1
|
Fu X, Zhang F, Dong X, Pu L, Feng Y, Xu Y, Gao F, Liang T, Kang J, Sun H, Hong T, Liu Y, Zhou H, Jiang J, Yin D, Hu X, Wang DZ, Ding J, Chen J. Adapting cytoskeleton-mitochondria patterning with myocyte differentiation by promyogenic PRR33. Cell Death Differ 2024:10.1038/s41418-024-01363-w. [PMID: 39147882 DOI: 10.1038/s41418-024-01363-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Coordinated cytoskeleton-mitochondria organization during myogenesis is crucial for muscle development and function. Our understanding of the underlying regulatory mechanisms remains inadequate. Here, we identified a novel muscle-enriched protein, PRR33, which is upregulated during myogenesis and acts as a promyogenic factor. Depletion of Prr33 in C2C12 represses myoblast differentiation. Genetic deletion of Prr33 in mice reduces myofiber size and decreases muscle strength. The Prr33 mutant mice also exhibit impaired myogenesis and defects in muscle regeneration in response to injury. Interactome and transcriptome analyses reveal that PRR33 regulates cytoskeleton and mitochondrial function. Remarkably, PRR33 interacts with DESMIN, a key regulator of cytoskeleton-mitochondria organization in muscle cells. Abrogation of PRR33 in myocytes substantially abolishes the interaction of DESMIN filaments with mitochondria, leading to abnormal intracellular accumulation of DESMIN and mitochondrial disorganization/dysfunction in myofibers. Together, our findings demonstrate that PRR33 and DESMIN constitute an important regulatory module coordinating mitochondrial organization with muscle differentiation.
Collapse
Affiliation(s)
- Xuyang Fu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Feng Zhang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Xiaoxuan Dong
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Linbin Pu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Yan Feng
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yang Xu
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Feng Gao
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Tian Liang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Jianmeng Kang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Hongke Sun
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Tingting Hong
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yunxia Liu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Hongmei Zhou
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jun Jiang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Deling Yin
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Xinyang Hu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Da-Zhi Wang
- University of South Florida Health Heart Institute, Center for Regenerative Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33602, USA
| | - Jian Ding
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Jinghai Chen
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China.
| |
Collapse
|
2
|
Triolo M, Baker N, Agarwal S, Larionov N, Podinić T, Khacho M. Optic atrophy 1 mediates muscle differentiation by promoting a metabolic switch via the supercomplex assembly factor SCAF1. iScience 2024; 27:109164. [PMID: 38414856 PMCID: PMC10897915 DOI: 10.1016/j.isci.2024.109164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/08/2023] [Accepted: 02/05/2024] [Indexed: 02/29/2024] Open
Abstract
Myogenic differentiation is integral for the regeneration of skeletal muscle following tissue damage. Though high-energy post-mitotic muscle relies predominantly on mitochondrial respiration, the importance of mitochondrial remodeling in enabling muscle differentiation and the players involved are not fully known. Here we show that the mitochondrial fusion protein OPA1 is essential for muscle differentiation. Our study demonstrates that OPA1 loss or inhibition, through genetic and pharmacological means, abolishes in vivo muscle regeneration and in vitro myotube formation. We show that both the inhibition and genetic deletion of OPA1 prevent the early onset metabolic switch required to drive myoblast differentiation. In addition, we observe an OPA1-dependent upregulation of the supercomplex assembly factor, SCAF1, at the onset of differentiation. Importantly, preventing the upregulation of SCAF1, through OPA1 loss or siRNA-mediated SCAF1 knockdown, impairs metabolic reprogramming and muscle differentiation. These findings reveal the integral role of OPA1 and mitochondrial reprogramming at the onset of myogenic differentiation.
Collapse
Affiliation(s)
- Matthew Triolo
- Department of Biochemistry, Microbiology and Immunology, Center for Neuromuscular Disease (CNMD), Ottawa Institute of Systems Biology (OISB), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nicole Baker
- Department of Biochemistry, Microbiology and Immunology, Center for Neuromuscular Disease (CNMD), Ottawa Institute of Systems Biology (OISB), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Soniya Agarwal
- Department of Biochemistry, Microbiology and Immunology, Center for Neuromuscular Disease (CNMD), Ottawa Institute of Systems Biology (OISB), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nikita Larionov
- Department of Biochemistry, Microbiology and Immunology, Center for Neuromuscular Disease (CNMD), Ottawa Institute of Systems Biology (OISB), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Tina Podinić
- Department of Biochemistry, Microbiology and Immunology, Center for Neuromuscular Disease (CNMD), Ottawa Institute of Systems Biology (OISB), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mireille Khacho
- Department of Biochemistry, Microbiology and Immunology, Center for Neuromuscular Disease (CNMD), Ottawa Institute of Systems Biology (OISB), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
3
|
Vardar Acar N, Özgül RK. A big picture of the mitochondria-mediated signals: From mitochondria to organism. Biochem Biophys Res Commun 2023; 678:45-61. [PMID: 37619311 DOI: 10.1016/j.bbrc.2023.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/02/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Mitochondria, well-known for years as the powerhouse and biosynthetic center of the cell, are dynamic signaling organelles beyond their energy production and biosynthesis functions. The metabolic functions of mitochondria, playing an important role in various biological events both in physiological and stress conditions, transform them into important cellular stress sensors. Mitochondria constantly communicate with the rest of the cell and even from other cells to the organism, transmitting stress signals including oxidative and reductive stress or adaptive signals such as mitohormesis. Mitochondrial signal transduction has a vital function in regulating integrity of human genome, organelles, cells, and ultimately organism.
Collapse
Affiliation(s)
- Neşe Vardar Acar
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - R Köksal Özgül
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
4
|
Hu B, Zhao C, Pan X, Wei H, Mo G, Xian M, Luo W, Nie Q, Li H, Zhang X. Local GHR roles in regulation of mitochondrial function through mitochondrial biogenesis during myoblast differentiation. Cell Commun Signal 2023; 21:148. [PMID: 37337300 DOI: 10.1186/s12964-023-01166-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/13/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Myoblast differentiation requires metabolic reprogramming driven by increased mitochondrial biogenesis and oxidative phosphorylation. The canonical GH-GHR-IGFs axis in liver exhibits a great complexity in response to somatic growth. However, the underlying mechanism of whether local GHR acts as a control valve to regulate mitochondrial function through mitochondrial biogenesis during myoblast differentiation remains unknown. METHODS We manipulated the GHR expression in chicken primary myoblast to investigate its roles in mitochondrial biogenesis and function during myoblast differentiation. RESULTS We reported that GHR is induced during myoblast differentiation. Local GHR promoted mitochondrial biogenesis during myoblast differentiation, as determined by the fluorescence intensity of Mito-Tracker Green staining and MitoTimer reporter system, the expression of mitochondrial biogenesis markers (PGC1α, NRF1, TFAM) and mtDNA encoded gene (ND1, CYTB, COX1, ATP6), as well as mtDNA content. Consistently, local GHR enhanced mitochondrial function during myoblast differentiation, as determined by the oxygen consumption rate, mitochondrial membrane potential, ATP level and ROS production. We next revealed that the regulation of mitochondrial biogenesis and function by GHR depends on IGF1. In terms of the underlying mechanism, we demonstrated that IGF1 regulates mitochondrial biogenesis via PI3K/AKT/CREB pathway. Additionally, GHR knockdown repressed myoblast differentiation. CONCLUSIONS In conclusion, our data corroborate that local GHR acts as a control valve to enhance mitochondrial function by promoting mitochondrial biogenesis via IGF1-PI3K/AKT/CREB pathway during myoblast differentiation. Video Abstract.
Collapse
Affiliation(s)
- Bowen Hu
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, Guangdong, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Changbin Zhao
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, Guangdong, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xiangchun Pan
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Haohui Wei
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, Guangdong, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Guodong Mo
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, Guangdong, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Mingjian Xian
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, Guangdong, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wen Luo
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, Guangdong, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Qinghua Nie
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, Guangdong, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Hongmei Li
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, Guangdong, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xiquan Zhang
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China.
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, Guangdong, China.
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
Odame E, Li L, Nabilla JA, Cai H, Xiao M, Ye J, Chen Y, Kyei B, Dai D, Zhan S, Cao J, Guo J, Zhong T, Wang L, Zhang H. miR-145-3p Inhibits MuSCs Proliferation and Mitochondria Mass via Targeting MYBL1 in Jianzhou Big-Eared Goats. Int J Mol Sci 2023; 24:ijms24098341. [PMID: 37176056 PMCID: PMC10179409 DOI: 10.3390/ijms24098341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/30/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Muscle growth and injury-induced regeneration are controlled by skeletal muscle satellite cells (MuSCs) through myogenesis in postnatal animals. Meanwhile, myogenesis is accompanied by mitochondrial function and enzyme activity. Nevertheless, the underlying molecular mechanisms involving non-coding RNAs including circular RNAs (circRNAs) and microRNAs (miRNAs) remain largely unsolved. Here, we explored the myogenic roles of miR-145-3p and MYBL1 on muscle development and mitochondrial mass. We noticed that overexpression of miR-145-3p inhibited MuSCs proliferation and reduced the number of viable cells. Meanwhile, deficiency of miR-145-3p caused by LNAantimiR-145-3p or an inhibitor retarded the differentiation of MuSCs. miR-145-3p altered the mitochondrial mass in MuSCs. Moreover, miR-145-3p targeted and negatively regulated the expression of CDR1as and MYBL1. The knockdown of the MYBL1 using ASO-2'MOE modification simulated the inhibitory function of miR-145-3p on cell proliferation. Additionally, MYBL1 mediated the regulation of miR-145-3p on Vexin, VCPIP1, COX1, COX2, and Pax7. These imply that CDR1as/miR-145-3p/MYBL1/COX1, COX2, VCPIP1/Vexin expression at least partly results in a reduction in mitochondrial mass and MuSCs proliferation. These novel findings confirm the importance of mitochondrial mass during myogenesis and the boosting of muscle/meat development in mammals.
Collapse
Affiliation(s)
- Emmanuel Odame
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Joshua Abdulai Nabilla
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - He Cai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Miao Xiao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiangfeng Ye
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuan Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Bismark Kyei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Dinghui Dai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Siyuan Zhan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiaxue Cao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiazhong Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Tao Zhong
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Linjie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
6
|
Lv S, Shen Q, Li H, Chen Q, Xie W, Li Y, Wang X, Ding G. Caloric restriction delays age-related muscle atrophy by inhibiting 11β-HSD1 to promote the differentiation of muscle stem cells. Front Med (Lausanne) 2023; 9:1027055. [PMID: 36687405 PMCID: PMC9849809 DOI: 10.3389/fmed.2022.1027055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/08/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction Calorie restriction (CR) is an important direction for the delay of sarcopenia in elderly individuals. However, the specific mechanisms of CR against aging are still unclear. Methods In this study, we used a CR model of elderly mice with muscle-specific 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) knockout mice and 11β-HSD1 overexpression mice to confirm that CR can delay muscle aging by inhibiting 11β-HSD1 which can transform inactive GC(cortisone) into active GC(cortisol). The ability of self-proliferation and differentiation into muscle fibers of these mouse muscle stem cells (MuSCs) was observed in vitro. Additionally, the mitochondrial function and mitochondrial ATP production capacity of MuSCs were measured by mitochondrial oxygen consumption. Results It was found that the 11β-HSD1 expression level was increased in age-related muscle atrophy. Overexpression of 11β-HSD1 led to muscle atrophy in young mice, and 11β-HSD1 knockout rescued age-related muscle atrophy. Moreover, CR in aged mice reduced the local effective concentration of glucocorticoid (GC) through 11β-HSD1, thereby promoting the mitochondrial function and differentiation ability of MuSCs. Conclusions Together, our findings highlight promising sarcopenia protection with 40% CR in older ages. Furthermore, we speculated that targeting an 11β-HSD1-dependent metabolic pathway may represent a novel strategy for developing therapeutics against age-related muscle atrophy.
