1
|
Lan XQ, Deng CJ, Wang QQ, Zhao LM, Jiao BW, Xiang Y. The role of TGF-β signaling in muscle atrophy, sarcopenia and cancer cachexia. Gen Comp Endocrinol 2024; 353:114513. [PMID: 38604437 DOI: 10.1016/j.ygcen.2024.114513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/24/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Skeletal muscle, comprising a significant proportion (40 to 50 percent) of total body weight in humans, plays a critical role in maintaining normal physiological conditions. Muscle atrophy occurs when the rate of protein degradation exceeds protein synthesis. Sarcopenia refers to age-related muscle atrophy, while cachexia represents a more complex form of muscle wasting associated with various diseases such as cancer, heart failure, and AIDS. Recent research has highlighted the involvement of signaling pathways, including IGF1-Akt-mTOR, MuRF1-MAFbx, and FOXO, in regulating the delicate balance between muscle protein synthesis and breakdown. Myostatin, a member of the TGF-β superfamily, negatively regulates muscle growth and promotes muscle atrophy by activating Smad2 and Smad3. It also interacts with other signaling pathways in cachexia and sarcopenia. Inhibition of myostatin has emerged as a promising therapeutic approach for sarcopenia and cachexia. Additionally, other TGF-β family members, such as TGF-β1, activin A, and GDF11, have been implicated in the regulation of skeletal muscle mass. Furthermore, myostatin cooperates with these family members to impair muscle differentiation and contribute to muscle loss. This review provides an overview of the significance of myostatin and other TGF-β signaling pathway members in muscular dystrophy, sarcopenia, and cachexia. It also discusses potential novel therapeutic strategies targeting myostatin and TGF-β signaling for the treatment of muscle atrophy.
Collapse
Affiliation(s)
- Xin-Qiang Lan
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Cheng-Jie Deng
- Department of Biochemistry and Molecular Biology, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Qi-Quan Wang
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Li-Min Zhao
- Senescence and Cancer Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Bao-Wei Jiao
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Yang Xiang
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China.
| |
Collapse
|
2
|
Ren P, Chen M, Li J, Lin Z, Yang C, Yu C, Zhang D, Liu Y. MYH1F promotes the proliferation and differentiation of chicken skeletal muscle satellite cells into myotubes. Anim Biotechnol 2023; 34:3074-3084. [PMID: 36244007 DOI: 10.1080/10495398.2022.2132953] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
In diploid organisms, interactions between alleles determine phenotypic variation. In previous experiments, only MYH1F was found to show both ASE (spatiotemporal allele-specific expression) and TRD (allelic transmission ratio distortion) characteristics in the pectoral muscle by comparing the genome-wide allele lists of hybrid populations (F1) of meat- and egg- type chickens. In addition, MYH1F is a member of the MYH gene family, which plays an important role in skeletal muscle and non-muscle cells of animals, but the specific expression and function of this gene in chickens are still unknown. Therefore, qRT-PCR was used to detect the expression of MYH1F in different tissues of chicken. Proliferation and differentiation of chicken skeletal muscle satellite cells (SMSCs) have been detected by transfection of MYH1F-specific small interfering RNA (siRNA). The results showed that the expression of MYH1F in chicken skeletal muscle was higher than that in other tissues. Combined with CCK-8 assay, EdU assay, immunofluorescence, and Western blot Assay, it was found that MYH1F knockdown could significantly suppress the proliferation of chicken SMSCs and depress the differentiation and fusion of the cells. These results suggest that MYH1F plays a critical role in myogenesis in poultry, which is of great significance for exploring the regulatory mechanisms of muscle development and improving animal productivity.
Collapse
Affiliation(s)
- Peng Ren
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Meiying Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Jingjing Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhongzhen Lin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Chaowu Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Chunlin Yu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Donghao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yiping Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Wei D, Zhang L, Raza SHA, Zhang J, Juan Z, Al-Amrah H, Al Abdulmonem W, Alharbi YM, Zhang G, Liang X. Interaction of C/EBPβ with SMAD2 and SMAD4 genes induces the formation of lipid droplets in bovine myoblasts. Funct Integr Genomics 2023; 23:191. [PMID: 37249689 DOI: 10.1007/s10142-023-01115-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/31/2023]
Abstract
As a key component of Transforming growth factor-β (TGF-β) pathway, Smad2 has many crucial roles in a variety of cellular processes, but it cannot bind DNA without complex formation with Smad4. In the present study, the molecular mechanism in the progress of myogenesis underlying transcriptional regulation of SMAD2 and SMAD4 had been clarified. The result showed the inhibition between SMAD2 and SMAD4, which promotes and inhibits bovine myoblast differentiation, respectively. Further, the characterization of promoter region of SMAD2 and SMAD4 was analyzed, and identified C/EBPβ directly bound to the core region of both SMAD2 and SMAD4 genes promoter and stimulated the transcriptional activity. However, C/EBPβ has lower expression in myoblasts which plays vital function in the transcriptional networks controlling adipogenesis, while the overexpression of C/EBPβ gene in myoblasts significantly increased SMAD2 and SMAD4 gene expression, induced the formation of lipid droplet in bovine myoblasts, and promoted the expression of adipogenesis-specific genes. Collectively, our results showed that C/EBPβ may play an important role in the trans-differentiation and dynamic equilibrium of myoblasts into adipocyte cells via promoting an increase in SMAD2 and SMAD4 gene levels. These results will provide an important basis for further understanding of the TGFβ pathway and C/EBPβ gene during myogenic differentiation.
Collapse
Affiliation(s)
- Dawei Wei
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Le Zhang
- Institute of Physical Education, Yan'an University, Yan'an, 716000, China
| | - Sayed Haidar Abbas Raza
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Nation-Local Joint Engineering Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, 510642, China
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Jiupan Zhang
- Institute of Animal Science, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan, 750021, China
| | - Zhao Juan
- College of Animal Science and Technology, South China Agricultural University, Guangzhou, 510642, China
| | - Hadba Al-Amrah
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, P.O. Box 6655, Buraidah, 51452, Kingdom of Saudi Arabia
| | - Yousef Mesfer Alharbi
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Guijie Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China.
| | - Xiaojun Liang
- Institute of Animal Science, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan, 750021, China.
| |
Collapse
|
4
|
Urban LA, Li J, Gundogdu G, Trinh A, Shao H, Nguyen T, Mauney JR, Downing TL. DNA Methylation Dynamics During Esophageal Epithelial Regeneration Following Repair with Acellular Silk Fibroin Grafts in Rat. Adv Biol (Weinh) 2023; 7:e2200160. [PMID: 36658732 PMCID: PMC10401397 DOI: 10.1002/adbi.202200160] [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: 06/07/2022] [Revised: 11/10/2022] [Indexed: 01/21/2023]
Abstract
Esophageal pathologies such as atresia and benign strictures often require surgical reconstruction with autologous tissues to restore organ continuity. Complications such as donor site morbidity and limited tissue availability have spurred the development of acellular grafts for esophageal tissue replacement. Acellular biomaterials for esophageal repair rely on the activation of intrinsic regenerative mechanisms to mediate de novo tissue formation at implantation sites. Previous research has identified signaling cascades involved in neoepithelial formation in a rat model of onlay esophagoplasty with acellular silk fibroin grafts, including phosphoinositide 3-kinase (PI3K), and protein kinase B (Akt) signaling. However, it is currently unknown how these mechanisms are governed by DNA methylation (DNAme) during esophageal wound healing processes. Reduced-representation bisulfite sequencing is performed to characterize temporal DNAme dynamics in host and regenerated tissues up to 1 week postimplantation. Overall, global hypermethylation is observed at postreconstruction timepoints and an inverse correlation between promoter DNAme and the expression levels of differentially expressed proteins during regeneration. Site-specific hypomethylation targets genes associated with immune activation, while hypermethylation occurs within gene bodies encoding PI3K-Akt signaling components during the tissue remodeling period. The data provide insight into the epigenetic mechanisms during esophageal regeneration following surgical repair with acellular grafts.
Collapse
Affiliation(s)
- Lauren A. Urban
- Department of Microbiology & Molecular Genetics, University of California Irvine; Irvine, California, USA
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California-Irvine, Irvine, CA 92697, USA
| | - Jiachun Li
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697, USA
| | - Gokhan Gundogdu
- Department of Urology, University of California, Irvine, Orange, CA, 92868, USA
| | - Annie Trinh
- Department of Microbiology & Molecular Genetics, University of California Irvine; Irvine, California, USA
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California-Irvine, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California-Irvine, Irvine, California 92697, USA
| | - Hanjuan Shao
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697, USA
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California-Irvine, Irvine, CA 92697, USA
| | - Travis Nguyen
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697, USA
| | - Joshua R. Mauney
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697, USA
- Department of Urology, University of California, Irvine, Orange, CA, 92868, USA
| | - Timothy L. Downing
- Department of Microbiology & Molecular Genetics, University of California Irvine; Irvine, California, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697, USA
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California-Irvine, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California-Irvine, Irvine, California 92697, USA
| |
Collapse
|
5
|
Marceca GP, Nigita G, Calore F, Croce CM. MicroRNAs in Skeletal Muscle and Hints on Their Potential Role in Muscle Wasting During Cancer Cachexia. Front Oncol 2020; 10:607196. [PMID: 33330108 PMCID: PMC7732629 DOI: 10.3389/fonc.2020.607196] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
Cancer-associated cachexia is a heterogeneous, multifactorial syndrome characterized by systemic inflammation, unintentional weight loss, and profound alteration in body composition. The main feature of cancer cachexia is represented by the loss of skeletal muscle tissue, which may or may not be accompanied by significant adipose tissue wasting. Such phenotypic alteration occurs as the result of concomitant increased myofibril breakdown and reduced muscle protein synthesis, actively contributing to fatigue, worsening of quality of life, and refractoriness to chemotherapy. According to the classical view, this condition is primarily triggered by interactions between specific tumor-induced pro-inflammatory cytokines and their cognate receptors expressed on the myocyte membrane. This causes a shift in gene expression of muscle cells, eventually leading to a pronounced catabolic condition and cell death. More recent studies, however, have shown the involvement of regulatory non-coding RNAs in the outbreak of cancer cachexia. In particular, the role exerted by microRNAs is being widely addressed, and several mechanistic studies are in progress. In this review, we discuss the most recent findings concerning the role of microRNAs in triggering or exacerbating muscle wasting in cancer cachexia, while mentioning about possible roles played by long non-coding RNAs and ADAR-mediated miRNA modifications.
