1
|
Nicholls A, Harris MB, Dewi L, Huang CY, Pang LN, Kung HJ, Chen LK, Kuo CH. Exercise-induced MyoD mRNA Expression in Young and Older Human Skeletal Muscle: A Systematic Review and Meta-Analysis. Sports Med 2025:10.1007/s40279-025-02207-4. [PMID: 40317450 DOI: 10.1007/s40279-025-02207-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Myoblast determination protein 1 (MyoD) is a master transcription factor that triggers myogenesis and drives muscle growth. OBJECTIVE The aim was to assess acute exercise-induced MyoD mRNA expression in skeletal muscle for young and older (age > 50) adults. DESIGN A meta-analysis and systematic review was conducted. METHODS A literature search was conducted for studies reporting MyoD mRNA changes in biopsied human muscle taken within 48 h after exercise. Fifty eligible studies with 822 participants (young 20-35 years; older 53-85 years) were included for meta-analysis. RESULTS Significant increases in MyoD mRNA expression in human skeletal muscle were observed 3-12 h post-exercise (standardized mean difference [SMD] = 1.39, p < 0.001), subsiding within 24-48 h (SMD = 0.47, p < 0.001). Older individuals showed a similar time pattern in MyoD mRNA expression post-exercise, but the response is weaker than in younger individuals. Intriguingly, resting levels of MyoD mRNA were higher in older individuals compared to younger individuals in most age-paired studies (SMD = 0.56, p < 0.01). Considering the decline in anabolic hormones during later life, this systematic review highlights age- and sex-related impacts on exercise-induced MyoD mRNA expression in human skeletal muscle, emphasizing the roles of sex hormones and insulin. CONCLUSION Pooled results from the eligible studies suggest a blunted exercise-induced increase in MyoD mRNA in skeletal muscle after age 50, likely due to elevated basal MyoD expression as a compensatory mechanism against persistent catabolic conditions in aging muscle. PROTOCOL REGISTRATION Registration number: CRD42023471840 (PROSPERO).
Collapse
Affiliation(s)
- Andrew Nicholls
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, 11153, Taiwan
| | - M Brennan Harris
- Department of Kinesiology and Health Science, College of William and Mary, Williamsburg, VA, USA
| | - Luthfia Dewi
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, 11153, Taiwan
- Department of Nutrition, Universitas Muhammadiyah Semarang, Semarang, 50273, Indonesia
| | - Chih-Yang Huang
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Cardiovascular and Mitochondria Related Disease Research Center, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Li-Ning Pang
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsing-Jien Kung
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Liang-Kung Chen
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Taipei Municipal Gan-Dau Hospital, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, 11153, Taiwan.
- Department of Kinesiology and Health Science, College of William and Mary, Williamsburg, VA, USA.
- School of Physical Education and Sports Science, Soochow University, Suzhou, China.
- Laboratory of Exercise Biochemistry, Education University of Hong Kong, Hong Kong, China.
| |
Collapse
|
2
|
Zhao X, Liu Y, Wang D, Li T, Xu Z, Li Z, Bai X, Wang Y. Role of GLP‑1 receptor agonists in sepsis and their therapeutic potential in sepsis‑induced muscle atrophy (Review). Int J Mol Med 2025; 55:74. [PMID: 40052580 PMCID: PMC11936484 DOI: 10.3892/ijmm.2025.5515] [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: 11/22/2024] [Accepted: 01/17/2025] [Indexed: 03/27/2025] Open
Abstract
Sepsis‑induced myopathy (SIM) is a common complication in intensive care units, which is often associated with adverse outcomes, primarily manifested as skeletal muscle weakness and atrophy. Currently, the management of SIM focuses on prevention strategies, as effective therapeutic options remain elusive. Glucagon‑like peptide‑1 (GLP‑1) receptor agonists (GLP‑1RAs) have garnered attention as hypoglycemic and weight‑loss agents, with an increasing body of research focusing on the extrapancreatic effects of GLP‑1. In preclinical settings, GLP‑1RAs exert protective effects against sepsis‑related multiple organ dysfunction through anti‑inflammatory and antioxidant mechanisms. Based on the existing research, we hypothesized that GLP‑1RAs may serve potential protective roles in the repair and regeneration of skeletal muscle affected by sepsis. The present review aimed to explore the relationship between GLP‑1RAs and sepsis, as well as their impact on muscle atrophy‑related myopathy. Furthermore, the potential mechanisms and therapeutic benefits of GLP‑1RAs are discussed in the context of muscle atrophy induced by sepsis.
Collapse
Affiliation(s)
- Xuan Zhao
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Dongfang Wang
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Tonghan Li
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhikai Xu
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhanfei Li
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiangjun Bai
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yuchang Wang
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
3
|
TIAN Y, BU H, WANG T, YANG D, ZHANG W, LIU T, ZHANG L, HUO Z. Efficacy of electro-acupuncture at "Weizhong" (BL40) on macrophage polarization in rats with injured lumbar multifidus. J TRADIT CHIN MED 2025; 45:335-347. [PMID: 40151120 PMCID: PMC11955755 DOI: 10.19852/j.cnki.jtcm.20220419.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/17/2022] [Indexed: 03/29/2025]
Abstract
OBJECTIVE To investigate the anti-inflammatory effect of electroacupuncture in rats with bupivacaine-induced lumbar multifidus injury and its underlying regulatory mechanism on macrophage polarization. METHODS A total of seventy-two Sprague-Dawley male rats were randomly divided into control, model, and electroacupuncture groups. Forty-eight rats categorized in model groups were injected 0.5% bupivacaine (BPVC) into the lumbar multifidus at the L4-L5 segment. Rats in the electroacupuncture groups received the intervention for 1, 2, 3 and 5 d, respectively. The degree of macrophage infiltration and change of M1/M2 polarization were observed based on hematoxylin and eosin staining, immunohistochemistry and immunofluorescence to evaluate the anti-inflammatory effect of electroacupuncture. Meanwhile, exosomal miRNA-sequencing and bioinformatics analysis predicted the pathways and biological processes related to inflammatory response and macrophage polarization regulated by electroacupuncture intervention. RESULTS BPVC injection induced the infiltration of local macrophages at the L4-L5 segment of lumbar multifidus. Comparison of mean IOD values with 2 d and 5 d post injury revealed the highest expression of CD68+ macrophages on day 3 post injury by immunohistochemistry. (P < 0.001, P < 0.001, respectively). Compared with the model group, the cell counts of iNOs+ CD68+ M1-macrophages were lower in the electroacupuncture group, while the positive percent of CD163+ CD206+ M2-macrophages was higher in the electroacupuncture group, on day 3 after BPVC injection (P < 0.001, P < 0.001, respectively). Moreover, the results of sequencing and bioinformatic analysis suggested that exosomal miRNAs were involved in the EA regulating macrophage polarization. CONCLUSIONS Electroacupuncture can promote macrophage polarization to reduce inflammation following lumbar multifidus muscular injury.