Collapse
Affiliation(s)
- Shan Lv
- Department of Geriatric Endocrinology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Qianjin Shen
- Department of Emergency Medicine, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qun Chen
- Department of Orthopedics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China,*Correspondence: Yusheng Li,
| | - Xiaodong Wang
- Department of Geriatric Endocrinology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China,Xiaodong Wang,
| | - Guoxian Ding
- Department of Geriatric Endocrinology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China,Guoxian Ding,
| |
Collapse
|
7
|
Yan Y, Li M, Lin J, Ji Y, Wang K, Yan D, Shen Y, Wang W, Huang Z, Jiang H, Sun H, Qi L. Adenosine monophosphate activated protein kinase contributes to skeletal muscle health through the control of mitochondrial function. Front Pharmacol 2022; 13:947387. [PMID: 36339617 PMCID: PMC9632297 DOI: 10.3389/fphar.2022.947387] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/06/2022] [Indexed: 11/26/2022] Open
Abstract
Skeletal muscle is one of the largest organs in the body and the largest protein repository. Mitochondria are the main energy-producing organelles in cells and play an important role in skeletal muscle health and function. They participate in several biological processes related to skeletal muscle metabolism, growth, and regeneration. Adenosine monophosphate-activated protein kinase (AMPK) is a metabolic sensor and regulator of systemic energy balance. AMPK is involved in the control of energy metabolism by regulating many downstream targets. In this review, we propose that AMPK directly controls several facets of mitochondrial function, which in turn controls skeletal muscle metabolism and health. This review is divided into four parts. First, we summarize the properties of AMPK signal transduction and its upstream activators. Second, we discuss the role of mitochondria in myogenesis, muscle atrophy, regeneration post-injury of skeletal muscle cells. Third, we elaborate the effects of AMPK on mitochondrial biogenesis, fusion, fission and mitochondrial autophagy, and discuss how AMPK regulates the metabolism of skeletal muscle by regulating mitochondrial function. Finally, we discuss the effects of AMPK activators on muscle disease status. This review thus represents a foundation for understanding this biological process of mitochondrial dynamics regulated by AMPK in the metabolism of skeletal muscle. A better understanding of the role of AMPK on mitochondrial dynamic is essential to improve mitochondrial function, and hence promote skeletal muscle health and function.
Collapse
Affiliation(s)
- Yan Yan
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Ming Li
- Department of Laboratory Medicine, Binhai County People’s Hospital Affiliated to Kangda College of Nanjing Medical University, Yancheng, China
| | - Jie Lin
- Department of Infectious Disease, Affiliated Hospital of Nantong University, Nantong, China
| | - Yanan Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Dajun Yan
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Wei Wang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Department of Pathology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Zhongwei Huang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Haiyan Jiang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| |
Collapse
|
8
|
Sun H, Shen L, Zhang P, Lin F, Ma J, Wu Y, Yu H, Sun L. Inhibition of High-Temperature Requirement Protein A2 Protease Activity Represses Myogenic Differentiation via UPRmt. Int J Mol Sci 2022; 23:ijms231911761. [PMID: 36233059 PMCID: PMC9569504 DOI: 10.3390/ijms231911761] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022] Open
Abstract
Skeletal muscles require muscle satellite cell (MuSC) differentiation to facilitate the replenishment and repair of muscle fibers. A key step in this process is called myogenic differentiation. The differentiation ability of MuSCs decreases with age and can result in sarcopenia. Although mitochondria have been reported to be involved in myogenic differentiation by promoting a bioenergetic remodeling, little is known about the interplay of mitochondrial proteostasis and myogenic differentiation. High-temperature-requirement protein A2 (HtrA2/Omi) is a protease that regulates proteostasis in the mitochondrial intermembrane space (IMS). Mice deficient in HtrA2 protease activity show a distinct phenotype of sarcopenia. To investigate the role of IMS proteostasis during myogenic differentiation, we treated C2C12 myoblasts with UCF101, a specific inhibitor of HtrA2 during differentiation process. A key step in this process is called myogenic differentiation. The differentiation ability of MuSCs decreases with age and can result in sarcopenia. Further, CHOP, p-eIF2α, and other mitochondrial unfolded protein response (UPRmt)-related proteins are upregulated. Therefore, we suggest that imbalance of mitochondrial IMS proteostasis acts via a retrograde signaling pathway to inhibit myogenic differentiation via the UPRmt pathway. These novel mechanistic insights may have implications for the development of new strategies for the treatment of sarcopenia.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huimei Yu
- Correspondence: (H.Y.); (L.S.); Tel.: +86-0431-8561-9495 (H.Y. & L.S.)
| | - Liankun Sun
- Correspondence: (H.Y.); (L.S.); Tel.: +86-0431-8561-9495 (H.Y. & L.S.)
| |
Collapse
|
9
|
Lv XH, Cong XX, Nan JL, Lu XM, Zhu QL, Shen J, Wang BB, Wang ZT, Zhou RY, Chen WA, Su L, Chen X, Li ZZ, Lin YN. Anti-diabetic drug canagliflozin hinders skeletal muscle regeneration in mice. Acta Pharmacol Sin 2022; 43:2651-2665. [PMID: 35217814 PMCID: PMC9525290 DOI: 10.1038/s41401-022-00878-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/22/2022] [Indexed: 12/28/2022] Open
Abstract
Canagliflozin is an antidiabetic medicine that inhibits sodium-glucose cotransporter 2 (SGLT2) in proximal tubules. Recently, it was reported to have several noncanonical effects other than SGLT2 inhibiting. However, the effects of canagliflozin on skeletal muscle regeneration remain largely unexplored. Thus, in vivo muscle contractile properties recovery in mice ischemic lower limbs following gliflozins treatment was evaluated. The C2C12 myoblast differentiation after gliflozins treatment was also assessed in vitro. As a result, both in vivo and in vitro data indicate that canagliflozin impairs intrinsic myogenic regeneration, thus hindering ischemic limb muscle contractile properties, fatigue resistance recovery, and tissue regeneration. Mitochondrial structure and activity are both disrupted by canagliflozin in myoblasts. Single-cell RNA sequencing of ischemic tibialis anterior reveals a decrease in leucyl-tRNA synthetase 2 (LARS2) in muscle stem cells attributable to canagliflozin. Further investigation explicates the noncanonical function of LARS2, which plays pivotal roles in regulating myoblast differentiation and muscle regeneration by affecting mitochondrial structure and activity. Enhanced expression of LARS2 restores the differentiation of canagliflozin-treated myoblasts, and accelerates ischemic skeletal muscle regeneration in canagliflozin-treated mice. Our data suggest that canagliflozin directly impairs ischemic skeletal muscle recovery in mice by downregulating LARS2 expression in muscle stem cells, and that LARS2 may be a promising therapeutic target for injured skeletal muscle regeneration.
Collapse
Affiliation(s)
- Xin-Huang Lv
- Research Institute of Experimental Neurobiology, Department of Neurology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiao-Xia Cong
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jin-Liang Nan
- Department of Pathology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xing-Mei Lu
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Qian-Li Zhu
- Wenzhou Municipal Key Cardiovascular Research Laboratory, Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Jian Shen
- Department of Pathology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Bei-Bei Wang
- Center of Cryo-Electron Microscopy, Zhejiang University, Hangzhou, 310058, China
| | - Zhi-Ting Wang
- Wenzhou Municipal Key Cardiovascular Research Laboratory, Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Ri-Yong Zhou
- Department of Anesthesiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Wei-An Chen
- Research Institute of Experimental Neurobiology, Department of Neurology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Lan Su
- Wenzhou Municipal Key Cardiovascular Research Laboratory, Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiao Chen
- Wenzhou Municipal Key Cardiovascular Research Laboratory, Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Zheng-Zheng Li
- Research Institute of Experimental Neurobiology, Department of Neurology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Yi-Nuo Lin
- Wenzhou Municipal Key Cardiovascular Research Laboratory, Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
10
|
Flierl A, Schriner SE, Hancock S, Coskun PE, Wallace DC. The mitochondrial adenine nucleotide transporters in myogenesis. Free Radic Biol Med 2022; 188:312-327. [PMID: 35714845 DOI: 10.1016/j.freeradbiomed.2022.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 01/06/2023]
Abstract
Adenine Nucleotide Translocator isoforms (ANTs) exchange ADP/ATP across the inner mitochondrial membrane, are also voltage-activated proton channels and regulate mitophagy and apoptosis. The ANT1 isoform predominates in heart and muscle while ANT2 is systemic. Here, we report the creation of Ant mutant mouse myoblast cell lines with normal Ant1 and Ant2 genes, deficient in either Ant1 or Ant2, and deficient in both the Ant1 and Ant2 genes. These cell lines are immortal under permissive conditions (IFN-γ + serum at 32 °C) permitting expansion but return to normal myoblasts that can be differentiated into myotubes at 37 °C. With this system we were able to complement our Ant1 mutant studies by demonstrating that ANT2 is important for myoblast to myotube differentiation and myotube mitochondrial respiration. ANT2 is also important in the regulation of mitochondrial biogenesis and antioxidant defenses. ANT2 is also associated with increased oxidative stress response and modulation for Ca++ sequestration and activation of the mitochondrial permeability transition (mtPTP) pore during cell differentiation.
Collapse
Affiliation(s)
- Adrian Flierl
- Center for Molecular and Mitochondrial Medicine and Genetics and the Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Samuel E Schriner
- Center for Molecular and Mitochondrial Medicine and Genetics and the Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Saege Hancock
- Center for Molecular and Mitochondrial Medicine and Genetics and the Department of Biological Chemistry, University of California, Irvine, CA, USA; Center for Mitochondrial and Epigenomic Medicine, Department of Pediatrics, Division of Human Genetics, Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Pinar E Coskun
- Center for Molecular and Mitochondrial Medicine and Genetics and the Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Douglas C Wallace
- Center for Molecular and Mitochondrial Medicine and Genetics and the Department of Biological Chemistry, University of California, Irvine, CA, USA; Center for Mitochondrial and Epigenomic Medicine, Department of Pediatrics, Division of Human Genetics, Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, PA, USA.
| |
Collapse
|
11
|
Cioffi F, Giacco A, Goglia F, Silvestri E. Bioenergetic Aspects of Mitochondrial Actions of Thyroid Hormones. Cells 2022; 11:cells11060997. [PMID: 35326451 PMCID: PMC8947633 DOI: 10.3390/cells11060997] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/04/2022] [Accepted: 03/13/2022] [Indexed: 02/07/2023] Open
Abstract
Much is known, but there is also much more to discover, about the actions that thyroid hormones (TH) exert on metabolism. Indeed, despite the fact that thyroid hormones are recognized as one of the most important regulators of metabolic rate, much remains to be clarified on which mechanisms control/regulate these actions. Given their actions on energy metabolism and that mitochondria are the main cellular site where metabolic transformations take place, these organelles have been the subject of extensive investigations. In relatively recent times, new knowledge concerning both thyroid hormones (such as the mechanisms of action, the existence of metabolically active TH derivatives) and the mechanisms of energy transduction such as (among others) dynamics, respiratory chain organization in supercomplexes and cristes organization, have opened new pathways of investigation in the field of the control of energy metabolism and of the mechanisms of action of TH at cellular level. In this review, we highlight the knowledge and approaches about the complex relationship between TH, including some of their derivatives, and the mitochondrial respiratory chain.
Collapse
|
12
|
Chabi B, Hennani H, Cortade F, Wrutniak-Cabello C. Characterization of mitochondrial respiratory complexes involved in the regulation of myoblast differentiation. Cell Biol Int 2021; 45:1676-1684. [PMID: 33764610 DOI: 10.1002/cbin.11602] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 03/14/2021] [Accepted: 03/21/2021] [Indexed: 11/10/2022]
Abstract
During myoblast differentiation, mitochondria undergo numerous changes that are necessary for the progression of the myogenic program. Notably, we previously showed that alteration in mitochondrial activity was able to control the expression of keys regulator of cell cycle withdrawal and terminal differentiation. Here, we assessed whether inhibition of one of the respiratory complexes was a key factor in the regulation of myogenic differentiation in C2C12 cells, and was associated with alteration in reactive oxygen species (ROS) production. C2C12 cells were treated from proliferation to differentiation with specific inhibitors of mitochondrial complexes at a concentration that were inhibiting respiration but not altering cell morphology. Proliferation was significantly repressed with inhibition of complexes I, II, and III, or mitochondrial protein synthesis (using Chloramphenicol treatment), while complex IV inhibition did not alter myoblast proliferation compared to control cells. Moreover, inhibition of complexes I and II altered cell cycle regulators, with p21 protein expression upregulated since proliferation and p27 protein expression reduced at differentiation. Myotubes formation and myogenin expression were blunted with complexes I and II inhibitors while MyoD protein expression was maintained, suggesting an alteration in its transcriptional activity. Finally, a decrease in overall ROS production was observed with continuous inhibition of mitochondrial complexes I-IV. In summary, our data provide evidence that complexes I and II may be the primary regulators of C2C12 myogenic differentiation. This occurs through specific regulation of myogenic rather than cell cycle regulators expression and ROS production at mitochondrial rather than cell level.
