1
|
Wu Y, Liu X, Fan Y, Zuo H, Niu X, Zuo B, Xu Z. MiR-34b Regulates Muscle Growth and Development by Targeting SYISL. Cells 2025; 14:379. [PMID: 40072107 PMCID: PMC11898696 DOI: 10.3390/cells14050379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Non-coding genes, such as microRNA and lncRNA, which have been widely studied, play an important role in the regulatory network of skeletal muscle development. However, the functions and mechanisms of most non-coding RNAs in skeletal muscle regulatory networks are unclear. This study investigated the function and mechanism of miR-34b in muscle growth and development. MiR-34b overexpression and interference tests were performed in C2C12 myoblasts and animal models. It was demonstrated that miR-34b significantly promoted mouse muscle growth and development in vivo, while miR-34b inhibited myoblast proliferation and promoted myoblast differentiation in vitro. Bioinformatics prediction using TargetScan for miRNA target identification and Bibiserv2 for potential miRNA-gene interaction analysis revealed a miR-34b binding site in the SYlSL sequence. The molecular mechanism of miR-34b regulating muscle growth and development was studied by co-transfection experiment, luciferase reporter gene detection, RNA immunoprecipitation, and RNA pull-down. MiR-34b can directly bind to SYISL and AGO2 proteins and regulate the expression of SYISL target genes p21 and MyoG by targeting SYISL, thereby regulating muscle growth and development. This study highlights that, as a novel regulator of myogenesis, miR-34b regulates muscle growth and development by targeting SYISL.
Collapse
Affiliation(s)
- Yuting Wu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiao Liu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yonghui Fan
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Hao Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaoyu Niu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Bo Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430068, China
| | - Zaiyan Xu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
2
|
Huang Y, Wang W, Fan X, Liu X, Liu W, Wang Z, Li Y, Yang Y, Tang Z. The miR-6240 target gene Igf2bp3 promotes myoblast fusion by enhancing myomaker mRNA stability. Cell Mol Biol Lett 2024; 29:152. [PMID: 39639214 PMCID: PMC11622686 DOI: 10.1186/s11658-024-00650-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 10/11/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Myoblast fusion plays a crucial role in myogenesis. Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) functions as an RNA N6-methyladenosine reader and exerts important roles in various biological processes. While our prior study suggested Igf2bp3 contributes to myogenesis, its molecular regulatory mechanism is largely unclear. METHODS Real-time quantitative polymerase chain reaction (RT-qPCR) and western blot were used for gene expression analysis. siRNA and CRISPRi technologies were conducted to knockdown the expression of Igf2bp3. CRISPR/Cas9 technology was performed to knockout Igf2bp3. The Igf2bp3 overexpression vector was designed using the pcDNA3.1(+) vector. Immunofluorescence detection was employed for subcellular localization and cell differentiation analysis. Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) assays were conducted for cell proliferation and fusion detection. The dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were utilized for regulatory mechanism analysis of Igf2bp3. RESULTS The overexpression of Igf2bp3 enhances myoblast fusion while knockdown of Igf2bp3 blocks the formation of myotubes. miR-6240 promotes myoblast proliferation while preventing myoblast differentiation and fusion by targeting the 3' untranslated rgion (UTR) of Igf2bp3. Notably, the impacts of miR-6240 mimics on myoblast proliferation, differentiation, and fusion can be effectively counteracted by the overexpression of Igf2bp3. Moreover, our findings elucidate a direct interaction between Igf2bp3 and the myoblast fusion factor myomaker (Mymk). Igf2bp3 binds to Mymk to enhance its mRNA stability. This interaction results in increased expression of Mymk and heightened myoblast fusion. CONCLUSIONS Our study unveils Igf2bp3 as a novel post-transcriptional regulator of myoblast fusion through the miR-6240/Mymk axis, significantly contributing to our understanding of skeletal muscle development.
Collapse
Affiliation(s)
- Yuxin Huang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention; College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Wei Wang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Xinhao Fan
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Xiaoqin Liu
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Weiwei Liu
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention; College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Zishuai Wang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Yixing Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention; College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi, China
| | - Yalan Yang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China.
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China.
| | - Zhonglin Tang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China.