Collapse
Affiliation(s)
- Yuan TIAN
- 1 Department of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
- 2 Department of Acupuncture-Moxibustion and Tuina, the Second Affiliated Hospital of Baotou Medical College, Inner Mongolia Autonomous Region, Baotou 014030, China
| | - He BU
- 2 Department of Acupuncture-Moxibustion and Tuina, the Second Affiliated Hospital of Baotou Medical College, Inner Mongolia Autonomous Region, Baotou 014030, China
| | - Tieshan WANG
- 3 Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | | | - Wei ZHANG
- 7 Department of Pathology, the First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Inner Mongolia 014010, China
| | - Tong LIU
- 5 the Fifth College of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou 510006, China
- 6 Department of Acupuncture and Rehabilitation, Guangdong Second Hospital of Traditional Chinese Medicine, Guangzhou 510095, China
| | - Li ZHANG
- 1 Department of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zejun HUO
- 8 Department of Chinese Medicine, Peking University 3rd Hospital, Beijing 100191, China
| |
Collapse
|
4
|
Li Y, Kawamura G, Dong Q, Li Q, Ozawa T. Near-Infrared Bioluminescence Assays for Protein-Protein Interactions and Cellular Membrane Fusion in Deep Tissues Using Split Akaluc Reconstitution. Anal Chem 2025; 97:6182-6191. [PMID: 40062634 DOI: 10.1021/acs.analchem.4c06986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Bioluminescence analysis using luciferase is an essential tool for studying biological processes in different cells. Split luciferase reconstitution is a technique that enables the analysis of biological events through the monitoring of protein-protein interactions. However, effective detection of cellular events in vivo remains challenging due to the limitation of light penetration into deep tissues and optical sensitivity. To address this, we developed a novel split luciferase reconstitution method using a near-infrared-emitting luciferase, Akaluc, and applied it to monitor two important biological events: G protein-coupled receptor (GPCR)/β-arrestin interactions and myogenic cell fusion in vivo. The developed split Akaluc reconstitution system demonstrated high sensitivity in detecting GPCR/β-arrestin interactions as well as myogenic cell fusion in vitro, enabling real-time insights into their temporal dynamics. Moreover, in vivo bioluminescence imaging successfully monitored GPCR/β-arrestin interactions in the mouse lung and the progression of myogenesis during mouse leg muscle regeneration. The split Akaluc reconstitution method will be a versatile tool for both in vitro and in vivo analyses of protein-protein interactions and cell fusion events. This system holds significant potential for advancing drug development, especially in the screening of GPCR-targeted therapeutic and myogenesis-promoting compounds in animal models.
Collapse
Affiliation(s)
- Yiling Li
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Genki Kawamura
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Qi Dong
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Qiaojing Li
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
5
|
Zhou J, Wu Y, Liu H, Tian W, Castanon RG, Bartlett A, Zhang Z, Yao G, Shi D, Clock B, Marcotte S, Nery JR, Liem M, Claffey N, Boggeman L, Barragan C, Drigo RAE, Weimer AK, Shi M, Cooper-Knock J, Zhang S, Snyder MP, Preissl S, Ren B, O’Connor C, Chen S, Luo C, Dixon JR, Ecker JR. Human Body Single-Cell Atlas of 3D Genome Organization and DNA Methylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.23.644697. [PMID: 40196612 PMCID: PMC11974725 DOI: 10.1101/2025.03.23.644697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Higher-order chromatin structure and DNA methylation are critical for gene regulation, but how these vary across the human body remains unclear. We performed multi-omic profiling of 3D genome structure and DNA methylation for 86,689 single nuclei across 16 human tissues, identifying 35 major and 206 cell subtypes. We revealed extensive changes in CG and non-CG methylation across almost all cell types and characterized 3D chromatin structure at an unprecedented cellular resolution. Intriguingly, extensive discrepancies exist between cell types delineated by DNA methylation and genome structure, indicating that the role of distinct epigenomic features in maintaining cell identity may vary by lineage. This study expands our understanding of the diversity of DNA methylation and chromatin structure and offers an extensive reference for exploring gene regulation in human health and disease.
