1
|
Sun Z, Cheng X, Wang Z, Qiao C, Qian H, Yuan T, Lv Z, Sun W, Zhang H, Liu Y, Lu Z, Lin J, Lai C, Wang Y, Yang X, Wang X, Meng J, Bao N. Single-nucleus transcriptomics reveals subsets of degenerative myonuclei after rotator cuff tear-induced muscle atrophy. Cell Prolif 2025; 58:e13763. [PMID: 39435630 PMCID: PMC11882757 DOI: 10.1111/cpr.13763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Rotator cuff tear (RCT) is the primary cause of shoulder pain and disability and frequently trigger muscle degeneration characterised by muscle atrophy, fatty infiltration and fibrosis. Single-nucleus RNA sequencing (snRNA-seq) was used to reveal the transcriptional changes in the supraspinatus muscle after RCT. Supraspinatus muscles were obtained from patients with habitual shoulder dislocation (n = 3) and RCT (n = 3). In response to the RCT, trajectory analysis showed progression from normal myonuclei to ANKRD1+ myonuclei, which captured atrophy-and fatty infiltration-related regulons (KLF5, KLF10, FOSL1 and BHLHE40). Transcriptomic alterations in fibro/adipogenic progenitors (FAPs) and muscle satellite cells (MuSCs) have also been studied. By predicting cell-cell interactions, we observed communication alterations between myofibers and muscle-resident cells following RCT. Our findings reveal the plasticity of muscle cells in response to RCT and offer valuable insights into the molecular mechanisms and potential therapeutic targets of RCT.
Collapse
Affiliation(s)
- Ziying Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xi Cheng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zheng Wang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Chenfeng Qiao
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| | - Hong Qian
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Tao Yuan
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zhongyang Lv
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Wenshuang Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Hanwen Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Yuan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zhihao Lu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Jintao Lin
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Chengteng Lai
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Yang Wang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xiaojiang Yang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xingyun Wang
- Hongqiao International Institute of Medicine, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Jia Meng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| | - Nirong Bao
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| |
Collapse
|
2
|
Jia H, Kaster N, Khan R, Ayari-Akkari A. The Roles of myomiRs in the Pathogenesis of Sarcopenia: From Literature to In Silico Analysis. Mol Biotechnol 2025:10.1007/s12033-025-01373-0. [PMID: 40025274 DOI: 10.1007/s12033-025-01373-0] [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: 10/22/2024] [Accepted: 12/30/2024] [Indexed: 03/04/2025]
Abstract
Senile sarcopenia is a condition of age-associated muscular disorder and is a significant health issue around the world. In the current review, we curated the information from the NCBI, PubMed, and Google Scholar literature and explored the non-genetic and genetic causes of senile sarcopenia. Interestingly, the myomiRs such as miR-1, miR-206, miR-133a, miR-133b, miR-208b, and miR-499 are skeletal muscle's critical structural and functional regulators. However, very scattered information is available regarding the roles of myomiRs in different skeletal muscle phenotypes through a diverse list of known target genes. Therefore, these pieces of information must be organized to focus on the conserved target genes and comparable effects of the myomiRs in regulating senile sarcopenia. Hence, in the present review, the roles of pathogenetic factors in regulating senile sarcopenia were highlighted. The literature was further curated for the roles of myomiRs such as hsa-miR-1-3p/206, hsa-miR-27-3p, hsa-miR-146-5p, and hsa-miR-499-5p and their target genes. Additionally, we used different bioinformatics tools and predicted target genes of the myomiRs and found the most critical target genes, shared pathways, and their standard functions in regulating muscle structure and functions. The information gathered in the current review will help the researchers to explore their possible therapeutic potential, especially the use of the myomiRs for the treatment of senile sarcopenia.
Collapse
Affiliation(s)
- Huanxia Jia
- Medical College of Xuchang University, No.1389, Xufan Road, Xuchang, 461000, Henan, People's Republic of China
| | - Nurgulsim Kaster
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, People's Republic of China.
- Faculty of Veterinary and Livestock Technology, S. Seifullin Kazakh Agro Technical University, Astana, Kazakhstan.
| | - Rajwali Khan
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, People's Republic of China.
- Department of Livestock Management, Breeding and Genetics, The University of Agriculture, Peshawar, Pakistan.
| | - Amel Ayari-Akkari
- Biology Department, College of Science, King Khalid University, P.O. Box 960, Abha, Saudi Arabia
| |
Collapse
|
3
|
Fessard A, Zavoriti A, Boyer N, Guillemaud J, Rahmati M, Del Carmine P, Gobet C, Chazaud B, Gondin J. Neuromuscular electrical stimulation training induces myonuclear accretion and hypertrophy in mice without overt signs of muscle damage and regeneration. Skelet Muscle 2025; 15:3. [PMID: 39910613 PMCID: PMC11796018 DOI: 10.1186/s13395-024-00372-0] [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: 12/19/2023] [Accepted: 12/23/2024] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Skeletal muscle is a plastic tissue that adapts to increased mechanical loading/contractile activity through fusion of muscle stem cells (MuSCs) with myofibers, a physiological process referred to as myonuclear accretion. However, it is still unclear whether myonuclear accretion is driven by increased mechanical loading per se, or occurs, at least in part, in response to muscle injury/regeneration. Here, we developed a non-damaging protocol to evaluate contractile activity-induced myonuclear accretion/hypertrophy in physiological conditions. METHODS Contractile activity was generated by applying repeated electrical stimuli over the mouse plantar flexor muscles. This method is commonly referred to as NeuroMuscular Electrical Simulation (NMES) in Human. Each NMES training session consisted of 80 isometric contractions delivered at ∼15% of maximal tetanic force to avoid muscle damage. C57BL/6J male mice were submitted to either a short (i.e., 6 sessions) or long (i.e., 12 sessions) individualized NMES training program while unstimulated mice were used as controls. Histological investigations were performed to assess the impact of NMES on MuSC number and status, myonuclei content and muscle tissue integrity, typology and size. RESULTS NMES led to a robust proliferation of MuSCs and myonuclear accretion in the absence of overt signs of muscle damage/regeneration. NMES-induced myonuclear accretion was specific to type IIB myofibers and was an early event preceding muscle hypertrophy inasmuch as a mild increase in myofiber cross-sectional area was only observed in response to the long-term NMES training protocol. CONCLUSION We conclude that NMES-induced myonuclear accretion and muscle hypertrophy are driven by a mild increase in mechanical loading in the absence of overt signs of muscle injury.
Collapse
Affiliation(s)
- Aurélie Fessard
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Aliki Zavoriti
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Natacha Boyer
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Jules Guillemaud
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Masoud Rahmati
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
- Department of Exercise Physiology, Faculty of Literature and Human Sciences, Lorestan University, Khoramabad, Iran
| | - Peggy Del Carmine
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Christelle Gobet
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France.
| |
Collapse
|
4
|
Boykin JR, Steiner JL, Laskin GR, Roberts MD, Vied C, Willis CRG, Etheridge T, Gordon BS. Comparative analysis of acute eccentric contraction-induced changes to the skeletal muscle transcriptome in young and aged mice and humans. Am J Physiol Regul Integr Comp Physiol 2025; 328:R45-R58. [PMID: 39495237 DOI: 10.1152/ajpregu.00224.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Adaptations to skeletal muscle following resistance exercise are due in part to changes to the skeletal muscle transcriptome. Although transcriptional changes in response to resistance exercise occur in young and aged muscles, aging alters this response. Rodent models have served great utility in defining regulatory factors that underscore the influence of mechanical load and aging on changes to skeletal muscle phenotype. Unilateral eccentric contractions in young and aged rodents are widely used to model resistance exercises in humans. However, the extent to which unilateral eccentric contractions in young and aged rodents mimic the transcriptional response in humans remains unknown. We reanalyzed two publicly available RNA sequencing datasets from young and aged mice and humans that were subjected to acute eccentric contractions to define key similarities and differences in the muscle transcriptional response following this exercise modality. The effect of aging on the number of contraction-sensitive genes, the distribution patterns of those genes into unique/common categories, and the cellular pathways associated with the differentially expressed genes (DEGs) were similar in mice and humans. However, there was little overlap between species when comparing specific contraction-sensitive DEGs within the same age group. There were strong intraspecies relationships for the common transcription factors predicted to influence the contraction-sensitive gene sets, whereas interspecies relationships were weak. Overall, these data demonstrate key similarities between mice and humans for the contraction-induced changes to the muscle transcriptome, but we posit species-specific responses exist and should be taken into consideration when attempting to translate rodent eccentric exercise models.NEW & NOTEWORTHY Acute eccentric muscle contractions in rodents are used to model resistance exercise in young and aged humans, including changes to the muscle transcriptome. This work defines the utility of the rodent model at mimicking the transcriptional features observed in young and aged humans.
Collapse
Affiliation(s)
- Jake R Boykin
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, Florida, United States
| | - Jennifer L Steiner
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, Florida, United States
- Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, Florida, United States
| | - Grant R Laskin
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, Florida, United States
| | - Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Cynthia Vied
- Translational Science Laboratory, Florida State University College of Medicine, Tallahassee, Florida, United States
| | - Craig R G Willis
- School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
| | - Timothy Etheridge
- Department of Sport and Health Sciences, University of Exeter, Exeter, United Kingdom
| | - Bradley S Gordon
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, Florida, United States
- Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
5
|
Sun C, Swoboda CO, Morales FM, Calvo C, Petrany MJ, Parameswaran S, VonHandorf A, Weirauch MT, Lepper C, Millay DP. Lineage tracing of nuclei in skeletal myofibers uncovers distinct transcripts and interplay between myonuclear populations. Nat Commun 2024; 15:9372. [PMID: 39477931 PMCID: PMC11526147 DOI: 10.1038/s41467-024-53510-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Multinucleated skeletal muscle cells need to acquire additional nuclei through fusion with activated skeletal muscle stem cells when responding to both developmental and adaptive growth stimuli. A fundamental question in skeletal muscle biology has been the reason underlying this need for new nuclei in cells that already harbor hundreds of nuclei. Here we utilize nuclear RNA-sequencing approaches and develop a lineage tracing strategy capable of defining the transcriptional state of recently fused nuclei and distinguishing this state from that of pre-existing nuclei. Our findings reveal the presence of conserved markers of newly fused nuclei both during development and after a hypertrophic stimulus in the adult. However, newly fused nuclei also exhibit divergent gene expression that is determined by the myogenic environment to which they fuse. Moreover, accrual of new nuclei through fusion is required for nuclei already resident in adult myofibers to mount a normal transcriptional response to a load-inducing stimulus. We propose a model of mutual regulation in the control of skeletal muscle development and adaptations, where newly fused and pre-existing myonuclear populations influence each other to maintain optimal functional growth.
Collapse
Affiliation(s)
- Chengyi Sun
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Casey O Swoboda
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Fabian Montecino Morales
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Cristofer Calvo
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael J Petrany
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sreeja Parameswaran
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew VonHandorf
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Christoph Lepper
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
6
|
Iwamori K, Kubota M, Zhang L, Kodama K, Kubo A, Kokubo H, Akimoto T, Fukada SI. Decreased number of satellite cells-derived myonuclei in both fast- and slow-twitch muscles in HeyL-KO mice during voluntary running exercise. Skelet Muscle 2024; 14:25. [PMID: 39449015 PMCID: PMC11515490 DOI: 10.1186/s13395-024-00357-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Skeletal muscles possess unique abilities known as adaptation or plasticity. When exposed to external stimuli, such as mechanical loading, both myofiber size and myonuclear number increase. Muscle stem cells, also known as muscle satellite cells (MuSCs), play vital roles in these changes. HeyL, a direct target of Notch signaling, is crucial for efficient muscle hypertrophy because it ensures MuSC proliferation in surgically overloaded muscles by inhibiting the premature differentiation. However, it remains unclear whether HeyL is essential for MuSC expansion in physiologically exercised muscles. Additionally, the influence of myofiber type on the requirement for HeyL in MuSCs within exercised muscles remains unclear. METHODS We used a voluntary wheel running model and HeyL-knockout mice to investigate the impact of HeyL deficiency on MuSC-derived myonuclei, MuSC behavior, muscle weight, myofiber size, and myofiber type in the running mice. RESULTS The number of new MuSC-derived myonuclei was significantly lower in both slow-twitch soleus and fast-twitch plantaris muscles from exercised HeyL-knockout mice than in control mice. However, expect for the frequency of Type IIb myofiber in plantaris muscle, exercised HeyL-knockout mice exhibited similar responses to control mice regarding myofiber size and type. CONCLUSIONS HeyL expression is crucial for MuSC expansion during physiological exercise in both slow and fast muscles. The frequency of Type IIb myofiber in plantaris muscle of HeyL-knockout mice was not significantly reduced compared to that of control mice. However, the absence of HeyL did not affect the increased size and frequency of Type IIa myofiber in plantaris muscles. In this model, no detectable changes in myofiber size or type were observed in the soleus muscles of either control or HeyL-knockout mice. These findings imply that the requirement for MuSCs in the wheel-running model is difficult to observe due to the relatively low degree of hypertrophy compared to surgically overloaded models.