Collapse
Affiliation(s)
- Béatrice Chabi
- DMEM, Université de Montpellier, INRAE, Montpellier, France
| | - Hanane Hennani
- DMEM, Université de Montpellier, INRAE, Montpellier, France
| | | | | |
Collapse
|
13
|
Lu Y, Mao J, Han X, Zhang W, Li Y, Liu Y, Li Q. Downregulated hypoxia-inducible factor 1α improves myoblast differentiation under hypoxic condition in mouse genioglossus. Mol Cell Biochem 2021; 476:1351-1364. [PMID: 33389500 DOI: 10.1007/s11010-020-03995-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 08/01/2020] [Indexed: 10/22/2022]
Abstract
The treatment of obstructive sleep apnea-hypopnea syndrome targets the narrow anatomic structure of the upper airway (UA) and lacks an effective therapy for UA dilator muscle dysfunction. Long-term hypoxia can cause damage to UA dilator muscles and trigger a vicious cycle. We previously confirmed that hypoxia-inducible factor 1α (HIF-1α) upregulation mediates muscle fatigue in hypoxia condition, but the underlying mechanism remains to be determined. The present study investigated the intrinsic mechanisms and related pathways of HIF-1α that affect myoblast differentiation, with an aim to search for compounds that have protective effects in hypoxic condition. Differentiation of myoblasts was induced under hypoxia, and we found that hypoxia significantly inhibits the differentiation of myoblasts, damages the ultrastructure of mitochondria, and reduces the expression of myogenin, PGC-1β and pAMPKα1. HIF-1α has a negative regulation effect on AMPK. Downregulation of HIF-1α increases the expression of the abovementioned proteins, promotes the differentiation of myoblasts, and protects mitochondrial integrity. In addition, mitochondrial biogenesis occurs during myogenic differentiation. Inhibition of the AMPK pathway inhibits mitochondrial biogenesis, decreases the level of PGC-1β, and increases apoptosis. Resveratrol dimer can reverse the mitochondrial damage induced by AMPK pathway inhibition and decrease myoblast apoptosis. Our results provided a regulatory mechanism for hypoxic injury in genioglossus which may contribute to the pathogenesis and treatment of OSAHS.
Collapse
Affiliation(s)
- Yun Lu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, No.356 East Beijing Road, Shanghai, 200001, China
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, 2 Tianjin Road, Shanghai, 200001, China
| | - Jiaqi Mao
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, 2 Tianjin Road, Shanghai, 200001, China
- Department of Endodontics, Stomatological Hospital, Hebei Medical University, 383 East Zhongshan Road, Shijiazhuang, 050017, China
| | - Xinxin Han
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, 2 Tianjin Road, Shanghai, 200001, China
| | - Weihua Zhang
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, No.356 East Beijing Road, Shanghai, 200001, China
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, 2 Tianjin Road, Shanghai, 200001, China
| | - Yuanyuan Li
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, 2 Tianjin Road, Shanghai, 200001, China
- Department of Pediatric Dentistry, Shanghai Stomatological Hospital, Fudan University, 356 East Beijing Road, Shanghai, 200001, China
| | - Yuehua Liu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, No.356 East Beijing Road, Shanghai, 200001, China.
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, 2 Tianjin Road, Shanghai, 200001, China.
| | - Qiang Li
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, No.356 East Beijing Road, Shanghai, 200001, China.
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, 2 Tianjin Road, Shanghai, 200001, China.
| |
Collapse
|
14
|
Cairns G, Thumiah-Mootoo M, Burelle Y, Khacho M. Mitophagy: A New Player in Stem Cell Biology. BIOLOGY 2020; 9:E481. [PMID: 33352783 PMCID: PMC7766552 DOI: 10.3390/biology9120481] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023]
Abstract
The fundamental importance of functional mitochondria in the survival of most eukaryotic cells, through regulation of bioenergetics, cell death, calcium dynamics and reactive oxygen species (ROS) generation, is undisputed. However, with new avenues of research in stem cell biology these organelles have now emerged as signaling entities, actively involved in many aspects of stem cell functions, including self-renewal, commitment and differentiation. With this recent knowledge, it becomes evident that regulatory pathways that would ensure the maintenance of mitochondria with state-specific characteristics and the selective removal of organelles with sub-optimal functions must play a pivotal role in stem cells. As such, mitophagy, as an essential mitochondrial quality control mechanism, is beginning to gain appreciation within the stem cell field. Here we review and discuss recent advances in our knowledge pertaining to the roles of mitophagy in stem cell functions and the potential contributions of this specific quality control process on to the progression of aging and diseases.
Collapse
Affiliation(s)
- George Cairns
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 7K4, Canada;
| | - Madhavee Thumiah-Mootoo
- Department of Cellular & Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Yan Burelle
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 7K4, Canada;
- Department of Cellular & Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Mireille Khacho
- Center for Neuromuscular Disease, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Ottawa Institute of Systems Biology (OISB), University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
15
|
Benlebna M, Balas L, Pessemesse L, Bonafos B, Fouret G, Pavlin L, Goustard B, Gaillet S, Durand T, Coudray C, Feillet-Coudray C, Casas F. FAHFAs Regulate the Proliferation of C2C12 Myoblasts and Induce a Shift toward a More Oxidative Phenotype in Mouse Skeletal Muscle. Int J Mol Sci 2020; 21:ijms21239046. [PMID: 33260741 PMCID: PMC7729663 DOI: 10.3390/ijms21239046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/22/2022] Open
Abstract
Branched fatty acid esters of hydroxy fatty acids (FAHFAs) are endogenous lipids reported to have antidiabetic and anti-inflammatory effects. Since skeletal muscle is a major target for insulin, the aim of this study is to explore for the first time the influence of several FAHFAs in C2C12 myoblasts and in skeletal muscle phenotype in mice. Here, we show that eleven FAHFAs belonging to different families inhibit C2C12 myoblast proliferation. In addition, all FAHFAs decreased mitochondrial cytochrome c oxidase activity without affecting reactive oxygen species production and the mitochondrial network. During C2C12 myoblasts differentiation, we found that two of the most active lipids, 9-PAHPA and 9-OAHPA, did not significantly affect the fusion index and the expression of myosin heavy chains. However, we found that three months’ intake of 9-PAHPA or 9-OAHPA in mice increased the expression of more oxidative myosin in skeletal muscle without affecting skeletal muscle mass, number, and mean fiber area, mitochondrial activity, and oxidative stress parameters. In conclusion, our study indicated that the eleven FAHFAs tested decreased the proliferation rate of C2C12 myoblasts, probably through the inhibition of mitochondrial activity. In addition, we found that 9-PAHPA or 9-OAHPA supplementation in mice induced a switch toward a more oxidative contractile phenotype of skeletal muscle. These data suggest that the increase in insulin sensitivity previously described for these two FAHFAs is of muscular origin.
Collapse
Affiliation(s)
- Melha Benlebna
- DMEM (Dynamique Musculaire et Métabolisme), INRAE, University Montpellier, 34060 Montpellier, France; (M.B.); (L.P.); (B.B.); (G.F.); (L.P.); (B.G.); (S.G.); (C.C.); (C.F.-C.)
| | - Laurence Balas
- IBMM (Institut des Biomolecules Max Mousseron), CNRS, ENSCM, University Montpellier, 34093 Montpellier, France; (L.B.); (T.D.)
| | - Laurence Pessemesse
- DMEM (Dynamique Musculaire et Métabolisme), INRAE, University Montpellier, 34060 Montpellier, France; (M.B.); (L.P.); (B.B.); (G.F.); (L.P.); (B.G.); (S.G.); (C.C.); (C.F.-C.)
| | - Béatrice Bonafos
- DMEM (Dynamique Musculaire et Métabolisme), INRAE, University Montpellier, 34060 Montpellier, France; (M.B.); (L.P.); (B.B.); (G.F.); (L.P.); (B.G.); (S.G.); (C.C.); (C.F.-C.)
| | - Gilles Fouret
- DMEM (Dynamique Musculaire et Métabolisme), INRAE, University Montpellier, 34060 Montpellier, France; (M.B.); (L.P.); (B.B.); (G.F.); (L.P.); (B.G.); (S.G.); (C.C.); (C.F.-C.)
| | - Laura Pavlin
- DMEM (Dynamique Musculaire et Métabolisme), INRAE, University Montpellier, 34060 Montpellier, France; (M.B.); (L.P.); (B.B.); (G.F.); (L.P.); (B.G.); (S.G.); (C.C.); (C.F.-C.)
| | - Bénédicte Goustard
- DMEM (Dynamique Musculaire et Métabolisme), INRAE, University Montpellier, 34060 Montpellier, France; (M.B.); (L.P.); (B.B.); (G.F.); (L.P.); (B.G.); (S.G.); (C.C.); (C.F.-C.)
| | - Sylvie Gaillet
- DMEM (Dynamique Musculaire et Métabolisme), INRAE, University Montpellier, 34060 Montpellier, France; (M.B.); (L.P.); (B.B.); (G.F.); (L.P.); (B.G.); (S.G.); (C.C.); (C.F.-C.)
| | - Thierry Durand
- IBMM (Institut des Biomolecules Max Mousseron), CNRS, ENSCM, University Montpellier, 34093 Montpellier, France; (L.B.); (T.D.)
| | - Charles Coudray
- DMEM (Dynamique Musculaire et Métabolisme), INRAE, University Montpellier, 34060 Montpellier, France; (M.B.); (L.P.); (B.B.); (G.F.); (L.P.); (B.G.); (S.G.); (C.C.); (C.F.-C.)
| | - Christine Feillet-Coudray
- DMEM (Dynamique Musculaire et Métabolisme), INRAE, University Montpellier, 34060 Montpellier, France; (M.B.); (L.P.); (B.B.); (G.F.); (L.P.); (B.G.); (S.G.); (C.C.); (C.F.-C.)
| | - François Casas
- DMEM (Dynamique Musculaire et Métabolisme), INRAE, University Montpellier, 34060 Montpellier, France; (M.B.); (L.P.); (B.B.); (G.F.); (L.P.); (B.G.); (S.G.); (C.C.); (C.F.-C.)
- Correspondence:
| |
Collapse
|
16
|
Levitt DE, Chalapati N, Prendergast MJ, Simon L, Molina PE. Ethanol-Impaired Myogenic Differentiation is Associated With Decreased Myoblast Glycolytic Function. Alcohol Clin Exp Res 2020; 44:2166-2176. [PMID: 32945016 PMCID: PMC7680427 DOI: 10.1111/acer.14453] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Myopathy affects nearly half of individuals with alcohol use disorder (AUD), and impaired skeletal muscle regenerative potential is a probable contributing factor. Previous findings from our laboratory indicate that chronic in vivo and in vitro ethanol (EtOH) treatment decreases myogenic potential of skeletal muscle myoblasts. Myogenesis, a highly coordinated process, requires shifts in cellular metabolic state allowing for myoblasts to proliferate and differentiate into mature myotubes. The objective of this study was to determine whether alcohol interferes with myoblast mitochondrial and glycolytic metabolism and impairs myogenic differentiation. METHODS Myoblasts were isolated from vastus lateralis muscle excised from alcohol-naïve adult male (n = 5) and female (n = 5) rhesus macaques. Myoblasts were proliferated for 3 days (day 0 differentiation; D0) and differentiated for 5 days (D5) with or without 50 mM EtOH. Metabolism was assessed using a mitochondrial stress test to measure oxygen consumption (OCR) and extracellular acidification (ECAR) rates at D0. Differentiation was examined at D5. Expression of mitochondrial and glycolytic genes and mitochondrial DNA (mtDNA) was measured at D0 and D5. RESULTS Ethanol significantly (p < 0.05) increased myoblast maximal OCR and decreased ECAR at D0, and decreased fusion index, myotubes per field, and total nuclei at D5. The EtOH-induced decrease in ECAR was associated with the EtOH-mediated decreases in fusion index and myotubes per field. EtOH did not alter the decrease in glycolytic gene expression and increase in mtDNA from D0 to D5. CONCLUSION During myoblast proliferation, EtOH decreased glycolytic metabolism and increased maximal OCR, suggesting that myoblast metabolic phenotype was dysregulated with EtOH. The EtOH-induced decrease in ECAR was associated with decreased differentiation. These findings suggest that EtOH-mediated shifts in metabolic phenotype may underlie impaired differentiation, which has important clinical implications for myogenesis in those affected by alcoholic myopathy.
