR-206) are highly elevated in the serum of patients with DMD and in dystrophin-deficient animal models. Furthermore, circulating myomiRs might be used as pharmacodynamic biomarkers, given that their levels can be restored towards wild-type levels following exon skipping therapy in dystrophic mice. The relationship between muscle pathology and extracellular myomiR release is complex, and incompletely understood. Here, we discuss current progress leading towards the clinical utility of extracellular miRNAs as putative DMD biomarkers, and their possible contribution to muscle physiology.
Collapse
Affiliation(s)
- Anna M L Coenen-Stass
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK; Institute of Neurology, Sobell Department of Motor Neuroscience and Movement Disorders, University College London, London, Queen Square, London, WC1N 3BG, UK
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK.
| | - Thomas C Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK; Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program, La Jolla, CA 92037, USA.
| |
Collapse
|
229
|
Translational Control of the Myogenic Program in Developing, Regenerating, and Diseased Skeletal Muscle. Curr Top Dev Biol 2017; 126:67-98. [PMID: 29305004 DOI: 10.1016/bs.ctdb.2017.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Translational control of genes that code for protein allows a cell to rapidly respond to changes in its environment, in part because translational control of gene expression does not depend on upstream events required to produce an mRNA molecule. The importance of translational control has been highlighted by studies concerning muscle development, regeneration, and disease. Translational control of specific mRNAs is achieved by microRNAs and RNA-binding proteins, which are particularly relevant to developmental myogenesis, where they ensure the stepwise differentiation of multipotent progenitors to committed myogenic progenitors that ultimately fuse into slow- or fast-type myofibers that make up skeletal muscle. The importance of translational control is also illustrated in muscle disease, where deregulated microRNA expression accelerates or delays progression of disease. Skeletal muscle is also unique for its remarkable capacity to regenerate after injury, which requires the activity of quiescent muscle stem cells, named satellite cells for their position underneath the basal lamina of the myofiber. Mitotically quiescent satellite cells are primed to activate the cell cycle and myogenic program, a unique feature that requires specific regulation of mRNA translation converging with pathways that regulate global protein synthesis. Emerging concepts in translational control of gene expression have shed light on multiple layers of control over the myogenic program. In parallel, the development and regeneration of skeletal muscle represents a unique, relevant, and highly defined context within which new concepts in translational control of gene expression should emerge.
Collapse
|
230
|
Sun H, Cai S, Zhang M, Zhao J, Wei S, Luo Y, Meng X, Zhou X, Li Y, Zhang W. MicroRNA-206 regulates vascular smooth muscle cell phenotypic switch and vascular neointimal formation. Cell Biol Int 2017; 41:739-748. [PMID: 28328152 DOI: 10.1002/cbin.10768] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/19/2017] [Indexed: 11/09/2022]
Abstract
MiR-206 has been found to play a critical role in skeletal muscle proliferation, differentiation, and regeneration. However, little is known about the function of miR-206 in vascular smooth muscle cells (VSMCs) biology. In this study, we will investigate its roles in phenotypic switching of VSMCs and neointimal lesion formation. First, we identified the expression of miR-206 in VSMCs treated with various concentrations of TGFβ1 and in rat carotid arteries after angioplasty by using qPCR. TGFβ1 inhibited the expression of miR-206 and TGFβ1 inhibitor induced miR-206 expression. In VSMCs of injured vascular walls, miR-206 expression was upregulated. Then, we overexpressed miR-206 using lentivirus Lv-rno-mir-206 and knocked down miR-206 using LV-rno-mir-206-inhibitor in rat carotid arteries after angioplasty. Overexpression of miR-206 resulted in decreasing SM22α expression in VSMCs in vitro and knockdown of miR-206 suppressed neointimal lesion formation in vivo. Finally, ZFP580 (zinc finger protein 580) was identified as the direct target of miR-206 in VSMCs by using luciferase report assay. The results indicate that miR-206 is involved in phenotypic switching of VSMCs and neointimal lesion formation after angioplasty through targeting ZFP580. These findings may provide a novel therapeutic target in post-angioplasty restenosis.