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China.
| |
Collapse
|
3
|
Wang T, Ran B, Luo Y, Ma J, Li J, Li P, Li M, Li D. Functional study of the ST6GAL2 gene regulating skeletal muscle growth and development. Heliyon 2024; 10:e37311. [PMID: 39296044 PMCID: PMC11407927 DOI: 10.1016/j.heliyon.2024.e37311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/21/2024] Open
Abstract
ST6GAL2, a member of the sialoglycosyltransferase family, primarily localizes within the cellular Golgi apparatus. However, the role of the ST6GAL2 gene in skeletal muscle growth and development remains elusive. In this study, the impact of the ST6GAL2 gene on the proliferation, differentiation, and apoptosis of primary chicken myoblasts at the cellular level was investigated. Quantitative fluorescent PCR was used to measure the expression levels of genes. Subsequently, using gene knockout mice, we assessed its effects on skeletal muscle growth and development in vivo. Our findings reveal that the ST6GAL2 gene promotes the expression of cell cycle and proliferation-related genes, including CCNB2 and PCNA, and apoptosis-related genes, such as Fas and Caspase-9. At the individual level, double knockout of ST6GAL2 inhibited the formation of both fast and slow muscle fibers in the quadriceps, extensor digitorum longus, and tibial anterior muscle, while promoting their formation in the gastrocnemius and soleus. These results collectively demonstrate that the ST6GAL2 gene facilitates the proliferation, apoptosis, and fusion processes of primary chicken myoblasts. Additionally, it promotes the enlargement of cross-sectional muscle fiber areas and regulates the formation of fast and slow muscle fibers at the individual level, albeit inhibiting muscle fusion. This study provides valuable insights into the role of the ST6GAL2 gene in promoting proliferation of skeletal muscle.
Collapse
Affiliation(s)
- Tao Wang
- School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Bo Ran
- Sichuan Animal Science Academy, Chengdu, 610066, China
| | - Yingyu Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jideng Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jing Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Penghao Li
- Jinxin Research Institute for Reproductive Medicine and Genetics, Chengdu Xi Nan Gynecological Hospital Co., Ltd., 66 Bisheng Road, Chengdu, 610000, China
| | - Mingzhou Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Diyan Li
- School of Pharmacy, Chengdu University, Chengdu, 610106, China
| |
Collapse
|
4
|
Zeng W, Meng Y, Nie L, Cheng C, Gao Z, Liu L, Zhu X, Chu W. The Regulatory Role of Myomaker in the Muscle Growth of the Chinese Perch ( Siniperca chuatsi). Animals (Basel) 2024; 14:2448. [PMID: 39272233 PMCID: PMC11394465 DOI: 10.3390/ani14172448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
The fusion of myoblasts is a crucial stage in the growth and development of skeletal muscle. Myomaker is an important myoblast fusion factor that plays a crucial role in regulating myoblast fusion. However, the function of Myomaker in economic fish during posthatching has been poorly studied. In this study, we found that the expression of Myomaker in the fast muscle of Chinese perch (Siniperca chuatsi) was higher than that in other tissues. To determine the function of Myomaker in fast muscle, Myomaker-siRNA was used to knockdown Myomaker in Chinese perch and the effect on muscle growth was determined. The results showed that the growth of Chinese perch was significantly decreased in the Myomaker-siRNA group. Furthermore, both the diameter of muscle fibers and the number of nuclei in single muscle fibers were significantly reduced in the Myomaker-siRNA group, whereas there was no significant difference in the number of BrdU-positive cells (proliferating cells) between the control and the Myomaker-siRNA groups. Together, these findings indicate that Myomaker may regulate growth of fast muscle in Chinese perch juveniles by promoting myoblast fusion rather than proliferation.
Collapse
Affiliation(s)
- Wei Zeng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Yangyang Meng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Lingtao Nie
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Congyi Cheng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Zexia Gao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Wuhan 430070, China
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Lusha Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Wuhan 430070, China
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xin Zhu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Wuying Chu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| |
Collapse
|
5
|
Feng L, Chen Z, Bian H. Skeletal muscle: molecular structure, myogenesis, biological functions, and diseases. MedComm (Beijing) 2024; 5:e649. [PMID: 38988494 PMCID: PMC11234433 DOI: 10.1002/mco2.649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024] Open
Abstract
Skeletal muscle is an important motor organ with multinucleated myofibers as its smallest cellular units. Myofibers are formed after undergoing cell differentiation, cell-cell fusion, myonuclei migration, and myofibril crosslinking among other processes and undergo morphological and functional changes or lesions after being stimulated by internal or external factors. The above processes are collectively referred to as myogenesis. After myofibers mature, the function and behavior of skeletal muscle are closely related to the voluntary movement of the body. In this review, we systematically and comprehensively discuss the physiological and pathological processes associated with skeletal muscles from five perspectives: molecule basis, myogenesis, biological function, adaptive changes, and myopathy. In the molecular structure and myogenesis sections, we gave a brief overview, focusing on skeletal muscle-specific fusogens and nuclei-related behaviors including cell-cell fusion and myonuclei localization. Subsequently, we discussed the three biological functions of skeletal muscle (muscle contraction, thermogenesis, and myokines secretion) and its response to stimulation (atrophy, hypertrophy, and regeneration), and finally settled on myopathy. In general, the integration of these contents provides a holistic perspective, which helps to further elucidate the structure, characteristics, and functions of skeletal muscle.