Collapse
Affiliation(s)
- Danielle E. Levitt
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Naveena Chalapati
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Matthew J. Prendergast
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Liz Simon
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Patricia E. Molina
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, LA
| |
Collapse
|
17
|
Bhattacharya D, Scimè A. Mitochondrial Function in Muscle Stem Cell Fates. Front Cell Dev Biol 2020; 8:480. [PMID: 32612995 PMCID: PMC7308489 DOI: 10.3389/fcell.2020.00480] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/22/2020] [Indexed: 01/25/2023] Open
Abstract
Mitochondria are crucial organelles that control cellular metabolism through an integrated mechanism of energy generation via oxidative phosphorylation. Apart from this canonical role, it is also integral for ROS production, fatty acid metabolism and epigenetic remodeling. Recently, a role for the mitochondria in effecting stem cell fate decisions has gained considerable interest. This is important for skeletal muscle, which exhibits a remarkable property for regeneration following injury, owing to satellite cells (SCs), the adult myogenic stem cells. Mitochondrial function is associated with maintaining and dictating SC fates, linked to metabolic programming during quiescence, activation, self-renewal, proliferation and differentiation. Notably, mitochondrial adaptation might take place to alter SC fates and function in the presence of different environmental cues. This review dissects the contribution of mitochondria to SC operational outcomes, focusing on how their content, function, dynamics and adaptability work to influence SC fate decisions.
Collapse
Affiliation(s)
| | - Anthony Scimè
- Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, ON, Canada
| |
Collapse
|
18
|
EDMD-Causing Emerin Mutant Myogenic Progenitors Exhibit Impaired Differentiation Using Similar Mechanisms. Cells 2020; 9:cells9061463. [PMID: 32549231 PMCID: PMC7349064 DOI: 10.3390/cells9061463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 11/17/2022] Open
Abstract
Mutations in the gene encoding emerin (EMD) cause Emery–Dreifuss muscular dystrophy (EDMD1), an inherited disorder characterized by progressive skeletal muscle wasting, irregular heart rhythms and contractures of major tendons. The skeletal muscle defects seen in EDMD are caused by failure of muscle stem cells to differentiate and regenerate the damaged muscle. However, the underlying mechanisms remain poorly understood. Most EDMD1 patients harbor nonsense mutations and have no detectable emerin protein. There are three EDMD-causing emerin mutants (S54F, Q133H, and Δ95–99) that localize correctly to the nuclear envelope and are expressed at wildtype levels. We hypothesized these emerin mutants would share in the disruption of key molecular pathways involved in myogenic differentiation. We generated myogenic progenitors expressing wildtype emerin and each EDMD1-causing emerin mutation (S54F, Q133H, Δ95–99) in an emerin-null (EMD−/y) background. S54F, Q133H, and Δ95–99 failed to rescue EMD−/y myogenic differentiation, while wildtype emerin efficiently rescued differentiation. RNA sequencing was done to identify pathways and networks important for emerin regulation of myogenic differentiation. This analysis significantly reduced the number of pathways implicated in EDMD1 muscle pathogenesis.
Collapse
|
19
|
Ciuffoli V, Lena AM, Gambacurta A, Melino G, Candi E. Myoblasts rely on TAp63 to control basal mitochondria respiration. Aging (Albany NY) 2019; 10:3558-3573. [PMID: 30487319 PMCID: PMC6286837 DOI: 10.18632/aging.101668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022]
Abstract
p53, with its family members p63 and p73, have been shown to promote myoblast differentiation by regulation of the function of the retinoblastoma protein and by direct activation of p21Cip/Waf1 and p57Kip2, promoting cell cycle exit. In previous studies, we have demonstrated that the TAp63γ isoform is the only member of the p53 family that accumulates during in vitro myoblasts differentiation, and that its silencing led to delay in myotube fusion. To better dissect the role of TAp63γ in myoblast physiology, we have generated both sh-p63 and Tet-On inducible TAp63γ clones. Gene array analysis of sh-p63 C2C7 clones showed a significant modulation of genes involved in proliferation and cellular metabolism. Indeed, we found that sh-p63 C2C7 myoblasts present a higher proliferation rate and that, conversely, TAp63γ ectopic expression decreases myoblasts proliferation, indicating that TAp63γ specifically contributes to myoblasts proliferation, independently of p53 and p73. In addition, sh-p63 cells have a defect in mitochondria respiration highlighted by a reduction in spare respiratory capacity and a decrease in complex I, IV protein levels. These results demonstrated that, beside contributing to cell cycle exit, TAp63γ participates to myoblasts metabolism control.
Collapse
Affiliation(s)
- Veronica Ciuffoli
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Alessandra Gambacurta
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy.,MRC-Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Eleonora Candi
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy.,IDI-IRCCS, Biochemistry laboratory, Rome, Italy
| |
Collapse
|
20
|
Regulation of mitochondrial activity controls the duration of skeletal muscle regeneration in response to injury. Sci Rep 2019; 9:12249. [PMID: 31439911 PMCID: PMC6706433 DOI: 10.1038/s41598-019-48703-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 08/05/2019] [Indexed: 12/20/2022] Open
Abstract
Thyroid hormone is a major regulator of skeletal muscle development and repair, and also a key regulator of mitochondrial activity. We have previously identified a 43 kDa truncated form of the nuclear T3 receptor TRα1 (p43) which stimulates mitochondrial activity and regulates skeletal muscle features. However, its role in skeletal muscle regeneration remains to be addressed. To this end, we performed acute muscle injury induced by cardiotoxin in mouse tibialis in two mouse models where p43 is overexpressed in or depleted from skeletal muscle. The measurement of muscle fiber size distribution at different time point (up to 70 days) upon injury lead us to unravel requirement of the p43 signaling pathway for satellite cells dependent muscle regeneration; strongly delayed in the absence of p43; whereas the overexpression of the receptor enhances of the regeneration process. In addition, we found that satellite cells derived from p43-Tg mice display higher proliferation rates when cultured in vitro when compared to control myoblasts, whereas p43-/- satellites shows reduced proliferation capacity. These finding strongly support that p43 plays an important role in vivo by controling the duration of skeletal muscle regeneration after acute injury, possibly through the regulation of mitochondrial activity and myoblasts proliferation.
Collapse
|
21
|
Liu X, Qu H, Zheng Y, Liao Q, Zhang L, Liao X, Xiong X, Wang Y, Zhang R, Wang H, Tong Q, Liu Z, Dong H, Yang G, Zhu Z, Xu J, Zheng H. Mitochondrial glycerol 3-phosphate dehydrogenase promotes skeletal muscle regeneration. EMBO Mol Med 2019; 10:emmm.201809390. [PMID: 30389681 PMCID: PMC6284384 DOI: 10.15252/emmm.201809390] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
While adult mammalian skeletal muscle is stable due to its post‐mitotic nature, muscle regeneration is still essential throughout life for maintaining functional fitness. During certain diseases, such as the modern pandemics of obesity and diabetes, the regeneration process becomes impaired, which leads to the loss of muscle function and contributes to the global burden of these diseases. However, the underlying mechanisms of the impairment are not well defined. Here, we identify mGPDH as a critical regulator of skeletal muscle regeneration. Specifically, it regulates myogenic markers and myoblast differentiation by controlling mitochondrial biogenesis via CaMKKβ/AMPK. mGPDH−/− attenuated skeletal muscle regeneration in vitro and in vivo, while mGPDH overexpression ameliorated dystrophic pathology in mdx mice. Moreover, in patients and animal models of obesity and diabetes, mGPDH expression in skeletal muscle was reduced, further suggesting a direct correlation between its abundance and muscular regeneration capability. Rescuing mGPDH expression in obese and diabetic mice led to a significant improvement in their muscle regeneration. Our study provides a potential therapeutic target for skeletal muscle regeneration impairment during obesity and diabetes.
Collapse
Affiliation(s)
- Xiufei Liu
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hua Qu
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yi Zheng
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qian Liao
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Linlin Zhang
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xiaoyu Liao
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xin Xiong
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuren Wang
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Rui Zhang
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hui Wang
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qiang Tong
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Virginia Health System, Charlottesville, VA, USA
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jing Xu
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hongting Zheng
- Translational Research Key Laboratory for Diabetes, Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
22
|
Chattergoon NN. Thyroid hormone signaling and consequences for cardiac development. J Endocrinol 2019; 242:T145-T160. [PMID: 31117055 PMCID: PMC6613780 DOI: 10.1530/joe-18-0704] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 05/20/2019] [Indexed: 01/10/2023]
Abstract
The fetal heart undergoes its own growth and maturation stages all while supplying blood and nutrients to the growing fetus and its organs. Immature contractile cardiomyocytes proliferate to rapidly increase and establish cardiomyocyte endowment in the perinatal period. Maturational changes in cellular maturation, size and biochemical capabilities occur, and require, a changing hormonal environment as the fetus prepares itself for the transition to extrauterine life. Thyroid hormone has long been known to be important for neuronal development, but also for fetal size and survival. Fetal circulating 3,5,3'-triiodothyronine (T3) levels surge near term in mammals and are responsible for maturation of several organ systems, including the heart. Growth factors like insulin-like growth factor-1 stimulate proliferation of fetal cardiomyocytes, while thyroid hormone has been shown to inhibit proliferation and drive maturation of the cells. Several cell signaling pathways appear to be involved in this complicated and coordinated process. The aim of this review was to discuss the foundational studies of thyroid hormone physiology and the mechanisms responsible for its actions as we speculate on potential fetal programming effects for cardiovascular health.
Collapse
Affiliation(s)
- Natasha N Chattergoon
- Center for Developmental Health, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
23
|
Saleh A, Subramaniam G, Raychaudhuri S, Dhawan J. Cytoplasmic sequestration of the RhoA effector mDiaphanous1 by Prohibitin2 promotes muscle differentiation. Sci Rep 2019; 9:8302. [PMID: 31165762 PMCID: PMC6549159 DOI: 10.1038/s41598-019-44749-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/23/2019] [Indexed: 02/06/2023] Open
Abstract
Muscle differentiation is controlled by adhesion and growth factor-dependent signalling through common effectors that regulate muscle-specific transcriptional programs. Here we report that mDiaphanous1, an effector of adhesion-dependent RhoA-signalling, negatively regulates myogenesis at the level of Myogenin expression. In myotubes, over-expression of mDia1ΔN3, a RhoA-independent mutant, suppresses Myogenin promoter activity and expression. We investigated mDia1-interacting proteins that may counteract mDia1 to permit Myogenin expression and timely differentiation. Using yeast two-hybrid and mass-spectrometric analysis, we report that mDia1 has a stage-specific interactome, including Prohibitin2, MyoD, Akt2, and β-Catenin, along with a number of proteosomal and mitochondrial components. Of these interacting partners, Prohibitin2 colocalises with mDia1 in cytoplasmic punctae in myotubes. We mapped the interacting domains of mDia1 and Phb2, and used interacting (mDia1ΔN3/Phb2 FL or mDia1ΔN3/Phb2-Carboxy) and non-interacting pairs (mDia1H + P/Phb2 FL or mDia1ΔN3/Phb2-Amino) to dissect the functional consequences of this partnership on Myogenin promoter activity. Co-expression of full-length as well as mDia1-interacting domains of Prohibitin2 reverse the anti-myogenic effects of mDia1ΔN3, while non-interacting regions do not. Our results suggest that Prohibitin2 sequesters mDia1, dampens its anti-myogenic activity and fine-tunes RhoA-mDia1 signalling to promote differentiation. Overall, we report that mDia1 is multi-functional signalling effector whose anti-myogenic activity is modulated by a differentiation-dependent interactome. The data have been deposited to the ProteomeXchange with identifier PXD012257.
Collapse
Affiliation(s)
- Amena Saleh
- Institute for Stem Cell Science & Regenerative Medicine, Bangalore, Karnataka, 560065, India
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
- Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Gunasekaran Subramaniam
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Swasti Raychaudhuri
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
| | - Jyotsna Dhawan
- Institute for Stem Cell Science & Regenerative Medicine, Bangalore, Karnataka, 560065, India.