Collapse
Affiliation(s)
- Gioacchino P Marceca
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giovanni Nigita
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Federica Calore
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
6
|
Jung HJ, Lee KP, Kwon KS, Suh Y. MicroRNAs in Skeletal Muscle Aging: Current Issues and Perspectives. J Gerontol A Biol Sci Med Sci 2020; 74:1008-1014. [PMID: 30215687 DOI: 10.1093/gerona/gly207] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle is one of the major organs responsible for body movements and metabolism making up approximately 40% of the total body mass. During aging, skeletal muscle exhibits a degenerative age-associated decline in mass and function termed sarcopenia. This age-associated dysfunction of skeletal muscle is a major criterion of morbidity, mortality, and overall declines of quality of life in the elderly people. Therefore, researchers have focused on identifying modulators of muscle aging process including messenger RNAs, proteins, and recently small noncoding RNAs such as microRNAs (miRNAs). In particular, miRNAs have been demonstrated to play a critical role in skeletal muscle development and homeostasis. Recent studies revealed that miRNAs were also involved in muscle aging processes and the rejuvenation of aged muscle by regulating important molecules and pathways of aging including insulin-like growth factors, nicotine-adenine dinucleotide (+)-dependent protein deacetylase sirtuin-1, telomerase reverse transcriptase, and transforming growth factor-β signaling pathway. Over the years, miRNAs have emerged as promising candidates for biomarkers of sarcopenia and targets for interventions to slow muscle aging. Here, we comprehensively review the current knowledge on the role of miRNAs in skeletal muscle aging and highlight their potential as biomarkers or therapeutic targets for skeletal muscle health.
Collapse
Affiliation(s)
- Hwa Jin Jung
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Kwang-Pyo Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Bio-Molecular Science, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Ki-Sun Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
7
|
Ding Z, Lin J, Sun Y, Cong S, Liu S, Zhang Y, Chen Q, Chen J. miR‐122‐5p negatively regulates the transforming growth factor‐β/Smad signaling pathway in skeletal muscle myogenesis. Cell Biochem Funct 2019; 38:231-238. [PMID: 31710120 DOI: 10.1002/cbf.3460] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/27/2019] [Accepted: 10/29/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Zheci Ding
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinrong Lin
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuang Cong
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shaohua Liu
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuhan Zhang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qingyan Chen
- Biology Department, Boston University, Boston, Massachusetts, USA
| | - Jiwu Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Chang CN, Kioussi C. Location, Location, Location: Signals in Muscle Specification. J Dev Biol 2018; 6:E11. [PMID: 29783715 PMCID: PMC6027348 DOI: 10.3390/jdb6020011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 12/15/2022] Open
Abstract
Muscles control body movement and locomotion, posture and body position and soft tissue support. Mesoderm derived cells gives rise to 700 unique muscles in humans as a result of well-orchestrated signaling and transcriptional networks in specific time and space. Although the anatomical structure of skeletal muscles is similar, their functions and locations are specialized. This is the result of specific signaling as the embryo grows and cells migrate to form different structures and organs. As cells progress to their next state, they suppress current sequence specific transcription factors (SSTF) and construct new networks to establish new myogenic features. In this review, we provide an overview of signaling pathways and gene regulatory networks during formation of the craniofacial, cardiac, vascular, trunk, and limb skeletal muscles.
Collapse
Affiliation(s)
- Chih-Ning Chang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
- Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, OR 97331, USA.
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
- Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
9
|
Powell D, Velleman S, Cowieson A, Singh M, Muir W. Influence of chick hatch time and access to feed on broiler muscle development. Poult Sci 2016; 95:1433-48. [DOI: 10.3382/ps/pew047] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/10/2016] [Indexed: 11/20/2022] Open
|
10
|
Peng G, Masood K, Gantz O, Sinha U. Neuromuscular electrical stimulation improves radiation-induced fibrosis through Tgf-Β1/MyoD homeostasis in head and neck cancer. J Surg Oncol 2016; 114:27-31. [DOI: 10.1002/jso.24265] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/01/2016] [Indexed: 11/05/2022]
Affiliation(s)
- Grace Peng
- Department of Otolaryngology-Head and Neck Surgery; University of Southern California; Los Angeles California
| | - Kamil Masood
- Department of Public Health; Keck School of Medicine; University of Southern California; Los Angeles California
| | - Oliver Gantz
- Department of Otolaryngology-Head and Neck Surgery; University of Southern California; Los Angeles California
| | - Uttam Sinha
- Department of Otolaryngology-Head and Neck Surgery; University of Southern California; Los Angeles California
| |
Collapse
|
11
|
Lee KP, Shin YJ, Panda AC, Abdelmohsen K, Kim JY, Lee SM, Bahn YJ, Choi JY, Kwon ES, Baek SJ, Kim SY, Gorospe M, Kwon KS. miR-431 promotes differentiation and regeneration of old skeletal muscle by targeting Smad4. Genes Dev 2015. [PMID: 26215566 PMCID: PMC4536309 DOI: 10.1101/gad.263574.115] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Lee et al. show that elevating miR-431 improved the myogenic capacity of old myoblasts, while inhibiting endogenous miR-431 lowered myogenesis. In an in vivo model of muscle regeneration following cardiotoxin injury, ectopic miR-431 injection greatly improved muscle regeneration and reduced SMAD4 levels. The myogenic capacity of myoblasts decreases in skeletal muscle with age. In addition to environmental factors, intrinsic factors are important for maintaining the regenerative potential of muscle progenitor cells, but their identities are largely unknown. Here, comparative analysis of microRNA (miRNA) expression profiles in young and old myoblasts uncovered miR-431 as a novel miRNA showing markedly reduced abundance in aged myoblasts. Importantly, elevating miR-431 improved the myogenic capacity of old myoblasts, while inhibiting endogenous miR-431 lowered myogenesis. Bioinformatic and biochemical analyses revealed that miR-431 directly interacted with the 3′ untranslated region (UTR) of Smad4 mRNA, which encodes one of the downstream effectors of TGF-β signaling. In keeping with the low levels of miR-431 in old myoblasts, SMAD4 levels increased in this myoblast population. Interestingly, in an in vivo model of muscle regeneration following cardiotoxin injury, ectopic miR-431 injection greatly improved muscle regeneration and reduced SMAD4 levels. Consistent with the finding that the mouse miR-431 seed sequence in the Smad4 3′ UTR is conserved in the human SMAD4 3′ UTR, inhibition of miR-431 also repressed the myogenic capacity of human skeletal myoblasts. Taken together, our results suggest that the age-associated miR-431 plays a key role in maintaining the myogenic ability of skeletal muscle with age.
Collapse
Affiliation(s)
- Kwang-Pyo Lee
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Yeo Jin Shin
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Department of Functional Genomics, Korea University of Science and Technology, Daejeon 305-333, Republic of Korea
| | - Amaresh C Panda
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Ji Young Kim
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Seung-Min Lee
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Young Jae Bahn
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Jeong Yi Choi
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Eun-Soo Kwon
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Su-Jin Baek
- Department of Functional Genomics, Korea University of Science and Technology, Daejeon 305-333, Republic of Korea; Genome Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Seon-Young Kim
- Department of Functional Genomics, Korea University of Science and Technology, Daejeon 305-333, Republic of Korea; Genome Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Myriam Gorospe
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Ki-Sun Kwon
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Department of Functional Genomics, Korea University of Science and Technology, Daejeon 305-333, Republic of Korea
| |
Collapse
|
12
|
Fu X, Wang H, Hu P. Stem cell activation in skeletal muscle regeneration. Cell Mol Life Sci 2015; 72:1663-77. [PMID: 25572293 PMCID: PMC4412728 DOI: 10.1007/s00018-014-1819-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/21/2014] [Accepted: 12/22/2014] [Indexed: 12/31/2022]
Abstract
Muscle stem cell (satellite cell) activation post muscle injury is a transient and critical step in muscle regeneration. It is regulated by physiological cues, signaling molecules, and epigenetic regulatory factors. The mechanisms that coherently turn on the complex activation process shortly after trauma are just beginning to be illuminated. In this review, we will discuss the current knowledge of satellite cell activation regulation.