Collapse
Affiliation(s)
- Lan‐Ting Feng
- Department of Cell Biology & National Translational Science Center for Molecular MedicineNational Key Laboratory of New Drug Discovery and Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhi‐Nan Chen
- Department of Cell Biology & National Translational Science Center for Molecular MedicineNational Key Laboratory of New Drug Discovery and Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Huijie Bian
- Department of Cell Biology & National Translational Science Center for Molecular MedicineNational Key Laboratory of New Drug Discovery and Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| |
Collapse
|
6
|
Shi M, Yang S, Zhao X, Sun D, Li Y, Yang J, Li M, Cai C, Guo X, Li B, Lu C, Cao G. Effect of LncRNA LOC106505926 on myogenesis and Lipogenesis of porcine primary cells. BMC Genomics 2024; 25:530. [PMID: 38816813 PMCID: PMC11137989 DOI: 10.1186/s12864-024-10422-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Skeletal muscle development and fat deposition have important effects on meat quality. The study of regulating skeletal muscle development and fat deposition is of great significance in improving the quality of carcass and meat. In the present study, whole transcriptome sequencing (including RNA-Seq and miRNA-Seq) was performed on the longissimus dorsi muscle (LDM) of Jinfen White pigs at 1, 90, and 180 days of age. RESULTS The results showed that a total of 245 differentially expressed miRNAs were screened in any two comparisons, which may be involved in the regulation of myogenesis. Among them, compared with 1-day-old group, miR-22-5p was significantly up-regulated in 90-day-old group and 180-day-old group. Functional studies demonstrated that miR-22-5p inhibited the proliferation and differentiation of porcine skeletal muscle satellite cells (PSCs). Pearson correlation coefficient analysis showed that long non-coding RNA (lncRNA) LOC106505926 and CXXC5 gene had strong negative correlations with miR-22-5p. The LOC106505926 and CXXC5 were proven to promote the proliferation and differentiation of PSCs, as opposed to miR-22-5p. In terms of mechanism, LOC106505926 functions as a molecular sponge of miR-22-5p to modulate the expression of CXXC5, thereby inhibits the differentiation of PSCs. In addition, LOC106505926 regulates the differentiation of porcine preadipocytes through direct binding with FASN. CONCLUSIONS Collectively, our results highlight the multifaceted regulatory role of LOC106505926 in controlling skeletal muscle and adipose tissue development in pigs and provide new targets for improving the quality of livestock products by regulating skeletal muscle development and fat deposition.
Collapse
Affiliation(s)
- Mingyue Shi
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Shuai Yang
- Shanxi Animal Husbandry Technology Extension Service Center, Taiyuan, 030001, China
| | - Xiaolei Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Di Sun
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Yifei Li
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Jingxian Yang
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Meng Li
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Chunbo Cai
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Xiaohong Guo
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Bugao Li
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Chang Lu
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China.
| | - Guoqing Cao
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China.
| |
Collapse
|
7
|
Koopmans PJ, Ismaeel A, Goljanek-Whysall K, Murach KA. The roles of miRNAs in adult skeletal muscle satellite cells. Free Radic Biol Med 2023; 209:228-238. [PMID: 37879420 PMCID: PMC10911817 DOI: 10.1016/j.freeradbiomed.2023.10.403] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/16/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023]
Abstract
Satellite cells are bona fide muscle stem cells that are indispensable for successful post-natal muscle growth and regeneration after severe injury. These cells also participate in adult muscle adaptation in several capacities. MicroRNAs (miRNAs) are post-transcriptional regulators of mRNA that are implicated in several aspects of stem cell function. There is evidence to suggest that miRNAs affect satellite cell behavior in vivo during development and myogenic progenitor behavior in vitro, but the role of miRNAs in adult skeletal muscle satellite cells is less studied. In this review, we provide evidence for how miRNAs control satellite cell function with emphasis on satellite cells of adult skeletal muscle in vivo. We first outline how miRNAs are indispensable for satellite cell viability and control the phases of myogenesis. Next, we discuss the interplay between miRNAs and myogenic cell redox status, senescence, and communication to other muscle-resident cells during muscle adaptation. Results from recent satellite cell miRNA profiling studies are also summarized. In vitro experiments in primary myogenic cells and cell lines have been invaluable for exploring the influence of miRNAs, but we identify a need for novel genetic tools to further interrogate how miRNAs control satellite cell behavior in adult skeletal muscle in vivo.