Collapse
Affiliation(s)
- Kanako Iwamori
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Manami Kubota
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Lidan Zhang
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
- Center for Medical Epigenetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 40016, China
| | - Kazuki Kodama
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Atsushi Kubo
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | - Hiroki Kokubo
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minamiku, Hiroshima, 734-8551, Japan
| | - Takayuki Akimoto
- Faculty of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, 359-1192, Saitama, Japan
| | - So-Ichiro Fukada
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Osaka, Japan.
| |
Collapse
|
7
|
Sun L, Luan J, Wang J, Li X, Zhang W, Ji X, Liu L, Wang R, Xu B. GEPREP: A comprehensive data atlas of RNA-seq-based gene expression profiles of exercise responses. JOURNAL OF SPORT AND HEALTH SCIENCE 2024; 14:100992. [PMID: 39341494 PMCID: PMC11863345 DOI: 10.1016/j.jshs.2024.100992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 06/11/2024] [Accepted: 07/31/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Physical activity can regulate and affect gene expression in multiple tissues and cells. Recently, with the development of next-generation sequencing, a large number of RNA-sequencing (RNA-seq)-based gene expression profiles about physical activity have been shared in public resources; however, they are poorly curated and underutilized. To tackle this problem, we developed a data atlas of such data through comprehensive data collection, curation, and organization. METHODS The data atlas, termed gene expression profiles of RNA-seq-based exercise responses (GEPREP), was built on a comprehensive collection of high-quality RNA-seq data on exercise responses. The metadata of each sample were manually curated. Data were uniformly processed and batch effects corrected. All the information was well organized in an easy-to-use website for free search, visualization, and download. RESULTS GEPREP now includes 69 RNA-seq datasets of pre- and post-exercise, comprising 26 human datasets (1120 samples) and 43 mouse datasets (1006 samples). Specifically, there were 977 (87.2 %) human samples of skeletal muscle and 143 (12.8 %) human samples of blood. There were also samples across 9 mice tissues with skeletal muscle (359, 35.7 %) and brain (280, 27.8 %) accounting for the main fractions. Metadata-including subject, exercise interventions, sampling sites, and post-processing methods-are also included. The metadata and gene expression profiles are freely accessible at http://www.geprep.org.cn/. CONCLUSION GEPREP is a comprehensive data atlas of RNA-seq-based gene expression profiles responding to exercise. With its reliable annotations and user-friendly interfaces, it has the potential to deepen our understanding of exercise physiology.
Collapse
Affiliation(s)
- Lei Sun
- School of Information Engineering, Yangzhou University, Yangzhou 225127, China; CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing 100101, China
| | - Jinwen Luan
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Jinbiao Wang
- School of Information Engineering, Yangzhou University, Yangzhou 225127, China
| | - Xiaoli Li
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Wenqian Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaohui Ji
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| | - Bingxiang Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
8
|
Saliu TP, Goh J, Kang G, Burke BI, Ismaeel A, McCarthy JJ. Satellite cell dynamics during skeletal muscle hypertrophy. Biochem Soc Trans 2024; 52:1921-1926. [PMID: 39136196 PMCID: PMC11660404 DOI: 10.1042/bst20240201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/26/2024]
Abstract
Skeletal muscle stem cells (MuSCs) display distinct behavior crucial for tissue maintenance and repair. Upon activation, MuSCs exhibit distinct modes of division: symmetric division, facilitating either self-renewal or differentiation, and asymmetric division, which dictates divergent cellular fates. This review explores the nuanced dynamics of MuSC division and the molecular mechanisms governing this behavior. Furthermore, it introduces a novel phenomenon observed in a subset of MuSCs under hypertrophic stimuli termed division-independent differentiation. Insights into the underlying mechanisms driving this process are discussed, alongside its broader implications for muscle physiology.
Collapse
Affiliation(s)
- Tolulope P. Saliu
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
| | - Jensen Goh
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
| | - Gyumin Kang
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
- Division of Biomedical Informatics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
| | - Benjamin I. Burke
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
| | - Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
| | - John J. McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
| |
Collapse
|
9
|
Hosoyama T, Kawai‐Takaishi M, Iida H, Yamamoto Y, Nakamichi Y, Watanabe T, Takemura M, Kato S, Uezumi A, Matsui Y. Lack of vitamin D signalling in mesenchymal progenitors causes fatty infiltration in muscle. J Cachexia Sarcopenia Muscle 2024; 15:907-918. [PMID: 38533539 PMCID: PMC11154772 DOI: 10.1002/jcsm.13448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/22/2024] [Accepted: 01/31/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Recent studies have indicated the importance of muscle quality in addition to muscle quantity in sarcopenia pathophysiology. Intramuscular adipose tissue (IMAT), which originates from mesenchymal progenitors (MPs) in adult skeletal muscle, is a key factor affecting muscle quality in older adults, suggesting that controlling IMAT formation is a promising therapeutic strategy for sarcopenia. However, the molecular mechanism underlying IMAT formation in older adults has not been clarified. We recently found that the vitamin D receptor (VDR) is highly expressed in MPs in comparison to myotubes (P = 0.028, N = 3), indicating a potential role of vitamin D signalling in MPs. In this study, we aimed to clarify the role of vitamin D signalling in MP kinetics, with a focus on adipogenesis. METHODS MPs isolated from mouse skeletal muscles were subjected to adipogenic differentiation conditions with or without vitamin D (1α,25(OH)2D3, 100 nM) for 7 days, and adipogenicity was evaluated based on adipogenic marker expression. For in vivo analysis, tamoxifen-inducible MP-specific VDR-deficient (VdrMPcKO) mice were newly developed to investigate whether lack of vitamin D signalling in MPs is involved in IMAT formation. To induce muscle atrophy, VdrMPcKO male mice were subjected to tenotomy of the gastrocnemius muscle, and then muscle weight, myofibre cross-sectional area, adipogenic marker expression, and fatty infiltration into the muscle were evaluated at 3 weeks after operation (N = 3-4). In addition, a vitamin D-deficient diet was provided to wild-type male mice (3 and 20 months of age, N = 5) for 3 months to investigate whether vitamin D deficiency causes IMAT formation. RESULTS Vitamin D treatment nearly completely inhibited adipogenesis of MPs through Runx1-mediated transcriptional modifications of early adipogenic factors such as PPARγ (P = 0.0031) and C/EBPα (P = 0.0027), whereas VDR-deficient MPs derived from VdrMPcKO mice differentiated into adipocytes even in the presence of vitamin D (P = 0.0044, Oil-Red O+ area). In consistency with in-vitro findings, VdrMPcKO mice and mice fed a vitamin D-deficient diet exhibited fat deposition in atrophied (P = 0.0311) and aged (P = 0.0216) skeletal muscle, respectively. CONCLUSIONS Vitamin D signalling is important to prevent fate decision of MPs towards the adipogenic lineage. As vitamin D levels decline with age, our data indicate that decreased vitamin D levels may be one of the causes of IMAT formation in older adults, and vitamin D signalling may be a novel therapeutic target for sarcopenia.
Collapse
Affiliation(s)
- Tohru Hosoyama
- Department of Musculoskeletal Disease, Research InstituteNational Center for Geriatrics and GerontologyObuJapan
| | - Minako Kawai‐Takaishi
- Department of Musculoskeletal Disease, Research InstituteNational Center for Geriatrics and GerontologyObuJapan
| | - Hiroki Iida
- Department of Musculoskeletal Disease, Research InstituteNational Center for Geriatrics and GerontologyObuJapan
- Department of Orthopaedic SurgeryNagoya University Graduate School of MedicineNagoyaJapan
| | - Yoko Yamamoto
- Department of Surgical OncologyThe University of TokyoTokyoJapan
| | - Yuko Nakamichi
- Institute for Oral ScienceMatsumoto Dental UniversityNaganoJapan
| | - Tsuyoshi Watanabe
- Center for Frailty and Locomotive SyndromeNational Center for Geriatrics and GerontologyObuJapan
- Department of Orthopaedic SurgeryNational Center for Geriatrics and GerontologyObuJapan
| | - Marie Takemura
- Center for Frailty and Locomotive SyndromeNational Center for Geriatrics and GerontologyObuJapan
| | - Shigeaki Kato
- Graduate School of Life Science and EngineeringIryo Sosei UniversityFukushimaJapan
- Research Institute of Innovative MedicineTokiwa FoundationFukushimaJapan
| | - Akiyoshi Uezumi
- Medical Institute of BioregulationKyushu UniversityFukuokaJapan
| | - Yasumoto Matsui
- Center for Frailty and Locomotive SyndromeNational Center for Geriatrics and GerontologyObuJapan
| |
Collapse
|
10
|
Lessard L, Saugues A, Gondin J, Mounier R, Kneppers A. Measurement of Myonuclear Accretion In Vitro and In Vivo. Methods Mol Biol 2024. [PMID: 38647863 DOI: 10.1007/7651_2024_540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Adult skeletal muscle stem cells (MuSC) are the regenerative precursors of myofibers and also have an important role in myofiber growth, adaptation, and maintenance by fusing to the myofibers-a process referred to as "myonuclear accretion." Due to a focus on MuSC function during regeneration, myofibers remain a largely overlooked component of the MuSC niche influencing MuSC fate. Here, we describe a method to directly measure the rate of myonuclear accretion in vitro and in vivo using ethynyl-2'-deoxyuridine (EdU)-based tracing of MuSC progeny. This method supports the dissection of MuSC intrinsic and myofiber-derived factors influencing myonuclear accretion as an alternative fate of MuSCs supporting myofiber homeostasis and plasticity.
Collapse
Affiliation(s)
- Lola Lessard
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Audrey Saugues
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Anita Kneppers
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France.
| |
Collapse
|
11
|
Zhang L, Saito H, Higashimoto T, Kaji T, Nakamura A, Iwamori K, Nagano R, Motooka D, Okuzaki D, Uezumi A, Seno S, Fukada SI. Regulation of muscle hypertrophy through granulin: Relayed communication among mesenchymal progenitors, macrophages, and satellite cells. Cell Rep 2024; 43:114052. [PMID: 38573860 DOI: 10.1016/j.celrep.2024.114052] [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: 06/07/2023] [Revised: 02/14/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024] Open
Abstract
Skeletal muscles exert remarkable regenerative or adaptive capacities in response to injuries or mechanical loads. However, the cellular networks underlying muscle adaptation are poorly understood compared to those underlying muscle regeneration. We employed single-cell RNA sequencing to investigate the gene expression patterns and cellular networks activated in overloaded muscles and compared these results with those observed in regenerating muscles. The cellular composition of the 4-day overloaded muscle, when macrophage infiltration peaked, closely resembled that of the 10-day regenerating muscle. In addition to the mesenchymal progenitor-muscle satellite cell (MuSC) axis, interactome analyses or targeted depletion experiments revealed communications between mesenchymal progenitors-macrophages and macrophages-MuSCs. Furthermore, granulin, a macrophage-derived factor, inhibited MuSC differentiation, and Granulin-knockout mice exhibited blunted muscle hypertrophy due to the premature differentiation of overloaded MuSCs. These findings reveal the critical role of granulin through the relayed communications of mesenchymal progenitors, macrophages, and MuSCs in facilitating efficient muscle hypertrophy.