Collapse
Affiliation(s)
- Xin Fu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | | | | |
Collapse
|
13
|
de Mello F, Streit DP, Sabin N, Gabillard JC. Dynamic expression of tgf-β2, tgf-β3 and inhibin βA during muscle growth resumption and satellite cell differentiation in rainbow trout (Oncorhynchus mykiss). Gen Comp Endocrinol 2015; 210:23-9. [PMID: 25449661 DOI: 10.1016/j.ygcen.2014.10.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/15/2014] [Accepted: 10/17/2014] [Indexed: 02/07/2023]
Abstract
Members of the TGF-β superfamily are involved in numerous cell functions; however, except for myostatin, their roles in the regulation of muscle growth in fish are completely unknown. We measured tgf-β1, tgf-β2, tgf-β3, inhibin βA (inh) and follistatin (fst) gene expression during muscle growth recovery following a fasting period. We observed that tgf-β1a and tgf-β2 expression were quickly down-regulated after refeeding and that tgf-β3 reached its highest level of expression 7days post-refeeding, mirroring myogenin expression. Inh βA1 mRNA levels decreased sharply after refeeding, in contrast to fst b2 expression, which peaked at day 2. No significant modification of expression was observed for tgf-β1a, tgf-β1b, tgf-β1c and tgf-β6 during refeeding. In vitro, tgf-β2 and inh βA1 expression decreased during the differentiation of satellite cells, whereas tgf-β3 expression increased following the same pattern as myogenin. Surprisingly, fst b1 and fst b2 expression decreased during differentiation, whereas no variation was observed in fst a1 and fst a2 expression levels. In vitro analyses also indicated that IGF1 treatment up-regulated tgf-β3, inh βA1 and myogenin expression, and that MSTN treatment increased fst b1 and fst b2 expression. In conclusion, we showed that the expression of tgf-β2, tgf-β3 and inh βA1 is dynamically regulated during muscle growth resumption and satellite cell differentiation, strongly suggesting that these genes have a role in the regulation of muscle growth.
Collapse
Affiliation(s)
- Fernanda de Mello
- Federal University of Rio Grande do Sul, Faculty of Agricultural Sciences, Research Group AQUAM, Aquaculture Sector, Department of Animal Science, Avenue Bento Gonçalves 7712, Agronomia, Porto Alegre 91540-000, RS, Brazil
| | - Danilo Pedro Streit
- Federal University of Rio Grande do Sul, Faculty of Agricultural Sciences, Research Group AQUAM, Aquaculture Sector, Department of Animal Science, Avenue Bento Gonçalves 7712, Agronomia, Porto Alegre 91540-000, RS, Brazil
| | - Nathalie Sabin
- INRA, UR1037 Laboratory of Fish Physiology and Genomic, Growth and Flesh Quality Group, campus de Beaulieu, 35000 Rennes, France
| | - Jean-Charles Gabillard
- INRA, UR1037 Laboratory of Fish Physiology and Genomic, Growth and Flesh Quality Group, campus de Beaulieu, 35000 Rennes, France.
| |
Collapse
|
14
|
Khanna N, Ge Y, Chen J. MicroRNA-146b promotes myogenic differentiation and modulates multiple gene targets in muscle cells. PLoS One 2014; 9:e100657. [PMID: 24956113 PMCID: PMC4067360 DOI: 10.1371/journal.pone.0100657] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/26/2014] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs are established as crucial modulators of skeletal myogenesis, but our knowledge about their identity and targets remains limited. In this study, we have identified microRNA-146b (miR-146b) as a novel regulator of skeletal myoblast differentiation. Following up on a previous microRNA profiling study, we establish that the expression of miR-146b is up-regulated during myoblast differentiation in vitro and muscle regeneration in vivo. Inhibition of miR-146b led to reduced myoblast differentiation, whereas overexpression of miR-146b enhanced differentiation. Computational prediction combined with gene expression information has revealed candidates for miR-146b targets in muscles. Among them, the expression of Smad4, Notch1, and Hmga2 are significantly suppressed by miR-146b overexpression in myocytes. In addition, expression levels of Smad4, Notch1 and Hmga2 are decreased during myoblast differentiation and muscle regeneration, inversely correlating to the levels of miR-146b. Importantly, inhibition of endogenous miR-146b prevents the down-regulation of Smad4, Notch1 and Hmga2 during differentiation. Furthermore, miR-146b directly targets the microRNA response elements (MREs) in the 3'UTR of those genes as assessed by reporter assays. Reporters with the seed regions of MREs mutated are insensitive to miR-146b, further confirming the specificity of targeting. In conclusion, miR-146b is a positive regulator of myogenic differentiation, possibly acting through multiple targets.
Collapse
Affiliation(s)
- Nidhi Khanna
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Yejing Ge
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Jie Chen
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
15
|
Zeng QJ, Wang LN, Shu G, Wang SB, Zhu XT, Gao P, Xi QY, Zhang YL, Zhang ZQ, Jiang QY. Decorin-induced proliferation of avian myoblasts involves the myostatin/Smad signaling pathway. Poult Sci 2014; 93:138-46. [PMID: 24570433 DOI: 10.3382/ps.2013-03300] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Decorin, a small leucine-rich proteoglycan as a component of the extracellular matrix, plays an important role in the skeletal muscle development. It has been reported that decorin promoted proliferation and differentiation of muscle cells by restraining myostatin activity in rodents. However, the effects and mechanisms of decorin on avian myoblast proliferation are not understood clearly. Thus, in our research, decorin overexpressing and knocking-down quail myoblast-7 (QM7) myoblasts were established to explore the effects of decorin on avian myoblast proliferation by flow cytometry. The results showed that overexpression of decorin enhanced the proliferation of QM7 myoblasts, which was accompanied by the upregulation of follistatin and primary muscle regulatory factors (i.e., myogenic factor 5, myogenic factor 1, myogenin), and downregulation of myostatin expression, as well as the decreased phosphorylation level of SMAD family member 3 (Smad3). In line with expectations, decorin RNAi displayed an opposite effect on the proliferation and gene expression pattern of QM7 cells. In conclusion, our in vitro studies suggested the decorin-mediated myostatin/Smad signaling pathway might be involved in the regulation of avian myoblast proliferation.
Collapse
Affiliation(s)
- Q J Zeng
- College of Animal Science, ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong, 510642, P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Cohen TV, Gnocchi VF, Cohen JE, Phadke A, Liu H, Ellis JA, Foisner R, Stewart CL, Zammit PS, Partridge TA. Defective skeletal muscle growth in lamin A/C-deficient mice is rescued by loss of Lap2α. Hum Mol Genet 2013; 22:2852-69. [PMID: 23535822 DOI: 10.1093/hmg/ddt135] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mutations in lamin A/C result in a range of tissue-specific disorders collectively called laminopathies. Of these, Emery-Dreifuss and Limb-Girdle muscular dystrophy 1B mainly affect striated muscle. A useful model for understanding both laminopathies and lamin A/C function is the Lmna(-/-) mouse. We found that skeletal muscle growth and muscle satellite (stem) cell proliferation were both reduced in Lmna(-/-) mice. Lamins A and C associate with lamina-associated polypeptide 2 alpha (Lap2α) and the retinoblastoma gene product, pRb, to regulate cell cycle exit. We found Lap2α to be upregulated in Lmna(-/-) myoblasts (MBs). To specifically test the contribution of elevated Lap2α to the phenotype of Lmna(-/-) mice, we generated Lmna(-/-)Lap2α(-/-) mice. Lifespan and body mass were increased in Lmna(-/-)Lap2α(-/-) mice compared with Lmna(-/-). Importantly, the satellite cell proliferation defect was rescued, resulting in improved myogenesis. Lmna(-/-) MBs also exhibited increased levels of Smad2/3, which were abnormally distributed in the cell and failed to respond to TGFβ1 stimulation as in control cells. However, using SIS3 to inhibit signaling via Smad3 reduced cell death and augmented MB fusion. Together, our results show that perturbed Lap2α/pRb and Smad2/3 signaling are important regulatory pathways mediating defective muscle growth in Lmna(-/-) mice, and that inhibition of either pathway alone or in combination can ameliorate this deleterious phenotype.
Collapse
Affiliation(s)
- Tatiana V Cohen
- Research Center for Genetic Medicine, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Dey BK, Gagan J, Yan Z, Dutta A. miR-26a is required for skeletal muscle differentiation and regeneration in mice. Genes Dev 2012; 26:2180-91. [PMID: 23028144 PMCID: PMC3465739 DOI: 10.1101/gad.198085.112] [Citation(s) in RCA: 164] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 08/15/2012] [Indexed: 12/15/2022]
Abstract
Multiple microRNAs are known to be induced during the differentiation of myoblasts to myotubes. Yet, experiments in animals have not provided clear evidence for the requirement of most of these microRNAs in myogenic differentiation in vivo. miR-26a is induced during skeletal muscle differentiation and is predicted to target a well-known inhibitor of differentiation, the transforming growth factor β/bone morphogenetic protein (TGF-β/BMP) signaling pathway. Here we show that exogenous miR-26a promotes differentiation of myoblasts, while inhibition of miR-26a by antisense oligonucleotides or by Tough-Decoys delays differentiation. miR-26a targets the transcription factors Smad1 and Smad4, critical for the TGF-β/BMP pathway, and expression of microRNA-resistant forms of these transcription factors inhibits differentiation. Injection of antagomirs specific to miR-26a into neonatal mice derepressed both Smad expression and activity and consequently inhibited skeletal muscle differentiation. In addition, miR-26a is induced during skeletal muscle regeneration after injury. Inhibiting miR-26a in the tibialis anterior muscles through the injection of adeno-associated virus expressing a Tough-Decoy targeting miR-26a prevents Smad down-regulation and delays regeneration. These findings provide evidence for the requirement of miR-26a for skeletal muscle differentiation and regeneration in vivo.