Collapse
Affiliation(s)
- Pieter Jan Koopmans
- Exercise Science Research Center, Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA; Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40506, USA
| | - Katarzyna Goljanek-Whysall
- School of Medicine, College of Medicine, Nursing, and Health Sciences, University of Galway, Galway, Ireland
| | - Kevin A Murach
- Exercise Science Research Center, Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA; Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, 72701, USA.
| |
Collapse
|
8
|
Ma J, Zhu Y, Zhou X, Zhang J, Sun J, Li Z, Jin L, Long K, Lu L, Ge L. miR-205 Regulates the Fusion of Porcine Myoblast by Targeting the Myomaker Gene. Cells 2023; 12:cells12081107. [PMID: 37190016 DOI: 10.3390/cells12081107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/10/2023] [Accepted: 03/31/2023] [Indexed: 05/17/2023] Open
Abstract
Skeletal muscle formation is an extremely important step in animal growth and development. Recent studies have found that TMEM8c (also known as Myomaker, MYMK), a muscle-specific transmembrane protein, can promote myoblast fusion and plays a key role in the normal development of skeletal muscle. However, the effect of Myomaker on porcine (Sus scrofa) myoblast fusion and the underlying regulatory mechanisms remain largely unknown. Therefore, in this study, we focused on the role and corresponding regulatory mechanism of the Myomaker gene during skeletal muscle development, cell differentiation, and muscle injury repair in pigs. We obtained the entire 3' UTR sequence of porcine Myomaker using the 3' RACE approach and found that miR-205 inhibited porcine myoblast fusion by targeting the 3' UTR of Myomaker. In addition, based on a constructed porcine acute muscle injury model, we discovered that both the mRNA and protein expression of Myomaker were activated in the injured muscle, while miR-205 expression was significantly inhibited during skeletal muscle regeneration. The negative regulatory relationship between miR-205 and Myomaker was further confirmed in vivo. Taken together, the present study reveals that Myomaker plays a role during porcine myoblast fusion and skeletal muscle regeneration and demonstrates that miR-205 inhibits myoblast fusion through targeted regulation of the expression of Myomaker.
Collapse
Affiliation(s)
- Jideng Ma
- Chongqing Academy of Animal Sciences, Chongqing 402460, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing 402460, China
- Chongqing Key Laboratory of Pig Industry Sciences, Chongqing 402460, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Zhu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiankun Zhou
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jinwei Zhang
- Chongqing Academy of Animal Sciences, Chongqing 402460, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing 402460, China
- Chongqing Key Laboratory of Pig Industry Sciences, Chongqing 402460, China
| | - Jing Sun
- Chongqing Academy of Animal Sciences, Chongqing 402460, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing 402460, China
- Chongqing Key Laboratory of Pig Industry Sciences, Chongqing 402460, China
| | - Zhengjie Li
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Long Jin
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Keren Long
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lu Lu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing 402460, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing 402460, China
- Chongqing Key Laboratory of Pig Industry Sciences, Chongqing 402460, China
- Technical Engineering Center for the Development and Utilization of Medical Animal Resources, Chongqing 402460, China
| |
Collapse
|
9
|
Sousa-Filho CPB, Silva V, Bolin AP, Rocha ALS, Otton R. Green tea actions on miRNAs expression – An update. Chem Biol Interact 2023; 378:110465. [PMID: 37004950 DOI: 10.1016/j.cbi.2023.110465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 04/01/2023]
Abstract
Compounds derived from plants have been widely studied in the context of metabolic diseases and associated clinical conditions. In this regard, although the effects of Camellia sinensis plant, from which various types of teas, such as green tea, originate, have been vastly reported in the literature, the mechanisms underlying these effects remain elusive. A deep search of the literature showed that green tea's action in different cells, tissues, and diseases is an open field in the research of microRNAs (miRNAs). miRNAs are important communicator molecules between cells in different tissues implicated in diverse cellular pathways. They have emerged as an important linkage between physiology and pathophysiology, raising the issue of polyphenols can act also by changing miRNA expression. miRNAs are short, non-coding endogenous RNA, which silence the gene functions by targeting messenger RNA (mRNA) through degradation or translation repression. Therefore, the aim of this review is to present the studies that show the main compounds of green tea modulating the expression of miRNAs in inflammation, adipose tissue, skeletal muscle, and liver. We provide an overview of a few studies that have tried to demonstrate the role of miRNAs associated with the beneficial effects of compounds from green tea. We have emphasized that there is still a considerable gap in the literature investigating the role and likely involvement of miRNAs in the extensive beneficial health effects of green tea compounds already described, indicating miRNAs as potential polyphenols' mediators with a promising field to be investigated.