Collapse
Affiliation(s)
- Huiyan Sun
- Department of Physiology and Pathophysiology, Logistics University of Chinese People's Armed Police Force, Huizhihuan Road 1, Dongli District, Tianjin, 300309, China
| | - Songzhi Cai
- Department of Cardiology, Affiliated Hospital, Logistics University of Chinese People's Armed Police Force, Chenglin Road 220, Dongli District, Tianjin, 300162, China
| | - Mei Zhang
- Department of Cardiology, Affiliated Hospital, Logistics University of Chinese People's Armed Police Force, Chenglin Road 220, Dongli District, Tianjin, 300162, China
| | - Juan Zhao
- Department of Physiology and Pathophysiology, Logistics University of Chinese People's Armed Police Force, Huizhihuan Road 1, Dongli District, Tianjin, 300309, China
| | - Shuping Wei
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Chenglin Road 220, Dongli District, Tianjin, 300162, China
| | - Yuyu Luo
- Department of Physiology and Pathophysiology, Logistics University of Chinese People's Armed Police Force, Huizhihuan Road 1, Dongli District, Tianjin, 300309, China
| | - Xiangyan Meng
- Department of Physiology and Pathophysiology, Logistics University of Chinese People's Armed Police Force, Huizhihuan Road 1, Dongli District, Tianjin, 300309, China
| | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Chenglin Road 220, Dongli District, Tianjin, 300162, China
| | - Yuming Li
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Chenglin Road 220, Dongli District, Tianjin, 300162, China
| | - Wencheng Zhang
- Department of Physiology and Pathophysiology, Logistics University of Chinese People's Armed Police Force, Huizhihuan Road 1, Dongli District, Tianjin, 300309, China.,Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Chenglin Road 220, Dongli District, Tianjin, 300162, China
| |
Collapse
|
231
|
Simon L, Ford SM, Song K, Berner P, Vande Stouwe C, Nelson S, Bagby GJ, Molina PE. Decreased myoblast differentiation in chronic binge alcohol-administered simian immunodeficiency virus-infected male macaques: role of decreased miR-206. Am J Physiol Regul Integr Comp Physiol 2017; 313:R240-R250. [PMID: 28637658 PMCID: PMC5625276 DOI: 10.1152/ajpregu.00146.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/07/2017] [Accepted: 06/21/2017] [Indexed: 01/05/2023]
Abstract
Skeletal muscle stem cells play a critical role in regeneration of myofibers. We previously demonstrated that chronic binge alcohol (CBA) markedly attenuates myoblast differentiation potential and myogenic gene expression. Muscle-specific microRNAs (miRs) are implicated in regulation of myogenic genes. The aim of this study was to determine whether myoblasts isolated from asymptomatic CBA-administered simian immunodeficiency virus (SIV)-infected macaques treated with antiretroviral therapy (ART) showed similar impairments and, if so, to elucidate potential underlying mechanisms. Myoblasts were isolated from muscle at 11 mo after SIV infection from CBA/SIV macaques and from time-matched sucrose (SUC)-treated SIV-infected (SUC/SIV) animals and age-matched controls. Myoblast differentiation and myogenic gene expression were significantly decreased in myoblasts from SUC/SIV and CBA/SIV animals compared with controls. SIV and CBA decreased muscle-specific miR-206 in plasma and muscle and SIV decreased miR-206 expression in myoblasts, with no statistically significant changes in other muscle-specific miRs. These findings were associated with a significant increase in histone deacetylase 4 (HDAC4) and decrease in myogenic enhancer factor 2C (MEF2C) expression in CBA/SIV muscle. Transfection with miR-206 inhibitor decreased myotube differentiation, increased expression of HDAC4, and decreased MEF2C, suggesting a critical role of miR-206 in myogenesis. Moreover, HDAC4 was confirmed to be a direct miR-206 target. These results support a mechanistic role for decreased miR-206 in suppression of myoblast differentiation resulting from chronic alcohol and SIV infection. The parallel changes in skeletal muscle and circulating levels of miR-206 warrant studies to establish the possible use of plasma miR-206 as an indicator of impaired muscle function.
Collapse
Affiliation(s)
- L Simon
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana;
- Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
| | - S M Ford
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - K Song
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - P Berner
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - C Vande Stouwe
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - S Nelson
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - G J Bagby
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - P E Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
| |
Collapse
|
232
|
Latimer M, Sabin N, Le Cam A, Seiliez I, Biga P, Gabillard JC. miR-210 expression is associated with methionine-induced differentiation of trout satellite cells. J Exp Biol 2017; 220:2932-2938. [PMID: 28576820 PMCID: PMC6514451 DOI: 10.1242/jeb.154484] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/30/2017] [Indexed: 01/20/2023]
Abstract
In fish, data on microRNAs (miRNAs) involved in myogenesis are scarce. In order to identify miRNAs involved in satellite cell differentiation, we used a methionine depletion/replenishment protocol to synchronize myogenic cell differentiation. Our results validated that methionine removal (72 h) from the medium strongly decreased myoD1 and myogenin expression, indicating differentiation arrest. In contrast, methionine replenishment rescued expression of myoD1 and myogenin, showing a resumption of differentiation. We performed a miRNA array analysis of myogenic cells under three conditions: presence of methionine for 72 h (control), absence of methionine for 72 h (Meth-) and absence of methionine for 48 h followed by 24 h of methionine replenishment (Meth-/+). A clustering analysis identified three clusters: cluster I corresponds to miRNA upregulated only in Meth-/+ conditions; cluster II corresponds to miRNA downregulated only in Meth-/+ conditions; cluster III corresponds to miRNAs with high expression in control, low expression in Meth- conditions and intermediate expression after methionine replenishment (Meth-/+). Cluster III was very interesting because it fitted with the data obtained for myoD1 and myogenin (supporting an involvement in differentiation) and contained seven miRNAs with muscle-related function (e.g. miR-133a) and one (miR-210) with unknown function. Based on our previously published miRNA repertoire ( Juanchich et al., 2016), we confirmed miR-133a was expressed only in white muscle and showed that miR-210 had strong expression in white muscle. We also showed that miR-210 expression was upregulated during differentiation of satellite cells, suggesting that miR-210 was potentially involved in the differentiation of satellite cells.