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India.
| |
Collapse
|
24
|
Leng X, Ji X, Hou Y, Settlage R, Jiang H. Roles of the proteasome and inhibitor of DNA binding 1 protein in myoblast differentiation. FASEB J 2019; 33:7403-7416. [PMID: 30865843 DOI: 10.1096/fj.201800574rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
This study was conducted to further understand the mechanism that controls myoblast differentiation, a key step in skeletal muscle formation. RNA sequencing of primary bovine myoblasts revealed many genes encoding the ubiquitin-proteasome system were up-regulated during myoblast differentiation. This up-regulation was accompanied by increased proteasomal activity. Treating myoblasts with the proteasome-specific inhibitor lactacystin impeded myoblast differentiation. Adenovirus-mediated overexpression of inhibitor of DNA binding 1 (ID1) protein inhibited myoblast differentiation too. Further experiments were conducted to determine whether the proteasome promotes myoblast differentiation by degrading ID1 protein. Both ID1 protein and mRNA expression decreased during myoblast differentiation. However, treating myoblasts with lactacystin reversed the decrease in ID1 protein but not in ID1 mRNA expression. Surprisingly, this reversal was not observed when myoblasts were also treated with the mRNA translation inhibitor cycloheximide. Direct incubation of ID1 protein with proteasomes from myoblasts did not show differentiation stage-associated degradation of ID1 protein. Furthermore, ubiquitinated ID1 protein was not detected in lactacystin-treated myoblasts. Overall, the results of this study suggest that, during myoblast differentiation, the proteasomal activity is up-regulated to further myoblast differentiation and that the increased proteasomal activity improves myoblast differentiation partly by inhibiting the synthesis, not the degradation, of ID1 protein.-Leng, X., Ji, X., Hou, Y., Settlage, R., Jiang, H. Roles of the proteasome and inhibitor of DNA binding 1 protein in myoblast differentiation.
Collapse
Affiliation(s)
- Xinyan Leng
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Xu Ji
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, USA.,College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China; and
| | - Yuguo Hou
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Robert Settlage
- Advanced Research Computing Unit, Division of Information Technology, Virginia Tech, Blacksburg, Virginia, USA
| | - Honglin Jiang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
25
|
Prince LM, Rand MD. Methylmercury exposure causes a persistent inhibition of myogenin expression and C2C12 myoblast differentiation. Toxicology 2018; 393:113-122. [PMID: 29104120 PMCID: PMC5757876 DOI: 10.1016/j.tox.2017.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/03/2017] [Accepted: 11/01/2017] [Indexed: 12/11/2022]
Abstract
Methylmercury (MeHg) is a ubiquitous environmental toxicant, best known for its selective targeting of the developing nervous system. MeHg exposure has been shown to cause motor deficits such as impaired gait and coordination, muscle weakness, and muscle atrophy, which have been associated with disruption of motor neurons. However, recent studies have suggested that muscle may also be a target of MeHg toxicity, both in the context of developmental myogenic events and of low-level chronic exposures affecting muscle wasting in aging. We therefore investigated the effects of MeHg on myotube formation, using the C2C12 mouse myoblast model. We found that MeHg inhibits both differentiation and fusion, in a concentration-dependent manner. Furthermore, MeHg specifically and persistently inhibits myogenin (MyoG), a transcription factor involved in myocyte differentiation, within the first six hours of exposure. MeHg-induced reduction in MyoG expression is contemporaneous with a reduction of a number of factors involved in mitochondrial biogenesis and mtDNA transcription and translation, which may implicate a role for mitochondria in mediating MeHg-induced change in the differentiation program. Unexpectedly, inhibition of myoblast differentiation with MeHg parallels inhibition of Notch receptor signaling. Our research establishes muscle cell differentiation as a target for MeHg toxicity, which may contribute to the underlying etiology of motor deficits with MeHg toxicity.
Collapse
Affiliation(s)
- Lisa M Prince
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Department of Environmental Medicine, Rochester, NY, 14642, USA.
| | - Matthew D Rand
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Department of Environmental Medicine, Rochester, NY, 14642, USA.
| |
Collapse
|
26
|
Wrutniak-Cabello C, Casas F, Cabello G. Thyroid Hormone Action: The p43 Mitochondrial Pathway. Methods Mol Biol 2018; 1801:163-181. [PMID: 29892824 DOI: 10.1007/978-1-4939-7902-8_14] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The possibility that several pathways are involved in the multiplicity of thyroid hormone physiological influences led to searches for the occurrence of T3 extra nuclear receptors. The existence of a direct T3 mitochondrial pathway is now well established. The demonstration that TRα1 mRNA encodes not only a nuclear thyroid hormone receptor but also two proteins imported into mitochondria with molecular masses of 43 and 28 kDa has provided new clues to understand the pleiotropic influence of iodinated hormones.The use of a T3 photo affinity label derivative (T3-PAL) allowed detecting two mitochondrial T3 binding proteins. In association with western blots using antibodies raised against the T3 nuclear receptor TRα1, mitochondrial T3 receptors were identified as truncated TRα1 forms. Import and in organello transcription experiments performed in isolated mitochondria led to the conclusion that p43 is a transcription factor of the mitochondrial genome, inducing changes in the mitochondrial/nuclear crosstalk. In vitro experiments indicated that this T3 mitochondrial pathway affects cell differentiation, apoptosis, and transformation. Generation of transgenic mice demonstrated the involvement of this mitochondrial pathway in the determination of muscle phenotype, glucose metabolism, and thermogenesis.
Collapse
|
27
|
Wrutniak-Cabello C, Casas F, Cabello G. Mitochondrial T3 receptor and targets. Mol Cell Endocrinol 2017; 458:112-120. [PMID: 28167126 DOI: 10.1016/j.mce.2017.01.054] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/28/2017] [Accepted: 01/31/2017] [Indexed: 12/25/2022]
Abstract
The demonstration that TRα1 mRNA encodes a nuclear thyroid hormone receptor and two proteins imported into mitochondria with molecular masses of 43 and 28 kDa has brought new clues to better understand the pleiotropic influence of iodinated hormones. If p28 activity remains unknown, p43 binds to T3 responsive elements occurring in the organelle genome, and, in the T3 presence, stimulates mitochondrial transcription and the subsequent synthesis of mitochondrial encoded proteins. This influence increases mitochondrial activity and through changes in the mitochondrial/nuclear cross talk affects important nuclear target genes regulating cell proliferation and differentiation, oncogenesis, or apoptosis. In addition, this pathway influences muscle metabolic and contractile phenotype, as well as glycaemia regulation. Interestingly, according to the process considered, p43 exerts opposite or cooperative effects with the well-known T3 pathway, thus allowing a fine tuning of the physiological influence of this hormone.
Collapse
Affiliation(s)
- Chantal Wrutniak-Cabello
- INRA, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France; Université de Montpellier, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France.
| | - François Casas
- INRA, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France; Université de Montpellier, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France
| | - Gérard Cabello
- INRA, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France; Université de Montpellier, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France
| |
Collapse
|
28
|
The Combination of Physical Exercise with Muscle-Directed Antioxidants to Counteract Sarcopenia: A Biomedical Rationale for Pleiotropic Treatment with Creatine and Coenzyme Q10. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7083049. [PMID: 29123615 PMCID: PMC5632475 DOI: 10.1155/2017/7083049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/13/2017] [Accepted: 08/23/2017] [Indexed: 12/21/2022]
Abstract
Sarcopenia represents an increasing public health risk due to the rapid aging of the world's population. It is characterized by both low muscle mass and function and is associated with mobility disorders, increased risk of falls and fractures, loss of independence, disabilities, and increased risk of death. Despite the urgency of the problem, the development of treatments for sarcopenia has lagged. Increased reactive oxygen species (ROS) production and decreased antioxidant (AO) defences seem to be important factors contributing to muscle impairment. Studies have been conducted to verify whether physical exercise and/or AOs could prevent and/or delay sarcopenia through a normalization of the etiologically relevant ROS imbalance. Despite the strong rationale, the results obtained were contradictory, particularly with regard to the effects of the tested AOs. A possible explanation might be that not all the agents included in the general heading of "AOs" could fulfill the requisites to counteract the complex series of events causing/accelerating sarcopenia: the combination of the muscle-directed antioxidants creatine and coenzyme Q10 with physical exercise as a biomedical rationale for pleiotropic prevention and/or treatment of sarcopenia is discussed.
Collapse
|
29
|
FTO is required for myogenesis by positively regulating mTOR-PGC-1α pathway-mediated mitochondria biogenesis. Cell Death Dis 2017; 8:e2702. [PMID: 28333151 PMCID: PMC5386528 DOI: 10.1038/cddis.2017.122] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 02/05/2023]
Abstract
Global germ line loss of fat mass- and obesity-associated (FTO) gene results in both the reduction of fat mass and lean mass in mice. The role of FTO in adipogenesis has been proposed, however, that in myogenesis has not. Skeletal muscle is the main component of body lean mass, so its connection with FTO physiologic significance need to be clarified. Here, we assessed the impact of FTO on murine skeletal muscle differentiation by in vitro and in vivo experiments. We found that FTO expression increased during myoblasts differentiation, while the silence of FTO inhibited the differentiation; in addition, skeletal muscle development was impaired in skeletal muscle FTO-deficient mice. Significantly, FTO-promoted myogenic differentiation was dependent on its m6A demethylase activity. Mechanically, we found that FTO downregulation suppressed mitochondria biogenesis and energy production, showing as the decreased mitochondria mass and mitochondrial DNA (mtDNA) content, the downregulated expression of mtDNA-encoding genes and peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) gene, together with declined ATP level. Moreover, the involvement of mTOR-PGC-1α pathway in the connection between FTO and muscle differentiation is displayed, since the expression of FTO affected the activity of mTOR and rapamycin blocked FTO-induced PGC-1α transcription, along with the parallel alteration pattern of FTO expression and mTOR phosphorylation during myoblasts differentiation. Summarily, our findings provide the first evidence for the contribution of FTO for skeletal muscle differentiation and a new insight to study the physiologic significance of RNA methylation.
Collapse
|
30
|
Abstract
The hypermetabolic effects of thyroid hormones (THs), the major endocrine regulators of metabolic rate, are widely recognized. Although, the cellular mechanisms underlying these effects have been extensively investigated, much has yet to be learned about how TH regulates diverse cellular functions. THs have a profound impact on mitochondria, the organelles responsible for the majority of cellular energy production, and several studies have been devoted to understand the respective importance of the nuclear and mitochondrial pathways for organelle activity. During the last decades, several new aspects of both THs (i.e., metabolism, transport, mechanisms of action, and the existence of metabolically active TH derivatives) and mitochondria (i.e., dynamics, respiratory chain organization in supercomplexes, and the discovery of uncoupling proteins other than uncoupling protein 1) have emerged, thus opening new perspectives to the investigation of the complex relationship between thyroid and the mitochondrial compartment. In this review, in the light of an historical background, we attempt to point out the present findings regarding thyroid physiology and the emerging recognition that mitochondrial dynamics as well as the arrangement of the electron transport chain in mitochondrial cristae contribute to the mitochondrial activity. We unravel the genomic and nongenomic mechanisms so far studied as well as the effects of THs on mitochondrial energetics and, principally, uncoupling of oxidative phosphorylation via various mechanisms involving uncoupling proteins. The emergence of new approaches to the question as to what extent and how the action of TH can affect mitochondria is highlighted. © 2016 American Physiological Society. Compr Physiol 6:1591-1607, 2016.
Collapse
Affiliation(s)
- Antonia Lanni
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università degli Studi di Napoli, Caserta, Italy
| | - Maria Moreno
- Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, Benevento, Italy
| | - Fernando Goglia
- Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, Benevento, Italy
| |
Collapse
|
31
|
Creatine Prevents the Structural and Functional Damage to Mitochondria in Myogenic, Oxidatively Stressed C2C12 Cells and Restores Their Differentiation Capacity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5152029. [PMID: 27610211 PMCID: PMC5005540 DOI: 10.1155/2016/5152029] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 06/29/2016] [Indexed: 11/18/2022]
Abstract
Creatine (Cr) is a nutritional supplement promoting a number of health benefits. Indeed Cr has been shown to be beneficial in disease-induced muscle atrophy, improve rehabilitation, and afford mild antioxidant activity. The beneficial effects are likely to derive from pleiotropic interactions. In accord with this notion, we previously demonstrated that multiple pleiotropic effects, including preservation of mitochondrial damage, account for the capacity of Cr to prevent the differentiation arrest caused by oxidative stress in C2C12 myoblasts. Given the importance of mitochondria in supporting the myogenic process, here we further explored the protective effects of Cr on the structure, function, and networking of these organelles in C2C12 cells differentiating under oxidative stressing conditions; the effects on the energy sensor AMPK, on PGC-1α, which is involved in mitochondrial biogenesis and its downstream effector Tfam were also investigated. Our results indicate that damage to mitochondria is crucial in the differentiation imbalance caused by oxidative stress and that the Cr-prevention of these injuries is invariably associated with the recovery of the normal myogenic capacity. We also found that Cr activates AMPK and induces an upregulation of PGC-1α expression, two events which are likely to contribute to the protection of mitochondrial quality and function.