Collapse
Affiliation(s)
- Lidan Zhang
- Center for Medical Epigenetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 40016, China; Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hayato Saito
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Tatsuyoshi Higashimoto
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takayuki Kaji
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Ayasa Nakamura
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Kanako Iwamori
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Ryoko Nagano
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan; Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi, Fukuoka 812-8582, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Akiyoshi Uezumi
- Division of Cell Heterogeneity, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, 3-1-1, Maidashi, Higashi, Fukuoka 812-8582, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - So-Ichiro Fukada
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
12
|
Espino-Gonzalez E, Tickle PG, Altara R, Gallagher H, Cheng CW, Engman V, Wood N, Justo da Silva GJ, Scalabrin M, Yu X, Zhong Z, Colman MA, Yuldasheva NY, Booz GW, Adams V, Pereira MG, Cataliotti A, Roberts LD, Egginton S, Bowen TS. Caloric Restriction Rejuvenates Skeletal Muscle Growth in Heart Failure With Preserved Ejection Fraction. JACC Basic Transl Sci 2024; 9:223-240. [PMID: 38510717 PMCID: PMC10950401 DOI: 10.1016/j.jacbts.2023.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 03/22/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major clinical problem, with limited treatments. HFpEF is characterized by a distinct, but poorly understood, skeletal muscle pathology, which could offer an alternative therapeutic target. In a rat model, we identified impaired myonuclear accretion as a mechanism for low myofiber growth in HFpEF following resistance exercise. Acute caloric restriction rescued skeletal muscle pathology in HFpEF, whereas cardiac therapies had no effect. Mechanisms regulating myonuclear accretion were dysregulated in patients with HFpEF. Overall, these findings may have widespread implications in HFpEF, indicating combined dietary with exercise interventions as a beneficial approach to overcome skeletal muscle pathology.
Collapse
Affiliation(s)
- Ever Espino-Gonzalez
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Peter G. Tickle
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Raffaele Altara
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Harrison Gallagher
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Chew W. Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine, University of Leeds, Leeds, United Kingdom
| | - Viktor Engman
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Nathanael Wood
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | | | - Mattia Scalabrin
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Xinyue Yu
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Ziyi Zhong
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Michael A. Colman
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Nadira Y. Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine, University of Leeds, Leeds, United Kingdom
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Volker Adams
- Heart Center Dresden, TU-Dresden, Dresden, Germany
| | - Marcelo G. Pereira
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Alessandro Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Lee D. Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine, University of Leeds, Leeds, United Kingdom
| | - Stuart Egginton
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - T. Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
13
|
Motohashi N, Minegishi K, Aoki Y. Inherited myogenic abilities in muscle precursor cells defined by the mitochondrial complex I-encoding protein. Cell Death Dis 2023; 14:689. [PMID: 37857600 PMCID: PMC10587152 DOI: 10.1038/s41419-023-06192-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/18/2023] [Accepted: 09/28/2023] [Indexed: 10/21/2023]
Abstract
Skeletal muscle comprises different muscle fibers, including slow- and fast-type muscles, and satellite cells (SCs), which exist in individual muscle fibers and possess different myogenic properties. Previously, we reported that myoblasts (MBs) from slow-type enriched soleus (SOL) had a high potential to self-renew compared with cells derived from fast-type enriched tibialis anterior (TA). However, whether the functionality of myogenic cells in adult muscles is attributed to the muscle fiber in which they reside and whether the characteristics of myogenic cells derived from slow- and fast-type fibers can be distinguished at the genetic level remain unknown. Global gene expression analysis revealed that the myogenic potential of MBs was independent of the muscle fiber type they reside in but dependent on the region of muscles they are derived from. Thus, in this study, proteomic analysis was conducted to clarify the molecular differences between MBs derived from TA and SOL. NADH dehydrogenase (ubiquinone) iron-sulfur protein 8 (Ndufs8), a subunit of NADH dehydrogenase in mitochondrial complex I, significantly increased in SOL-derived MBs compared with that in TA-derived cells. Moreover, the expression level of Ndufs8 in MBs significantly decreased with age. Gain- and loss-of-function experiments revealed that Ndufs8 expression in MBs promoted differentiation, self-renewal, and apoptosis resistance. In particular, Ndufs8 suppression in MBs increased p53 acetylation, followed by a decline in NAD/NADH ratio. Nicotinamide mononucleotide treatment, which restores the intracellular NAD+ level, could decrease p53 acetylation and increase myogenic cell self-renewal ability in vivo. These results suggested that the functional differences in MBs derived from SOL and TA governed by the mitochondrial complex I-encoding gene reflect the magnitude of the decline in SC number observed with aging, indicating that the replenishment of NAD+ is a possible approach for improving impaired cellular functions caused by aging or diseases.
Collapse
Affiliation(s)
- Norio Motohashi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan.
| | - Katsura Minegishi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan.
| |
Collapse
|
14
|
Roberts MD, McCarthy JJ, Hornberger TA, Phillips SM, Mackey AL, Nader GA, Boppart MD, Kavazis AN, Reidy PT, Ogasawara R, Libardi CA, Ugrinowitsch C, Booth FW, Esser KA. Mechanisms of mechanical overload-induced skeletal muscle hypertrophy: current understanding and future directions. Physiol Rev 2023; 103:2679-2757. [PMID: 37382939 PMCID: PMC10625844 DOI: 10.1152/physrev.00039.2022] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Mechanisms underlying mechanical overload-induced skeletal muscle hypertrophy have been extensively researched since the landmark report by Morpurgo (1897) of "work-induced hypertrophy" in dogs that were treadmill trained. Much of the preclinical rodent and human resistance training research to date supports that involved mechanisms include enhanced mammalian/mechanistic target of rapamycin complex 1 (mTORC1) signaling, an expansion in translational capacity through ribosome biogenesis, increased satellite cell abundance and myonuclear accretion, and postexercise elevations in muscle protein synthesis rates. However, several lines of past and emerging evidence suggest that additional mechanisms that feed into or are independent of these processes are also involved. This review first provides a historical account of how mechanistic research into skeletal muscle hypertrophy has progressed. A comprehensive list of mechanisms associated with skeletal muscle hypertrophy is then outlined, and areas of disagreement involving these mechanisms are presented. Finally, future research directions involving many of the discussed mechanisms are proposed.
Collapse
Affiliation(s)
- Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gustavo A Nader
- Department of Kinesiology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States
| | - Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Andreas N Kavazis
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Paul T Reidy
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Riki Ogasawara
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Cleiton A Libardi
- MUSCULAB-Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| | - Carlos Ugrinowitsch
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Karyn A Esser
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
15
|
Sun C, Swoboda CO, Petrany MJ, Parameswaran S, VonHandorf A, Weirauch MT, Lepper C, Millay DP. Lineage tracing of newly accrued nuclei in skeletal myofibers uncovers distinct transcripts and interplay between nuclear populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554609. [PMID: 37662191 PMCID: PMC10473681 DOI: 10.1101/2023.08.24.554609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Multinucleated skeletal muscle cells have an obligatory need to acquire additional nuclei through fusion with activated skeletal muscle stem cells when responding to both developmental and adaptive growth stimuli. A fundamental question in skeletal muscle biology has been the reason underlying this need for new nuclei in syncytial cells that already harbor hundreds of nuclei. To begin to answer this long-standing question, we utilized nuclear RNA-sequencing approaches and developed a lineage tracing strategy capable of defining the transcriptional state of recently fused nuclei and distinguishing this state from that of pre-existing nuclei. Our findings reveal the presence of conserved markers of newly fused nuclei both during development and after a hypertrophic stimulus in the adult. However, newly fused nuclei also exhibit divergent gene expression that is determined by the myogenic environment to which they fuse. Moreover, accrual of new nuclei through fusion is required for nuclei already resident in adult myofibers to mount a normal transcriptional response to a load-inducing stimulus. We propose a model of mutual regulation in the control of skeletal muscle development and adaptations, where newly fused and pre-existing myonuclear populations influence each other to maintain optimal functional growth.
Collapse
Affiliation(s)
- Chengyi Sun
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Casey O. Swoboda
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Michael J. Petrany
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Sreeja Parameswaran
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew VonHandorf
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T. Weirauch
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Christoph Lepper
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Douglas P. Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
16
|
Yu Z, Xu X, Ai N, Wang K, Zhang P, Li X, LiuFu S, Liu X, Jiang J, Gu J, Gao N, Ma H. Integrated analysis of circRNA, lncRNA, miRNA and mRNA to reveal the ceRNA regulatory network of postnatal skeletal muscle development in Ningxiang pig. Front Cell Dev Biol 2023; 11:1185823. [PMID: 37465009 PMCID: PMC10350537 DOI: 10.3389/fcell.2023.1185823] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/22/2023] [Indexed: 07/20/2023] Open
Abstract
Introduction: The development of skeletal muscle is regulated by regulatory factors of genes and non-coding RNAs (ncRNAs). Methods: The objective of this study was to understand the transformation of muscle fiber type in the longissimus dorsi muscle of male Ningxiang pigs at four different growth stages (30, 90, 150, and 210 days after birth, n = 3) by histological analysis and whole transcriptome sequencing. Additionally, the study investigated the expression patterns of various RNAs involved in muscle fiber transformation and constructed a regulatory network for competing endogenous RNA (ceRNA) that includes circular RNA (circRNA)/long non-coding RNA (lncRNA)-microRNA (miRNA)-messenger RNA (mRNA). Results: Histomorphology analysis showed that the diameter of muscle fiber reached its maximum at 150 days after birth. The slow muscle fiber transformation showed a pattern of initial decrease followed by an increase. 29,963 circRNAs, 2,683 lncRNAs, 986 miRNAs and 22,411 mRNAs with expression level ≥0 were identified by whole transcriptome sequencing. Furthermore, 642 differentially expressed circRNAs (DEc), 505 differentially expressed lncRNAs (DEl), 316 differentially expressed miRNAs (DEmi) and 6,090 differentially expressed mRNAs (DEm) were identified by differential expression analysis. Functions of differentially expressed mRNA were identified by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). GO enrichment analysis indicates that 40 known genes and 6 new genes are associated with skeletal muscle development. Additionally, KEGG analysis shows that these genes regulate skeletal muscle development via MAPK, FoxO, Hedgehog, PI3K-Akt, Notch, VEGF and other signaling pathways. Through protein-protein interaction (PPI) and transcription factor prediction (TFP), the action mode of skeletal muscle-related genes was explored. PPI analysis showed that there were stable interactions among 19 proteins, meanwhile, TFP analysis predicted 22 transcription factors such as HMG20B, MYF6, MYOD1 and MYOG, and 12 of the 19 interacting proteins were transcription factors. The regulatory network of ceRNA related to skeletal muscle development was constructed based on the correlation of various RNA expression levels and the targeted binding characteristics with miRNA. The regulatory network included 31 DEms, 59 miRNAs, 667 circRNAs and 224 lncRNAs. conclusion: Overall, the study revealed the role of ceRNA regulatory network in the transformation of skeletal muscle fiber types in Ningxiang pigs, which contributes to the understanding of ceRNA regulatory network in Ningxiang pigs during the skeletal muscle development period.