Collapse
Affiliation(s)
| | - Victoria Silva
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Anaysa Paola Bolin
- Department of Pharmacology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | | | - Rosemari Otton
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil.
| |
Collapse
|
10
|
Palma-Flores C, Zárate-Segura PB, Hernández-Hernández JM, de los Santos S, Tejeda-Gómez AS, Cano-Martínez LJ, Canto P, Garcia-Rebollar JO, Coral-Vázquez RM. (−)-Epicatechin modulates the expression of myomiRs implicated in exercise response in mouse skeletal muscle. Gene X 2023; 849:146907. [DOI: 10.1016/j.gene.2022.146907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/01/2022] [Accepted: 09/19/2022] [Indexed: 11/30/2022] Open
|
11
|
Chen S, Wu Q, Wang Y, Xu J, Wang Y, Luo X. miR-491-5p Inhibits Emilin 1 to Promote Fibroblasts Proliferation and Fibrosis in Gluteal Muscle Contracture via TGF-β1/Smad2 Pathway. Physiol Res 2022; 71:285-295. [DOI: 10.33549/physiolres.934804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Gluteal muscle contracture (GMC) is a chronic fibrotic disease of gluteal muscles due to multiple etiologies. Emilin 1 plays a determinant role in fibers formation, but its role in the progression of GMC remains unclear. The present study was aimed to search for the predictive role and regulatory mechanism of Emilin 1 on GMC. Here, Protein and mRNA expression of Emilin 1 were decreased in GMC tissues compared to normal muscle tissues. Using the analysis of target prediction, Emilin 1 was observed to be a potential downstream sponge of miR-491-5p. In comparison to Emilin 1, miR-491-5p showed an aberrant elevation in GMC tissues, which was further proven to have a negative correlation with Emilin 1. The direct binding of miR-491-5p to Emilin 1 mRNA was confirmed by luciferase reporter gene assay, and miR-491-5p mimics inhibited, while miR-491-5p inhibitor promoted the protein expression and secretion of Emilin 1 in contraction bands (CB) fibroblasts. Additionally, miR-491-5p mimics promoted the expression of cyclin-dependent kinase 2 and cyclin D1 and the proliferation of CB fibroblasts, which could be reversed by Emilin 1 overexpression. Mechanistically, miR-491-5p mimics possibly activated transforming growth factor β1 (TGF-β1)/Smad3 signal cascade via binding to 3’-untranslated region of Emilin 1 mRNA, thereby promoting the progression of fibrosis of CB fibroblasts. Collectively, miR-491-5p inhibited Emilin 1 expression, and subsequently promoted CB fibroblasts proliferation and fibrosis via activating TGF-β1/Smad3 signal axis. MiR-491-5p might be a potentially effective biomarker for predicting GMC, providing a novel therapeutic strategy for GMC.
Collapse
Affiliation(s)
- S Chen
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | | | | | | | | | | |
Collapse
|
12
|
Sutandyo N. The role of microRNA in cancer cachexia and muscle wasting: A review article. CASPIAN JOURNAL OF INTERNAL MEDICINE 2021; 12:124-128. [PMID: 34012527 PMCID: PMC8111806 DOI: 10.22088/cjim.12.2.124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Almost half of cancer patients experience cachexia syndrome. Cachexic patients are at risk of increased side effects of chemotherapy, reduced tolerance to chemotherapy drugs, longer duration of treatment period, and decreased quality of life. Cancer cachexia is a multifactorial syndrome. Micro ribonucleic acid (miRNA), a "non-coding RNA", is considered to be a risk factor of cachexia and muscle wasting in cancer patients. miRNA has a role in affecting protein regulation, associated with different inflammatory and disease pathways. miRNA can also affect cytokines or directly change the regulation of metabolism that lead to cachexia. In this review, we want to focus on the pathophysiology to give a better understanding about the role of miRNA in the development of cancer cachexia. Based on various pathways of miRNA in cancer cachexia, it can be a potential target for therapeutic strategies. Improved knowledge about miRNA can give the opportunity to develop new treatment in the management of cancer cachexia.
Collapse
Affiliation(s)
- Noorwati Sutandyo
- Hematology and Medical Oncology Division, Dharmais National Cancer Centre Hospital
| |
Collapse
|
13
|
Dong YH, Zhou CJ, Zhang MY, Tao J, Zhang XM, An L, Zhang J, Yang J, Liu DJ, Cang M. MiR-455-5p monitors myotube morphogenesis by targeting mylip. J Cell Biochem 2021; 122:442-455. [PMID: 33399227 DOI: 10.1002/jcb.29873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 11/10/2022]
Abstract
As a posttranscriptional regulatory factor, microRNA (miRNA) plays an important role in the formation of myotubes. However, little is known about the mechanism of miRNA regulating myotube morphogenesis. Here, we aimed to characterize the function of miR-455-5p in myotube morphogenesis by inducing differentiation in C2C12 myoblasts containing murine Mylip fragments with the miR-455-5p target sequence. We found that miR-455-5p overexpression promoted the differentiation and hypertrophy of myotubes, while miR-455-5p inhibition led to the failure of myotube differentiation and formation of short myotubes. Furthermore, we demonstrated that miR-455-5p directly targeted the Mylip 3'-untranslated region, which plays a key role in monitoring myotube morphogenesis. Interestingly, the expression and function of Mylip were opposite to those of miR-455-5p during myogenesis. Our data uncovered novel miR-455-5p targets and established a functional link between Mylip and myotube morphogenesis. Understanding the involvement of Mylip in myotube morphogenesis provides insight into the function of the gene regulatory network.