Collapse
Affiliation(s)
- Bijan K. Dey
- Department of Biochemistry and Molecular Genetics
| | | | - Zhen Yan
- Department of Medicine and Pharmacology
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | |
Collapse
|
18
|
Olguín HC, Pisconti A. Marking the tempo for myogenesis: Pax7 and the regulation of muscle stem cell fate decisions. J Cell Mol Med 2012; 16:1013-25. [PMID: 21615681 PMCID: PMC4365881 DOI: 10.1111/j.1582-4934.2011.01348.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Post-natal growth and regeneration of skeletal muscle is highly dependent on a population of resident myogenic precursors known as satellite cells. Transcription factors from the Pax gene family, Pax3 and Pax7, are critical for satellite cell biogenesis, survival and potentially self-renewal; however, the underlying molecular mechanisms remain unsolved. This is particularly true in the case of Pax7, which appears to regulate myogenesis at multiple levels. Accordingly, recent data have highlighted the importance of a functional relationship between Pax7 and the MyoD family of muscle regulatory transcription factors during normal muscle formation and disease. Here we will critically review key findings suggesting that Pax7 may play a dual role by promoting resident muscle progenitors to commit to the skeletal muscle lineage while preventing terminal differentiation, thus keeping muscle progenitors poised to differentiate upon environmental cues. In addition, potential regulatory mechanisms for the control of Pax7 activity will be proposed.
Collapse
Affiliation(s)
- Hugo C Olguín
- Departamento Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | |
Collapse
|
19
|
Fakhfakh R, Lamarre Y, Skuk D, Tremblay JP. Losartan Enhances the Success of Myoblast Transplantation. Cell Transplant 2012; 21:139-52. [PMID: 21535912 DOI: 10.3727/096368911x576045] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Duchenne muscular dystrophy is a recessive X-linked genetic disease caused by dystrophin gene mutations. Cell therapy can be a potential approach aiming to introduce a functional dystrophin in the dystrophic patient myofibers. However, this strategy produced so far limited results. Transforming growth factor-β (TGF-β) is a negative regulator of skeletal muscle development and is responsible for limiting myogenic regeneration. The combination of TGF-β signaling inhibition with myoblast transplantation can be an effective therapeutic approach in dystrophin-deficient patients. Our aim was to verify whether the success of human myoblast transplantation in immunodeficient dystrophic mice is enhanced with losartan, a molecule that downregulates TGF-β expression. In vitro, blocking TGF-β activity with losartan increased proliferation and fusion and decreased apoptosis in human myoblasts. In vivo, human myoblasts were transplanted in mice treated with oral losartan. Immunodetection of human dystrophin in tibialis anterior cross sections 1 month posttransplantation revealed more human dystrophin-positive myofibers in these mice than in nontreated dystrophic mice. Thus, blocking the TGF-β signal with losartan treatment improved the success of myoblast transplantation probably by increasing myoblast proliferation and fusion, decreasing macrophage activation, and changing the expression of myogenic regulator factors.
Collapse
Affiliation(s)
- Raouia Fakhfakh
- Unité de recherche de recherche en Génétique Humaine, Centre de recherche de CHUL, CHUQ, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Yann Lamarre
- Unité de recherche de recherche en Génétique Humaine, Centre de recherche de CHUL, CHUQ, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Daniel Skuk
- Unité de recherche de recherche en Génétique Humaine, Centre de recherche de CHUL, CHUQ, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Jacques P. Tremblay
- Unité de recherche de recherche en Génétique Humaine, Centre de recherche de CHUL, CHUQ, Faculté de médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
20
|
Inhibiting TGF-β activity improves respiratory function in mdx mice. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2611-21. [PMID: 21641384 DOI: 10.1016/j.ajpath.2011.02.024] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 01/26/2011] [Accepted: 02/03/2011] [Indexed: 11/23/2022]
Abstract
Respiratory function is the main cause of mortality in patients with Duchenne muscular dystrophy (DMD). Elevated levels of TGF-β play a key role in the pathophysiology of DMD. To determine whether therapeutic attenuation of TGF-β signaling improves respiratory function, mdx mice were treated from 2 weeks of age to 2 months or 9 months of age with either 1D11 (a neutralizing antibody to all three isoforms of TGF-β), losartan (an angiotensin receptor antagonist), or a combination of the two agents. Respiratory function was measured in nonanesthetized mice by plethysmography. The 9-month-old mdx mice had elevated Penh values and decreased breathing frequency, due primarily to decreased inspiratory flow rate. All treatments normalized Penh values and increased peak inspiratory flow, leading to decreased inspiration times and breathing frequency. Additionally, forelimb grip strength was improved after 1D11 treatment at both 2 and 9 months of age, whereas, losartan improved grip strength only at 2 months. Decreased serum creatine kinase levels (significant improvement for all groups), increased diaphragm muscle fiber density, and decreased hydroxyproline levels (significant improvement for 1D11 only) also suggested improved muscle function after treatment. For all endpoints, 1D11 was equivalent or superior to losartan; coadministration of the two agents was not superior to 1D11 alone. In conclusion, TGF-β antagonism may be a useful therapeutic approach for treating DMD patients.
Collapse
|
21
|
Secretome Analysis of Skeletal Myogenesis Using SILAC and Shotgun Proteomics. INTERNATIONAL JOURNAL OF PROTEOMICS 2011; 2011:329467. [PMID: 22084683 PMCID: PMC3200090 DOI: 10.1155/2011/329467] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 01/26/2011] [Indexed: 12/18/2022]
Abstract
Myogenesis, the formation of skeletal muscle, is a multistep event that commences with myoblast proliferation, followed by cell-cycle arrest, and finally the formation of multinucleated myotubes via fusion of mononucleated myoblasts. Each step is orchestrated by well-documented intracellular factors, such as cytoplasmic signalling molecules and nuclear transcription factors. Regardless, the key step in getting a more comprehensive understanding of the regulation of myogenesis is to explore the extracellular factors that are capable of eliciting the downstream intracellular factors. This could further provide valuable insight into the acute cellular response to extrinsic cues in maintaining normal muscle development. In this paper, we survey the intracellular factors that respond to extracellular cues that are responsible for the cascades of events during myogenesis: myoblast proliferation, cell-cycle arrest of myoblasts, and differentiation of myoblasts into myotubes. This focus on extracellular perspective of muscle development illustrates our mass spectrometry-based proteomic approaches to identify differentially expressed secreted factors during skeletal myogenesis.
Collapse
|
22
|
Chan CYX, Masui O, Krakovska O, Belozerov VE, Voisin S, Ghanny S, Chen J, Moyez D, Zhu P, Evans KR, McDermott JC, Siu KWM. Identification of differentially regulated secretome components during skeletal myogenesis. Mol Cell Proteomics 2011; 10:M110.004804. [PMID: 21343469 DOI: 10.1074/mcp.m110.004804] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myogenesis is a well-characterized program of cellular differentiation that is exquisitely sensitive to the extracellular milieu. Systematic characterization of the myogenic secretome (i.e. the ensemble of secreted proteins) is, therefore, warranted for the identification of novel secretome components that regulate both the pluripotency of these progenitor mesenchymal cells, and also their commitment and passage through the differentiation program. Previously, we have successfully identified 26 secreted proteins in the mouse skeletal muscle cell line C2C12 (1). In an effort to attain a more comprehensive picture of the regulation of myogenesis by its extracellular milieu, quantitative profiling employing stable isotope labeling by amino acids in cell culture was implemented in conjunction with two parallel high throughput online reverse phase liquid chromatography-tandem mass spectrometry systems. In summary, 34 secreted proteins were quantified, 30 of which were shown to be differentially expressed during muscle development. Intriguingly, our analysis has revealed several novel up- and down-regulated secretome components that may have critical biological relevance for both the maintenance of pluripotency and the passage of cells through the differentiation program. In particular, the altered regulation of secretome components, including follistatin-like protein-1, osteoglycin, spondin-2, and cytokine-induced apoptosis inhibitor-1, along with constitutively expressed factors, such as fibulin-2, illustrate dynamic changes in the secretome that take place when differentiation to a specific lineage occurs.
Collapse
Affiliation(s)
- C Y X'avia Chan
- Department of Biology, York University, 4700 Keele Street, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Winbanks CE, Wang B, Beyer C, Koh P, White L, Kantharidis P, Gregorevic P. TGF-beta regulates miR-206 and miR-29 to control myogenic differentiation through regulation of HDAC4. J Biol Chem 2011; 286:13805-14. [PMID: 21324893 DOI: 10.1074/jbc.m110.192625] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRs) are emerging as prominent players in the regulation of many biological processes, including myogenic commitment and skeletal muscle formation. Members of the TGF-β family can influence the proliferation and myogenic differentiation of cells, although it is presently not clear what role miRNAs play in the TGF-β-mediated control of myogenic differentiation. Here, we demonstrate in the myogenic C2C12 cell line, and in primary muscle cells, that miR-206 and miR-29-two miRs that act on transcriptional events implicated in muscle differentiation are down-regulated by TGF-β. We further demonstrate that TGF-β treatment of myogenic cells is associated with increased expression of histone deacetylase 4 (HDAC4), a key inhibitor of muscle differentiation that has been identified as a target for regulation by miR-206 and miR-29. We confirmed that increased expression of miR-206 and miR-29 resulted in the translational repression of HDAC4 in the presence or absence of TGF-β via interaction with the HDAC4 3'-untranslated region. Importantly, we found that miR-206 and miR-29 can attenuate the inhibitory actions of TGF-β on myogenic differentiation. Furthermore, we present evidence that the mechanism by which miR-206 and miR-29 can inhibit the TGF-β-mediated up-regulation of HDAC4 is via the inhibition of Smad3 expression, a transducer of TGF-β signaling. These findings identify a novel mechanism of interaction between TGF-β and miR-206 and -29 in the regulation of myogenic differentiation through HDAC4.