Collapse
Affiliation(s)
- Zonggang Yu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xueli Xu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Nini Ai
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Kaiming Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Peiwen Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xintong Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Sui LiuFu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xiaolin Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Jun Jiang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Jingjing Gu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Ning Gao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Haiming Ma
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
17
|
Yang BA, Larouche JA, Sabin KM, Fraczek PM, Parker SCJ, Aguilar CA. Three-dimensional chromatin re-organization during muscle stem cell aging. Aging Cell 2023; 22:e13789. [PMID: 36727578 PMCID: PMC10086523 DOI: 10.1111/acel.13789] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/29/2022] [Accepted: 01/11/2023] [Indexed: 02/03/2023] Open
Abstract
Age-related skeletal muscle atrophy or sarcopenia is a significant societal problem that is becoming amplified as the world's population continues to increase. The regeneration of damaged skeletal muscle is mediated by muscle stem cells, but in old age muscle stem cells become functionally attenuated. The molecular mechanisms that govern muscle stem cell aging encompass changes across multiple regulatory layers and are integrated by the three-dimensional organization of the genome. To quantitatively understand how hierarchical chromatin architecture changes during muscle stem cell aging, we generated 3D chromatin conformation maps (Hi-C) and integrated these datasets with multi-omic (chromatin accessibility and transcriptome) profiles from bulk populations and single cells. We observed that muscle stem cells display static behavior at global scales of chromatin organization during aging and extensive rewiring of local contacts at finer scales that were associated with variations in transcription factor binding and aberrant gene expression. These data provide insights into genome topology as a regulator of molecular function in stem cell aging.
Collapse
Affiliation(s)
- Benjamin A. Yang
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Biointerfaces InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Jacqueline A. Larouche
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Biointerfaces InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Kaitlyn M. Sabin
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Biointerfaces InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Paula M. Fraczek
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Biointerfaces InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Stephen C. J. Parker
- Program in Cellular and Molecular BiologyUniversity of MichiganAnn ArborMichiganUSA
- Department of Computational Medicine & BioinformaticsUniversity of MichiganAnn ArborMichiganUSA
- Department of Human GeneticsUniversity of MichiganAnn ArborMichiganUSA
| | - Carlos A. Aguilar
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Biointerfaces InstituteUniversity of MichiganAnn ArborMichiganUSA
- Program in Cellular and Molecular BiologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
18
|
Tomasch J, Maleiner B, Hromada C, Szwarc-Hofbauer D, Teuschl-Woller AH. Cyclic Tensile Stress Induces Skeletal Muscle Hypertrophy and Myonuclear Accretion in a 3D Model. Tissue Eng Part A 2023; 29:257-268. [PMID: 36606693 DOI: 10.1089/ten.tea.2022.0182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle is highly adaptive to mechanical stress due to its resident stem cells and the pronounced level of myotube plasticity. Herein, we study the adaptation to mechanical stress and its underlying molecular mechanisms in a tissue-engineered skeletal muscle model. We subjected differentiated 3D skeletal muscle-like constructs to cyclic tensile stress using a custom-made bioreactor system, which resulted in immediate activation of stress-related signal transducers (Erk1/2, p38). Cell cycle re-entry, increased proliferation, and onset of myogenesis indicated subsequent myoblast activation. Furthermore, elevated focal adhesion kinase and β-catenin activity in mechanically stressed constructs suggested increased cell adhesion and migration. After 3 days of mechanical stress, gene expression of the fusogenic markers MyoMaker and MyoMixer, myotube diameter, myonuclear accretion, as well as S6 activation, were significantly increased. Our results highlight that we established a promising tool to study sustained adaptation to mechanical stress in healthy, hypertrophic, or regenerating skeletal muscle.
Collapse
Affiliation(s)
- Janine Tomasch
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Babette Maleiner
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Carina Hromada
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Dorota Szwarc-Hofbauer
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andreas H Teuschl-Woller
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
19
|
Fujimaki S, Ono Y. Murine Models of Tenotomy-Induced Mechanical Overloading and Tail-Suspension-Induced Mechanical Unloading. Methods Mol Biol 2023; 2640:207-215. [PMID: 36995597 DOI: 10.1007/978-1-0716-3036-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Skeletal muscle is a highly plastic tissue that can alter its mass and strength in response to mechanical stimulation, such as overloading and unloading, which lead to muscle hypertrophy and atrophy, respectively. Mechanical loading in the muscle influences muscle stem cell dynamics, including activation, proliferation, and differentiation. Although experimental models of mechanical overloading and unloading have been widely used for the investigation of the molecular mechanisms regulating muscle plasticity and stem cell function, few studies have described the methods in detail. Here, we describe the appropriate procedures for tenotomy-induced mechanical overloading and tail-suspension-induced mechanical unloading, which are the most common and simple methods to induce muscle hypertrophy and atrophy in mouse models.
Collapse
Affiliation(s)
- Shin Fujimaki
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
20
|
Drummer DJ, Lavin KM, Graham ZA, O'Bryan SM, McAdam JS, Lixandrão ME, Seay R, Aban I, Siegel HJ, Ghanem E, Singh JA, Bonfitto A, Antone J, Reiman R, Hutchins E, Van Keuren-Jensen K, Schutzler SE, Barnes CL, Ferrando AA, Bridges SL, Bamman MM. Muscle transcriptomic circuits linked to periarticular physiology in end-stage osteoarthritis. Physiol Genomics 2022; 54:501-513. [PMID: 36278270 PMCID: PMC9762959 DOI: 10.1152/physiolgenomics.00092.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/08/2022] [Accepted: 10/20/2022] [Indexed: 02/01/2023] Open
Abstract
The ability of individuals with end-stage osteoarthritis (OA) to functionally recover from total joint arthroplasty is highly inconsistent. The molecular mechanisms driving this heterogeneity have yet to be elucidated. Furthermore, OA disproportionately impacts females, suggesting a need for identifying female-specific therapeutic targets. We profiled the skeletal muscle transcriptome in females with end-stage OA (n = 20) undergoing total knee or hip arthroplasty using RNA-Seq. Single-gene differential expression (DE) analyses tested for DE genes between skeletal muscle overlaying the surgical (SX) joint and muscle from the contralateral (CTRL) leg. Network analyses were performed using Pathway-Level Information ExtractoR (PLIER) to summarize genes into latent variables (LVs), i.e., gene circuits, and link them to biological pathways. LV differences in SX versus CTRL muscle and across sources of muscle tissue (vastus medialis, vastus lateralis, or tensor fascia latae) were determined with ANOVA. Linear models tested for associations between LVs and muscle phenotype on the SX side (inflammation, function, and integrity). DE analysis revealed 360 DE genes (|Log2 fold-difference| ≥ 1, FDR ≤ 0.05) between the SX and CTRL limbs, many associated with inflammation and lipid metabolism. PLIER analyses revealed circuits associated with protein degradation and fibro-adipogenic cell gene expression. Muscle inflammation and function were linked to an LV associated with endothelial cell gene expression highlighting a potential regulatory role of endothelial cells within skeletal muscle. These findings may provide insight into potential therapeutic targets to improve OA rehabilitation before and/or following total joint replacement.
Collapse
Affiliation(s)
- Devin J Drummer
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kaleen M Lavin
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- Florida Institute for Human and Machine Cognition, Pensacola, Florida
| | - Zachary A Graham
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- Florida Institute for Human and Machine Cognition, Pensacola, Florida
- Birmingham VA Medical Center, Birmingham, Alabama
| | - Samia M O'Bryan
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jeremy S McAdam
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- Florida Institute for Human and Machine Cognition, Pensacola, Florida
| | - Manoel E Lixandrão
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Regina Seay
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Inmaculada Aban
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Herrick J Siegel
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Orthopaedic Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Elie Ghanem
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Orthopaedic Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jasvinder A Singh
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham VA Medical Center, Birmingham, Alabama
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Arthritis, Musculoskeletal, Bone, and Autoimmunity Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anna Bonfitto
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, Arizona
| | - Jerry Antone
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, Arizona
| | - Rebecca Reiman
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, Arizona
| | - Elizabeth Hutchins
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, Arizona
| | | | - Scott E Schutzler
- Department of Geriatrics and Center for Translational Research in Aging and Longevity, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - C Lowry Barnes
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Arny A Ferrando
- Department of Geriatrics and Center for Translational Research in Aging and Longevity, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - S Louis Bridges
- Department of Medicine, Hospital for Special Surgery, New York, New York
- Division of Rheumatology, Weill Cornell Medical Center, New York, New York
| | - Marcas M Bamman
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- Florida Institute for Human and Machine Cognition, Pensacola, Florida
| |
Collapse
|
21
|
Bernard C, Zavoriti A, Pucelle Q, Chazaud B, Gondin J. Role of macrophages during skeletal muscle regeneration and hypertrophy-Implications for immunomodulatory strategies. Physiol Rep 2022; 10:e15480. [PMID: 36200266 PMCID: PMC9535344 DOI: 10.14814/phy2.15480] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/12/2022] [Indexed: 06/16/2023] Open
Abstract
Skeletal muscle is a plastic tissue that regenerates ad integrum after injury and adapts to raise mechanical loading/contractile activity by increasing its mass and/or myofiber size, a phenomenon commonly refers to as skeletal muscle hypertrophy. Both muscle regeneration and hypertrophy rely on the interactions between muscle stem cells and their neighborhood, which include inflammatory cells, and particularly macrophages. This review first summarizes the role of macrophages in muscle regeneration in various animal models of injury and in response to exercise-induced muscle damage in humans. Then, the potential contribution of macrophages to skeletal muscle hypertrophy is discussed on the basis of both animal and human experiments. We also present a brief comparative analysis of the role of macrophages during muscle regeneration versus hypertrophy. Finally, we summarize the current knowledge on the impact of different immunomodulatory strategies, such as heat therapy, cooling, massage, nonsteroidal anti-inflammatory drugs and resolvins, on skeletal muscle regeneration and their potential impact on muscle hypertrophy.
Collapse
Affiliation(s)
- Clara Bernard
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Aliki Zavoriti
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Quentin Pucelle
- Université de Versailles Saint‐Quentin‐En‐YvelinesVersaillesFrance
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| |
Collapse
|
22
|
Fukada SI, Higashimoto T, Kaneshige A. Differences in muscle satellite cell dynamics during muscle hypertrophy and regeneration. Skelet Muscle 2022; 12:17. [PMID: 35794679 PMCID: PMC9258228 DOI: 10.1186/s13395-022-00300-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/29/2022] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle homeostasis and function are ensured by orchestrated cellular interactions among several types of cells. A noticeable aspect of skeletal muscle biology is the drastic cell-cell communication changes that occur in multiple scenarios. The process of recovering from an injury, which is known as regeneration, has been relatively well investigated. However, the cellular interplay that occurs in response to mechanical loading, such as during resistance training, is poorly understood compared to regeneration. During muscle regeneration, muscle satellite cells (MuSCs) rebuild multinuclear myofibers through a stepwise process of proliferation, differentiation, fusion, and maturation, whereas during mechanical loading-dependent muscle hypertrophy, MuSCs do not undergo such stepwise processes (except in rare injuries) because the nuclei of MuSCs become directly incorporated into the mature myonuclei. In this review, six specific examples of such differences in MuSC dynamics between regeneration and hypertrophy processes are discussed.