Collapse
Affiliation(s)
- Mary Latimer
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nathalie Sabin
- INRA, UR1037 Laboratoire de Physiologie et Génomique des Poissons, 35000 Rennes, France
| | - Aurélie Le Cam
- INRA, UR1037 Laboratoire de Physiologie et Génomique des Poissons, 35000 Rennes, France
| | - Iban Seiliez
- INRA-UPPA, UMR1419 Nutrition Métabolisme Aquaculture, F-64310 St-Pée-sur-Nivelle, France
| | - Peggy Biga
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
233
|
Abstract
Exosome-dependent intercellular communication is an emerging signaling mechanism involved in tissue repair and regeneration. Now in this issue of Cell Stem Cell, Fry et al. (2016) show that muscle stem cells communicate with fibrogenic cells by exosomal trafficking of microRNA-206 to regulate extracellular matrix deposition and muscle tissue remodeling.
Collapse
|
234
|
Velleman SG, Song Y. Development and Growth of the Avian Pectoralis Major (Breast) Muscle: Function of Syndecan-4 and Glypican-1 in Adult Myoblast Proliferation and Differentiation. Front Physiol 2017; 8:577. [PMID: 28848451 PMCID: PMC5550705 DOI: 10.3389/fphys.2017.00577] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/25/2017] [Indexed: 12/19/2022] Open
Abstract
Muscle fiber number is determined around the time hatch with continued posthatch muscle growth being mediated by the adult myoblast, satellite cell, population of cells. Satellite cells are dynamic in their expression of proteins including the cell membrane associated proteoglycans, syndecan-4 and glypican-1. These proteoglycans play roles in organizing the extracellular environment in the satellite cell niche, cytoskeletal structure, cell-to-cell adhesion, satellite cell migration, and signal transduction. This review article focuses on syndecan-4 and glypican-1 as both are capable of regulating satellite cell responsiveness to fibroblast growth factor 2. Fibroblast growth factor 2 is a potent stimulator of muscle cell proliferation and a strong inhibitor of differentiation. Proteoglycans are composed of a central core protein defined functional domains, and covalently attached glycosaminoglycans and N-glycosylation chains. The functional association of these components with satellite cell function is discussed as well as an emerging role for microRNA regulation of syndecan-4 and glypican-1.
Collapse
Affiliation(s)
- Sandra G Velleman
- Department of Animal Sciences, The Ohio State UniversityWooster, OH, United States
| | - Yan Song
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical SchoolBoston, MA, United States
| |
Collapse
|
235
|
von Haehling S, Ebner N, Dos Santos MR, Springer J, Anker SD. Role of microRNAs in wasting in heart failure. Nat Rev Cardiol 2017; 14:566. [PMID: 28770867 DOI: 10.1038/nrcardio.2017.123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
| | - Nicole Ebner
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
| | - Marcelo R Dos Santos
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, D-37075 Göttingen, Germany.,Heart Institute (InCor), University of Sao Paulo Medical School, Dr. Arnaldo Avenue, 455 Cerqueira César, 01246903 Sao Paulo, Brazil
| | - Jochen Springer
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
| | - Stefan D Anker
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, D-37075 Göttingen, Germany.,Division of Cardiology and Metabolism: Heart Failure, Cachexia and Sarcopenia, Department of Internal Medicine and Cardiology, Brandenburg Center for Regenerative Therapies, Charité Medical School, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
236
|
D’Souza RF, Markworth JF, Aasen KMM, Zeng N, Cameron-Smith D, Mitchell CJ. Acute resistance exercise modulates microRNA expression profiles: Combined tissue and circulatory targeted analyses. PLoS One 2017; 12:e0181594. [PMID: 28750051 PMCID: PMC5531502 DOI: 10.1371/journal.pone.0181594] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/03/2017] [Indexed: 12/17/2022] Open
Abstract
A subset of short non-coding RNAs, microRNAs (miRs), have been identified in the regulation of skeletal muscle hypertrophy and atrophy. Expressed within cells, miRs are also present in circulation (c-miR) and have a putative role in cross-tissue signalling. The aim of this study was to examine the impact of a single bout of high intensity resistance exercise (RE) on skeletal muscle and circulatory miRs harvested simultaneously. Resistance trained males (n = 9, 24.6 ± 4.9 years) undertook a single bout of high volume RE with venous blood and muscle biopsies collected before, 2 and 4hr post-exercise. Real time polymerase chain reaction (Rt-PCR) analyses was performed on 30 miRs that have previously been shown to be required for skeletal muscle function. Of these, 6 miRs were significantly altered within muscle following exercise; miR-23a, -133a, -146a, -206, -378b and 486. Analysis of these same miRs in circulation demonstrated minimal alterations with exercise, although c-miR-133a (~4 fold, p = 0.049) and c-miR-149 (~2.4 fold; p = 0.006) were increased 4hr post-exercise. Thus a single bout of RE results in the increased abundance of a subset of miRs within the skeletal muscle, which was not evident in plasma. The lack a qualitative agreement in the response pattern of intramuscular and circulating miR expression suggests the analysis of circulatory miRs is not reflective of the miR responses within skeletal muscle after exercise.