Collapse
Affiliation(s)
- Jingtian Zhou
- Arc Institute, Palo Alto, CA, USA
- Genomic Analysis Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA, USA
| | - Yue Wu
- Genomic Analysis Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Hanqing Liu
- Genomic Analysis Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- Society of Fellows, Harvard University, Cambridge, MA, USA
| | - Wei Tian
- Genomic Analysis Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Rosa G Castanon
- Genomic Analysis Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Anna Bartlett
- Genomic Analysis Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zuolong Zhang
- School of Software, Henan University, Kaifeng, Henan, China
| | - Guocong Yao
- School of Computer and Information Engineering, Henan University, Kaifeng, Henan, China
| | - Dengxiaoyu Shi
- Genomic Analysis Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ben Clock
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Samantha Marcotte
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Joseph R. Nery
- Genomic Analysis Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michelle Liem
- Flow Cytometry Core Facility, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Naomi Claffey
- Flow Cytometry Core Facility, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Lara Boggeman
- Flow Cytometry Core Facility, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Cesar Barragan
- Genomic Analysis Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Rafael Arrojo e Drigo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Center for Computational Systems Biology, Vanderbilt University, Nashville, TN
- Diabetes Research and Training Center (DRTC), Vanderbilt University Medical Center, Nashville, TN, 37235
| | - Annika K. Weimer
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Minyi Shi
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Sai Zhang
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
- Departments of Biostatistics & Biomedical Engineering, Genetics Institute, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Sebastian Preissl
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Pharmaceutical Sciences, Pharmacology & Toxicology, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Bing Ren
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Carolyn O’Connor
- Flow Cytometry Core Facility, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Shengbo Chen
- School of Software, Nanchang University, Nanchang, Jiangxi, China
| | - Chongyuan Luo
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
| | - Jesse R. Dixon
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Joseph R. Ecker
- Genomic Analysis Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA, USA
| |
Collapse
|
6
|
Egerman MA, Zhang Y, Donne R, Xu J, Gadi A, McEwen C, Salmon H, Xiong K, Bai Y, Germino M, Barringer K, Jimenez Y, Del Pilar Molina-Portela M, Shavlakadze T, Glass DJ. ActRII or BMPR ligands inhibit skeletal myoblast differentiation, and BMPs promote heterotopic ossification in skeletal muscles in mice. Skelet Muscle 2025; 15:4. [PMID: 39994804 PMCID: PMC11853584 DOI: 10.1186/s13395-025-00373-7] [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: 07/17/2024] [Accepted: 01/26/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Prior studies suggested that canonical Activin Receptor II (ActRII) and BMP receptor (BMPR) ligands can have opposing, distinct effects on skeletal muscle depending in part on differential downstream SMAD activation. It was therefore of interest to test ActRII ligands versus BMP ligands in settings of muscle differentiation and in vivo. METHODS AND RESULTS In human skeletal muscle cells, both ActRII ligands and BMP ligands inhibited myogenic differentiation: ActRII ligands in a SMAD2/3-dependent manner, and BMP ligands via SMAD1/5. Surprisingly, a neutralizing ActRIIA/B antibody mitigated the negative effects of both classes of ligands, indicating that some BMPs act at least partially through the ActRII receptors in skeletal muscle. Gene expression analysis showed that both ActRII and BMP ligands repress muscle differentiation genes in human myoblasts and myotubes. In mice, hepatic BMP9 over-expression induced liver toxicity, caused multi-organ wasting, and promoted a pro-atrophy gene signature despite elevated SMAD1/5 signaling in skeletal muscle. Local overexpression of BMP7 or BMP9, achieved by intramuscular AAV delivery, induced heterotopic ossification. Elevated SMAD1/5 signaling with increased expression of BMP target genes was also observed in sarcopenic muscles of old rats. CONCLUSIONS The canonical ActRII ligand-SMAD2/3 and BMP ligand-SMAD1/5 axes can both block human myoblast differentiation. Our observations further demonstrate the osteoinductive function of BMP ligands while pointing to a potential relevancy of blocking the BMP-SMAD1/5 axis in the setting of therapeutic anti-ActRIIA/B inhibition.
Collapse
Affiliation(s)
- Marc A Egerman
- Aging/Age-Related Disorders, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Yuhong Zhang
- Aging/Age-Related Disorders, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Romain Donne
- Aging/Age-Related Disorders, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Jianing Xu
- Aging/Age-Related Disorders, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Abhilash Gadi
- Aging/Age-Related Disorders, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Corissa McEwen
- Aging/Age-Related Disorders, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Hunter Salmon
- Aging/Age-Related Disorders, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Kun Xiong
- Molecular Profiling, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Yu Bai
- Molecular Profiling, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Mary Germino
- Imaging Sciences, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Kevin Barringer
- Inflammation & Immune Diseases, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Yasalp Jimenez
- Inflammation & Immune Diseases, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | - Tea Shavlakadze
- Aging/Age-Related Disorders, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - David J Glass
- Aging/Age-Related Disorders, 777 Old Saw Mill River Road, Regeneron Pharmaceuticals, Tarrytown, NY, USA.
| |
Collapse
|
7
|
Guo L, Jin K, Sun Q, Zhang C, Chen X, Geng Z. Adiponectin regulates proliferation and differentiation of chicken skeletal muscle satellite cells via ERK1/2 and p38 signaling pathways. Poult Sci 2025; 104:104813. [PMID: 39823838 PMCID: PMC11786077 DOI: 10.1016/j.psj.2025.104813] [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: 11/14/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025] Open
Abstract
Skeletal muscle satellite cells (SMSCs) are critical for postnatal skeletal muscle growth and regeneration. Adiponectin plays a pivotal role in regulating muscle glucose uptake and fatty acid metabolism. However, its function in the proliferation and differentiation of chicken SMSCs remains poorly understood. In this study, we investigated the effects of adiponectin on the proliferation and differentiation of in vitro cultured chicken SMSCs. Our results demonstrated that adiponectin promoted SMSCs proliferation while inhibiting myogenic differentiation and inducing adipogenic differentiation. RNA-seq analysis revealed enrichment of the MAPK signaling pathway, suggesting its potential involvement in the regulation of adiponectin on SMSCs activity. Western blot analysis revealed that adiponectin activated ERK1/2 phosphorylation and inhibited p38 phosphorylation during the process of the inhibition on myogenic differentiation in chicken SMSCs. Furthermore, suppression of ERK1/2 signaling with U0126 or activation of p38 signaling with SSK1 reversed the downregulated expression of myogenic differentiation marker MyHC, MyOD1, and MyOG induced by adiponectin. These findings validated that adiponectin impeded myogenic differentiation through activation of ERK1/2 and inhibition of p38 signaling pathways. Additionally, activation of p38 signaling pathway reduced the increased percentage of EdU-positive cells induced by adiponectin. Collectively, these findings demonstrated that adiponectin impedes myogenic differentiation of SMSCs through activating ERK1/2 and inhibiting p38 signaling pathways, while promoting proliferation by inhibiting p38 signaling pathway.