Collapse
Affiliation(s)
- So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Tatsuyoshi Higashimoto
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Akihiro Kaneshige
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, 569-1125, Japan
| |
Collapse
|
23
|
Yu W, Yang MK, Sung DJ, Park TJ, Kim M, Ntigura E, Kim SH, Kim B, Park SW, Bae YM. Differential expression profiles of miRNA in the serum of sarcopenic rats. Biochem Biophys Rep 2022; 30:101251. [PMID: 35313645 PMCID: PMC8933690 DOI: 10.1016/j.bbrep.2022.101251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/07/2022] [Accepted: 03/12/2022] [Indexed: 11/30/2022] Open
Abstract
As the geriatric population and life expectancy increase, the interest in preventing geriatric diseases, such as sarcopenia, is increasing. However, the causes of sarcopenia are unclear, and current diagnostic methods for sarcopenia are unreliable. We hypothesized that the changes in the expression of certain miRNAs may be associated with the pathophysiology of sarcopenia. Herein, we analyzed the miRNA expression profiles in the blood of young (3-months-old) healthy rats, old sarcopenic (17-months-old) rats, and age-matched (17-months-old) control rats. The changes in miRNA expression levels were analyzed using Bowtie 2 software. A total of 523 miRNAs were detected in the rat serum. Using scatter plots and clustering heatmap data, we found 130 miRNAs that were differentially expressed in sarcopenic rats (>2-fold change) compared to the expression in young healthy and age-matched control rats. With a threshold of >5-fold change, we identified 14 upregulated miRNAs, including rno-miR-133b-3p, rno-miR-133a-3p, rno-miR-133c, rno-miR-208a-3p, and rno-miR434-5p among others in the serum of sarcopenic rats. A protein network map based on these 14 miRNAs identified the genes involved in skeletal muscle differentiation, among which Notch1, Egr2, and Myocd represented major nodes. The data obtained in this study are potentially useful for the early diagnosis of sarcopenia and for the identification of novel therapeutic targets for the treatment and/or prevention of sarcopenia. Early diagnosis of sarcopenia and discovery of therapeutic targets are necessary. We determined the significantly increased miRNAs in the serum of sarcopenic rats. They include rno-miR-133b-3p, 133a-3p, 133c, 208a-3p, and miR434-5p. Network mapping from the miRNA expression profiles helps identify target proteins. Notch, Egr, and Myocd genes were suggested as targets for sarcopenia treatment.
Collapse
Affiliation(s)
- Wonjong Yu
- Department of Physical Therapy, Eulji University, South Korea
| | - Min-kyu Yang
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, South Korea
| | - Dong Jun Sung
- Department of Sport and Health Studies, College of Biomedical and Health Science, Konkuk University, Chungju, Republic of Korea
| | - Tae Jun Park
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, South Korea
| | - Myungchul Kim
- Department of Physical Therapy, Eulji University, South Korea
| | - Eustache Ntigura
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, South Korea
- Department of Preventive and Community Dentistry, School of Dentistry, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | - Sung Hea Kim
- Department of Cardiology, Konkuk University School of Medicine, Seoul, South Korea
| | - Bokyung Kim
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, South Korea
| | - Sang Woong Park
- Department of Emergency Medical Services, Eulji University, South Korea
- Corresponding author.
| | - Young Min Bae
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, South Korea
- Corresponding author.
| |
Collapse
|
24
|
Kaneshige A, Kaji T, Saito H, Higashimoto T, Nakamura A, Kurosawa T, Ikemoto-Uezumi M, Uezumi A, Fukada SI. Detection of muscle stem cell-derived myonuclei in murine overloaded muscles. STAR Protoc 2022; 3:101307. [PMID: 35463471 PMCID: PMC9018451 DOI: 10.1016/j.xpro.2022.101307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Akihiro Kaneshige
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takayuki Kaji
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hayato Saito
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Tatsuyoshi Higashimoto
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Ayasa Nakamura
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Tamaki Kurosawa
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medical Sciences, Graduate School of Agriculture and Life Sciences, Tokyo University, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Madoka Ikemoto-Uezumi
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Akiyoshi Uezumi
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - So-ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
- Corresponding author
| |
Collapse
|
25
|
Gioftsidi S, Relaix F, Mourikis P. The Notch signaling network in muscle stem cells during development, homeostasis, and disease. Skelet Muscle 2022; 12:9. [PMID: 35459219 PMCID: PMC9027478 DOI: 10.1186/s13395-022-00293-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/16/2022] [Indexed: 01/22/2023] Open
Abstract
Skeletal muscle stem cells have a central role in muscle growth and regeneration. They reside as quiescent cells in resting muscle and in response to damage they transiently amplify and fuse to produce new myofibers or self-renew to replenish the stem cell pool. A signaling pathway that is critical in the regulation of all these processes is Notch. Despite the major differences in the anatomical and cellular niches between the embryonic myotome, the adult sarcolemma/basement-membrane interphase, and the regenerating muscle, Notch signaling has evolved to support the context-specific requirements of the muscle cells. In this review, we discuss the diverse ways by which Notch signaling factors and other modifying partners are operating during the lifetime of muscle stem cells to establish an adaptive dynamic network.
Collapse
Affiliation(s)
- Stamatia Gioftsidi
- Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Mondor Institute for Biomedical Research (IMRB), F-94010, Créteil, France
| | - Frederic Relaix
- Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Mondor Institute for Biomedical Research (IMRB), F-94010, Créteil, France
- EnvA, IMRB, F-94700, Maisons-Alfort, France
- Etablissement Français du Sang (EFS), IMRB, F-94010, Creteil, France
- Assistance Publique-Hôpitaux de Paris, Hopital Mondor, Service d'Histologie, F-94010, Creteil, France
| | - Philippos Mourikis
- Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Mondor Institute for Biomedical Research (IMRB), F-94010, Créteil, France.
| |
Collapse
|
26
|
Ato S, Fukada SI, Kokubo H, Ogasawara R. Implication of satellite cell behaviors in capillary growth via VEGF expression-independent mechanism in response to mechanical loading in HeyL-null mice. Am J Physiol Cell Physiol 2022; 322:C275-C282. [PMID: 35020502 DOI: 10.1152/ajpcell.00343.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 01/04/2022] [Accepted: 01/04/2022] [Indexed: 11/22/2022]
Abstract
Angiogenesis and muscle satellite cell (SC)-mediated myonuclear accretion are considered essential for the robust response of contraction-induced muscle hypertrophy. Moreover, both myonucleus and SCs are physically adjacent to capillaries and are the major sites for the expression of proangiogenic factors, such as VEGF, in the skeletal muscle. Thus, events involving the addition of new myonuclei via activation of SCs may play an important role in angiogenesis during muscle hypertrophy. However, the relevance among myonuclei number, capillary supply, and angiogenesis factor is not demonstrated. The Notch effector HeyL is specifically expressed in SCs in the skeletal muscle and is crucial for SC proliferation by inhibiting MyoD in overload-induced muscle hypertrophy. Here, we tested whether the addition of new myonuclei by SC in overloaded muscle is associated with angiogenic adaptation by reanalyzing skeletal muscle from HeyL-knockout (KO) mice, which show blunted responses of SC proliferation, myonucleus addition, and overload-induced muscle hypertrophy. Reanalysis confirmed blunted SC proliferation and myonuclear accretion in the plantaris muscle of HeyL-KO mice 9 wk after synergist ablation. Interestingly, the increase in capillary-to-fiber ratio observed in wild-type (WT) mice was impaired in HeyL-KO mice. In both WT and HeyL-KO mice, the expression of VEGFA and VEGFB was similarly increased in response to overload. In addition, the expression pattern of TSP-1, a negative regulator of angiogenesis, was also not changed between WT and HeyL-KO mice. Collectively, these results suggest that SCs activation-myonuclear accretion plays a crucial role in angiogenesis during overload-induced muscle hypertrophy via independent of angiogenesis regulators.
Collapse
Affiliation(s)
- Satoru Ato
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Hiroki Kokubo
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Riki Ogasawara
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| |
Collapse
|
27
|
Lahmann I, Zhang Y, Baum K, Wolf J, Birchmeier C. An oscillatory network controlling self-renewal of skeletal muscle stem cells. Exp Cell Res 2021; 409:112933. [PMID: 34793773 DOI: 10.1016/j.yexcr.2021.112933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/04/2021] [Accepted: 11/14/2021] [Indexed: 12/11/2022]
Abstract
The balance between proliferation and differentiation of muscle stem cells is tightly controlled, ensuring the maintenance of a cellular pool needed for muscle growth and repair. Muscle stem cells can proliferate, they can generate differentiating cells, or they self-renew to produce new stem cells. Notch signaling plays a crucial role in this process. Recent studies revealed that expression of the Notch effector HES1 oscillates in activated muscle stem cells. The oscillatory expression of HES1 periodically represses transcription from the genes encoding the myogenic transcription factor MYOD and the Notch ligand DLL1, thereby driving MYOD and DLL1 oscillations. This oscillatory network allows muscle progenitor cells and activated muscle stem cells to remain in a proliferative and 'undecided' state, in which they can either differentiate or self-renew. When HES1 is downregulated, MYOD oscillations become unstable and are replaced by sustained expression, which drives the cells into terminal differentiation. During development and regeneration, proliferating stem cells contact each other and the stability of the oscillatory expression depends on regular DLL1 inputs provided by neighboring cells. In such communities of cells that receive and provide Notch signals, the appropriate timing of DLL1 inputs is important, as sustained DLL1 cannot replace oscillatory DLL1. Thus, in cell communities, DLL1 oscillations ensure the appropriate balance between self-renewal and differentiation. In summary, oscillations in myogenic cells are an important example of dynamic gene expression determining cell fate.
Collapse
Affiliation(s)
- Ines Lahmann
- Neurowissenschaftliches Forschungszentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology/Signal Transduction Group, 13125, Berlin, Germany
| | - Yao Zhang
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology/Signal Transduction Group, 13125, Berlin, Germany
| | - Katharina Baum
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Mathematical Modelling of Cellular Processes, 13125, Berlin, Germany; New address: Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, 14482, Potsdam, Germany
| | - Jana Wolf
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Mathematical Modelling of Cellular Processes, 13125, Berlin, Germany; Free University Berlin, Department of Mathematics and Computer Science, Arnimallee 14, 14195, Berlin, Germany
| | - Carmen Birchmeier
- Neurowissenschaftliches Forschungszentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology/Signal Transduction Group, 13125, Berlin, Germany.
| |
Collapse
|
28
|
Gerrard JC, Hay JP, Adams RN, Williams JC, Huot JR, Weathers KM, Marino JS, Arthur ST. Current Thoughts of Notch's Role in Myoblast Regulation and Muscle-Associated Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182312558. [PMID: 34886282 PMCID: PMC8657396 DOI: 10.3390/ijerph182312558] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022]
Abstract
The evolutionarily conserved signaling pathway Notch is unequivocally essential for embryogenesis. Notch’s contribution to the muscle repair process in adult tissue is complex and obscure but necessary. Notch integrates with other signals in a functional antagonist manner to direct myoblast activity and ultimately complete muscle repair. There is profound recent evidence describing plausible mechanisms of Notch in muscle repair. However, the story is not definitive as evidence is slowly emerging that negates Notch’s importance in myoblast proliferation. The purpose of this review article is to examine the prominent evidence and associated mechanisms of Notch’s contribution to the myogenic repair phases. In addition, we discuss the emerging roles of Notch in diseases associated with muscle atrophy. Understanding the mechanisms of Notch’s orchestration is useful for developing therapeutic targets for disease.
Collapse
Affiliation(s)
- Jeffrey C. Gerrard
- Department of Applied Physiology, Health and Clinical Sciences, University of North Carolina-Charlotte, Charlotte, NC 28223, USA; (J.C.G.); (J.P.H.); (R.N.A.); (J.C.W.III); (K.M.W.); (J.S.M.)
| | - Jamison P. Hay
- Department of Applied Physiology, Health and Clinical Sciences, University of North Carolina-Charlotte, Charlotte, NC 28223, USA; (J.C.G.); (J.P.H.); (R.N.A.); (J.C.W.III); (K.M.W.); (J.S.M.)
| | - Ryan N. Adams
- Department of Applied Physiology, Health and Clinical Sciences, University of North Carolina-Charlotte, Charlotte, NC 28223, USA; (J.C.G.); (J.P.H.); (R.N.A.); (J.C.W.III); (K.M.W.); (J.S.M.)
| | - James C. Williams
- Department of Applied Physiology, Health and Clinical Sciences, University of North Carolina-Charlotte, Charlotte, NC 28223, USA; (J.C.G.); (J.P.H.); (R.N.A.); (J.C.W.III); (K.M.W.); (J.S.M.)
| | - Joshua R. Huot
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Kaitlin M. Weathers
- Department of Applied Physiology, Health and Clinical Sciences, University of North Carolina-Charlotte, Charlotte, NC 28223, USA; (J.C.G.); (J.P.H.); (R.N.A.); (J.C.W.III); (K.M.W.); (J.S.M.)
| | - Joseph S. Marino
- Department of Applied Physiology, Health and Clinical Sciences, University of North Carolina-Charlotte, Charlotte, NC 28223, USA; (J.C.G.); (J.P.H.); (R.N.A.); (J.C.W.III); (K.M.W.); (J.S.M.)
| | - Susan T. Arthur
- Department of Applied Physiology, Health and Clinical Sciences, University of North Carolina-Charlotte, Charlotte, NC 28223, USA; (J.C.G.); (J.P.H.); (R.N.A.); (J.C.W.III); (K.M.W.); (J.S.M.)