Collapse
Affiliation(s)
- Yan-Hua Dong
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Cheng-Jie Zhou
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Meng-Yuan Zhang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Jin Tao
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Xiao-Meng Zhang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Lu An
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Ju Zhang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Jie Yang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Dong-Jun Liu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Ming Cang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia, China
| |
Collapse
|
14
|
Li J, Su T, Zou C, Luo W, Shi G, Chen L, Fang C, Li C. Long Non-coding RNA H19 Regulates Porcine Satellite Cell Differentiation Through miR-140-5p/ SOX4 and DBN1. Front Cell Dev Biol 2020; 8:518724. [PMID: 33324629 PMCID: PMC7723966 DOI: 10.3389/fcell.2020.518724] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 09/29/2020] [Indexed: 12/14/2022] Open
Abstract
The H19 gene promotes skeletal muscle differentiation in mice, but the regulatory models and mechanisms of myogenesis regulated by H19 are largely unknown in pigs. Therefore, the regulatory modes of H19 in the differentiation of porcine skeletal muscle satellite cells (PSCs) need to be determined. We observed that H19 gene silencing could decrease the expressions of the myogenin (MYOG) gene, myogenic differentiation (MYOD), and myosin heavy chain (MYHC) in PSCs. Therefore, we constructed and sequenced 12 cDNA libraries of PSCs after knockdown of H19 at two differentiation time points to analyze the transcriptome differences. A total of 11,419 differentially expressed genes (DEGs) were identified. Among these DEGs, we found through bioinformatics analysis and protein interaction experiment that SRY-box transcription factor 4 (SOX4) and Drebrin 1 (DBN1) were the key genes in H19-regulated PSC differentiation. Functional analysis shows that SOX4 and DBN1 promote PSC differentiation. Mechanistically, H19 regulates PSC differentiation through two different pathways. On the one hand, H19 functions as a molecular sponge of miR-140-5p, which inhibits the differentiation of PSCs, thereby modulating the derepression of SOX4. On the other hand, H19 regulates PSC differentiation through directly binding with DBN1. Furthermore, MYOD binds to the promoters of H19 and DBN1. The knockdown of MYOD inhibits the expression of H19 and DBN1. We determined the function of H19 and provided a molecular model to elucidate H19’s role in regulating PSC differentiation.
Collapse
Affiliation(s)
- Jingxuan Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China.,Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Tao Su
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Cheng Zou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Wenzhe Luo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Gaoli Shi
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Lin Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Chengchi Fang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production of Hubei Province, Wuhan, China
| | - Changchun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production of Hubei Province, Wuhan, China
| |
Collapse
|
15
|
Chen B, You W, Wang Y, Shan T. The regulatory role of Myomaker and Myomixer-Myomerger-Minion in muscle development and regeneration. Cell Mol Life Sci 2020; 77:1551-1569. [PMID: 31642939 PMCID: PMC11105057 DOI: 10.1007/s00018-019-03341-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022]
Abstract
Skeletal muscle plays essential roles in motor function, energy, and glucose metabolism. Skeletal muscle formation occurs through a process called myogenesis, in which a crucial step is the fusion of mononucleated myoblasts to form multinucleated myofibers. The myoblast/myocyte fusion is triggered and coordinated in a muscle-specific way that is essential for muscle development and post-natal muscle regeneration. Many molecules and proteins have been found and demonstrated to have the capacity to regulate the fusion of myoblast/myocytes. Interestingly, two newly discovered muscle-specific membrane proteins, Myomaker and Myomixer (also called Myomerger and Minion), have been identified as fusogenic regulators in vertebrates. Both Myomaker and Myomixer-Myomerger-Minion have the capacity to directly control the myogenic fusion process. Here, we review and discuss the latest studies related to these two proteins, including the discovery, structure, expression pattern, functions, and regulation of Myomaker and Myomixer-Myomerger-Minion. We also emphasize and discuss the interaction between Myomaker and Myomixer-Myomerger-Minion, as well as their cooperative regulatory roles in cell-cell fusion. Moreover, we highlight the areas for exploration of Myomaker and Myomixer-Myomerger-Minion in future studies and consider their potential application to control cell fusion for cell-therapy purposes.
Collapse
Affiliation(s)
- Bide Chen
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, China.
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China.