Collapse
Affiliation(s)
- Liping Guo
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, 130 Changjiang West Rd., Hefei 230036, PR China
| | - Kaiming Jin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Qi Sun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Chenchao Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Xiongyong Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, 130 Changjiang West Rd., Hefei 230036, PR China.
| | - Zhaoyu Geng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, 130 Changjiang West Rd., Hefei 230036, PR China
| |
Collapse
|
8
|
Cruz VA, Marques RS, Kvamme K, Limede AC, Cidrini FAA, Cidrini IA, dos Santos Nascimento K, Mackey SJ, Cooke RF, Farmer C, Heldt J. Effects of maternal Cu, Mn, and Zn supplementation from different sources on physiological and productive responses of cows and their offspring. J Anim Sci 2025; 103:skae391. [PMID: 39742412 PMCID: PMC11725650 DOI: 10.1093/jas/skae391] [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: 09/30/2024] [Accepted: 12/31/2024] [Indexed: 01/03/2025] Open
Abstract
Seventy-two nonlactating, pregnant Angus cows (initial body weight [BW] = 637 ± 13 kg; body condition score [BCS] = 5.5 ± 0.07 yr; and age = 6.1 ± 0.57; exposed to artificial insemination = 54 and bull = 18) were ranked by pregnancy type, BW, and BCS and assigned to receive a supplement containing: 1) Cu, Mn, and Zn sulfate source (INR; n = 24), 2) Cu, Mn, and Zn organic-complexed source (ORG; n = 24), or 3) Cu, Mn, and Zn hydroxychloride source (HDX; n = 24). From day 11 (days of gestation 195 ± 5.1) to calving, cows were allocated to rangeland pasture. From day 0 until calving, cows were gathered and fed the treatments thrice a week. Cow BW and BCS were recorded, and blood was collected on days 11 and 10, upon calving, and at weaning. Liver biopsies were performed in all cows on day 10 and upon calving (cows and calves). Longissimus muscle (LM) biopsies were performed, and blood was collected in all calves upon calving. Calves were weaned on day 260, backgrounded for 99 d, and then sent to a commercial feedyard. Calves blood samples were collected on days 245, 260, 264, 268, 275, 280, and 288. No differences were detected (P ≥ 0.31) for cow BW and BCS changes among treatments during gestation, and mineral sources did not improve (P ≥ 0.16) mineral liver concentrations of Zn, Mn, Se, and Co at parturition. However, there was a tendency (P = 0.07) for HDX to have improved liver Cu concentration vs. ORG and INR cows. Cows fed HDX and ORG had improved (P ≤ 0.03) BCS at weaning, and BCS changed from parturition to weaning vs. INR cows. No treatment differences were detected (P ≥ 0.21) for calf birth measurements, IgG levels, mRNA expression of hepatic enzymes, or LM genes associated with muscle and adipose tissue development. At weaning and during the backgrounding, no treatment differences were detected (P ≥ 0.21) for offspring performance, health outcomes, plasma cortisol, serum nonesterified fatty acids, and serum β-hydroxybutyrate. However, a tendency for a treatment × day interaction was detected (P = 0.07) for haptoglobin concentrations, which was reduced (P < 0.01) in calves from cows supplemented with HDX vs. calves from cows supplemented with ORG and INR 15 d after weaning. No treatment effects were noted (P ≥ 0.35) for final BW, feedyard average daily gain, and carcass traits between treatment groups. Overall, different sources of Cu, Mn, and Zn supplemented to late-gestating beef cows yielded similar cow-calf productive responses from birth to finishing.
Collapse
Affiliation(s)
- Vinicius Alves Cruz
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Rodrigo S Marques
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Keenan Kvamme
- Department of Animal and Range Sciences, Montana State University, Bozeman, MT 59717, USA
| | - Arnaldo Cintra Limede
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | | | | | | | - Shea J Mackey
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
| | - Reinaldo F Cooke
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | | | | |
Collapse
|
9
|
Cruz VA, Marques RS, Kvamme K, Limede AC, Cidrini FAA, Cidrini IA, Nascimento KS, Harvey KM, Llarena JFC, Cooke RF. Effects of omega-3 polyunsaturated fatty acid supplementation to first-calf beef heifers during late gestation on offspring physiology and performance. J Anim Sci 2025; 103:skaf091. [PMID: 40126062 PMCID: PMC12032581 DOI: 10.1093/jas/skaf091] [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: 01/31/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025] Open
Abstract
Forty-four pregnant Angus first-calf heifers (initial body weight [BW] = 490 ± 8 kg, body condition score [BCS] = 5.4 ± 0.06) were selected and assigned to the experiment at the end of their second trimester of gestation. From day 0 until calving, first-calf heifers were gathered and individually fed 3 times a week with dry distillers' grain (DDG; 3.5 kg/heifer per feeding; as-fed basis) + treatments. Treatments were 1) 234 g/heifer per feeding of Ca salts of PUFA based on eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids (OMG, Strata; Virtus Nutrition LLC, Corcoran, CA; n = 22), or 2) 234 g/heifer per feeding of Ca salts of saturated and monosaturated fatty acids based on palmitic and oleic acids (CON; EnerGII, Virtus Nutrition; n = 22). From day -11 (d of gestation 192 ± 5.1) to calving, heifers were allocated to rangeland pasture. First-calf heifer BW and BCS were recorded (days -10 and -9), and blood was collected on day -10, and upon calving. Calves were weaned on day 260, and preconditioned from days 260 to 302, and feedlot was received from days 303 to 350. No differences were detected (P ≥ 0.33) for heifer BW or BCS changes at calving. A tendency for greater concentration of colostrum IgG (P = 0.08) was observed in heifers supplemented with OMG vs. CON cohorts. There were no differences (P ≥ 0.31) among treatments at calving for calving rate, birth BW, heart girth, and % of male calves born. Calves from OMG heifers had greater (P = 0.04) plasma IgG concentration compared to CON calves. No differences were observed (P ≥ 0.11) for weaning rate and age, birth to weaning average daily gain (ADG), weaning weight, liver enzymes, and serum antibodies against respiratory viruses. During the preconditioning, OMG calves had greater (P ≤ 0.05) final BW and ADG compared with calves born to CON heifers. These differences in performance remained (P ≤ 0.05) throughout the receiving phase. A treatment × day interaction was detected (P = 0.05) for plasma cortisol concentration, which was greater (P < 0.01) for calves born to OMG first-calf heifers on days 260 and 263 and lower (P < 0.01) on day 306 compared to CON cohorts. Supplementing omega-3 fatty acids to late-gestating first-calf beef heifers enhances IgG concentrations in both heifers and their offspring. While offspring growth from birth to weaning remained unaffected, OMG calves demonstrated increased weight during the preconditioning and receiving phases, suggesting potential long-term growth benefits.