- Correspondence:
| |
Collapse
|
29
|
Kaneshige A, Kaji T, Zhang L, Saito H, Nakamura A, Kurosawa T, Ikemoto-Uezumi M, Tsujikawa K, Seno S, Hori M, Saito Y, Matozaki T, Maehara K, Ohkawa Y, Potente M, Watanabe S, Braun T, Uezumi A, Fukada SI. Relayed signaling between mesenchymal progenitors and muscle stem cells ensures adaptive stem cell response to increased mechanical load. Cell Stem Cell 2021; 29:265-280.e6. [PMID: 34856120 DOI: 10.1016/j.stem.2021.11.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 09/24/2021] [Accepted: 11/09/2021] [Indexed: 11/29/2022]
Abstract
Adaptation to mechanical load, leading to enhanced force and power output, is a characteristic feature of skeletal muscle. Formation of new myonuclei required for efficient muscle hypertrophy relies on prior activation and proliferation of muscle stem cells (MuSCs). However, the mechanisms controlling MuSC expansion under conditions of increased load are not fully understood. Here we demonstrate that interstitial mesenchymal progenitors respond to mechanical load and stimulate MuSC proliferation in a surgical mouse model of increased muscle load. Mechanistically, transcriptional activation of Yes-associated protein 1 (Yap1)/transcriptional coactivator with PDZ-binding motif (Taz) in mesenchymal progenitors results in local production of thrombospondin-1 (Thbs1), which, in turn, drives MuSC proliferation through CD47 signaling. Under homeostatic conditions, however, CD47 signaling is insufficient to promote MuSC proliferation and instead depends on prior downregulation of the Calcitonin receptor. Our results suggest that relayed signaling between mesenchymal progenitors and MuSCs through a Yap1/Taz-Thbs1-CD47 pathway is critical to establish the supply of MuSCs during muscle hypertrophy.
Collapse
Affiliation(s)
- Akihiro Kaneshige
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan; Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan; Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takayuki Kaji
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Lidan Zhang
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hayato Saito
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Ayasa Nakamura
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Tamaki Kurosawa
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi, Tokyo 173-0015, Japan; Laboratory of Veterinary Pharmacology, Department of Veterinary Medical Sciences, Graduate School of Agriculture and Life Sciences, Tokyo University, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Madoka Ikemoto-Uezumi
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi, Tokyo 173-0015, Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Masatoshi Hori
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medical Sciences, Graduate School of Agriculture and Life Sciences, Tokyo University, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yasuyuki Saito
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takashi Matozaki
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Kazumitsu Maehara
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Michael Potente
- Angiogenesis & Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; Berlin Institute of Health at Charité (BIH) - Universitätsmedizin Berlin, 13125 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Shuichi Watanabe
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Akiyoshi Uezumi
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi, Tokyo 173-0015, Japan.
| | - So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
30
|
Fukada SI, Ito N. Regulation of muscle hypertrophy: Involvement of the Akt-independent pathway and satellite cells in muscle hypertrophy. Exp Cell Res 2021; 409:112907. [PMID: 34793776 DOI: 10.1016/j.yexcr.2021.112907] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 10/04/2021] [Accepted: 10/29/2021] [Indexed: 12/25/2022]
Abstract
Skeletal muscles are composed of multinuclear cells called myofibers and have unique abilities, one of which is plasticity. In response to the mechanical load induced by physical activity, skeletal muscle exerts several local adaptations, including an increase in myofiber size and myonuclear number, known as muscle hypertrophy. Protein synthesis and muscle satellite cells (MuSCs) are mainly responsible for these adaptations. However, the upstream signaling pathways that promote protein synthesis remain controversial. Further, the necessity of MuSCs in muscle hypertrophy is also a highly debated issue. In this review, we summarized the insulin-like growth factor 1 (IGF-1)/Akt-independent activation of mammalian target of rapamycin (mTOR) signaling in muscle hypertrophy and the involvement of mTOR signaling in age-related loss of skeletal muscle function and mass and in sarcopenia. The roles and behaviors of MuSCs, characteristics of new myonuclei in muscle hypertrophy, and their relevance to sarcopenia have also been updated in this review.
Collapse
Affiliation(s)
- So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
| | - Naoki Ito
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation (IBRI), Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, Japan
| |
Collapse
|
31
|
Mori T, Ato S, Knudsen JR, Henriquez-Olguin C, Li Z, Wakabayashi K, Suginohara T, Higashida K, Tamura Y, Nakazato K, Jensen TE, Ogasawara R. c-Myc overexpression increases ribosome biogenesis and protein synthesis independent of mTORC1 activation in mouse skeletal muscle. Am J Physiol Endocrinol Metab 2021; 321:E551-E559. [PMID: 34423683 DOI: 10.1152/ajpendo.00164.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
High-intensity muscle contractions (HiMCs) are known to increase c-Myc expression that is known to stimulate ribosome biogenesis and protein synthesis in most cells. However, although c-Myc mRNA transcription and c-Myc mRNA translation have been shown to be upregulated following resistance exercise concomitantly with increased ribosome biogenesis, this connection has not been tested directly. We investigated the effect of adeno-associated virus (AAV)-mediated c-Myc overexpression, with or without fasting or percutaneous electrical stimulation-induced HiMC, on ribosome biogenesis and protein synthesis in adult mouse skeletal muscles. AAV-mediated overexpression of c-Myc in mouse skeletal muscles for 2 wk increased the DNA polymerase subunit POL1 mRNA, 45S-pre-rRNA, total RNA, and muscle protein synthesis without altering mechanistic target of rapamycin complex 1 (mTORC1) signaling under both ad libitum and fasted conditions. RNA-sequencing (RNA-seq) analyses revealed that c-Myc overexpression mainly regulated ribosome biogenesis-related biological processes. The protein synthesis response to c-Myc overexpression mirrored the response with HiMC. No additional effect of combining c-Myc overexpression and HiMC was observed. Our results suggest that c-Myc overexpression is sufficient to stimulate skeletal muscle ribosome biogenesis and protein synthesis without activation of mTORC1. Therefore, the HiMC-induced increase in c-Myc may contribute to ribosome biogenesis and increased protein synthesis following HiMC.NEW & NOTEWORTHY Resistance exercise is known to increase c-Myc expression, which is known to stimulate ribosome biogenesis and protein synthesis in a variety of cells. However, whether the increase in c-Myc stimulates ribosome biogenesis and protein synthesis in skeletal muscles remains unknown. We found that c-Myc overexpression is sufficient to stimulate skeletal muscle ribosome biogenesis and protein synthesis without activation of mTORC1.
Collapse
Affiliation(s)
- Takahiro Mori
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | - Satoru Ato
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | - Jonas R Knudsen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Microsystems Laboratory 2, Institute of Microengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Carlos Henriquez-Olguin
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Zhencheng Li
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Koki Wakabayashi
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | - Takeshi Suginohara
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | | | - Yuki Tamura
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo, Japan
| | - Koichi Nakazato
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo, Japan
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Riki Ogasawara
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Murach KA, Fry CS, Dupont-Versteegden EE, McCarthy JJ, Peterson CA. Fusion and beyond: Satellite cell contributions to loading-induced skeletal muscle adaptation. FASEB J 2021; 35:e21893. [PMID: 34480776 PMCID: PMC9293230 DOI: 10.1096/fj.202101096r] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022]
Abstract
Satellite cells support adult skeletal muscle fiber adaptations to loading in numerous ways. The fusion of satellite cells, driven by cell-autonomous and/or extrinsic factors, contributes new myonuclei to muscle fibers, associates with load-induced hypertrophy, and may support focal membrane damage repair and long-term myonuclear transcriptional output. Recent studies have also revealed that satellite cells communicate within their niche to mediate muscle remodeling in response to resistance exercise, regulating the activity of numerous cell types through various mechanisms such as secretory signaling and cell-cell contact. Muscular adaptation to resistance and endurance activity can be initiated and sustained for a period of time in the absence of satellite cells, but satellite cell participation is ultimately required to achieve full adaptive potential, be it growth, function, or proprioceptive coordination. While significant progress has been made in understanding the roles of satellite cells in adult muscle over the last few decades, many conclusions have been extrapolated from regeneration studies. This review highlights our current understanding of satellite cell behavior and contributions to adaptation outside of regeneration in adult muscle, as well as the roles of satellite cells beyond fusion and myonuclear accretion, which are gaining broader recognition.
Collapse
Affiliation(s)
- Kevin A Murach
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, Arkansas, USA.,Cell and Molecular Biology Program, University of Arkansas, Fayetteville, Arkansas, USA
| | - Christopher S Fry
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Esther E Dupont-Versteegden
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
33
|
Fukada SI, Nakamura A. Exercise/Resistance Training and Muscle Stem Cells. Endocrinol Metab (Seoul) 2021; 36:737-744. [PMID: 34372625 PMCID: PMC8419599 DOI: 10.3803/enm.2021.401] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 07/29/2021] [Indexed: 01/10/2023] Open
Abstract
Skeletal muscle has attracted attention as endocrine organ, because exercise-dependent cytokines called myokines/exerkines are released from skeletal muscle and are involved in systemic functions. While, local mechanical loading to skeletal muscle by exercise or resistance training alters myofiber type and size and myonuclear number. Skeletal muscle-resident stem cells, known as muscle satellite cells (MuSCs), are responsible for the increased number of myonuclei. Under steady conditions, MuSCs are maintained in a mitotically quiescent state but exit from that state and start to proliferate in response to high physical activity. Alterations in MuSC behavior occur when myofibers are damaged, but the lethal damage to myofibers does not seem to evoke mechanical loading-dependent MuSC activation and proliferation. Given that MuSCs proliferate without damage, it is unclear how the different behaviors of MuSCs are controlled by different physical activities. Recent studies demonstrated that myonuclear number reflects the size of myofibers; hence, it is crucial to know the properties of MuSCs and the mechanism of myonuclear accretion by MuSCs. In addition, the elucidation of mechanical load-dependent changes in muscle resident cells, including MuSCs, will be necessary for the discovery of new myokines/exerkines and understating skeletal muscle diseases.
Collapse
Affiliation(s)
- So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Ayasa Nakamura
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| |
Collapse
|
34
|
Li Y, Li X, Gao Y, Huang C, Lin D. NMR-Based Metabolomic Analysis for the Effects of α-Ketoglutarate Supplementation on C2C12 Myoblasts in Different Energy States. Molecules 2021; 26:1841. [PMID: 33805924 PMCID: PMC8037044 DOI: 10.3390/molecules26071841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 11/17/2022] Open
Abstract
α-Ketoglutarate (AKG) is attracting much attention from researchers owing to its beneficial effects on anti-aging and cancer suppression, and, more recently, in nutritional supplements. Given that glucose is the main source of energy to maintain normal physiological functions of skeletal muscle, the effects of AKG supplementation for improving muscle performance are closely related to the glucose level in skeletal muscle. The differences of AKG-induced effects in skeletal muscle between two states of normal energy and energy deficiency are unclear. Furthermore, AKG-induced metabolic changes in skeletal muscles in different energy states also remain elusive. Here, we assessed the effects of AKG supplementation on mouse C2C12 myoblast cells cultured both in normal medium (Nor cells) and in low-glucose medium (Low cells), which were used to mimic two states of normal energy and energy deficiency, respectively. We further performed NMR-based metabolomic analysis to address AKG-induced metabolic changes in Nor and Low cells. AKG supplementation significantly promoted the proliferation and differentiation of cells in the two energy states through glutamine metabolism, oxidative stress, and energy metabolism. Under normal culture conditions, AKG up-regulated the intracellular glutamine level, changed the cellular energy status, and maintained the antioxidant capacity of cells. Under low-glucose culture condition, AKG served as a metabolic substrate to reduce the glutamine-dependence of cells, remarkably enhanced the antioxidant capacity of cells and significantly elevated the intracellular ATP level, thereby ensuring the normal growth and metabolism of cells in the state of energy deficiency. Our results provide a mechanistic understanding of the effects of AKG supplements on myoblasts in both normal energy and energy deficiency states. This work may be beneficial to the exploitation of AKG applications in clinical treatments and nutritional supplementations.