Collapse
Affiliation(s)
- Catherine E Winbanks
- Division of Metabolism and Obesity, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 8008, Australia
| | | | | | | | | | | | | |
Collapse
|
24
|
Ciemerych MA, Archacka K, Grabowska I, Przewoźniak M. Cell cycle regulation during proliferation and differentiation of mammalian muscle precursor cells. Results Probl Cell Differ 2011; 53:473-527. [PMID: 21630157 DOI: 10.1007/978-3-642-19065-0_20] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Proliferation and differentiation of muscle precursor cells are intensively studied not only in the developing mouse embryo but also using models of skeletal muscle regeneration or analyzing in vitro cultured cells. These analyses allowed to show the universality of the cell cycle regulation and also uncovered tissue-specific interplay between major cell cycle regulators and factors crucial for the myogenic differentiation. Examination of the events accompanying proliferation and differentiation leading to the formation of functional skeletal muscle fibers allows understanding the molecular basis not only of myogenesis but also of skeletal muscle regeneration. This chapter presents the basis of the cell cycle regulation in proliferating and differentiating muscle precursor cells during development and after muscle injury. It focuses at major cell cycle regulators, myogenic factors, and extracellular environment impacting on the skeletal muscle.
Collapse
Affiliation(s)
- Maria A Ciemerych
- Department of Cytology, Institute of Zoology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland.
| | | | | | | |
Collapse
|
25
|
Zhang L, Ran L, Garcia GE, Wang XH, Han S, Du J, Mitch WE. Chemokine CXCL16 regulates neutrophil and macrophage infiltration into injured muscle, promoting muscle regeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:2518-27. [PMID: 19893053 DOI: 10.2353/ajpath.2009.090275] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Only a few specific chemokines that mediate interactions between inflammatory and satellite cells in muscle regeneration have been identified. The chemokine CXCL16 differs from other chemokines because it has both a transmembrane region and active, soluble chemokine forms. Indeed, we found increased expression of CXCL16 and its receptor, CXCR6, in regenerating myofibers. Muscle regeneration in CXCL16-deficient (CXCL16KO) mice was severely impaired compared with regeneration in wild-type mice. In addition, there was decreased MyoD and myogenin expression in regenerating muscle in CXCL16KO mice, indicating impaired satellite cell proliferation and differentiation. After 1 month, new myofibers in CXCL16KO mice remained significantly smaller than those in muscle of wild-type mice. To understand how CXCL16 regulates muscle regeneration, we examined cells infiltrating injured muscle. There were more infiltrating neutrophils and fewer macrophages in injured muscle of CXCL16KO mice compared with events in wild-type mice. Moreover, absence of CXCL16 led to different expression of cytokines/chemokines in injured muscles: mRNAs of macrophage-inflammatory protein (MIP)-1alpha, MIP-1beta, and MIP-2 were increased, whereas regulated on activation normal T cell expressed and secreted, T-cell activation-3, and monocyte chemoattractant protein-1 mRNAs were lower compared with results in muscles of wild-type mice. Impaired muscle regeneration in CXCL16KO mice also resulted in fibrosis, which was linked to transforming growth factor-beta1 expression. Thus, CXCL16 expression is a critical mediator of muscle regeneration, and it suppresses the development of fibrosis.
Collapse
Affiliation(s)
- Liping Zhang
- Nephrology Division, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Li X, McFarland DC, Velleman SG. Transforming growth factor-beta1-induced satellite cell apoptosis in chickens is associated with beta1 integrin-mediated focal adhesion kinase activation. Poult Sci 2009; 88:1725-34. [PMID: 19590089 DOI: 10.3382/ps.2008-00534] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transforming growth factor-beta1 (TGF-beta1) induces apoptosis in many types of cells. The cell adhesion receptor, beta1 integrin subunit, prevents apoptosis and may be involved in TGF-beta1-induced muscle cell apoptosis. In the current study, chicken primary satellite cells, myogenic precursors, were used to investigate the apoptotic effect of TGF-beta1 on muscle cells. The data from the current study showed that the addition of exogenous TGF-beta1 reduced beta1 integrin expression and altered its localization. Treatment of the satellite cells with TGF-beta1 increased the number of apoptotic cells indicated by annexin-V using flow cytometry. The number of caspase-positive cells was increased in the TGF-beta1-treated immunostained cells, which supported that TGF-beta1 induced satellite cell apoptosis. It has been shown that beta1 integrin is involved in muscle cell survival. In response to the activation of beta1 integrin, focal adhesion kinase (FAK) phosphorylates tyrosine at residue 397 and activates cell survival signal transduction. The phosphorylation of FAK was significantly reduced from 30 min to 4 h after TGF-beta1 treatment during both satellite cell proliferation and differentiation. These data suggested that the apoptotic effect of TGF-beta1 on satellite cells is likely associated with a beta1 integrin-mediated FAK signaling pathway during satellite cell proliferation and differentiation.
Collapse
Affiliation(s)
- X Li
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA
| | | | | |
Collapse
|
27
|
Abstract
Lipases are acyl hydrolases that represent a diverse group of enzymes present in organisms ranging from prokaryotes to humans. This article focuses on an evolutionarily related family of extracellular lipases that include lipoprotein lipase, hepatic lipase and endothelial lipase. As newly synthesized proteins, these lipases undergo a series of co- and post-translational maturation steps occurring in the endoplasmic reticulum, including glycosylation and glycan processing, and protein folding and subunit assembly. This article identifies and discusses mechanisms that direct early and late events in lipase folding and assembly. Lipase maturation employs the two general chaperone systems operating in the endoplasmic reticulum, as well as a recently identified lipase-specific chaperone termed lipase maturation factor 1. We propose that the two general chaperone systems act in a coordinated manner early in lipase maturation in order to help create partially folded monomers; lipase maturation factor 1 then facilitates final monomer folding and subunit assembly into fully functional homodimers. Once maturation is complete, the lipases exit the endoplasmic reticulum and are secreted to extracellular sites, where they carry out a number of functions related to lipoprotein and lipid metabolism.
Collapse
Affiliation(s)
- Mark H Doolittle
- VA Greater Los Angeles, Healthcare System, 11301 Wilshire Blvd, Bldg 113, Rm 312, Los Angeles, CA 90073, USA, Tel.: +1 661 433 6349
| | | |
Collapse
|
28
|
Li X, Velleman S. Effect of transforming growth factor-β1 on embryonic and posthatch muscle growth and development in normal and low score normal chicken. Poult Sci 2009; 88:265-75. [DOI: 10.3382/ps.2008-00234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
29
|
Kim HJ, Archer E, Escobedo N, Tapscott SJ, Unguez GA. Inhibition of mammalian muscle differentiation by regeneration blastema extract of Sternopygus macrurus. Dev Dyn 2008; 237:2830-43. [PMID: 18816861 DOI: 10.1002/dvdy.21702] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Tissue regeneration through stem cell activation and/or cell dedifferentiation is widely distributed across the animal kingdom. By comparison, regeneration in mammals is poor and this may reflect a limited dedifferentiation potential of mature cells. Because mammalian myotubes can dedifferentiate in the presence of newt blastema extract, the present study tested the dedifferentiation induction capability of the blastema from the teleost Sternopygus macrurus (SmBE). Our in vitro data showed that SmBE did not induce cell cycle reentry of myonuclei in myotubes. Instead, SmBE caused myotubes to detach and time-lapse imaging analyses characterized the cellular events before their detachment. Furthermore, SmBE enhanced myoblast proliferation and reversibly inhibited their differentiation. These data suggest the presence of protein factors in SmBE that regulate mammalian muscle physiology and differentiation, but do not support the conservation of a dedifferentiation induction capability by the blastema of S. macrurus.
Collapse
Affiliation(s)
- Hyun-Jung Kim
- Biology Department, New Mexico State University, Las Cruces, New Mexico, USA
| | | | | | | | | |
Collapse
|
30
|
Li X, McFarland DC, Velleman SG. Effect of Smad3-mediated transforming growth factor-beta1 signaling on satellite cell proliferation and differentiation in chickens. Poult Sci 2008; 87:1823-33. [PMID: 18753451 DOI: 10.3382/ps.2008-00133] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transforming growth factor-beta1 (TGF-beta1) is a potent inhibitor of muscle cell proliferation and differentiation. The TGF-beta1 signal is carried by Smad proteins into the cell nucleus, resulting in the regulation of the expression of key myogenic regulatory factors including MyoD and myogenin during myogenesis. However, to date, the molecular mechanism of the inhibition by Smad-mediated TGF-beta1 signaling on the function of the myogenic regulatory factors has not been well understood. The present study was designed to investigate the effect of TGF-beta1 on satellite cell proliferation and differentiation by a Smad3-dependent signaling pathway. A chicken line, low score normal (LSN) with reduced muscling and upregulated TGF-beta1 expression, was used and compared with a normal chicken line. In LSN satellite cell cultures, both MyoD and myogenin expression was significantly decreased compared with the normal cells. Furthermore, in response to exogenous TGF-beta1, the normal satellite cells had a significant decrease in both MyoD and myogenin expression, which suggests that TGF-beta1 inhibited MyoD and myogenin expression, resulting in decreased satellite cell proliferation and differentiation. The expression of Smad3 and Smad7, key proteins of the Smad family, was greater in the LSN cultures than that measured in the normal culture. The addition of TGF-beta1 reduced Smad3 expression, but did not affect the expression of Smad7. The reduction of Smad3 in response to TGF-beta1 suggests that a negative regulatory feedback is likely involved in LSN satellite cell proliferation and differentiation. The overexpression of Smad3 inhibited both MyoD and myogenin expression in normal and LSN satellite cells. In contrast, the underexpression of Smad3 increased the expression of MyoD and myogenin in the LSN cells. However, in the normal cells, only myogenin expression was increased by Smad3 overexpression, but not MyoD. These data together suggest that LSN satellite cells are more responsive to a Smad3-dependent TGF-beta1 signaling pathway than normal satellite cells, and a Smad3-independent pathway is also likely involved in the regulation of satellite cell proliferation and differentiation.