Collapse
Affiliation(s)
- Vinicius A Cruz
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Rodrigo S Marques
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Keenan Kvamme
- Department of Animal and Range Sciences, Montana State University, Bozeman, MT 59717, USA
| | - Arnaldo Cintra Limede
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | | | - Iorrano Andrade Cidrini
- Universidade Estadual Paulista “Júlio de Mesquita Filho” (UNESP), Jaboticabal, São Paulo, Brazil
| | - Keuven S Nascimento
- Universidade Estadual Paulista “Júlio de Mesquita Filho” (UNESP), Jaboticabal, São Paulo, Brazil
| | - Kelsey M Harvey
- Prairie Research Unit, Mississippi State University, Prairie, MS 39756, USA
| | | | - Reinaldo F Cooke
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
| |
Collapse
|
10
|
Hogarth MW, Kurukunda MP, Ismat K, Uapinyoying P, Jaiswal JK. Exploring the therapeutic potential of fibroadipogenic progenitors in muscle disease. J Neuromuscul Dis 2025; 12:22143602241298545. [PMID: 39973455 PMCID: PMC11949306 DOI: 10.1177/22143602241298545] [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] [Indexed: 02/21/2025]
Abstract
Skeletal muscle relies on its inherent self-repair ability to withstand continuous mechanical damage. Myofiber-intrinsic processes facilitate the repair of damage to sarcolemma and sarcomeres, but it is the coordinated interaction between muscle-resident satellite and stromal cells that are crucial in the regeneration of muscles to replace the lost muscle fibers. Fibroadipogenic progenitors (FAPs), are muscle-resident mesenchymal cells that are notable for their role in creating the dynamic stromal niche required to support long-term muscle homeostasis and regeneration. While FAP-mediated extracellular matrix formation and the establishment of a homeostatic muscle niche are essential for maintaining muscle health, excessive accumulation of FAPs and their aberrant differentiation leads to the fibrofatty degeneration that is a hallmark of myopathies and muscular dystrophies. Recent advancements, including single-cell RNA sequencing and in vivo analysis of FAPs, are providing deeper insights into the functions and specialization of FAPs, shedding light on their roles in both health and disease. This review will explore the above insights, discussing how FAP dysregulation contributes to muscle diseases. It will offer a concise overview of potential therapeutic interventions targeting FAPs to restore disrupted interactions among FAPs and muscle-resident cells, ultimately addressing degenerative muscle loss in neuromuscular diseases.
Collapse
Affiliation(s)
- Marshall W Hogarth
- Center for Genetic Medicine Research, Children’s National Research Institute, Washington, DC, U.S.A
| | - Medha P Kurukunda
- Center for Genetic Medicine Research, Children’s National Research Institute, Washington, DC, U.S.A
| | - Karim Ismat
- Center for Genetic Medicine Research, Children’s National Research Institute, Washington, DC, U.S.A
| | - Prech Uapinyoying
- Center for Genetic Medicine Research, Children’s National Research Institute, Washington, DC, U.S.A
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, U.S.A
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children’s National Research Institute, Washington, DC, U.S.A
- Department of Genomics and Precision Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, U.S.A
| |
Collapse
|
11
|
Feng S, Zhou H, Lin X, Zhu S, Chen H, Zhou H, Wang R, Wang P, Shao X, Wang J. Exercise promotes skeletal muscle growth in adolescents via modulating Mettl3-mediated m6A methylation of MyoD in muscle satellite cells. Cell Mol Biol Lett 2024; 29:150. [PMID: 39633280 PMCID: PMC11616192 DOI: 10.1186/s11658-024-00670-x] [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: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Exercise exerts positive impacts on skeletal muscle health and homeostasis. Emerging evidence suggests that m6A methylation is involved in various physiological processes. However, the impact of exercise on adolescent skeletal muscle growth and the underlying epigenetic mechanisms remain poorly understood. METHODS The lower-limb skeletal muscles were harvested from exercise and control groups to compare the skeletal muscle growth in adolescents. mRNA sequencing was conducted to explore the mechanisms underlying enhanced skeletal muscle growth following exercise. The effects and mechanisms of Mettl3-mediated m6A methylation on adolescent skeletal muscle growth were investigated using muscle satellite cell (MuSC)-specific Mettl3 knockout (KO) mice. The potential function of MyoD for skeletal muscle growth in adolescents was explored by phenotypes after overexpression and evaluation of in vivo myogenesis. Additionally, the effects of the methyl donor betaine on adolescent skeletal muscle growth were investigated in vitro and in vivo. RESULTS Exercise could promote skeletal muscle growth in adolescents. Sequencing data analysis and confirmation assays uncovered that exercise significantly increased Mettl3-mediated m6A methylation and elevated the expression levels of activation marker MyoD in MuSCs. Establishment of MuSC-specific Mettl3 KO mice further demonstrated that Mettl3-mediated m6A methylation in MyoD contributed to skeletal muscle growth during adolescence. Mettl3-mediated m6A methylation regulated MyoD mRNA stability at the posttranscriptional level in MuSCs, with a functional site at 234 bp A. Increased expression of MyoD could contribute to myogenesis of adolescent MuSCs. Furthermore, the methyl donor betaine could enhance MyoD expression, contributing to MuSCs activation and skeletal muscle growth in adolescents by boosting m6A methylation levels. CONCLUSIONS Exercise promoted skeletal muscle growth in adolescents through facilitating MyoD mRNA stability of MuSCs in a Mettl3-mediated m6A-dependent manner. The methyl donor betaine could be a potential alternative to exercise for promoting adolescent skeletal muscle growth by directly augmenting the global levels of m6A methylation. These findings may provide a theoretical foundation for encouraging daily fitness exercise and ensuring healthy growth in adolescents.