Collapse
Affiliation(s)
| | - James F. Markworth
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- Department of Orthopedic Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
| | | | - Nina Zeng
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | | | - Cameron J. Mitchell
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- * E-mail:
| |
Collapse
|
237
|
Margolis LM, Rivas DA, Pasiakos SM, McClung JP, Ceglia L, Fielding RA. Upregulation of circulating myomiR following short-term energy restriction is inversely associated with whole body protein synthesis. Am J Physiol Regul Integr Comp Physiol 2017; 313:R298-R304. [PMID: 28659285 DOI: 10.1152/ajpregu.00054.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/22/2017] [Accepted: 06/26/2017] [Indexed: 01/05/2023]
Abstract
The objective of the present investigation was to determine whether energy restriction (ER) influences expression of skeletal muscle-specific microRNA (miRNA) in circulation (c-myomiR) and whether changes in c-myomiR are associated with rates of whole body protein synthesis. Sixteen older (64 ± 2 yr) overweight (28.5 ± 1.2 kg/m2) men enrolled in this 35-day controlled feeding trial. A 7-day weight maintenance (WM) period was followed by 28 days of 30% ER. Whole body protein turnover was determined from [15N]glycine enrichments in 24-h urine collections, and c-myomiR (miR-1-3p, miR-133a-3p, miR-133b, and miR-206) expression was assessed from serum samples by RT-quantitative PCR upon completion of the WM and ER periods. Participants lost 4.4 ± 0.3 kg body mass during ER (P < 0.05). After 28 days of ER, miR-133a and miR-133b expression was upregulated (P < 0.05) compared with WM. When all four c-myomiR were grouped as c-myomiR score (sum of the median fold change of all myomiR), overall expression of c-myomiR was higher (P < 0.05) at ER than WM. Backward linear regression analysis of whole body protein synthesis and breakdown and carbohydrate, fat, and protein oxidation determined protein synthesis to be the strongest predictor of c-myomiR score. An inverse association (P < 0.05) was observed with ER c-myomiR score and whole body protein synthesis (r = -0.729, r2 = -0.530). Findings from the present investigation provide evidence that upregulation of c-myomiR expression profiles in response to short-term ER is associated with lower rates of whole body protein synthesis.
Collapse
Affiliation(s)
- Lee M Margolis
- Nutrition, Exercise, Physiology, and Sarcopenia Laboratory, United States Department of Agriculture Jean Mayer Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts
| | - Donato A Rivas
- Nutrition, Exercise, Physiology, and Sarcopenia Laboratory, United States Department of Agriculture Jean Mayer Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts
| | - Stefan M Pasiakos
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts
| | - James P McClung
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts
| | - Lisa Ceglia
- Bone Metabolism Laboratory, United States Department of Agriculture Jean Mayer Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts; and.,Division of Endocrinology, Diabetes, and Metabolism, Tufts Medical Center, Boston, Massachusetts
| | - Roger A Fielding
- Nutrition, Exercise, Physiology, and Sarcopenia Laboratory, United States Department of Agriculture Jean Mayer Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts;
| |
Collapse
|
238
|
Margolis LM, Dawson-Hughes B, Rivas DA, Ezzyat Y, Fielding RA, Ceglia L. Effects of Potassium Bicarbonate Supplements on Circulating microRNA Expression. J Endocr Soc 2017; 1:1015-1026. [PMID: 29264553 PMCID: PMC5686674 DOI: 10.1210/js.2017-00106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/08/2017] [Indexed: 01/31/2023] Open
Abstract
Several studies suggest that neutralizing acid load in the diet with alkali had favorable effects on intermediate markers of musculoskeletal health. We examined whether alkali supplementation with potassium bicarbonate [(KHCO3); 81 mmol/d; n = 12] vs placebo (n = 12) for 84 days altered serum microRNAs, potential biomarkers associated with innumerable biological processes including bone and muscle metabolism. Serum microRNAs, urinary net acid excretion (UNAE), urinary N-telopeptide (UNTX), urinary calcium (UCa), urinary nitrogen (UN), glomerular filtration rate, serum procollagen type 1 amino-terminal propeptide (P1NP), serum insulin-like growth factor-1 (IGF-1), and its serum binding protein IGFBP3 were measured at baseline and day 84. Baseline characteristics and measurements were similar in the two treatment groups. Eighty-four-day changes in UNAE differed by group (KHCO3, -47 ± 9 mmol; placebo, -5 ± 5 mmol; P < 0.01). KHCO3 significantly reduced UNTX, UCa, and serum P1NP but did not affect UN, serum IGF-1, or IGFBP3 levels compared with placebo over 84 days. Fold change in serum circulating microRNA (c-miR)-133b differed significantly by group (KHCO3, 2.26 ± 0.85; placebo, -1.23 ± 0.69; P < 0.01); there was a similar trend in c-miR-21-5p. Fold changes in c-miR-133b and c-miR-21-5p were inversely associated with changes in UNAE and UNTX; fold change in c-miR-21-5p was inversely associated with change in UCa, with a similar trend with c-miR-133b. In summary, reducing renal acid load with KHCO3 was associated with increased expressions of c-miR-133b and c-miR-21-5p. Furthermore, increases in c-miRNA-133b and c-miR-21-5p were inversely associated with bone resorption markers UNTX and UCa consistent with potential beneficial effects on bone in older adults. However, the broader significance of c-miRNAs as musculoskeletal biomarkers is still under investigation, and larger studies are needed to verify these preliminary results.