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China.
| |
Collapse
|
16
|
NADPH Oxidase 4 Contributes to Myoblast Fusion and Skeletal Muscle Regeneration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3585390. [PMID: 31827673 PMCID: PMC6885834 DOI: 10.1155/2019/3585390] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/24/2019] [Accepted: 10/08/2019] [Indexed: 01/28/2023]
Abstract
Myoblast fusion is an essential step in skeletal muscle development and regeneration. NADPH oxidase 4 (Nox4) regulates cellular processes such as proliferation, differentiation, and survival by producing reactive oxygen species (ROS). Insulin-like growth factor 1 induces muscle hypertrophy via Nox4, but its function in myoblast fusion remains elusive. Here, we report a ROS-dependent role of Nox4 in myoblast differentiation. Regenerating muscle fibers after injury by cardiotoxin had a lower cross-sectional area in Nox4-knockout (KO) mice than myofibers in wild-type (WT) mice. Diameters and fusion index values of myotubes differentiated from Nox4-KO primary myoblasts were significantly lower than those of myotubes derived from WT myoblasts. However, no difference was observed in the differentiation index and expression of MyoD, myogenin, and myosin heavy chain 3 (MHC) between KO and WT myotubes. The decreased fusion index was also observed during differentiation of primary myoblasts and C2C12 cells with suppressed Nox4 expression. In contrast, in C2C12 cells overexpressing Nox4, the fusion index was increased, whereas the differentiation index and MHC and myogenin protein expression were not affected compared to control. Interestingly, the expression of myomaker (Tmem8c), a fusogenic protein that controls myoblast fusion, was reduced in Nox4-knockdown C2C12 cells. The myomaker expression level was proportional to the cellular ROS level, which was regulated by of Nox4 expression level. These results suggests that Nox4 contributes to myoblast fusion, possibly through the regulation of myomaker expression via ROS production, and that Nox4-dependent ROS may promote skeletal muscle regeneration and growth.
Collapse
|
17
|
Huang Y, Wu S, Zhang J, Wen H, Zhang M, He F. Methylation status and expression patterns of myomaker gene play important roles in postnatal development in the Japanese flounder (Paralichthys olivaceus). Gen Comp Endocrinol 2019; 280:104-114. [PMID: 31002826 DOI: 10.1016/j.ygcen.2019.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/28/2019] [Accepted: 04/16/2019] [Indexed: 01/01/2023]
Abstract
Myomaker is a membrane protein that plays a crucial role in the fusion of myoblasts during muscle growth. DNA methylation, a significant factor, regulates gene expression. The aim of this study was to examine the methylation and mRNA expression patterns of the myomaker gene during 8 different postnatal developmental stages in the Japanese flounder (L: 7 days post hatch (dph); M1: 21 dph; M2: 28 dph; M3: 35 dph; J1: 90 dph; J2: 180 dph; A1: 24 months; A2: 36 months). Muscle tissue samples were taken from Japanese flounder at different postnatal development stages to measure the extent of DNA methylation and gene expression. Methylation level in the promoter and exon 1 of myomaker was measured using bisulfite sequencing, and the relative expression of myomaker during each developmental stage was measured by quantitative PCR. The relative expression levels of myomaker were up-regulated from stages L to M2, M3 to J2, and methylation of myomaker was negatively correlated with mRNA expression. Furthermore, the CpG site located at -26 bp in the promoter was the lowest methylated region in all developmental stages. These results offer a basis for understanding the mechanism by which myomaker regulates muscle formation during postnatal development.
Collapse
Affiliation(s)
- Yajuan Huang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China
| | - Shuxian Wu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China
| | - Jingru Zhang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China
| | - Haishen Wen
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China
| | - Meizhao Zhang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China
| | - Feng He
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, China.
| |
Collapse
|
18
|
Wang J, Tan J, Qi Q, Yang L, Wang Y, Zhang C, Hu L, Chen H, Fang X. miR-487b-3p Suppresses the Proliferation and Differentiation of Myoblasts by Targeting IRS1 in Skeletal Muscle Myogenesis. Int J Biol Sci 2018; 14:760-774. [PMID: 29910686 PMCID: PMC6001677 DOI: 10.7150/ijbs.25052] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/29/2018] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs are endogenous, small non-coding RNAs that can play critical gene-regulatory roles during skeletal muscle development and are highly conserved. miR-487b-3p is expressed in muscle, and the detailed mechanism by which it regulates myoblast proliferation and differentiation has not been explored. Here, we found that miR-487b-3p expression was significantly higher in goat muscle tissues than in other tissues and was higher in fetal goat muscle tissues than in mature goat tissues, suggesting that miR-487b-3p has an important effect on skeletal muscle myogenesis. Functional studies showed that miR-487b-3p overexpression significantly suppressed C2C12 myoblast proliferation and differentiation, which was accompanied by the down-regulation of functional genes related to proliferation (MyoD, Pax7 and PCNA) and differentiation (Myf5, MyoG and Mef2c), whereas the inhibition of miR-487b-3p accelerated C2C12 myoblast proliferation and differentiation and was accompanied by the up-regulation of functional genes. Using Target-Scan and David, we found that miR-487b-3p targeted the 3'-UTR of IRS1, an essential regulator in the PI3K/Akt and MAPK/Erk pathways. We then confirmed the targeting of IRS1 by miR-487b-3p using dual-luciferase assays, RT-qPCR and western blotting. Furthermore, IRS1 silencing markedly inhibited proliferation and differentiation in cultured C2C12 myoblasts, confirming the important role of IRS1 in myogenesis. These results reveal an IRS1-mediated regulatory link between miR-487b-3p and the PI3K/Akt and MAPK/Erk pathways during skeletal muscle myogenesis.