Collapse
Affiliation(s)
- Shujing Feng
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Hao Zhou
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingzuan Lin
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
| | - Siyuan Zhu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Huifang Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Han Zhou
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Peng Wang
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiexiang Shao
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jianhua Wang
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
12
|
Zhe Y, Wu Z, Yasenjian S, Zhong J, Jiang H, Zhang M, Chai Z, Xin J. Effect of NR1D1 on the proliferation and differentiation of yak skeletal muscle satellite cells. Front Vet Sci 2024; 11:1428117. [PMID: 39559540 PMCID: PMC11571325 DOI: 10.3389/fvets.2024.1428117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024] Open
Abstract
The severe conditions at high altitudes, where yaks inhabit, contribute to delayed muscular growth and compromised tenderness of their muscle tissue. Myosatellite cells are responsible for the growth and regeneration of skeletal muscle after birth and have the potential to proliferate and differentiate, its development is closely related to meat quality, and the nuclear receptor gene NR1D1 is involved in muscle formation and skeletal muscle regulation. Therefore, in order to understand the effect of NR1D1 on muscle satellite cells, we identified the mRNA expression levels of marker genes specifically expressed in muscle satellite cells at different stages to determine the type of cells isolated. Eventually, we successfully constructed a primary cell line of yak muscle satellite cells. Then we constructed NR1D1 overexpression vector and interference RNA, and introduced them into isolated yak skeletal muscle satellite cells. We performed qPCR, CCK8, and fluorescence-specific to detect the expression of genes or abundance of proteins as markers of cell proliferation and differentiation. Compared with those in the control group, the expression levels of proliferation marker genes KI-67, CYCLIND1, and CYCLINA were significantly inhibited after NR1D1 overexpression, which was also supported by the CCK-8 test, whereas differentiation marker genes MYOD, MYOG, and MYF5 were significantly inhibited. Fluorescence-specific staining showed that KI-67 protein abundance and the number of microfilaments both decreased, while the opposite trend was observed after NR1D1 interference. In conclusion, we confirmed that NR1D1 inhibited the proliferation and differentiation of yak skeletal muscle satellite cells, which provides a theoretical basis for further research on the effect of NR1D1 on improving meat quality traits and meat production performance of yaks.
Collapse
Affiliation(s)
- Yuqi Zhe
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Zhijuan Wu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Sibinuer Yasenjian
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Hui Jiang
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Institute of Animal Science and Veterinary Research, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Ming Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Zhixin Chai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Jinwei Xin
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Institute of Animal Science and Veterinary Research, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| |
Collapse
|
13
|
Lu Y, Walji T, Ravaux B, Pandey P, Yang C, Li B, Luvsanjav D, Lam KH, Zhang R, Luo Z, Zhou C, Habela CW, Snapper SB, Li R, Goldhamer DJ, Schmidtke DW, Pan D, Svitkina TM, Chen EH. Spatiotemporal coordination of actin regulators generates invasive protrusions in cell-cell fusion. Nat Cell Biol 2024; 26:1860-1877. [PMID: 39487253 DOI: 10.1038/s41556-024-01541-5] [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: 06/26/2023] [Accepted: 09/11/2024] [Indexed: 11/04/2024]
Abstract
Invasive membrane protrusions play a central role in a variety of cellular processes. Unlike filopodia, invasive protrusions are mechanically stiff and propelled by branched actin polymerization. However, how branched actin filaments are organized to create finger-like invasive protrusions is unclear. Here, by examining the mammalian fusogenic synapse, where invasive protrusions are generated to promote cell membrane juxtaposition and fusion, we have uncovered the mechanism underlying invasive protrusion formation. We show that two nucleation-promoting factors for the Arp2/3 complex, WAVE and N-WASP, exhibit different localization patterns in the protrusions. Whereas WAVE is closely associated with the plasma membrane at the leading edge of the protrusive structures, N-WASP is enriched with WIP along the actin bundles in the shafts of the protrusions. During protrusion initiation and growth, the Arp2/3 complex nucleates branched actin filaments to generate low-density actin clouds in which the large GTPase dynamin organizes the new branched actin filaments into bundles, followed by actin-bundle stabilization by WIP, the latter functioning as an actin-bundling protein. Disruption of any of these components results in defective protrusions and failed myoblast fusion in cultured cells and mouse embryos. Together, our study has revealed the intricate spatiotemporal coordination between two nucleation-promoting factors and two actin-bundling proteins in building invasive protrusions at the mammalian fusogenic synapse and has general implications in understanding invasive protrusion formation in cellular processes beyond cell-cell fusion.
Collapse
Affiliation(s)
- Yue Lu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tezin Walji
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Benjamin Ravaux
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pratima Pandey
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Changsong Yang
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Bing Li
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Delgermaa Luvsanjav
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kevin H Lam
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Ruihui Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhou Luo
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chuanli Zhou
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christa W Habela
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Scott B Snapper
- Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Rong Li
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - David J Goldhamer
- Department of Molecular and Cell Biology, University of Connecticut Stem Cell Institute, Storrs, CT, USA
| | - David W Schmidtke
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth H Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
14
|
Zhang S, Lu H, Lou H, Shi Y, Liu D, Chen Q. An efficient serum-free medium for ex vivo expansion of myoblasts from Larimichthys crocea for cultured meat production. Food Res Int 2024; 196:115073. [PMID: 39614562 DOI: 10.1016/j.foodres.2024.115073] [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: 05/31/2024] [Revised: 07/31/2024] [Accepted: 09/09/2024] [Indexed: 12/01/2024]
Abstract
The large-scale production of cultured meat has been restricted by high cost of serum, batch difference and unclear composition, thus it is important to develop serum-free media for cell growth. Here we obtained a serum-free medium (SFM), which was suitable for ex vivo culturing of myoblasts from larimichthys crocea. It offered simple composition and high effectiveness that supporting myoblast attachment, expansion and differentiation. Our findings revealed that the optimal SFM consisted of DMEM/F12 base medium, 5 mg/mL bovine serum albumin (BSA), 20 mmol HEPES, 1 % penicillin/streptomycin mixture (PS) and the exogenous additive hydrocortisone. The living cells of SFM could occupy about 65 % of the serum-containing medium (SM). Additionally, differentiation assays showed that myoblasts cultured without serum still differentiated into more muscle tubes, with a higher fusion index than those cultured with serum. Finally, transcriptome sequencing was conducted to demonstrate that hydrocortisone might act as a ligand binding to G protein-coupled receptors, triggering the downstream PI3K/AKT signaling pathway and leading to cell proliferation. Our findings will provide a basis for large-scale cell expansion and cell culture meat production in the future.