Collapse
Affiliation(s)
- Lee M Margolis
- Nutrition, Exercise, Physiology, and Sarcopenia Laboratory, Jean Mayer Human Nutrition Research Center on Aging, US Department of Agriculture, Tufts University, Boston, Massachusetts 02111
| | - Bess Dawson-Hughes
- Bone Metabolism Laboratory, Jean Mayer Human Nutrition Research Center on Aging, US Department of Agriculture, Tufts University, Boston, Massachusetts 02111
| | - Donato A Rivas
- Nutrition, Exercise, Physiology, and Sarcopenia Laboratory, Jean Mayer Human Nutrition Research Center on Aging, US Department of Agriculture, Tufts University, Boston, Massachusetts 02111
| | - Yassine Ezzyat
- Nutrition, Exercise, Physiology, and Sarcopenia Laboratory, Jean Mayer Human Nutrition Research Center on Aging, US Department of Agriculture, Tufts University, Boston, Massachusetts 02111
| | - Roger A Fielding
- Nutrition, Exercise, Physiology, and Sarcopenia Laboratory, Jean Mayer Human Nutrition Research Center on Aging, US Department of Agriculture, Tufts University, Boston, Massachusetts 02111
| | - Lisa Ceglia
- Bone Metabolism Laboratory, Jean Mayer Human Nutrition Research Center on Aging, US Department of Agriculture, Tufts University, Boston, Massachusetts 02111
| |
Collapse
|
239
|
Schroeder M, Jakovcevski M, Polacheck T, Lebow M, Drori Y, Engel M, Ben-Dor S, Chen A. A Methyl-Balanced Diet Prevents CRF-Induced Prenatal Stress-Triggered Predisposition to Binge Eating-like Phenotype. Cell Metab 2017; 25:1269-1281.e6. [PMID: 28576418 DOI: 10.1016/j.cmet.2017.05.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 01/30/2017] [Accepted: 05/08/2017] [Indexed: 12/31/2022]
Abstract
Binge eating (BE) is a common aberrant form of eating behavior, characterized by overconsumption of food in a brief period of time. Recurrent episodes of BE constitute the BE disorder, which mostly affects females and is associated with early-life adversities. Here, we show that corticotropin releasing factor (CRF)-induced prenatal stress (PNS) in late gestation predisposes female offspring to BE-like behavior that coincides with hypomethylation of hypothalamic miR-1a and downstream dysregulation of the melanocortin system through Pax7/Pax3. Moreover, exposing the offspring to a methyl-balanced diet during adolescence prevents the dysregulation and predisposition from being triggered. We demonstrate that gestational programming, per se, will not lead to BE-like behavior, but pre-existing alterations due to prenatal programming are revealed only when challenged during adolescence. We provide experimental evidence for long-term epigenetic abnormalities stemming from PNS in predisposing female offspring to BE disorder as well as a potential non-invasive prevention strategy.
Collapse
Affiliation(s)
- Mariana Schroeder
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Mira Jakovcevski
- Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Tamar Polacheck
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Maya Lebow
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Yonat Drori
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Mareen Engel
- Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Shifra Ben-Dor
- Bioinformatics and Biological Computing Unit, Biological Services, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany.
| |
Collapse
|
240
|
Velleman S, Harding R. Regulation of turkey myogenic satellite cell migration by MicroRNAs miR-128 and miR-24. Poult Sci 2017; 96:1910-1917. [DOI: 10.3382/ps/pew434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/26/2016] [Indexed: 12/13/2022] Open
|
241
|
Differential expression profile of miRNAs in porcine muscle and adipose tissue during development. Gene 2017; 618:49-56. [DOI: 10.1016/j.gene.2017.04.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/20/2017] [Accepted: 04/07/2017] [Indexed: 12/25/2022]
|
242
|
Lu C, Wu J, Xiong S, Zhang X, Zhang J, Mei J. MicroRNA-203a regulates fast muscle differentiation by targeting dmrt2a in zebrafish embryos. Gene 2017; 625:49-54. [PMID: 28483596 DOI: 10.1016/j.gene.2017.05.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 04/26/2017] [Accepted: 05/04/2017] [Indexed: 01/25/2023]
Abstract
Dmrt2b (doublesex and mab-3 related transcription factor 2b) has been revealed to be involved in zebrafish slow muscle development. However, the function of dmrt2a, a paralogue gene of dmrt2b, remains unclear during zebrafish muscle development. Here, we demonstrated that knockdown of dmrt2a resulted in severe developmental defects, and caused downregulation of fast muscle marker myhz-2 and upregulation of slow muscle marker myhz-5, respectively. It is known that microRNAs (miRNAs) control many biological events including muscle development. Dmrt2a was predicted to be a target gene of miR-203, which was further verified by luciferase reporter assay, since miR-203a was found to directly reduce the expression of dmrt2a by binding to the seed sequence of its 3'UTR. After miR-203a injection into zebrafish embryos, the expression of dmrt2a was significantly inhibited. Similar to the effect of dmrt2a knockdown, miR-203a overexpression led to downregulation of myhz-2 and upregulation of myhz-5. Our studies indicated that miR-203a directly regulated dmrt2a expression to control fast and slow muscle differentiation, while overexpression of miR-203a or knockdown of dmrt2a will impair fast muscle development and promote slow muscle development.