Collapse
Affiliation(s)
- Yantong Li
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| | - Xiaoyuan Li
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| | - Yifeng Gao
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361021, China
| | - Donghai Lin
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| |
Collapse
|
35
|
Zhang L, Kubota M, Nakamura A, Kaji T, Seno S, Uezumi A, Andersen DC, Jensen CH, Fukada SI. Dlk1 regulates quiescence in calcitonin receptor-mutant muscle stem cells. Stem Cells 2021; 39:306-317. [PMID: 33295098 DOI: 10.1002/stem.3312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/20/2020] [Indexed: 12/30/2022]
Abstract
Muscle stem cells, also called muscle satellite cells (MuSCs), are responsible for skeletal muscle regeneration and are sustained in an undifferentiated and quiescent state under steady conditions. The calcitonin receptor (CalcR)-protein kinase A (PKA)-Yes-associated protein 1 (Yap1) axis is one pathway that maintains quiescence in MuSCs. Although CalcR signaling in MuSCs has been identified, the critical CalcR signaling targets are incompletely understood. Here, we show the relevance between the ectopic expression of delta-like non-canonical Notch ligand 1 (Dlk1) and the impaired quiescent state in CalcR-conditional knockout (cKO) MuSCs. Dlk1 expression was rarely detected in both quiescent and proliferating MuSCs in control mice, whereas Dlk1 expression was remarkably increased in CalcR-cKO MuSCs at both the mRNA and protein levels. It is noteworthy that all Ki67+ non-quiescent CalcR-cKO MuSCs express Dlk1, and non-quiescent CalcR-cKO MuSCs are enriched in the Dlk1+ fraction by cell sorting. Using mutant mice, we demonstrated that PKA-activation or Yap1-depletion suppressed Dlk1 expression in CalcR-cKO MuSCs, which suggests that the CalcR-PKA-Yap1 axis inhibits the expression of Dlk1 in quiescent MuSCs. Moreover, the loss of Dlk1 rescued the quiescent state in CalcR-cKO MuSCs, which indicates that the ectopic expression of Dlk1 disturbs quiescence in CalcR-cKO. Collectively, our results suggest that ectopically expressed Dlk1 is responsible for the impaired quiescence in CalcR-cKO MuSCs.
Collapse
Affiliation(s)
- Lidan Zhang
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Manami Kubota
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Ayasa Nakamura
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Takayuki Kaji
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Suita, Osaka, Japan
| | - Akiyoshi Uezumi
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Itabashi, Tokyo, Japan
| | - Ditte Caroline Andersen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense C, Denmark
- Clinical Institute, University of Southern Denmark, Odense C, Denmark
| | - Charlotte Harken Jensen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense C, Denmark
| | - So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
36
|
Jabre S, Hleihel W, Coirault C. Nuclear Mechanotransduction in Skeletal Muscle. Cells 2021; 10:cells10020318. [PMID: 33557157 PMCID: PMC7913907 DOI: 10.3390/cells10020318] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle is composed of multinucleated, mature muscle cells (myofibers) responsible for contraction, and a resident pool of mononucleated muscle cell precursors (MCPs), that are maintained in a quiescent state in homeostatic conditions. Skeletal muscle is remarkable in its ability to adapt to mechanical constraints, a property referred as muscle plasticity and mediated by both MCPs and myofibers. An emerging body of literature supports the notion that muscle plasticity is critically dependent upon nuclear mechanotransduction, which is transduction of exterior physical forces into the nucleus to generate a biological response. Mechanical loading induces nuclear deformation, changes in the nuclear lamina organization, chromatin condensation state, and cell signaling, which ultimately impacts myogenic cell fate decisions. This review summarizes contemporary insights into the mechanisms underlying nuclear force transmission in MCPs and myofibers. We discuss how the cytoskeleton and nuclear reorganizations during myogenic differentiation may affect force transmission and nuclear mechanotransduction. We also discuss how to apply these findings in the context of muscular disorders. Finally, we highlight current gaps in knowledge and opportunities for further research in the field.
Collapse
Affiliation(s)
- Saline Jabre
- Sorbonne Université, INSERM UMRS-974 and Institut de Myologie, 75013 Paris, France;
- Department of Biology, Faculty of Arts and Sciences, Holy Spirit University of Kasik (USEK), Jounieh 446, Lebanon;
| | - Walid Hleihel
- Department of Biology, Faculty of Arts and Sciences, Holy Spirit University of Kasik (USEK), Jounieh 446, Lebanon;
- Department of Basic Health Sciences, Faculty of Medicine, Holy Spirit University of Kaslik (USEK), Jounieh 446, Lebanon
| | - Catherine Coirault
- Sorbonne Université, INSERM UMRS-974 and Institut de Myologie, 75013 Paris, France;
- Correspondence:
| |
Collapse
|
37
|
Cramer AAW, Prasad V, Eftestøl E, Song T, Hansson KA, Dugdale HF, Sadayappan S, Ochala J, Gundersen K, Millay DP. Nuclear numbers in syncytial muscle fibers promote size but limit the development of larger myonuclear domains. Nat Commun 2020; 11:6287. [PMID: 33293533 PMCID: PMC7722938 DOI: 10.1038/s41467-020-20058-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 10/30/2020] [Indexed: 12/11/2022] Open
Abstract
Mammalian cells exhibit remarkable diversity in cell size, but the factors that regulate establishment and maintenance of these sizes remain poorly understood. This is especially true for skeletal muscle, comprised of syncytial myofibers that each accrue hundreds of nuclei during development. Here, we directly explore the assumed causal relationship between multinucleation and establishment of normal size through titration of myonuclear numbers during mouse neonatal development. Three independent mouse models, where myonuclear numbers were reduced by 75, 55, or 25%, led to the discovery that myonuclei possess a reserve capacity to support larger functional cytoplasmic volumes in developing myofibers. Surprisingly, the results revealed an inverse relationship between nuclei numbers and reserve capacity. We propose that as myonuclear numbers increase, the range of transcriptional return on a per nuclear basis in myofibers diminishes, which accounts for both the absolute reliance developing myofibers have on nuclear accrual to establish size, and the limits of adaptability in adult skeletal muscle.
Collapse
Affiliation(s)
- Alyssa A W Cramer
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Vikram Prasad
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Einar Eftestøl
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Taejeong Song
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Kenth-Arne Hansson
- Department of Biosciences, University of Oslo, Oslo, Norway
- Center for Integrative Neuroplasticity (CINPLA), Department of Biosciences, University of Oslo, Oslo, Norway
| | - Hannah F Dugdale
- Center of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Sakthivel Sadayappan
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Julien Ochala
- Center of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Randall Center for Cell and Molecular Biophysics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, Guy's Campus, King's College London, London, UK
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| |
Collapse
|
38
|
Kobayashi Y, Tanaka T, Mulati M, Ochi H, Sato S, Kaldis P, Yoshii T, Okawa A, Inose H. Cyclin-Dependent Kinase 1 Is Essential for Muscle Regeneration and Overload Muscle Fiber Hypertrophy. Front Cell Dev Biol 2020; 8:564581. [PMID: 33163487 PMCID: PMC7591635 DOI: 10.3389/fcell.2020.564581] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/18/2020] [Indexed: 12/27/2022] Open
Abstract
Satellite cell proliferation is an essential step in proper skeletal muscle development and muscle regeneration. However, the mechanisms regulating satellite cell proliferation are relatively unknown compared to the knowledge associated with the differentiation of satellite cells. Moreover, it is still unclear whether overload muscle fiber hypertrophy is dependent on satellite cell proliferation. In general, cell proliferation is regulated by the activity of cell cycle regulators, such as cyclins and cyclin-dependent kinases (CDKs). Despite recent reports on the function of CDKs and CDK inhibitors in satellite cells, the physiological role of Cdk1 in satellite cell proliferation remains unknown. Herein, we demonstrate that Cdk1 regulates satellite cell proliferation, muscle regeneration, and muscle fiber hypertrophy. Cdk1 is highly expressed in myoblasts and is downregulated upon myoblast differentiation. Inhibition of CDK1 activity inhibits myoblast proliferation. Deletion of Cdk1 in satellite cells leads to inhibition of muscle recovery after muscle injury due to reduced satellite cell proliferation in vivo. Finally, we provide direct evidence that Cdk1 expression in satellite cells is essential for overload muscle fiber hypertrophy in vivo. Collectively, our results demonstrate that Cdk1 is essential for myoblast proliferation, muscle regeneration, and muscle fiber hypertrophy. These findings could help to develop treatments for refractory muscle injuries and muscle atrophy, such as sarcopenia.
Collapse
Affiliation(s)
- Yutaka Kobayashi
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomoyuki Tanaka
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mieradilli Mulati
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroki Ochi
- Department of Rehabilitation for Movement Functions, National Rehabilitation Center for Persons with Disabilities, Research Institute, Tokorozawa, Japan
| | - Shingo Sato
- Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Philipp Kaldis
- Department of Clinical Sciences, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Toshitaka Yoshii
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Okawa
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Inose
- Department of Orthopedic and Trauma Research, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
39
|
Vitamin D Inhibits Myogenic Cell Fusion and Expression of Fusogenic Genes. Nutrients 2020; 12:nu12082192. [PMID: 32717927 PMCID: PMC7469064 DOI: 10.3390/nu12082192] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/18/2020] [Accepted: 07/18/2020] [Indexed: 01/02/2023] Open
Abstract
Vitamin D, a fat-soluble vitamin, is an important nutrient for tissue homeostasis and is recently gaining attention for its role in sarcopenia. Although several studies have focused on the role of vitamin D in muscle homeostasis, the molecular mechanism underlying its action on skeletal muscle remains unclear. This study investigated the role of vitamin D in myogenesis and muscle fiber maintenance in an immortalized mouse myogenic cell line. A high concentration of active vitamin D, 1α,25(OH)2D3, decreased the expression of myogenic regulatory factors (MRFs), myf5 and myogenin in proliferating myoblasts. In addition, high concentration of vitamin D reduced myoblast-to-myoblast and myoblast-to-myotube fusion through the inhibition of Tmem8c (myomaker) and Gm7325 (myomerger), which encode muscle-specific fusion-related micropeptides. A similar inhibitory effect of vitamin D was also observed in immortalized human myogenic cells. A high concentration of vitamin D also induced hypertrophy of multinucleated myotubes by stimulating protein anabolism. The results from this study indicated that vitamin D had both positive and negative effects on muscle homeostasis, such as in muscle regeneration and myofiber maintenance. Elderly individuals face a higher risk of falling and suffering fractures; hence, administration of vitamin D for treating fractures in the elderly could actually promote fusion impairment and, consequently, severe defects in muscle regeneration. Therefore, our results suggest that vitamin D replacement therapy should be used for prevention of age-related muscle loss, rather than for treatment of sarcopenia.