Collapse
Affiliation(s)
- Shengliang Zhang
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Hongyun Lu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Hanghang Lou
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Ying Shi
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Donghong Liu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Qihe Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
15
|
Sun L, Yuan C, An X, Kong L, Zhang D, Chen B, Lu Z, Liu J. Delta-like noncanonical notch ligand 2 regulates the proliferation and differentiation of sheep myoblasts through the Wnt/β-catenin signaling pathway. J Cell Physiol 2024; 239:e31385. [PMID: 39030845 DOI: 10.1002/jcp.31385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/25/2024] [Accepted: 07/05/2024] [Indexed: 07/22/2024]
Abstract
This study delved into the role of delta-like noncanonical notch ligand 2 (DLK2) in the cell cycle, proliferation, apoptosis, and differentiation of myoblasts, as well as its interaction with the classical Wnt/β-catenin signaling pathway in regulating myoblast function. The research revealed that upregulation of DLK2 in myoblasts during the proliferation phase enhanced myoblast proliferation, facilitated cell cycle progression, and reduced apoptosis. Conversely, downregulation of DLK2 expression using siRNA during the differentiation phase promoted myoblast hypertrophy and fusion, suppressed the expression of muscle fiber degradation factors, and expedited the differentiation process. DLK2 regulates myoblasts function by influencing the expression of various factors associated with the Wnt/β-catenin signaling pathway, including CTNNB1, FZD1, FZD6, RSPO1, RSPO4, WNT4, WNT5A, and adenomatous polyposis coli. In essence, DLK2, with the involvement of the Wnt/β-catenin signaling pathway, plays a crucial regulatory role in the cell cycle, proliferation, apoptosis, and differentiation of myoblasts.
Collapse
Affiliation(s)
- Lixia Sun
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Chao Yuan
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xuejiao An
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Lingying Kong
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Dan Zhang
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Bowen Chen
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zengkui Lu
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jianbin Liu
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
- Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
16
|
Ong CZL, Nasir NJM, Welsch RE, Tucker-Kellogg L, Rajapakse JC. Quantification of muscle fiber malformations using edge detection to investigate chronic muscle pressure ulcers. FRONTIERS IN BIOINFORMATICS 2024; 4:1450146. [PMID: 39497739 PMCID: PMC11532102 DOI: 10.3389/fbinf.2024.1450146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/30/2024] [Indexed: 11/07/2024] Open
Abstract
Background Microscopy of regenerated tissue shows different morphologies between the healing of acute wounds and chronic wounds. This difference can be seen manually by biologists, but computational methods are needed to automate the characterization of morphology and regenerative quality in regenerated muscle tissue. Results From the detected edge segments, we computed several imaging biomarkers of interest, such as median tortuosity, number of edge segments normalized by area, median edge segment distance and interquartile range of orientation angles of edge segments of the microscope images of successful and unsuccessful muscle regeneration. We observed that muscle fibers in saline-treated pressure ulcers had a larger interquartile range of orientation angles of the edge segments (p = 0.05) and shorter edge segment distances (p = 0.003) compared to those of acute cardiotoxin injuries. Conclusion Our edge detection method was able to identify statistically significant differences in some of the imaging biomarkers between saline-treated pressure ulcers and cardiotoxin injuries, suggesting that chronic pressure ulcers have increased muscle fiber malformations compared to cardiotoxin injuries.
Collapse
Affiliation(s)
- Charlene Z. L. Ong
- Health Informatics Lab, College of Computing and Data Science, Nanyang Technological University, Singapore, Singapore
| | - N. Jannah M. Nasir
- Centre for Computational Biology and Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Roy E. Welsch
- Sloan School of Management Operations Research and Statistics, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Lisa Tucker-Kellogg
- Centre for Computational Biology and Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Jagath C. Rajapakse
- Health Informatics Lab, College of Computing and Data Science, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
17
|
Qiu J, Yue F, Kim KH, Chen X, Khedr MA, Chen J, Gu L, Ren J, Ferreira CR, Ellis J, Kuang S. Overexpression of CPT1A disrupts the maintenance and regenerative function of muscle stem cells. FASEB J 2024; 38:e70071. [PMID: 39382025 PMCID: PMC11486317 DOI: 10.1096/fj.202400947r] [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: 04/26/2024] [Revised: 09/02/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
The skeletal muscle satellite cells (SCs) mediate regeneration of myofibers upon injury. As they switch from maintenance (quiescence) to regeneration, their relative reliance on glucose and fatty acid metabolism alters. To explore the contribution of mitochondrial fatty acid oxidation (FAO) pathway to SCs and myogenesis, we examined the role of carnitine palmitoyltransferase 1A (CPT1A), the rate-limiting enzyme of FAO. CPT1A is highly expressed in quiescent SCs (QSCs) compared with activated and proliferating SCs, and its expression level decreases during myogenic differentiation. Myod1Cre-driven overexpression (OE) of Cpt1a in embryonic myoblasts (Cpt1aMTG) reduces muscle weight, grip strength, and contractile force without affecting treadmill endurance of adult mice. Adult Cpt1aMTG mice have reduced number of SC, impairing muscle regeneration and promoting lipid infiltration. Similarly, Pax7CreER-driven, tamoxifen-inducible Cpt1a-OE in QSCs of adult muscles (Cpt1aPTG) leads to depletion of SCs and compromises muscle regeneration. The reduced proliferation of Cpt1a-OE SCs is associated with elevated level of acyl-carnitine, and acyl-carnitine treatment impedes proliferation of wildtype SCs. These findings indicate that aberrant level of CPT1A elevates acyl-carnitine to impair the maintenance, proliferation and regenerative function of SCs.
Collapse
Affiliation(s)
- Jiamin Qiu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
- These authors contributed equally to this work
| | - Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA
- These authors contributed equally to this work
| | - Kun Ho Kim
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Xiyue Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | | - Jingjuan Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Lijie Gu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Junxiao Ren
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Christina R. Ferreira
- Purdue Metabolite Profiling Facility, Purdue University, West, Lafayette, IN 47907, USA
| | - Jessica Ellis
- Department of Physiology and East Carolina Diabetes and Obesity Institute, Brody School of Medicine at East Carolina University Greenville, NC 27834, USA
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
- Purdue University Institute for Cancer Research, West Lafayette, IN 47907, USA
- Departments of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
18
|
Lu Y, Walji T, Pandey P, Zhou C, Habela CW, Snapper SB, Li R, Chen EH. Branched actin polymerization drives invasive protrusion formation to promote myoblast fusion during skeletal muscle regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.30.615960. [PMID: 39416162 PMCID: PMC11482830 DOI: 10.1101/2024.09.30.615960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Skeletal muscle regeneration is a multistep process involving the activation, proliferation, differentiation, and fusion of muscle stem cells, known as satellite cells. The fusion of satellite cell-derived mononucleated muscle cells (SCMs) is indispensable for the generation of multinucleated, contractile myofibers during muscle repair. However, the molecular and cellular mechanisms underlying SCM fusion during muscle regeneration remain poorly understood. In this study, we uncovered an essential role for branched actin polymerization in SCM fusion. Using conditional knockouts of the Arp2/3 complex and its actin nucleation-promoting factors, N-WASP and WAVE, we demonstrated that branched actin polymerization is required for the SCM fusion, but not for satellite cell proliferation, differentiation, and migration. We showed that the N-WASP and WAVE complexes have partially redundant functions in regulating SCM fusion. Furthermore, we showed that branched actin polymerization is essential for generating invasive protrusions at the fusogenic synapses in SCMs. Taken together, our study has identified new components of the myoblast fusion machinery in skeletal muscle regeneration and demonstrated a critical role for branched actin-propelled invasive protrusions in this process.