Collapse
Affiliation(s)
- Chang Lu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Huazhong Agricultural University, Wuhan 430070, China
| | - Junjie Wu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Huazhong Agricultural University, Wuhan 430070, China
| | - Shuting Xiong
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Huazhong Agricultural University, Wuhan 430070, China
| | - Xuemei Zhang
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Huazhong Agricultural University, Wuhan 430070, China
| | - Jin Zhang
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Huazhong Agricultural University, Wuhan 430070, China
| | - Jie Mei
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
243
|
McCormick R, Goljanek-Whysall K. MicroRNA Dysregulation in Aging and Pathologies of the Skeletal Muscle. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 334:265-308. [PMID: 28838540 DOI: 10.1016/bs.ircmb.2017.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skeletal muscle is one of the biggest organs of the body with important mechanistic and metabolic functions. Muscle homeostasis is controlled by environmental, genetic, and epigenetic factors. Indeed, MiRNAs, small noncoding RNAs robust regulators of gene expression, have and have been shown to regulate muscle homeostasis on several levels: through controlling myogenesis, muscle growth (hypertrophy) and atrophy, as well as interactions of muscle with other tissues. Given the large number of MiRNA target genes and the important role of MiRNAs in most physiological processes and various diseases, MiRNAs may have an enormous potential as therapeutic targets against numerous disorders, including pathologies of muscle. The purpose of this review is to present the current knowledge of the role of MiRNAs in skeletal muscle homeostasis and pathologies and the potential of MiRNAs as therapeutics for skeletal muscle wasting, with particular focus on the age- and disease-related loss of muscle mass and function.
Collapse
Affiliation(s)
- Rachel McCormick
- Musculoskeletal Biology II, Centre for Integrated Research into Musculoskeletal Aging, Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, United Kingdom.
| | - Katarzyna Goljanek-Whysall
- Musculoskeletal Biology II, Centre for Integrated Research into Musculoskeletal Aging, Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, United Kingdom.
| |
Collapse
|
244
|
Zhai L, Wu R, Han W, Zhang Y, Zhu D. miR-127 enhances myogenic cell differentiation by targeting S1PR3. Cell Death Dis 2017; 8:e2707. [PMID: 28358363 PMCID: PMC5386531 DOI: 10.1038/cddis.2017.128] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/22/2017] [Accepted: 02/22/2017] [Indexed: 12/25/2022]
Abstract
MicroRNAs (miRNAs) have recently been implicated in muscle stem cell function. miR-127 is known to be predominantly expressed in skeletal muscle, but its roles in myogenic differentiation and muscle regeneration are unknown. Here, we show that miR-127 is upregulated during C2C12 and satellite cell (SC) differentiation and, by establishing C2C12 cells stably expressing miR-127, demonstrate that overexpression of miR-127 in C2C12 cells enhances myogenic cell differentiation. To investigate the function of miR-127 during muscle development and regeneration in vivo, we generated miR-127 transgenic mice. These mice exhibited remarkably accelerated muscle regeneration compared with wild-type mice by promoting SC differentiation. Mechanistically, we demonstrated that the gene encoding sphingosine-1-phosphate receptor 3 (S1PR3), a G-protein-coupled receptor for sphingosine-1-phosphate, is a target of miR-127 required for its function in promoting myogenic cell differentiation. Importantly, overexpression of miR-127 in muscular dystrophy model mdx mice considerably ameliorated the disease phenotype. Thus, our findings suggest that miR-127 may serve as a potential therapeutic target for the treatment of skeletal muscle disease in humans.
Collapse
Affiliation(s)
- Lili Zhai
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, PR China
| | - Rimao Wu
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, PR China
| | - Wanhong Han
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, PR China
| | - Yong Zhang
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, PR China
| | - Dahai Zhu
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, PR China
| |
Collapse
|
245
|
Wang H, Shi L, Liang T, Wang B, Wu W, Su G, Wei J, Li P, Huang R. MiR-696 Regulates C2C12 Cell Proliferation and Differentiation by Targeting CNTFRα. Int J Biol Sci 2017; 13:413-425. [PMID: 28529450 PMCID: PMC5436562 DOI: 10.7150/ijbs.17508] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/06/2017] [Indexed: 12/28/2022] Open
Abstract
Micro-696 (miR-696) has been previously known as an exercise related miRNA, which has a profound role in fatty acid oxidation and mitochondrial biogenesis of skeletal muscle. However, its role in skeletal myoblast proliferation and differentiation is still unclear. In this study, we found that miR-696 expressed highly in skeletal muscle and reduced during C2C12 myoblasts differentiation. MiR-696 overexpression repressed C2C12 myoblast proliferation and myofiber formation, while knockdown of endogenous miR-696 expression showed opposite results. During myogenesis, we observed an inversed expression pattern between miR-696 and CNTFRα in vitro, and demonstrated that miR-696 could specifically target CNTFRα and repress the expression of CNTFRα. Additionally, we further found that knockdown of CNTFRα suppressed the proliferation and differentiation of C2C12 cells. Taking all things together, we propose a novel insight that miR-696 down-regulates C2C12 cell myogenesis by inhibiting CNTFRα expression.