Collapse
Affiliation(s)
- X Li
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster 44691, USA
| | | | | |
Collapse
|
31
|
Kollias HD, McDermott JC. Transforming growth factor-beta and myostatin signaling in skeletal muscle. J Appl Physiol (1985) 2007; 104:579-87. [PMID: 18032576 DOI: 10.1152/japplphysiol.01091.2007] [Citation(s) in RCA: 223] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The superfamily of transforming growth factor-beta (TGF-beta) cytokines has been shown to have profound effects on cellular proliferation, differentiation, and growth. Recently, there have been major advances in our understanding of the signaling pathway(s) conveying TGF-beta signals to the nucleus to ultimately control gene expression. One tissue that is potently influenced by TGF-beta superfamily signaling is skeletal muscle. Skeletal muscle ontogeny and postnatal physiology have proven to be exquisitely sensitive to the TGF-beta superfamily cytokine milieu in various animal systems from mice to humans. Recently, major strides have been made in understanding the role of TGF-beta and its closely related family member, myostatin, in these processes. In this overview, we will review recent advances in our understanding of the TGF-beta and myostatin signaling pathways and, in particular, focus on the implications of this signaling pathway for skeletal muscle development, physiology, and pathology.
Collapse
Affiliation(s)
- Helen D Kollias
- Department of Neurology, Johns Hopkins Hospital, Baltimore, MD, USA
| | | |
Collapse
|
32
|
Abstract
Somatic stem cell populations participate in the development and regeneration of their host tissues. Skeletal muscle is capable of complete regeneration due to stem cells that reside in skeletal muscle and nonmuscle stem cell populations. However, in severe myopathic diseases such as Duchenne Muscular Dystrophy, this regenerative capacity is exhausted. In the present review, studies will be examined that focus on the origin, gene expression, and coordinated regulation of stem cell populations to highlight the regenerative capacity of skeletal muscle and emphasize the challenges for this field. Intense interest has focused on cell-based therapies for chronic, debilitating myopathic diseases. Future studies that enhance our understanding of stem cell biology and repair mechanisms will provide a platform for therapeutic applications directed toward these chronic, life-threatening diseases.
Collapse
Affiliation(s)
- Xiaozhong Shi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | |
Collapse
|
33
|
Sterrenburg E, Turk R, 't Hoen PAC, van Deutekom JCT, Boer JM, van Ommen GJB, den Dunnen JT. Large-scale gene expression analysis of human skeletal myoblast differentiation. Neuromuscul Disord 2004; 14:507-18. [PMID: 15336692 DOI: 10.1016/j.nmd.2004.03.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2004] [Revised: 03/16/2004] [Accepted: 03/24/2004] [Indexed: 10/26/2022]
Abstract
To study pathways involved in human skeletal myogenesis, we profiled gene expression in human primary myoblast cells derived from three individuals using both oligonucleotide and cDNA microarrays. Following stringent statistical testing (false-positive rate 0.4%), we identified 146 genes differentially expressed over time. Interestingly, 86 of these genes have not been reported to be involved in myogenesis in mouse cell lines. This demonstrates the additional value of human primary cell cultures in the study of muscle differentiation. Many of the identified genes play a role in muscle regeneration, indicating the close relationship of this process with muscle development. In addition, we found overlap with expression profiling studies in muscle from Duchenne muscular dystrophy patients, confirming ongoing muscle regeneration in Duchenne muscular dystrophy. Further study of these genes can bring new insights into the process of muscle differentiation, and they are candidate genes for neuromuscular disorders with an as yet unidentified cause.
Collapse
Affiliation(s)
- Ellen Sterrenburg
- Center for Human and Clinical Genetics, Leiden University Medical Center, Wassenaarseweg 72, 2333 AL Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
34
|
Füchtbauer EM. Inhibition of skeletal muscle development: less differentiation gives more muscle. Results Probl Cell Differ 2003; 38:143-61. [PMID: 12132393 DOI: 10.1007/978-3-540-45686-5_7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The fact that stem cells have to be protected from premature differentiation is true for many organs in the developing embryo and the adult organism. However, there are several arguments that this is particularly important for (skeletal) muscle. There are some evolutionary arguments that muscle is a "default" pathway for mesodermal cells, which has to be actively prevented in order to allow cells to differentiate into other tissues. Myogenic cells originate from very small areas of the embryo where only a minor portion of these cells is supposed to differentiate. Differentiated muscle fibres are unconditionally post-mitotic, leaving undifferentiated stem cells as the only source of regeneration. The mechanical usage of muscle and its superficial location in the vertebrate body makes regeneration a frequently used mechanism. Looking at the different inhibitory mechanisms that have been found within the past 10 or so years, it appears as if evolution has taken this issue very serious. At all possible levels we find regulatory mechanisms that help to fine tune the differentiation of myogenic cells. Secreted molecules specifying different populations of somitic cells, diffusing or membrane-bound signals among fellow myoblasts, modulating molecules within the extracellular matrix and last, but not least, a changing set of activating and repressing cofactors. We have come a long way from the simple model of MyoD just to be turned on at the right time in the right cell.
Collapse
Affiliation(s)
- Ernst-Martin Füchtbauer
- Institute of Molecular and Structural Biology, Aarhus University, C.F. Møllers Allé, Bygn. 130, Arhus C, Denmark
| |
Collapse
|
35
|
Liu D, Black BL, Derynck R. TGF-beta inhibits muscle differentiation through functional repression of myogenic transcription factors by Smad3. Genes Dev 2001; 15:2950-66. [PMID: 11711431 PMCID: PMC312830 DOI: 10.1101/gad.925901] [Citation(s) in RCA: 303] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Transforming growth factor-beta (TGF-beta) is a potent inhibitor of skeletal muscle differentiation, but the molecular mechanism and signaling events that lead to this inhibition are poorly characterized. Here we show that the TGF-beta intracellular effector Smad3, but not Smad2, mediates the inhibition of myogenic differentiation in MyoD-expressing C3H10T1/2 cells and C2C12 myoblasts by repressing the activity of the MyoD family of transcriptional factors. The Smad3-mediated repression was directed at the E-box sequence motif within muscle gene enhancers and the bHLH region of MyoD, the domain required for its association with E-protein partners such as E12 and E47. The repression could be overcome by supplying an excess of E12, and covalent tethering of E47 to MyoD rendered the E-box-dependent transcriptional activity refractory to the effects of Smad3 and TGF-beta. Smad3 physically interacted with the HLH domain of MyoD, and this interaction correlated with the ability of Smad3 to interfere with MyoD/E protein heterodimerization and binding of MyoD complexes to oligomerized E-box sites. Together, these results reveal a model for how TGF-beta, through Smad3-mediated transcriptional repression, inhibits myogenic differentiation.
Collapse
Affiliation(s)
- D Liu
- Department of Growth and Development, University of California at San Francisco, 94143, USA
| | | | | |
Collapse
|
36
|
Abstract
Adult skeletal muscle has a remarkable ability to regenerate following myotrauma. Because adult myofibers are terminally differentiated, the regeneration of skeletal muscle is largely dependent on a small population of resident cells termed satellite cells. Although this population of cells was identified 40 years ago, little is known regarding the molecular phenotype or regulation of the satellite cell. The use of cell culture techniques and transgenic animal models has improved our understanding of this unique cell population; however, the capacity and potential of these cells remain ill-defined. This review will highlight the origin and unique markers of the satellite cell population, the regulation by growth factors, and the response to physiological and pathological stimuli. We conclude by highlighting the potential therapeutic uses of satellite cells and identifying future research goals for the study of satellite cell biology.
Collapse
Affiliation(s)
- T J Hawke
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | |
Collapse
|
37
|
Bouché M, Canipari R, Melchionna R, Willems D, Senni MI, Molinaro M. TGF-beta autocrine loop regulates cell growth and myogenic differentiation in human rhabdomyosarcoma cells. FASEB J 2000; 14:1147-58. [PMID: 10834937 DOI: 10.1096/fasebj.14.9.1147] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Transforming growth factor beta (TGF) is a well-known inhibitor of myogenic differentiation as well as an autocrine product of rhabdomyosarcoma cells. We studied the role of the TGF-beta autocrine loop in regulating growth and myogenic differentiation in the human rhabdomyosarcoma cell line, RD. We previously reported that the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA) induces growth arrest and myogenic differentiation in these cells, which constitutively express muscle regulatory factors. We show that TPA inhibits the activation of secreted latent TGF-beta, thus decreasing the concentration of active TGF-beta to which the cells are exposed. This event is mediated by the TPA-induced alteration of the uPA/PAI serine-protease system. Complete removal of TGF-beta, mediated by the ectopic expression of a soluble type II TGF-beta receptor dominant negative cDNA, induces growth arrest, but does not trigger differentiation. In contrast, a reduction in the TGF-beta concentration, to a range of 0.14-0.20 x 10(-2) ng/ml (which is similar to that measured in TPA-treated cells), mimics TPA-induced differentiation. Taken together, these data demonstrate that cell growth and suppression of differentiation in rhabdomyosarcoma cells require overproduction of active TGF-beta; furthermore, they show that a 'critical' concentration of TGF-beta is necessary for myogenic differentiation to occur, whereas myogenesis is abolished below and above this concentration. By impairing the TGF-beta autocrine loop, TPA stabilizes the factor concentration within the range compatible for differentiation to occur. In contrast, in human primary muscle cells a much higher concentration of exogenous TGF-beta is required for the differentiation inhibitory effect and TPA inhibits differentiation in these cells probably through a TGF-beta independent mechanism. These data thus clarify the mechanism underlying the multiple roles of TGF-beta in the regulation of both the transformed and differentiated phenotype.