Collapse
|
40
|
Murach KA, McCarthy JJ, Peterson CA, Dungan CM. Making Mice Mighty: recent advances in translational models of load-induced muscle hypertrophy. J Appl Physiol (1985) 2020; 129:516-521. [PMID: 32673155 DOI: 10.1152/japplphysiol.00319.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The ability to genetically manipulate mice allows for gain- and loss-of-function in vivo, making them an ideal model for elucidating mechanisms of skeletal muscle mass regulation. Combining genetic models with mechanical muscle loading enables identification of specific factors involved in the hypertrophic response as well as the ability to test the requirement of those factors for adaptation, thereby informing performance and therapeutic interventions. Until recently, approaches for inducing mechanically mediated muscle hypertrophy (i.e., resistance-training analogs) have been limited and considered "nontranslatable" to humans. This mini-review outlines recent translational advances in loading-mediated strategies for inducing muscle hypertrophy in mice, and highlights the advantages and disadvantages of each method. The skeletal muscle field is poised for new breakthroughs in understanding mechanisms regulating load-induced muscle growth given the numerous murine tools that have very recently been described.
Collapse
Affiliation(s)
- Kevin A Murach
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Cory M Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
41
|
Masschelein E, D'Hulst G, Zvick J, Hinte L, Soro-Arnaiz I, Gorski T, von Meyenn F, Bar-Nur O, De Bock K. Exercise promotes satellite cell contribution to myofibers in a load-dependent manner. Skelet Muscle 2020; 10:21. [PMID: 32646489 PMCID: PMC7346400 DOI: 10.1186/s13395-020-00237-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/15/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Satellite cells (SCs) are required for muscle repair following injury and are involved in muscle remodeling upon muscular contractions. Exercise stimulates SC accumulation and myonuclear accretion. To what extent exercise training at different mechanical loads drive SC contribution to myonuclei however is unknown. RESULTS By performing SC fate tracing experiments, we show that 8 weeks of voluntary wheel running increased SC contribution to myofibers in mouse plantar flexor muscles in a load-dependent, but fiber type-independent manner. Increased SC fusion however was not exclusively linked to muscle hypertrophy as wheel running without external load substantially increased SC fusion in the absence of fiber hypertrophy. Due to nuclear propagation, nuclear fluorescent fate tracing mouse models were inadequate to quantify SC contribution to myonuclei. Ultimately, by performing fate tracing at the DNA level, we show that SC contribution mirrors myonuclear accretion during exercise. CONCLUSIONS Collectively, mechanical load during exercise independently promotes SC contribution to existing myofibers. Also, due to propagation of nuclear fluorescent reporter proteins, our data warrant caution for the use of existing reporter mouse models for the quantitative evaluation of satellite cell contribution to myonuclei.
Collapse
Affiliation(s)
- Evi Masschelein
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Gommaar D'Hulst
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Joel Zvick
- Department Health Sciences and Technology, Laboratory of Regenerative and Movement Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Laura Hinte
- Department Health Sciences and Technology, Laboratory of Nutrition and Metabolic Epigenetics, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Inés Soro-Arnaiz
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Tatiane Gorski
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ferdinand von Meyenn
- Department Health Sciences and Technology, Laboratory of Nutrition and Metabolic Epigenetics, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ori Bar-Nur
- Department Health Sciences and Technology, Laboratory of Regenerative and Movement Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Katrien De Bock
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
| |
Collapse
|
42
|
Murach KA, Vechetti IJ, Van Pelt DW, Crow SE, Dungan CM, Figueiredo VC, Kosmac K, Fu X, Richards CI, Fry CS, McCarthy JJ, Peterson CA. Fusion-Independent Satellite Cell Communication to Muscle Fibers During Load-Induced Hypertrophy. FUNCTION 2020; 1:zqaa009. [PMID: 32864621 PMCID: PMC7448100 DOI: 10.1093/function/zqaa009] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 01/06/2023] Open
Abstract
The "canonical" function of Pax7+ muscle stem cells (satellite cells) during hypertrophic growth of adult muscle fibers is myonuclear donation via fusion to support increased transcriptional output. In recent years, however, emerging evidence suggests that satellite cells play an important secretory role in promoting load-mediated growth. Utilizing genetically modified mouse models of delayed satellite cell fusion and in vivo extracellular vesicle (EV) tracking, we provide evidence for satellite cell communication to muscle fibers during hypertrophy. Myogenic progenitor cell-EV-mediated communication to myotubes in vitro influences extracellular matrix (ECM)-related gene expression, which is congruent with in vivo overload experiments involving satellite cell depletion, as well as in silico analyses. Satellite cell-derived EVs can transfer a Cre-induced, cytoplasmic-localized fluorescent reporter to muscle cells as well as microRNAs that regulate ECM genes such as matrix metalloproteinase 9 (Mmp9), which may facilitate growth. Delayed satellite cell fusion did not limit long-term load-induced muscle hypertrophy indicating that early fusion-independent communication from satellite cells to muscle fibers is an underappreciated aspect of satellite cell biology. We cannot exclude the possibility that satellite cell-mediated myonuclear accretion is necessary to maintain prolonged growth, specifically in the later phases of adaptation, but these data collectively highlight how EV delivery from satellite cells can directly contribute to mechanical load-induced muscle fiber hypertrophy, independent of cell fusion to the fiber.
Collapse
Affiliation(s)
- Kevin A Murach
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Ivan J Vechetti
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Douglas W Van Pelt
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Samuel E Crow
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Cory M Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Vandre C Figueiredo
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Kate Kosmac
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Xu Fu
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher I Richards
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher S Fry
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
43
|
Fukada SI, Akimoto T, Sotiropoulos A. Role of damage and management in muscle hypertrophy: Different behaviors of muscle stem cells in regeneration and hypertrophy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118742. [PMID: 32417255 DOI: 10.1016/j.bbamcr.2020.118742] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/07/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
Skeletal muscle is a dynamic tissue with two unique abilities; one is its excellent regenerative ability, due to the activity of skeletal muscle-resident stem cells named muscle satellite cells (MuSCs); and the other is the adaptation of myofiber size in response to external stimulation, intrinsic factors, or physical activity, which is known as plasticity. Low physical activity and some disease conditions lead to the reduction of myofiber size, called atrophy, whereas hypertrophy refers to the increase in myofiber size induced by high physical activity or anabolic hormones/drugs. MuSCs are essential for generating new myofibers during regeneration and the increase in new myonuclei during hypertrophy; however, there has been little investigation of the molecular mechanisms underlying MuSC activation, proliferation, and differentiation during hypertrophy compared to those of regeneration. One reason is that 'degenerative damage' to myofibers during muscle injury or upon hypertrophy (especially overloaded muscle) is believed to trigger similar activation/proliferation of MuSCs. However, evidence suggests that degenerative damage of myofibers is not necessary for MuSC activation/proliferation during hypertrophy. When considering MuSC-based therapy for atrophy, including sarcopenia, it will be indispensable to elucidate MuSC behaviors in muscles that exhibit non-degenerative damage, because degenerated myofibers are not present in the atrophied muscles. In this review, we summarize recent findings concerning the relationship between MuSCs and hypertrophy, and discuss what remains to be discovered to inform the development and application of relevant treatments for muscle atrophy.
Collapse
Affiliation(s)
- So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
| | | | - Athanassia Sotiropoulos
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France
| |
Collapse
|
44
|
Zhu M, Chen G, Yang Y, Yang J, Qin B, Gu L. miR‑217‑5p regulates myogenesis in skeletal muscle stem cells by targeting FGFR2. Mol Med Rep 2020; 22:850-858. [PMID: 32626929 PMCID: PMC7339560 DOI: 10.3892/mmr.2020.11133] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/04/2020] [Indexed: 02/07/2023] Open
Abstract
MicroRNA-217-5p (miR-217-5p) has been implicated in cell proliferation; however, its role in skeletal muscle stem cells (SkMSCs) remains unknown. The present study aimed to explore the roles of miR‑217‑5p in the biological characteristics of SkMSCs. SkMSCs were identified by cell surface markers using flow cytometry. The present study observed that miR‑217‑5p mimics accelerated the proliferation and suppressed the differentiation in SkMSCs. In addition, the results of the present study revealed that fibroblast growth factor receptor 2 (FGFR2) was a target of miR‑217‑5p, as miR‑217‑5p bound directly to the 3'‑untranslated region of FGFR2 mRNA, resulting in increased FGFR2 mRNA and protein levels. In addition, the present study suppressed the expression of FGFR2 in SkMSCs using a selective FGFR inhibitor AZD4547 and detected the efficiency of inhibition by reverse transcription‑quantitative PCR and western blotting. miR‑217‑5p levels were positively associated with FGFR2 expression, which was upregulated and accelerated the proliferation of SkMSCs compared with that of the miR‑NC group. Collectively, these results demonstrated that miR‑217‑5p may act as a myogenesis promoter in SkMSCs by directly targeting FGFR2 and may regulate the myogenesis of these cells.
Collapse
Affiliation(s)
- Menghai Zhu
- Department of Orthopedic Trauma and Microsurgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Gang Chen
- Department of Orthopedic Trauma and Microsurgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yi Yang
- Department of Orthopedic Trauma and Microsurgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jiantao Yang
- Department of Orthopedic Trauma and Microsurgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Bengang Qin
- Department of Orthopedic Trauma and Microsurgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Liqiang Gu
- Department of Orthopedic Trauma and Microsurgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
45
|
Pakvasa M, Haravu P, Boachie-Mensah M, Jones A, Coalson E, Liao J, Zeng Z, Wu D, Qin K, Wu X, Luo H, Zhang J, Zhang M, He F, Mao Y, Zhang Y, Niu C, Wu M, Zhao X, Wang H, Huang L, Shi D, Liu Q, Ni N, Fu K, Lee MJ, Wolf JM, Athiviraham A, Ho SS, He TC, Hynes K, Strelzow J, El Dafrawy M, Reid RR. Notch signaling: Its essential roles in bone and craniofacial development. Genes Dis 2020; 8:8-24. [PMID: 33569510 PMCID: PMC7859553 DOI: 10.1016/j.gendis.2020.04.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/25/2020] [Accepted: 04/03/2020] [Indexed: 02/08/2023] Open
Abstract
Notch is a cell–cell signaling pathway that is involved in a host of activities including development, oncogenesis, skeletal homeostasis, and much more. More specifically, recent research has demonstrated the importance of Notch signaling in osteogenic differentiation, bone healing, and in the development of the skeleton. The craniofacial skeleton is complex and understanding its development has remained an important focus in biology. In this review we briefly summarize what recent research has revealed about Notch signaling and the current understanding of how the skeleton, skull, and face develop. We then discuss the crucial role that Notch plays in both craniofacial development and the skeletal system, and what importance it may play in the future.
Collapse
Affiliation(s)
- Mikhail Pakvasa
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA.,Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Pranav Haravu
- Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Michael Boachie-Mensah
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Alonzo Jones
- Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Elam Coalson
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Junyi Liao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Departments of Orthopaedic Surgery, Gastrointestinal Surgery, Obstetrics and Gynecology, and Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory and Diagnostic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Di Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Kevin Qin
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Xiaoxing Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Departments of Orthopaedic Surgery, Gastrointestinal Surgery, Obstetrics and Gynecology, and Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Huaxiu Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Jing Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Departments of Orthopaedic Surgery, Gastrointestinal Surgery, Obstetrics and Gynecology, and Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Meng Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Orthopaedic Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, PR China
| | - Fang He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Departments of Orthopaedic Surgery, Gastrointestinal Surgery, Obstetrics and Gynecology, and Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Yukun Mao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430072, PR China
| | - Yongtao Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266061, PR China
| | - Changchun Niu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Laboratory Diagnostic Medicine, Chongqing General Hospital, Chongqing, 400021, PR China
| | - Meng Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Institute of Bone and Joint Research, and the Department of Orthopaedic Surgery, The Second Hospitals of Lanzhou University, Gansu, Lanzhou, 730030, PR China
| | - Xia Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266061, PR China
| | - Hao Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory and Diagnostic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Linjuan Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Departments of Orthopaedic Surgery, Gastrointestinal Surgery, Obstetrics and Gynecology, and Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Deyao Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430072, PR China
| | - Qing Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Spine Surgery, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, PR China
| | - Na Ni
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory and Diagnostic Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Kai Fu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Departments of Orthopaedic Surgery and Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430072, PR China
| | - Michael J Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Sherwin S Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Kelly Hynes
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Mostafa El Dafrawy
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|