Collapse
Affiliation(s)
- Han Wang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lei Shi
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Tingting Liang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - BinBin Wang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - WangJun Wu
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Guosheng Su
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - Julong Wei
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Pinghua Li
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ruihua Huang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
246
|
Koganti PP, Wang J, Cleveland B, Ma H, Weber GM, Yao J. Estradiol regulates expression of miRNAs associated with myogenesis in rainbow trout. Mol Cell Endocrinol 2017; 443:1-14. [PMID: 28011237 DOI: 10.1016/j.mce.2016.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/14/2016] [Accepted: 12/13/2016] [Indexed: 10/20/2022]
Abstract
17β-Estradiol (E2) is a steroid hormone that negatively affects muscle growth in rainbow trout, but the mechanism associated with this response is not fully understood. To better characterize the effects of E2 on muscle, we identified differentially regulated microRNAs (miRNAs) and muscle atrophy-related transcripts in juvenile rainbow trout exposed to E2. Small RNA-Seq analysis of E2-treated vs. control muscle identified 36 differentially expressed miRNAs including those known to be involved in myogenesis, cell cycle, apoptosis, and cell death. Some important myogenic miRNAs, such as miR-133 and miR-206, are upregulated while others like miR-145 and miR-499, are downregulated. Gene Ontology analysis of the target genes regulated by the miRNAs involved in atrophy and cell cycle indicates that E2 influence leads to expansion of quiescent myogenic precursor cell population to address atrophying mature muscle in rainbow trout during sexual development.
Collapse
Affiliation(s)
- Prasanthi P Koganti
- Genetics and Developmental Biology, West Virginia University, Morgantown, WV, United States
| | - Jian Wang
- Genetics and Developmental Biology, West Virginia University, Morgantown, WV, United States
| | - Beth Cleveland
- USDA/ARS, National Center for Cool and Cold Water Aquaculture Research, Kearneysville, WV, United States
| | - Hao Ma
- USDA/ARS, National Center for Cool and Cold Water Aquaculture Research, Kearneysville, WV, United States
| | - Gregory M Weber
- USDA/ARS, National Center for Cool and Cold Water Aquaculture Research, Kearneysville, WV, United States
| | - Jianbo Yao
- Genetics and Developmental Biology, West Virginia University, Morgantown, WV, United States.
| |
Collapse
|
247
|
Gonçalves TJ, Armand AS. Non-coding RNAs in skeletal muscle regeneration. Noncoding RNA Res 2017; 2:56-67. [PMID: 30159421 PMCID: PMC6096429 DOI: 10.1016/j.ncrna.2017.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/23/2017] [Accepted: 03/23/2017] [Indexed: 01/09/2023] Open
Abstract
Following injury, skeletal muscles can regenerate from muscle specific stem cells, called satellite cells. Quiescent in uninjured muscles, satellite cells become activated, proliferate and differentiate into myotubes. Muscle regeneration occurs following distinct main overlapping phases, including inflammation, regeneration and maturation of the regenerated myofibers. Each step of muscle regeneration is orchestrated through complex signaling networks and gene regulatory networks, leading to the expression of specific set of genes in each concerned cell type. Apart from the well-established transcriptional mechanisms involving the myogenic regulatory factors of the MyoD family, increasing data indicate that each step of muscle regeneration is controlled by a wide range of non-coding RNAs. In this review, we discuss the role of two classes of non-coding RNAs (microRNAs and long non-coding RNAs) in the inflammatory, regeneration and maturation steps of muscle regeneration.