Collapse
|
32
|
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
|
33
|
Grefte S, Wagenaars JAL, Jansen R, Willems PHGM, Koopman WJH. Rotenone inhibits primary murine myotube formation via Raf-1 and ROCK2. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1606-14. [PMID: 25827955 DOI: 10.1016/j.bbamcr.2015.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/04/2015] [Accepted: 03/19/2015] [Indexed: 11/30/2022]
Abstract
Rotenone (ROT) is a widely used inhibitor of complex I (CI), the first complex of the mitochondrial oxidative phosphorylation (OXPHOS) system. However, particularly at high concentrations ROT was also described to display off-target effects. Here we studied how ROT affected in vitro primary murine myotube formation. We demonstrate that myotube formation is specifically inhibited by ROT (10-100nM), but not by piericidin A (PA; 100nM), another CI inhibitor. At 100nM, both ROT and PA fully blocked myoblast oxygen consumption. Knock-down of Rho-associated, coiled-coil containing protein kinase 2 (ROCK2) and, to a lesser extent ROCK1, prevented the ROT-induced inhibition of myotube formation. Moreover, the latter was reversed by inhibiting Raf-1 activity. In contrast, ROT-induced inhibition of myotube formation was not prevented by knock-down of RhoA. Taken together, our results support a model in which ROT reduces primary myotube formation independent of its inhibitory effect on CI-driven mitochondrial ATP production, but via a mechanism primarily involving the Raf-1/ROCK2 pathway.
Collapse
Affiliation(s)
- Sander Grefte
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jori A L Wagenaars
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Renate Jansen
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter H G M Willems
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Werner J H Koopman
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
34
|
Mesquita-Ferrari RA, Alves AN, de Oliveira Cardoso V, Artilheiro PP, Bussadori SK, Rocha LA, Nunes FD, Fernandes KPS. Low-level laser irradiation modulates cell viability and creatine kinase activity in C2C12 muscle cells during the differentiation process. Lasers Med Sci 2015; 30:2209-13. [PMID: 25616713 DOI: 10.1007/s10103-015-1715-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 01/09/2015] [Indexed: 12/16/2022]
Abstract
Low-level laser irradiation (LLLI) is increasingly used to treat musculoskeletal disorders, with satisfactory results described in the literature. Skeletal muscle satellite cells play a key role in muscle regeneration. The aim of the present study was to evaluate the effect of LLLI on cell viability, creatine kinase (CK) activity, and the expression of myogenic regulatory factors in C2C12 myoblasts during the differentiation process. C2C12 cells were cultured in Dulbecco's modified Eagle's medium (DMEM) containing 2% horse serum and submitted to irradiation with GaAlAs diode laser (wavelength, 780 nm; output power, 10 mW; energy density, 5 J/cm2). Cell viability and the expression of myogenic regulatory factors were assessed 24, 48, and 72 h after irradiation by 3-(4,5-dimethylthiazol-2-yl)-2,5,-diphenyltetrazolium bromide (MTT) assay and quantitative real-time polymerase chain reaction (RT-qPCR), respectively. CK activity was analyzed at 24 and 72 h. An increase in cell viability was found in the laser group in comparison to the control group at all evaluation times. CK activity was significantly increased in the laser group at 72 h. Myogenin messenger RNA (mRNA) demonstrated a tendency toward an increase in the laser group, but the difference in comparison to the control group was non-significant. In conclusion, LLLI was able to modulate cell viability and CK activity in C2C12 myoblasts during the differentiation process.
Collapse
Affiliation(s)
- Raquel Agnelli Mesquita-Ferrari
- Postgraduate Program in Rehabilitation Sciences and Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235/249, Liberdade, São Paulo, SP, 01504-001, Brazil.
| | - Agnelo Neves Alves
- Postgraduate Program in Rehabilitation Sciences and Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235/249, Liberdade, São Paulo, SP, 01504-001, Brazil
| | - Vinicius de Oliveira Cardoso
- Postgraduate Program in Rehabilitation Sciences and Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235/249, Liberdade, São Paulo, SP, 01504-001, Brazil
| | - Paola Pelegrineli Artilheiro
- Postgraduate Program in Rehabilitation Sciences and Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235/249, Liberdade, São Paulo, SP, 01504-001, Brazil
| | - Sandra Kalil Bussadori
- Postgraduate Program in Rehabilitation Sciences and Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235/249, Liberdade, São Paulo, SP, 01504-001, Brazil
| | - Lilia Alves Rocha
- Departament of Molecular Pathology, School of Dentistry, University of São Paulo, Av. Professor Lineu Prestes, 2227, Cidade Universitária, São Paulo, 05508-000, SP, Brazil
| | - Fábio Daumas Nunes
- Department of Oral Pathology, School of Dentistry, University of São Paulo, Av. Professor Lineu Prestes, 2227, Cidade Universitária, São Paulo, 05508-000, SP, Brazil
| | - Kristianne Porta Santos Fernandes
- Postgraduate Program in Rehabilitation Sciences and Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235/249, Liberdade, São Paulo, SP, 01504-001, Brazil
| |
Collapse
|
35
|
Peng WH, Lee YC, Chau YP, Lu KS, Kung HN. Short-term exposure of zebrafish embryos to arecoline leads to retarded growth, motor impairment, and somite muscle fiber changes. Zebrafish 2014; 12:58-70. [PMID: 25549301 DOI: 10.1089/zeb.2014.1010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The areca nut-chewing habit is common in Southeast Asia, India, and Taiwan, and arecoline is the most abundant and potent component in the areca nut. The effects of arecoline on birth defects have been explored in many species, including chicken, mice, and zebrafish. The effects of arecoline on embryos after long-term exposure are well established; however, the effects of short-term embryo exposure to arecoline are not understood. Using zebrafish, we study the effects of short-term exposure of arecoline on embryos to model the human habit of areca nut-chewing during early pregnancy. Arecoline, at concentrations from 0.001% to 0.04%, was administered to zebrafish embryos from 4 to 24 hours post fertilization. The morphological changes, survival rates, body length, and skeletal muscle fiber structure were then investigated by immunohistochemistry, confocal microscopy, and conventional electron microscopy. With exposure of embryos to increasing arecoline concentrations, we observed a significant decline in the hatching and survival rates, general growth retardation, lower locomotor activity, and swimming ability impairment. Immunofluorescent staining demonstrated a loose arrangement of myosin heavy chains, and ultrastructural observations revealed altered myofibril arrangement and swelling of the mitochondria. In addition, the results of flow-cytometry and JC-1 staining to assay mitochondria activity, as well as reverse transcription-polymerase chain reaction analyses of functional gene expression, revealed mitochondrial dysfunctions after exposure to arecoline. We confirmed that short-term arecoline exposure resulted in retarded embryonic development and decreased locomotor activity due to defective somitic skeletal muscle development and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Wei-Hau Peng
- 1 Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University , Taipei, Taiwan
| | | | | | | | | |
Collapse
|
36
|
Abdel Khalek W, Cortade F, Ollendorff V, Lapasset L, Tintignac L, Chabi B, Wrutniak-Cabello C. SIRT3, a mitochondrial NAD⁺-dependent deacetylase, is involved in the regulation of myoblast differentiation. PLoS One 2014; 9:e114388. [PMID: 25489948 PMCID: PMC4260865 DOI: 10.1371/journal.pone.0114388] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 11/06/2014] [Indexed: 12/13/2022] Open
Abstract
Sirtuin 3 (SIRT3), one of the seven mammalian sirtuins, is a mitochondrial NAD+-dependent deacetylase known to control key metabolic pathways. SIRT3 deacetylases and activates a large number of mitochondrial enzymes involved in the respiratory chain, in ATP production, and in both the citric acid and urea cycles. We have previously shown that the regulation of myoblast differentiation is tightly linked to mitochondrial activity. Since SIRT3 modulates mitochondrial activity, we decide to address its role during myoblast differentiation. For this purpose, we first investigated the expression of endogenous SIRT3 during C2C12 myoblast differentiation. We further studied the impact of SIRT3 silencing on both the myogenic potential and the mitochondrial activity of C2C12 cells. We showed that SIRT3 protein expression peaked at the onset of myoblast differentiation. The inhibition of SIRT3 expression mediated by the stable integration of SIRT3 short inhibitory RNA (SIRT3shRNA) in C2C12 myoblasts, resulted in: 1) abrogation of terminal differentiation - as evidenced by a marked decrease in the myoblast fusion index and a significant reduction of Myogenin, MyoD, Sirtuin 1 and Troponin T protein expression - restored upon MyoD overexpression; 2) a decrease in peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and citrate synthase protein expression reflecting an alteration of mitochondrial density; and 3) an increased production of reactive oxygen species (ROS) mirrored by the decreased activity of manganese superoxide dismutase (MnSOD). Altogether our data demonstrate that SIRT3 mainly regulates myoblast differentiation via its influence on mitochondrial activity.
Collapse
Affiliation(s)
- Waed Abdel Khalek
- INRA, UMR866 Dynamique Musculaire et Métabolisme, F-34060 Montpellier - Université Montpellier 1, F-34000 Montpellier - Université Montpellier 2, Montpellier, France
| | - Fabienne Cortade
- INRA, UMR866 Dynamique Musculaire et Métabolisme, F-34060 Montpellier - Université Montpellier 1, F-34000 Montpellier - Université Montpellier 2, Montpellier, France
| | - Vincent Ollendorff
- INRA, UMR866 Dynamique Musculaire et Métabolisme, F-34060 Montpellier - Université Montpellier 1, F-34000 Montpellier - Université Montpellier 2, Montpellier, France
| | - Laure Lapasset
- IGMM, Institut de Génétique Moléculaire de Montpellier, CNRS-UMR5535, Montpellier France - Université Montpellier 1, F-34000 Montpellier - Université Montpellier 2, Montpellier, France
| | - Lionel Tintignac
- INRA, UMR866 Dynamique Musculaire et Métabolisme, F-34060 Montpellier - Université Montpellier 1, F-34000 Montpellier - Université Montpellier 2, Montpellier, France
- Biozentrum, University of Basel, Basel, Switzerland
| | - Béatrice Chabi
- INRA, UMR866 Dynamique Musculaire et Métabolisme, F-34060 Montpellier - Université Montpellier 1, F-34000 Montpellier - Université Montpellier 2, Montpellier, France
| | - Chantal Wrutniak-Cabello
- INRA, UMR866 Dynamique Musculaire et Métabolisme, F-34060 Montpellier - Université Montpellier 1, F-34000 Montpellier - Université Montpellier 2, Montpellier, France
- * E-mail:
| |
Collapse
|
37
|
Pessemesse L, Lepourry L, Bouton K, Levin J, Cabello G, Wrutniak-Cabello C, Casas F. p28, a truncated form of TRα1 regulates mitochondrial physiology. FEBS Lett 2014; 588:4037-43. [PMID: 25263706 DOI: 10.1016/j.febslet.2014.09.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 09/12/2014] [Accepted: 09/15/2014] [Indexed: 12/13/2022]
Abstract
We have previously identified in mitochondria two truncated forms of the T3 nuclear receptor TRα1, with molecular weights of 43kDa (p43) and 28kDa (p28) respectively located in the matrix and in the inner membrane. Previously, we have demonstrated that p43 stimulates mitochondrial transcription and protein synthesis in the presence of T3. Here we report that p28 is targeted into the organelle in a T3-dependent manner and displays an affinity for T3 higher than the nuclear receptor. We tried to generate mice overexpressing p28 using the human α-skeletal actin promoter, however we found an early embryonic lethality that was probably linked to a transient expression of p28 in trophoblast giant cells. This could be partly explained by the observation that overexpression of p28 in human fibroblasts induced alterations of mitochondrial physiology.