Collapse
MESH Headings
- Animals
- Aprotinin/pharmacology
- Autocrine Communication/drug effects
- Cell Differentiation/drug effects
- Cell Division/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- Humans
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Mutation/genetics
- Myosin Heavy Chains/metabolism
- Pepstatins/pharmacology
- Plasminogen/metabolism
- Plasminogen Inactivators/metabolism
- Protein Precursors/genetics
- Protein Precursors/metabolism
- Protein Processing, Post-Translational/drug effects
- Protein Serine-Threonine Kinases
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Rhabdomyosarcoma/metabolism
- Rhabdomyosarcoma/pathology
- Tetradecanoylphorbol Acetate/pharmacology
- Transfection
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/pharmacology
- Tumor Cells, Cultured
- Urokinase-Type Plasminogen Activator/antagonists & inhibitors
- Urokinase-Type Plasminogen Activator/metabolism
Collapse
Affiliation(s)
- M Bouché
- Department of Histology and Medical Embryology, University of Rome 'La Sapienza', 00161, Rome, Italy.
| | | | | | | | | | | |
Collapse
|
38
|
Adi S, Cheng ZQ, Zhang PL, Wu NY, Mellon SH, Rosenthal SM. Opposing early inhibitory and late stimulatory effects of insulin-like growth factor-I on myogenin gene transcription. J Cell Biochem 2000. [DOI: 10.1002/1097-4644(20000915)78:4<617::aid-jcb11>3.0.co;2-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Layne MD, Farmer SR. Tumor necrosis factor-alpha and basic fibroblast growth factor differentially inhibit the insulin-like growth factor-I induced expression of myogenin in C2C12 myoblasts. Exp Cell Res 1999; 249:177-87. [PMID: 10328964 DOI: 10.1006/excr.1999.4465] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tumor necrosis factor-alpha (TNF-alpha) plays a role in several disease states such as sepsis, cachexia, and non-insulin-dependent diabetes. TNF-alpha interferes with insulin signaling and inhibits differentiation-specific gene expression in adipose tissue and skeletal muscle. We have examined the mechanisms by which TNF-alpha, in comparison to basic fibroblast growth factor (bFGF), inhibits the insulin-like growth factor-I (IGF-I)-induced differentiation of C2C12 myoblasts. Adhesion of quiescent, suspended myoblasts to collagen in high concentrations of IGF-I (10 nM) induced these cells to proliferate during the initial 24 h postplating and in so doing transiently inhibited the expression of myogenin, an essential transcription factor controlling myoblast differentiation. Low doses of IGF-I (1 nM) were minimally mitogenic and enhanced muscle-specific gene expression. Quiescent myoblasts treated with bFGF in combination with IGF-I did not express myogenin, but expressed proliferating cell nuclear antigen and underwent DNA synthesis. In contrast, TNF-alpha in the presence or absence of 1 nM IGF-I, did not stimulate DNA synthesis in myoblasts. However, TNF-alpha inhibited myogenin mRNA and protein expression. Expression of the cyclin-dependent kinase inhibitor p21 correlated with myogenin expression and myoblast differentiation, but not with growth arrest. These results indicate that both TNF-alpha and bFGF inhibit myogenin expression but differentially influence myoblast proliferation.
Collapse
Affiliation(s)
- M D Layne
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | |
Collapse
|
40
|
Yamane A, Takahashi K, Mayo M, Vo H, Shum L, Zeichner-David M, Slavkin HC. Induced expression of myoD, myogenin and desmin during myoblast differentiation in embryonic mouse tongue development. Arch Oral Biol 1998; 43:407-16. [PMID: 9681116 DOI: 10.1016/s0003-9969(98)00018-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Significant progress has been made in defining mechanisms governing myogenesis at the transcriptional levels, but the extracellular signal-transduction pathways involved in myogenesis are not as yet defined. The developing mouse tongue provides a model for the regulation of myogenesis during precise time periods in embryogenesis. The molecular cues that regulate the close-range autocrine and/or paracrine signalling processes required for the fast-twitch complex tongue musculature are not known. This study was designed to test the hypothesis that transforming growth factor-alpha (TGF alpha) controls myogenesis in embryonic mouse tongue through the induction of myogenic regulatory factors such as myoD, myf5, myogenin and MRF4/myf6/herculin. To test this hypothesis, the effects of exogenous TGF alpha on the transcription of myoD, myf5, myogenin, MRF4 and desmin were examined in tongue samples from embryonic day-10.5 mandibular explants cultured in serum-free, chemically defined medium and then processed for competitive, reverse transcription-polymerase chain reaction. TGF alpha induced myoD, myogenin and desmin expression. Treatment with 20 and 40 ng/ml TGF alpha decreased or downregulated myf5 mRNA. MRF4 was not detected in the explants. TGF alpha apparently induces the early developmental stages of myogenesis through sequential upregulation of myoD and myogenin, downregulation of myf5 and corresponding significant increases in muscle-specific gene expression such as desmin transcription.
Collapse
Affiliation(s)
- A Yamane
- Department of Pharmacology, School of Dental Medicine, Tsurumi University, Yokohama, Japan
| | | | | | | | | | | | | |
Collapse
|
41
|
Winter B, Kautzner I, Issinger OG, Arnold HH. Two putative protein kinase CK2 phosphorylation sites are important for Myf-5 activity. Biol Chem 1997; 378:1445-56. [PMID: 9461343 DOI: 10.1515/bchm.1997.378.12.1445] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Myf-5, a member of a family of muscle-specific transcription factors, is important for myogenic cell determination and differentiation. Here, we report that Myf-5 protein constitutes a substrate for phosphorylation in vitro by protein kinase CK2. We identified two potential phosphorylation sites at serine49 and serine133, both of which seem to be necessary for Myf-5 activity. Mutants which can no longer be phosphorylated fail to transactivate E-box-dependent reporter genes and act as trans-dominant repressors of wild-type Myf-5. Normal activity can be restored by replacing the serine residues with glutamate suggesting that a negative charge at these sites is obligatory for Myf-5 activity. Although serine133 is part of helix 2 which mediates dimerization, we find no evidence for impaired DNA-binding or heterodimerization of the Ser-Ala133 mutant. Some serine49 mutations exhibit reduced nuclear localization and/or protein stability. Our data suggest that CK2-mediated phosphorylation of Myf-5 is required for Myf-5 activity.
Collapse
Affiliation(s)
- B Winter
- Department of Cell and Molecular Biology, University of Braunschweig, Germany
| | | | | | | |
Collapse
|
42
|
Gerber AN, Klesert TR, Bergstrom DA, Tapscott SJ. Two domains of MyoD mediate transcriptional activation of genes in repressive chromatin: a mechanism for lineage determination in myogenesis. Genes Dev 1997; 11:436-50. [PMID: 9042858 DOI: 10.1101/gad.11.4.436] [Citation(s) in RCA: 230] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Genetic studies have demonstrated that MyoD and Myf5 establish the skeletal muscle lineage, whereas myogenin mediates terminal differentiation, yet the molecular basis for this distinction is not understood. We show that MyoD can remodel chromatin at binding sites in muscle gene enhancers and activate transcription at previously silent loci. TGF-beta, basic-FGF, and sodium butyrate blocked MyoD-mediated chromatin reorganization and the initiation of transcription. In contrast, TGF-beta and sodium butyrate did not block transcription when added after chromatin remodeling had occurred. MyoD and Myf-5 were 10-fold more efficient than myogenin at activating genes in regions of transcriptionally silent chromatin. Deletion mutagenesis of the MyoD protein demonstrated that the ability to activate endogenous genes depended on two regions: a region rich in cysteine and histidine residues between the acidic activation domain and the bHLH domain, and a second region in the carboxyl terminus of the protein. Neither region has been shown previously to regulate gene transcription and both have domains that are conserved in the Myf5 protein. Our results establish a mechanism for chromatin modeling in the skeletal muscle lineage and define domains of MyoD, independent of the activation domain, that participate in chromatin reorganization.
Collapse
Affiliation(s)
- A N Gerber
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | | | | |
Collapse
|
43
|
Rohrer DK, Blau HM. Defective myogenesis in NFB-s mutant associated with a saturable suppression of MYF5 activity. SOMATIC CELL AND MOLECULAR GENETICS 1996; 22:349-61. [PMID: 9039845 DOI: 10.1007/bf02369892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Myogenic cell lines have proved to be useful tools for investigating the molecular mechanisms that control cellular differentiation. NFB-s is a mutant myogenic cell line which fails to differentiate in vitro, and can repress differentiation in normal myogenic cells when fused to form heterokaryons. The NFB-s cell line was used here to study the molecular mechanisms underlying such myogenic repression. Using muscle-specific reporter genes, we show that NFB-s cells fail to activate fully the muscle differentiation program at a transcriptional level, although muscle-specific transcription can be enhanced by regulators of differentiation such as pertussis toxin. Paradoxically we find that the myogenic regulator myf5 is expressed at constitutively high levels in NFB-s cells, and retains DNA binding activity. Expression plasmids encoding NFB-derived myf5 cDNA can rescue the myogenic phenotype in NFB-s cells, demonstrating that a threshold level of positive regulators must be reached before the myogenic program is activated. Thus, the dominant negative phenotype does not appear to result from defective myf5, but is due to a dosage-dependent saturable mechanism that interferes with myf5 function. These studies demonstrate that the stoichiometric ratio of positive and negative regulators is critical for determining the myogenic differentiation state.