Collapse
Affiliation(s)
- Tristan J.M. Gonçalves
- Institut Necker-Enfants Malades, Inserm, U1151, 14 rue Maria Helena Vieira Da Silva, CS 61431, Paris, F-75014, France
- INSERM UMRS 1124, 45 rue des Saints-Pères, F-75270 Paris cedex 06, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne-Sophie Armand
- Institut Necker-Enfants Malades, Inserm, U1151, 14 rue Maria Helena Vieira Da Silva, CS 61431, Paris, F-75014, France
- INSERM UMRS 1124, 45 rue des Saints-Pères, F-75270 Paris cedex 06, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
248
|
Amirouche A, Jahnke VE, Lunde JA, Koulmann N, Freyssenet DG, Jasmin BJ. Muscle-specific microRNA-206 targets multiple components in dystrophic skeletal muscle representing beneficial adaptations. Am J Physiol Cell Physiol 2017; 312:C209-C221. [DOI: 10.1152/ajpcell.00185.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 12/06/2016] [Accepted: 12/21/2016] [Indexed: 01/31/2023]
Abstract
Over the last several years, converging lines of evidence have indicated that miR-206 plays a pivotal role in promoting muscle differentiation and regeneration, thereby potentially impacting positively on the progression of neuromuscular disorders, including Duchenne muscular dystrophy (DMD). Despite several studies showing the regulatory function of miR-206 on target mRNAs in skeletal muscle cells, the effects of overexpression of miR-206 in dystrophic muscles remain to be established. Here, we found that miR-206 overexpression in mdx mouse muscles simultaneously targets multiple mRNAs and proteins implicated in satellite cell differentiation, muscle regeneration, and at the neuromuscular junction. Overexpression of miR-206 also increased the levels of several muscle-specific mRNAs/proteins, while enhancing utrophin A expression at the sarcolemma. Finally, we also observed that the increased expression of miR-206 in dystrophin-deficient mouse muscle decreased the production of proinflammatory cytokines and infiltration of macrophages. Taken together, our results show that miR-206 acts as a pleiotropic regulator that targets multiple key mRNAs and proteins expected to provide beneficial adaptations in dystrophic muscle, thus highlighting its therapeutic potential for DMD.
Collapse
Affiliation(s)
- Adel Amirouche
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| | - Vanessa E. Jahnke
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
| | - John A. Lunde
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| | - Nathalie Koulmann
- Institut de Recherche Biomédicale des Armées, Département Environnements Opérationnels, Bretigny-sur-Orge, France
| | - Damien G. Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
| | - Bernard J. Jasmin
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| |
Collapse
|
249
|
Jia X, Ouyang H, Abdalla BA, Xu H, Nie Q, Zhang X. miR-16 controls myoblast proliferation and apoptosis through directly suppressing Bcl2 and FOXO1 activities. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:674-684. [PMID: 28258011 DOI: 10.1016/j.bbagrm.2017.02.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 02/25/2017] [Accepted: 02/27/2017] [Indexed: 12/18/2022]
Abstract
Myogenesis mainly involves several steps including myoblast proliferation, differentiation, apoptosis and fusion. Except for muscle specific regulators, few miRNAs were proved to coordinate this complex process. Here, we reported that miR-16 inhibited myoblast proliferation and promoted myoblast apoptosis by directly targeting Bcl2 and FOXO1. The expression level of miR-16 was significantly decreased in the hypertrophic pectoral muscle compared to the normal pectoral muscle in chicken. In vitro, elevating miR-16 significantly inhibited myoblast proliferation and promoted myoblast apoptosis, resulting in about 11.2% cells arrested in G1 phase and 12.3% apoptotic cells in the early stage. Bioinformatic and biochemical analyses revealed Bcl2 and FOXO1 as direct targets of miR-16. Consist to the effect of miR-16 on myogenesis, specific inhibition of Bcl2 or FOXO1 significantly suppressed myoblast proliferation and induced myoblast apoptosis, indicating that both Bcl2 and FOXO1 contributed to miR-16 regulatory function in myogenesis. Interestingly, FOXO1, as the core target, mediated multiple growth-related pathways induced by miR-16 such as PI3K-AKT-MAPK and PI3K-AKT-mTOR. Chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-seq) revealed that 234 annotated genes bound by FOXO1 in the early-differentiated myoblasts, which were significantly enriched in myogenic proliferation, death and hypotrophy. Altogether, we proposed that miR-16 acted as a coordinated mediator to suppress myogenesis in avian through the control of myoblast proliferation and apoptosis. These findings have provided a novel mechanism whereby miR-16 represses Bcl2 and FOXO1 expression to maintain myoblast growth and skeletal muscle mass.
Collapse
Affiliation(s)
- Xinzheng Jia
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Hongjia Ouyang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Bahareldin Ali Abdalla
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Haiping Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Qinghua Nie
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China.
| | - Xiquan Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| |
Collapse
|
250
|
Terena SML, Fernandes KPS, Bussadori SK, Deana AM, Mesquita-Ferrari RA. Systematic review of the synergist muscle ablation model for compensatory hypertrophy. Rev Assoc Med Bras (1992) 2017; 63:164-172. [DOI: 10.1590/1806-9282.63.02.164] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 06/26/2016] [Indexed: 11/21/2022] Open
Abstract
Summary Objective: The aim was to evaluate the effectiveness of the experimental synergists muscle ablation model to promote muscle hypertrophy, determine the period of greatest hypertrophy and its influence on muscle fiber types and determine differences in bilateral and unilateral removal to reduce the number of animals used in this model. Method: Following the application of the eligibility criteria for the mechanical overload of the plantar muscle in rats, nineteen papers were included in the review. Results: The results reveal a greatest hypertrophy occurring between days 12 and 15, and based on the findings, synergist muscle ablation is an efficient model for achieving rapid hypertrophy and the contralateral limb can be used as there was no difference between unilateral and bilateral surgery, which reduces the number of animals used in this model. Conclusion: This model differs from other overload models (exercise and training) regarding the characteristics involved in the hypertrophy process (acute) and result in a chronic muscle adaptation with selective regulation and modification of fast-twitch fibers in skeletal muscle. This is an efficient and rapid model for compensatory hypertrophy.
Collapse
|