Collapse
Affiliation(s)
- Laurence Pessemesse
- INRA, UMR866 Dynamique Musculaire et Métabolisme, 2 place Viala, F-34060 Montpellier, France; Université Montpellier I et II, F-34060 Montpellier, France
| | - Laurence Lepourry
- INRA, UMR866 Dynamique Musculaire et Métabolisme, 2 place Viala, F-34060 Montpellier, France; Université Montpellier I et II, F-34060 Montpellier, France
| | - Katia Bouton
- INRA, UMR866 Dynamique Musculaire et Métabolisme, 2 place Viala, F-34060 Montpellier, France; Université Montpellier I et II, F-34060 Montpellier, France
| | - Jonathan Levin
- INRA, UMR866 Dynamique Musculaire et Métabolisme, 2 place Viala, F-34060 Montpellier, France; Université Montpellier I et II, F-34060 Montpellier, France
| | - Gérard Cabello
- INRA, UMR866 Dynamique Musculaire et Métabolisme, 2 place Viala, F-34060 Montpellier, France; Université Montpellier I et II, F-34060 Montpellier, France
| | - Chantal Wrutniak-Cabello
- INRA, UMR866 Dynamique Musculaire et Métabolisme, 2 place Viala, F-34060 Montpellier, France; Université Montpellier I et II, F-34060 Montpellier, France
| | - François Casas
- INRA, UMR866 Dynamique Musculaire et Métabolisme, 2 place Viala, F-34060 Montpellier, France; Université Montpellier I et II, F-34060 Montpellier, France.
| |
Collapse
|
38
|
Sondag GR, Salihoglu S, Lababidi SL, Crowder DC, Moussa FM, Abdelmagid SM, Safadi FF. Osteoactivin induces transdifferentiation of C2C12 myoblasts into osteoblasts. J Cell Physiol 2014; 229:955-66. [PMID: 24265122 DOI: 10.1002/jcp.24512] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 11/18/2013] [Indexed: 12/19/2022]
Abstract
Osteoactivin (OA) is a novel osteogenic factor important for osteoblast differentiation and function. Previous studies showed that OA stimulates matrix mineralization and transcription of osteoblast specific genes required for differentiation. OA plays a role in wound healing and its expression was shown to increase in post fracture calluses. OA expression was reported in muscle as OA is upregulated in cases of denervation and unloading stress. The regulatory mechanisms of OA in muscle and bone have not yet been determined. In this study, we examined whether OA plays a role in transdifferentiation of C2C12 myoblast into osteoblasts. Infected C2C12 with a retroviral vector overexpressing OA under the CMV promoter were able to transdifferentiate from myoblasts into osteoblasts. Immunofluorescence analysis showed that skeletal muscle marker MF-20 was severely downregulated in cells overexpressing OA and contained significantly less myotubes compared to uninfected control. C2C12 myoblasts overexpressing OA showed an increase in expression of bone specific markers such as alkaline phosphatase and alizarin red staining, and also showed an increase in Runx2 protein expression. We also detected increased levels of phosphorylated focal adhesion kinase (FAK) in C2C12 myoblasts overexpressing OA compared to control. Taken together, our results suggest that OA is able to induce transdifferentiation of myoblasts into osteoblasts through increasing levels of phosphorylated FAK.
Collapse
Affiliation(s)
- Gregory R Sondag
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, Ohio; School of Biomedical Sciences, Kent State University, Kent, Ohio
| | | | | | | | | | | | | |
Collapse
|
39
|
Bertrand C, Blanchet E, Pessemesse L, Annicotte JS, Feillet-Coudray C, Chabi B, Levin J, Fajas L, Cabello G, Wrutniak-Cabello C, Casas F. Mice lacking the p43 mitochondrial T3 receptor become glucose intolerant and insulin resistant during aging. PLoS One 2013; 8:e75111. [PMID: 24098680 PMCID: PMC3787095 DOI: 10.1371/journal.pone.0075111] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 08/09/2013] [Indexed: 01/29/2023] Open
Abstract
Thyroid hormones (TH) play an important regulatory role in energy expenditure regulation and are key regulators of mitochondrial activity. We have previously identified a mitochondrial triiodothyronine (T3) receptor (p43) which acts as a mitochondrial transcription factor of the organelle genome, which leads in vitro and in vivo, to a stimulation of mitochondrial biogenesis. Recently, we generated mice carrying a specific p43 invalidation. At 2 months of age, we reported that p43 depletion in mice induced a major defect in insulin secretion both in vivo and in isolated pancreatic islets, and a loss of glucose-stimulated insulin secretion. The present study was designed to determine whether p43 invalidation influences life expectancy and modulates blood glucose and insulin levels as well as glucose tolerance or insulin sensitivity during aging. We report that from 4 months old onwards, mice lacking p43 are leaner than wild-type mice. p43−/− mice also have a moderate reduction of life expectancy compared to wild type. We found no difference in blood glucose levels, excepted at 24 months old where p43−/− mice showed a strong hyperglycemia in fasting conditions compared to controls animals. However, the loss of glucose-stimulated insulin secretion was maintained whatever the age of mice lacking p43. If up to 12 months old, glucose tolerance remained unchanged, beyond this age p43−/− mice became increasingly glucose intolerant. In addition, if up to 12 months old p43 deficient animals were more sensitive to insulin, after this age we observed a loss of this capacity, culminating in 24 months old mice with a decreased sensitivity to the hormone. In conclusion, we demonstrated that during aging the depletion of the mitochondrial T3 receptor p43 in mice progressively induced an increased glycemia in the fasted state, glucose intolerance and an insulin-resistance several features of type-2 diabetes.
Collapse
Affiliation(s)
- Christelle Bertrand
- INRA, Institut National de la Recherche Agronomique, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France; Université Montpellier 1, Montpellier, France; Université Montpellier 2, Montpellier, France
| | - Emilie Blanchet
- IGMM, Institut de Génétique Moléculaire de Montpellier, CNRS-UMR5535, Montpellier France; Université Montpellier 1, Montpellier, France; Université Montpellier 2, Montpellier France
| | - Laurence Pessemesse
- INRA, Institut National de la Recherche Agronomique, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France; Université Montpellier 1, Montpellier, France; Université Montpellier 2, Montpellier, France
| | - Jean Sébastien Annicotte
- IGMM, Institut de Génétique Moléculaire de Montpellier, CNRS-UMR5535, Montpellier France; Université Montpellier 1, Montpellier, France; Université Montpellier 2, Montpellier France
| | - Christine Feillet-Coudray
- INRA, Institut National de la Recherche Agronomique, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France; Université Montpellier 1, Montpellier, France; Université Montpellier 2, Montpellier, France
| | - Béatrice Chabi
- INRA, Institut National de la Recherche Agronomique, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France; Université Montpellier 1, Montpellier, France; Université Montpellier 2, Montpellier, France
| | - Jonathan Levin
- INRA, Institut National de la Recherche Agronomique, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France; Université Montpellier 1, Montpellier, France; Université Montpellier 2, Montpellier, France
| | - Lluis Fajas
- IGMM, Institut de Génétique Moléculaire de Montpellier, CNRS-UMR5535, Montpellier France; Université Montpellier 1, Montpellier, France; Université Montpellier 2, Montpellier France
| | - Gérard Cabello
- INRA, Institut National de la Recherche Agronomique, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France; Université Montpellier 1, Montpellier, France; Université Montpellier 2, Montpellier, France
| | - Chantal Wrutniak-Cabello
- INRA, Institut National de la Recherche Agronomique, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France; Université Montpellier 1, Montpellier, France; Université Montpellier 2, Montpellier, France
| | - François Casas
- INRA, Institut National de la Recherche Agronomique, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France; Université Montpellier 1, Montpellier, France; Université Montpellier 2, Montpellier, France
- * E-mail:
| |
Collapse
|
40
|
Fumel B, Roy S, Fouchécourt S, Livera G, Parent AS, Casas F, Guillou F. Depletion of the p43 mitochondrial T3 receptor increases Sertoli cell proliferation in mice. PLoS One 2013; 8:e74015. [PMID: 24040148 PMCID: PMC3767600 DOI: 10.1371/journal.pone.0074015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/26/2013] [Indexed: 01/01/2023] Open
Abstract
Among T3 receptors, TRα1 is ubiquitous and its deletion or a specific expression of a dominant-negative TRα1 isoform in Sertoli cell leads to an increase in testis weight and sperm production. The identification of a 43-kDa truncated form of the nuclear receptor TRα1 (p43) in the mitochondrial matrix led us to test the hypothesis that this mitochondrial transcription factor could regulate Sertoli cell proliferation. Here we report that p43 depletion in mice increases testis weight and sperm reserve. In addition, we found that p43 deletion increases Sertoli cell proliferation in postnatal testis at 3 days of development. Electron microscopy studies evidence an alteration of mitochondrial morphology observed specifically in Sertoli cells of p43−/− mice. Moreover, gene expression studies indicate that the lack of p43 in testis induced an alteration of the mitochondrial-nuclear cross-talk. In particular, the up-regulation of Cdk4 and c-myc pathway in p43−/− probably explain the extended proliferation recorded in Sertoli cells of these mice. Our finding suggests that T3 limits post-natal Sertoli cell proliferation mainly through its mitochondrial T3 receptor p43.
Collapse
Affiliation(s)
- Betty Fumel
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France
- CNRS, UMR7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France
- Université François Rabelais de Tours, Tours, France
- IFCE, Nouzilly, France
| | - Stéphanie Roy
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Sophie Fouchécourt
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Gabriel Livera
- Université Paris Diderot, Sorbonne Paris Cité, INSERM U967, CEA/DSV/iRCM/SCSR Laboratoire de Développement des Gonades, Fontenay-Aux-Roses, France
| | - Anne-Simone Parent
- Developmental Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, CHU Sart Tilman, Liège, Belgium
| | - François Casas
- INRA, UMR 866 Dynamique Musculaire et métabolisme, Montpellier, France
- Université de Montpellier 1 et 2, Montpellier, France
| | - Florian Guillou
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France
- * E-mail:
| |
Collapse
|
41
|
Skeletal muscle PGC-1α controls whole-body lactate homeostasis through estrogen-related receptor α-dependent activation of LDH B and repression of LDH A. Proc Natl Acad Sci U S A 2013; 110:8738-43. [PMID: 23650363 DOI: 10.1073/pnas.1212976110] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) controls metabolic adaptations. We now show that PGC-1α in skeletal muscle drives the expression of lactate dehydrogenase (LDH) B in an estrogen-related receptor-α-dependent manner. Concomitantly, PGC-1α reduces the expression of LDH A and one of its regulators, the transcription factor myelocytomatosis oncogene. PGC-1α thereby coordinately alters the composition of the LDH complex and prevents the increase in blood lactate during exercise. Our results show how PGC-1α actively coordinates lactate homeostasis and provide a unique molecular explanation for PGC-1α-mediated muscle adaptations to training that ultimately enhance exercise performance and improve metabolic health.
Collapse
|
42
|
Mitochondria as a potential regulator of myogenesis. ScientificWorldJournal 2013; 2013:593267. [PMID: 23431256 PMCID: PMC3574753 DOI: 10.1155/2013/593267] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 01/16/2013] [Indexed: 12/24/2022] Open
Abstract
Recent studies have shown that mitochondria play a role in the regulation of myogenesis. Indeed, the abundance, morphology, and functional properties of mitochondria become altered when the myoblasts differentiate into myotubes. For example, mitochondrial mass/volume, mtDNA copy number, and mitochondrial respiration are markedly increased after the onset of myogenic differentiation. Besides, mitochondrial enzyme activity is also increased, suggesting that the metabolic shift from glycolysis to oxidative phosphorylation as the major energy source occurs during myogenic differentiation. Several lines of evidence suggest that impairment of mitochondrial function and activity blocks myogenic differentiation. However, yet little is known about the molecular mechanisms underlying the regulation of myogenesis by mitochondria. Understanding how mitochondria are involved in myogenesis will provide a valuable insight into the underlying mechanisms that regulate the maintenance of cellular homeostasis. Here, we will summarize the current knowledge regarding the role of mitochondria as a potential regulator of myogenesis.
Collapse
|
43
|
Carazo A, Levin J, Casas F, Seyer P, Grandemange S, Busson M, Pessemesse L, Wrutniak-Cabello C, Cabello G. Protein sequences involved in the mitochondrial import of the 3,5,3′-L-triiodothyronine receptor p43. J Cell Physiol 2012; 227:3768-77. [DOI: 10.1002/jcp.24085] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
44
|
Ge X, Vajjala A, McFarlane C, Wahli W, Sharma M, Kambadur R. Lack of Smad3 signaling leads to impaired skeletal muscle regeneration. Am J Physiol Endocrinol Metab 2012; 303:E90-102. [PMID: 22535746 DOI: 10.1152/ajpendo.00113.2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Smad3 is a key intracellular signaling mediator for both transforming growth factor-β and myostatin, two major regulators of skeletal muscle growth. Previous published work has revealed pronounced muscle atrophy together with impaired satellite cell functionality in Smad3-null muscles. In the present study, we have further validated a role for Smad3 signaling in skeletal muscle regeneration. Here, we show that Smad3-null mice had incomplete recovery of muscle weight and myofiber size after muscle injury. Histological/immunohistochemical analysis suggested impaired inflammatory response and reduced number of activated myoblasts during the early stages of muscle regeneration in the tibialis anterior muscle of Smad3-null mice. Nascent myofibers formed after muscle injury were also reduced in number. Moreover, Smad3-null regenerated muscle had decreased oxidative enzyme activity and impaired mitochondrial biogenesis, evident by the downregulation of the gene encoding mitochondrial transcription factor A, a master regulator of mitochondrial biogenesis. Consistent with known Smad3 function, reduced fibrotic tissue formation was also seen in regenerated Smad3-null muscle. In conclusion, Smad3 deficiency leads to impaired muscle regeneration, which underscores an essential role of Smad3 in postnatal myogenesis. Given the negative role of myostatin during muscle regeneration, the increased expression of myostatin observed in Smad3-null muscle may contribute to the regeneration defects.