Collapse
Affiliation(s)
- Jian Wang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Jiaoyan Tan
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Qi Qi
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Lingzhi Yang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Yanhong Wang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Chunlei Zhang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Linyong Hu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, 810001, China
| | - Hong Chen
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Xingtang Fang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| |
Collapse
|
19
|
Ultimo S, Zauli G, Martelli AM, Vitale M, McCubrey JA, Capitani S, Neri LM. Influence of physical exercise on microRNAs in skeletal muscle regeneration, aging and diseases. Oncotarget 2018; 9:17220-17237. [PMID: 29682218 PMCID: PMC5908319 DOI: 10.18632/oncotarget.24991] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/06/2018] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle is a dynamic tissue with remarkable plasticity and its growth and regeneration are highly organized, with the activation of specific transcription factors, proliferative pathways and cytokines. The decline of skeletal muscle tissue with age, is one of the most important causes of functional loss of independence in older adults. Maintaining skeletal muscle function throughout the lifespan is a prerequisite for good health and independent living. Physical activity represents one of the most effective preventive agents for muscle decay in aging. Several studies have underlined the importance of microRNAs (miRNAs) in the control of myogenesis and of skeletal muscle regeneration and function. In this review, we reported an overview and recent advances about the role of miRNAs expressed in the skeletal muscle, miRNAs regulation by exercise in skeletal muscle, the consequences of different physical exercise training modalities in the skeletal muscle miRNA profile, their regulation under pathological conditions and the role of miRNAs in age-related muscle wasting. Specific miRNAs appear to be involved in response to different types of exercise and therefore to play an important role in muscle fiber identity and myofiber gene expression in adults and elder population. Understanding the roles and regulation of skeletal muscle miRNAs during muscle regeneration may result in new therapeutic approaches in aging or diseases with impaired muscle function or re-growth.
Collapse
Affiliation(s)
- Simona Ultimo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marco Vitale
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,CoreLab, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, USA
| | - Silvano Capitani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
20
|
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
|
21
|
Blondelle J, Pais de Barros JP, Pilot-Storck F, Tiret L. Targeted Lipidomic Analysis of Myoblasts by GC-MS and LC-MS/MS. Methods Mol Biol 2017; 1668:39-60. [PMID: 28842901 DOI: 10.1007/978-1-4939-7283-8_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Lipids represent ∼10% of the cell dry mass and play essential roles in membrane composition and physical properties, energy storage, and signaling pathways. In the developing or the regenerating skeletal muscle, modifications in the content or the flipping between leaflets of membrane lipid components can modulate the fusion capacity of myoblasts, thus constituting one of the regulatory mechanisms underlying myofiber growth. Recently, few genes controlling these qualitative and quantitative modifications have started to be unraveled. The precise functional characterization of these genes requires both qualitative and quantitative evaluations of a global lipid profile. Here, we describe a lipidomic protocol using mass spectrometry, allowing assessing the content of fatty acids, glycerophospholipids, and cholesterol in the routinely used C2C12 mouse myoblast cell line, or in primary cultures of mouse myoblasts.
Collapse
Affiliation(s)
- Jordan Blondelle
- Institut Mondor de Recherche Biomédicale (IMRB), U955-E10 Biologie du Système Neuromusculaire, Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort (EnvA), Maisons-Alfort, France
- Department of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | - Jean-Paul Pais de Barros
- Plateforme de Lipidomique-uBourgogne, INSERM UMR1231/LabEx LipSTIC, UFR des Sciences de Santé - Bâtiment B3, Dijon, France
| | - Fanny Pilot-Storck
- Institut Mondor de Recherche Biomédicale (IMRB), U955-E10 Biologie du Système Neuromusculaire, Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort (EnvA), Maisons-Alfort, France
| | - Laurent Tiret
- Institut Mondor de Recherche Biomédicale (IMRB), U955-E10 Biologie du Système Neuromusculaire, Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort (EnvA), Maisons-Alfort, France.
| |
Collapse
|