Collapse
Affiliation(s)
- D K Rohrer
- Department of Molecular Pharmacology, Stanford University Medical Center, California 94305-5332, USA
| | | |
Collapse
|
44
|
Groisman R, Masutani H, Leibovitch MP, Robin P, Soudant I, Trouche D, Harel-Bellan A. Physical interaction between the mitogen-responsive serum response factor and myogenic basic-helix-loop-helix proteins. J Biol Chem 1996; 271:5258-64. [PMID: 8617811 DOI: 10.1074/jbc.271.9.5258] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Terminal differentiation of muscle cells results in opposite effects on gene promoters: muscle-specific promoters, which are repressed during active proliferation of myoblasts, are turned on, whereas at least some proliferation-associated promoters, such as c-fos, which are active during cell division, are turned off. MyoD and myogenin, transcription factors from the basic-helix-loop-helix (bHLH) family, are involved in both processes, up-regulating muscle genes and down-regulating c-fos. On the other hand, the serum response factor (SRF) is involved in the activation of muscle-specific genes, such as c-fos, as well as in the up-regulation of a subset of genes that are responsive to mitogens. Upon terminal differentiation, the activity of these various transcription factors could be modulated by the formation of distinct protein-protein complexes. Here, we have investigated the hypothesis that the function of SRF and/or MyoD and myogenin could be modulated by a physical association between these transcription factors. We show that myogenin from differentiating myoblasts specifically binds to SRF. In vitro analysis, using the glutathione S-transferase pull-down assay, indicates that SRF-myogenin interactions occur only with myogenin-E12 heterodimers and not with isolated myogenin. A physical interaction between myogenin, E12, and SRF could also be demonstrated in vivo using a triple-hybrid approach in yeast. Glutathione S-transferase pull-down analysis of various mutants of the proteins demonstrated that the bHLH domain of myogenin and that of E12 were necessary and sufficient for the interaction to be observed. Specific binding to SRF was also seen with MyoD. In contrast, Id, a natural inhibitor of myogenic bHLH proteins, did not bind SRF in any of the situations tested. These data suggest that SRF, on one hand, and myogenic bHLH, on the other, could modulate each other's activity through the formation of a heterotrimeric complex.
Collapse
Affiliation(s)
- R Groisman
- Laboratoire de Biologie des Tumeurs Humaines, CNRS URA 1156, Institut Gustave Roussy, 39 rue Camille Desmoulins, 94805 Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
45
|
Affiliation(s)
- J M Venuti
- Department of Anatomy and Cell Biology, Columbia College of Physicians and Surgeons, New York, New York 10032, USA
| | | |
Collapse
|
46
|
Trouche D, Masutani H, Groisman R, Robin P, Lenormand JL, Harel-Bellan A. Myogenin binds to and represses c-fos promoter. FEBS Lett 1995; 361:140-4. [PMID: 7698311 DOI: 10.1016/0014-5793(95)00140-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Myogenin (a member of the myogenic basic helix-loop-helix transcription factor family) seems to be the main effector of proliferation repression, a crucial step which precedes muscle cell terminal differentiation during muscle development. Proliferation repression most likely occurs through inhibition of proliferation-associated genes such as the proto-oncogene, c-fos. Here, we demonstrate that myogenin binds to an E-box located in the main element of the c-fos promoter, the serum response element (SRE). Results from co-transfection experiments indicate that myogenin acts as a repressor for the SRE. Our data suggest that myogenin could play a role in c-fos inhibition at the onset of muscle cell terminal differentiation.
Collapse
Affiliation(s)
- D Trouche
- Laboratoire de Biologie des Tumeurs Humaines, CNRS URA 1156, Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | |
Collapse
|
47
|
Jiang CK, Tomić-Canić M, Lucas DJ, Simon M, Blumenberg M. TGF beta promotes the basal phenotype of epidermal keratinocytes: transcriptional induction of K#5 and K#14 keratin genes. Growth Factors 1995; 12:87-97. [PMID: 8679251 DOI: 10.3109/08977199509028955] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
TGFbeta is an important regulator of epidermal keratinocyte function because it suppresses cell proliferation, while it induces synthesis of extracellular matrix proteins and their cells surface receptors. To examine whether TGFbeta affects synthesis of intracellular proteins as well, specifically the transcription of keratin genes, we transfected a series of DNA constructs that contain keratin gene promoters into human epidermal keratinocytes. The transfected cells were grown in the presence and absence of TGFbeta. We found that TGFbeta specifically induces transcription controlled by the promoters of K#5 and K#14 keratin genes, markers of basal cells. No other keratin gene promoters were induced. The effect of TGFbeta is concentration-dependent, can be demonstrated in HeLa cells, does not depend on keratinocyte growth conditions and can be elicited by both TGFbeta1 and TGFbeta2. We conclude that TGFbeta promotes the basal cell phenotype in stratified epithelia such as the epidermis.
Collapse
Affiliation(s)
- C K Jiang
- Ronald O. Perelman Department of Dermatology, New York University Medical Center, N.Y. 10016, USA
| | | | | | | | | |
Collapse
|
48
|
Vandromme M, Carnac G, Gauthier-Rouvière C, Fesquet D, Lamb N, Fernandez A. Nuclear import of the myogenic factor MyoD requires cAMP-dependent protein kinase activity but not the direct phosphorylation of MyoD. J Cell Sci 1994; 107 ( Pt 2):613-20. [PMID: 8207083 DOI: 10.1242/jcs.107.2.613] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
MyoD is a nuclear phosphoprotein that belongs to the family of myogenic regulatory factors and acts in the transcriptional activation of muscle-specific genes. We have investigated the role of cAMP-dependent protein kinase (A-kinase) in modulating the nuclear locale of MyoD. Purified MyoD protein microinjected into the cytoplasm of rat embryo fibroblasts is rapidly translocated into the nucleus. Inhibition of A-kinase activity through injection of the specific inhibitory peptide PKI prevents this nuclear localisation. This inhibition of nuclear location is specifically reversed by injection of purified A-kinase catalytic subunit, showing the requirement for A-kinase in the nuclear import of MyoD. Site-directed mutagenesis of all the putative sites for A-kinase-dependent phosphorylation on MyoD, substituting serine or threonine residues for the non-phosphorylatable amino acid alanine, had no effect on nuclear import of mutated MyoD. These data exclude the possibility that the effect of A-kinase on the nuclear translocation of MyoD is mediated by direct phosphorylation of MyoD and imply that A-kinase operates through phosphorylation of components involved in the nuclear transport of MyoD.
Collapse
Affiliation(s)
- M Vandromme
- Cell Biology Unit, CRBM, CNRS-INSERM, Montpellier, France
| | | | | | | | | | | |
Collapse
|
49
|
Bradham DM, in der Wiesche B, Precht P, Balakir R, Horton W. Transrepression of type II collagen by TGF-beta and FGF is protein kinase C dependent and is mediated through regulatory sequences in the promoter and first intron. J Cell Physiol 1994; 158:61-8. [PMID: 8263029 DOI: 10.1002/jcp.1041580109] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Transforming growth factor beta and basic fibroblast growth factor are multipotential factors found in bone and cartilage that may be involved in both the proliferation and differentiation of chondrocytes. It was previously reported that TGF-beta plus FGF caused a modulation of chondrocyte phenotype that included the down-regulation of steady-state level of the collagen II transcript. In this report, the results of nuclear run-off data indicate that repression of transcript initiation from the collagen II gene is the primary mechanism involved in the growth factor induced inhibition. Transient transfection assays with CAT expression vectors containing portions of the collagen II gene show that the TGF-beta/FGF induced transrepression requires a region in the first intron previously reported to have transcriptional enhancer activity and to bind chondrocyte nuclear proteins. In addition, silencer elements in the promoter also appear to play a role. Protein data as well as transient transfection experiments indicate that the activation of protein kinase C is necessary for the growth factor-induced down-regulation of collagen II expression. These studies suggest that a cascade initiating with PKC activation is responsible for modifying transcription factors that interact with regulatory sequences in the collagen II gene. A detailed understanding of the factors involved in cartilage-specific gene regulation in chondrocytes would facilitate development of therapeutic protocols for the repair of degenerated cartilage in diseases such as osteoarthritis.
Collapse
Affiliation(s)
- D M Bradham
- National Institutes of Health/National Institute on Aging/Gerontology Research Center, Baltimore, Maryland 21224
| | | | | | | | | |
Collapse
|
50
|
Hughes SM, Taylor JM, Tapscott SJ, Gurley CM, Carter WJ, Peterson CA. Selective accumulation of MyoD and myogenin mRNAs in fast and slow adult skeletal muscle is controlled by innervation and hormones. Development 1993; 118:1137-47. [PMID: 8269844 DOI: 10.1242/dev.118.4.1137] [Citation(s) in RCA: 287] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Each of the myogenic helix-loop-helix transcription factors (MyoD, Myogenin, Myf-5, and MRF4) is capable of activating muscle-specific gene expression, yet distinct functions have not been ascribed to the individual proteins. We report here that MyoD and Myogenin mRNAs selectively accumulate in hindlimb muscles of the adult rat that differ in contractile properties: MyoD is prevalent in fast twitch and Myogenin in slow twitch muscles. The distribution of MyoD and Myogenin transcripts also differ within a single muscle and correlate with the proportions of fast glycolytic and slow oxidative muscle fibres, respectively. Furthermore, the expression of a transgene consisting of a muscle-specific cis-regulatory region from the myoD gene controlling lacZ was primarily associated with the fast glycolytic fibres. Alteration of the fast/slow fibre type distribution by thyroid hormone treatment or by cross-reinnervation resulted in a corresponding alteration in the MyoD/Myogenin mRNA expression pattern. These findings show that the expression of specific myogenic helix-loop-helix regulators is under the control of innervation and humoral factors and may mediate differential control of contractile protein gene expression in adult muscle.
Collapse
Affiliation(s)
- S M Hughes
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock
| | | | | | | | | | | |
Collapse
|