Collapse
MESH Headings
- Animals
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Fibrosis
- Gene Expression Regulation
- Macrophages/immunology
- Male
- Mice
- Mice, Knockout
- Mitochondria, Muscle/metabolism
- Mitochondrial Proteins/genetics
- Mitochondrial Proteins/metabolism
- Muscle, Skeletal/immunology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Atrophy/immunology
- Muscular Atrophy/metabolism
- Muscular Atrophy/pathology
- Myoblasts, Skeletal/enzymology
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/pathology
- Myogenic Regulatory Factors/genetics
- Myogenic Regulatory Factors/metabolism
- Myostatin/genetics
- Myostatin/metabolism
- Necrosis
- Neutrophil Infiltration
- RNA, Messenger/metabolism
- Satellite Cells, Skeletal Muscle/enzymology
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/pathology
- Signal Transduction
- Smad3 Protein/genetics
- Smad3 Protein/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Xiaojia Ge
- School of Biological Sciences, Nanyang Technological University, Singapore
| | | | | | | | | | | |
Collapse
|
45
|
Cambier L, Rassam P, Chabi B, Mezghenna K, Gross R, Eveno E, Auffray C, Wrutniak-Cabello C, Lajoix AD, Pomiès P. M19 modulates skeletal muscle differentiation and insulin secretion in pancreatic β-cells through modulation of respiratory chain activity. PLoS One 2012; 7:e31815. [PMID: 22363741 PMCID: PMC3282743 DOI: 10.1371/journal.pone.0031815] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 01/13/2012] [Indexed: 11/18/2022] Open
Abstract
Mitochondrial dysfunction due to nuclear or mitochondrial DNA alterations contributes to multiple diseases such as metabolic myopathies, neurodegenerative disorders, diabetes and cancer. Nevertheless, to date, only half of the estimated 1,500 mitochondrial proteins has been identified, and the function of most of these proteins remains to be determined. Here, we characterize the function of M19, a novel mitochondrial nucleoid protein, in muscle and pancreatic β-cells. We have identified a 13-long amino acid sequence located at the N-terminus of M19 that targets the protein to mitochondria. Furthermore, using RNA interference and over-expression strategies, we demonstrate that M19 modulates mitochondrial oxygen consumption and ATP production, and could therefore regulate the respiratory chain activity. In an effort to determine whether M19 could play a role in the regulation of various cell activities, we show that this nucleoid protein, probably through its modulation of mitochondrial ATP production, acts on late muscle differentiation in myogenic C2C12 cells, and plays a permissive role on insulin secretion under basal glucose conditions in INS-1 pancreatic β-cells. Our results are therefore establishing a functional link between a mitochondrial nucleoid protein and the modulation of respiratory chain activities leading to the regulation of major cellular processes such as myogenesis and insulin secretion.
Collapse
Affiliation(s)
- Linda Cambier
- CNRS UMR5237, Centre de Recherche en Biochimie Macromoléculaire, Montpellier, France
- Université Montpellier 1, Montpellier, France
- Université Montpellier 2, Montpellier, France
| | - Patrice Rassam
- CNRS UMR5237, Centre de Recherche en Biochimie Macromoléculaire, Montpellier, France
- Université Montpellier 1, Montpellier, France
- Université Montpellier 2, Montpellier, France
| | - Béatrice Chabi
- INRA UMR866, Dynamique Musculaire et Métabolisme, Montpellier, France
- Université Montpellier 1, Montpellier, France
- Université Montpellier 2, Montpellier, France
| | - Karima Mezghenna
- CNRS UMR5232, Centre for Pharmacology and Innovation in Diabetes, Montpellier, France
- Université Montpellier 1, Montpellier, France
| | - René Gross
- CNRS UMR5232, Centre for Pharmacology and Innovation in Diabetes, Montpellier, France
- Université Montpellier 1, Montpellier, France
| | - Eric Eveno
- Genexpress, Functional Genomics and Systems Biology for Health, CNRS Institute of Biological Sciences, Villejuif, France
| | - Charles Auffray
- Genexpress, Functional Genomics and Systems Biology for Health, CNRS Institute of Biological Sciences, Villejuif, France
| | - Chantal Wrutniak-Cabello
- INRA UMR866, Dynamique Musculaire et Métabolisme, Montpellier, France
- Université Montpellier 1, Montpellier, France
- Université Montpellier 2, Montpellier, France
| | - Anne-Dominique Lajoix
- CNRS UMR5232, Centre for Pharmacology and Innovation in Diabetes, Montpellier, France
- Université Montpellier 1, Montpellier, France
| | - Pascal Pomiès
- CNRS UMR5237, Centre de Recherche en Biochimie Macromoléculaire, Montpellier, France
- Université Montpellier 1, Montpellier, France
- Université Montpellier 2, Montpellier, France
- INSERM U1046, Physiologie et Médecine Expérimentale du Coeur et des Muscles, Montpellier, France
- * E-mail:
| |
Collapse
|
46
|
Pessemesse L, Schlernitzauer A, Sar C, Levin J, Grandemange S, Seyer P, Favier FB, Kaminski S, Cabello G, Wrutniak-Cabello C, Casas F. Depletion of the p43 mitochondrial T3 receptor in mice affects skeletal muscle development and activity. FASEB J 2011; 26:748-56. [PMID: 22109994 DOI: 10.1096/fj.11-195933] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In vertebrates, skeletal muscle myofibers display different contractile and metabolic properties associated with different mitochondrial content and activity. We have previously identified a mitochondrial triiodothyronine receptor (p43) regulating mitochondrial transcription and mitochondrial biogenesis. When overexpressed in skeletal muscle, it increases mitochondrial DNA content, stimulates mitochondrial respiration, and induces a shift in the metabolic and contractile features of muscle fibers toward a slower and more oxidative phenotype. Here we show that a p43 depletion in mice decreases mitochondrial DNA replication and respiratory chain activity in skeletal muscle in association with the induction of a more glycolytic muscle phenotype and a decrease of capillary density. In addition, p43(-/-) mice displayed a significant increase in muscle mass relative to control animals and had an improved ability to use lipids. Our findings establish that the p43 mitochondrial receptor strongly affects muscle mass and the metabolic and contractile features of myofibers and provides evidence that this receptor mediates, in part, the influence of thyroid hormone in skeletal muscle.
Collapse
Affiliation(s)
- Laurence Pessemesse
- Institut National de Recherche Agronomique (INRA), Unité Mixte de Recherche (UMR)866 Dynamique Musculaire et Métabolisme, Montpellier, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Blanchet E, Bertrand C, Annicotte JS, Schlernitzauer A, Pessemesse L, Levin J, Fouret G, Feillet-Coudray C, Bonafos B, Fajas L, Cabello G, Wrutniak-Cabello C, Casas F. Mitochondrial T3 receptor p43 regulates insulin secretion and glucose homeostasis. FASEB J 2011; 26:40-50. [PMID: 21914860 DOI: 10.1096/fj.11-186841] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Thyroid hormone is a major determinant of energy expenditure and a key regulator of mitochondrial activity. We have previously identified a mitochondrial triiodothyronine receptor (p43) that acts as a mitochondrial transcription factor of the organelle genome, which leads, in vitro and in vivo, to a stimulation of mitochondrial biogenesis. Here we generated mice specifically lacking p43 to address its physiological influence. We found that p43 is required for normal glucose homeostasis. The p43(-/-) mice had a major defect in insulin secretion both in vivo and in isolated pancreatic islets and a loss of glucose-stimulated insulin secretion. Moreover, a high-fat/high-sucrose diet elicited more severe glucose intolerance than that recorded in normal animals. In addition, we observed in p43(-/-) mice both a decrease in pancreatic islet density and in the activity of complexes of the respiratory chain in isolated pancreatic islets. These dysfunctions were associated with a down-regulation of the expression of the glucose transporter Glut2 and of Kir6.2, a key component of the K(ATP) channel. Our findings establish that p43 is an important regulator of glucose homeostasis and pancreatic β-cell function and provide evidence for the first time of a physiological role for a mitochondrial endocrine receptor.
Collapse
Affiliation(s)
- Emilie Blanchet
- UMR866 Dynamique Musculaire et Métabolisme, Université Montpellier 1, Montpellier, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Seyer P, Grandemange S, Rochard P, Busson M, Pessemesse L, Casas F, Cabello G, Wrutniak-Cabello C. P43-dependent mitochondrial activity regulates myoblast differentiation and slow myosin isoform expression by control of Calcineurin expression. Exp Cell Res 2011; 317:2059-71. [PMID: 21664352 DOI: 10.1016/j.yexcr.2011.05.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 05/17/2011] [Accepted: 05/19/2011] [Indexed: 11/18/2022]
Abstract
We have previously shown that mitochondrial protein synthesis regulates myoblast differentiation, partly through the control of c-Myc expression, a cellular oncogene regulating myogenin expression and myoblast withdrawal from the cell cycle. In this study we provide evidence of the involvement of Calcineurin in this regulation. In C2C12 myoblasts, inhibition of mitochondrial protein synthesis by chloramphenicol decreases Calcineurin expression. Conversely, stimulation of this process by overexpressing the T3 mitochondrial receptor (p43) increases Calcineurin expression. Moreover, expression of a constitutively active Calcineurin (ΔCN) stimulates myoblast differentiation, whereas a Calcineurin antisense has the opposite effect. Lastly, ΔCN expression or stimulation of mitochondrial protein synthesis specifically increases slow myosin heavy chain expression. In conclusion, these data clearly suggest that, partly via Calcineurin expression, mitochondrial protein synthesis is involved in muscle development through the control of myoblast differentiation and probably the acquisition of the contractile and metabolic phenotype of muscle fibres.
Collapse
Affiliation(s)
- Pascal Seyer
- UMR 866 Différenciation Cellulaire et Croissance (INRA-UMI-UMII), Unité d'Endocrinologie Cellulaire, Institut National de la Recherche Agronomique (INRA), 2 Place Viala, 34060 Montpellier Cedex 1, France
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Barbieri E, Battistelli M, Casadei L, Vallorani L, Piccoli G, Guescini M, Gioacchini AM, Polidori E, Zeppa S, Ceccaroli P, Stocchi L, Stocchi V, Falcieri E. Morphofunctional and Biochemical Approaches for Studying Mitochondrial Changes during Myoblasts Differentiation. J Aging Res 2011; 2011:845379. [PMID: 21629710 PMCID: PMC3100678 DOI: 10.4061/2011/845379] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 02/15/2011] [Accepted: 03/04/2011] [Indexed: 12/16/2022] Open
Abstract
This study describes mitochondrial behaviour during the C2C12 myoblast differentiation program and proposes a proteomic approach to mitochondria integrated with classical morphofunctional and biochemical analyses. Mitochondrial ultrastructure variations were determined by transmission electron microscopy; mitochondrial mass and membrane potential were analysed by Mitotracker Green and JC-1 stains and by epifluorescence microscope. Expression of PGC1α, NRF1α, and Tfam genes controlling mitochondrial biogenesis was studied by real-time PCR. The mitochondrial functionality was tested by cytochrome c oxidase activity and COXII expression. Mitochondrial proteomic profile was also performed. These assays showed that mitochondrial biogenesis and activity significantly increase in differentiating myotubes. The proteomic profile identifies 32 differentially expressed proteins, mostly involved in oxidative metabolism, typical of myotubes formation. Other notable proteins, such as superoxide dismutase (MnSOD), a cell protection molecule, and voltage-dependent anion-selective channel protein (VDAC1) involved in the mitochondria-mediated apoptosis, were found to be regulated by the myogenic process. The integration of these approaches represents a helpful tool for studying mitochondrial dynamics, biogenesis, and functionality in comparative surveys on mitochondrial pathogenic or senescent satellite cells.
Collapse
Affiliation(s)
- Elena Barbieri
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via I Maggetti, 26, 61029 Urbino (PU), Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Muscle regeneration occurs to coincide with mitochondrial biogenesis. Mol Cell Biochem 2010; 349:139-47. [DOI: 10.1007/s11010-010-0668-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 11/15/2010] [Indexed: 01/04/2023]
|