Collapse
Affiliation(s)
- Yue Lu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tezin Walji
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pratima Pandey
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chuanli Zhou
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christa W. Habela
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Scott B. Snapper
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
| | - Rong Li
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Elizabeth H. Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
19
|
Huang N, Zou K, Zhong Y, Luo Y, Wang M, Xiao L. Hotspots and trends in satellite cell research in muscle regeneration: A bibliometric visualization and analysis from 2010 to 2023. Heliyon 2024; 10:e37529. [PMID: 39309858 PMCID: PMC11415684 DOI: 10.1016/j.heliyon.2024.e37529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Background The incidence of muscle atrophy or sports injuries is increasing with time and population aging, thereby attracting considerable attention to muscle generation research. Muscle satellite cells, which play an important role in this process, lack comprehensive literature regarding their use for muscle regeneration. Hence, this study aimed to analyze the hotspots and trends in satellite cell research from 2010 to 2023, providing a reference for muscle regeneration research. Methods Studies on satellite cells' role in muscle regeneration from 2010 to 2023 were retrieved from the Web of Science Core Collection. Using CiteSpace and VOSviewer, we analyzed annual publications, authors and co-citing authors, countries and institutions, journals and co-citing journals, co-citing references, and keywords. Results From 2010 to 2023, 1468 papers were retrieved, indicating an overall increasing trend in the number of annual publications related to satellite cells in muscle regeneration. The United States had the highest number of publications, while the Institut National de la Santé et de la Recherche Médicale was the institution with the most publications. Among journals, " PloS One" had the highest number of published papers, and "Cell" emerged as the most co-cited journal. A total of 7425 authors were involved, with Michael A. Rudnicki being the author with the highest number of publications and the most co-cited author. The most cited reference was "Satellite cells and the muscle stem cell niche." Among keywords, "satellite cells" was the most common, with "heterogeneity" having the highest centrality. Frontier themes included "Duchenne muscular dystrophy," "skeletal muscle," "in-vivo," "muscle regeneration," "mice," "muscle atrophy," "muscle fibers," "inflammation," " mesenchymal stem cells," and "satellite cell." Conclusion This study presents the current status and trends in satellite cell research on muscle regeneration from 2010 to 2023 using bibliometric analyses, providing valuable insights into numerous future research directions.
Collapse
Affiliation(s)
- Nan Huang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Intelligent Rehabilitation Technology Innovation Center, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Kang Zou
- Department of Critical Care Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Yanbiao Zhong
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Yun Luo
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Maoyuan Wang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Li Xiao
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| |
Collapse
|
20
|
Shen E, Piao M, Li Y, Wu Y, Li S, Lee SH, Jin L, Lee KY. CMTM3 Suppresses Proliferation and Osteogenic Transdifferentiation of C2C12 Myoblasts through p53 Upregulation. Cells 2024; 13:1352. [PMID: 39195242 PMCID: PMC11352514 DOI: 10.3390/cells13161352] [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: 07/18/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
CKLF-like MARVEL transmembrane domain-containing 3 (CMTM3), a member of the CMTM family that is closely related to tumor occurrence and progression, plays crucial roles in the immune system, cardiovascular system, and male reproductive system. Recently, CMTM3 has emerged as a potential target for treating diseases related to bone formation. However, additional studies are needed to understand the mechanisms by which CMTM3 regulates the process of osteogenic differentiation. In this study, we observed a significant downregulation of Cmtm3 expression during the transdifferentiation of C2C12 myoblasts into osteoblasts induced by BMP4. Cmtm3 overexpression suppressed proliferation and osteogenic differentiation in BMP4-induced C2C12 cells, whereas its knockdown conversely facilitated the process. Mechanistically, Cmtm3 overexpression upregulated both the protein and mRNA levels of p53 and p21. Conversely, Cmtm3 knockdown exerted the opposite effects. Additionally, we found that Cmtm3 interacts with p53 and increases protein stability by inhibiting proteasome-mediated ubiquitination and degradation. Notably, Trp53 downregulation abrogated the inhibitory effect of Cmtm3 on BMP4-induced proliferation and osteogenic differentiation of C2C12 myoblasts. Collectively, our findings provide key insights into the role of CMTM3 in regulating myoblast proliferation and transdifferentiation into osteoblasts, highlighting its significance in osteogenesis research.
Collapse
Affiliation(s)
- Enzhao Shen
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (E.S.); (M.P.); (Y.L.)
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (S.L.)
| | - Meiyu Piao
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (E.S.); (M.P.); (Y.L.)
| | - Yuankuan Li
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (E.S.); (M.P.); (Y.L.)
| | - Yuecheng Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (S.L.)
| | - Sihang Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (S.L.)
| | - Sung Ho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (E.S.); (M.P.); (Y.L.)
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (S.L.)
| | - Kwang Youl Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (E.S.); (M.P.); (Y.L.)
| |
Collapse
|
21
|
Yang Y, Huang K, Jiang H, Wang S, Xu X, Liu Y, Liu Q, Wei M, Li Z. Unveiling the role of circRBBP7 in myoblast proliferation and differentiation: A novel regulator of muscle development. FASEB J 2024; 38:e23808. [PMID: 38994637 DOI: 10.1096/fj.202302599rr] [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: 12/18/2023] [Revised: 06/13/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024]
Abstract
Muscle development is a multistep process regulated by diverse gene networks, and circRNAs are considered novel regulators mediating myogenesis. Here, we systematically analyzed the role and underlying regulatory mechanisms of circRBBP7 in myoblast proliferation and differentiation. Results showed that circRBBP7 has a typical circular structure and encodes a 13 -kDa protein. By performing circRBBP7 overexpression and RNA interference, we found that the function of circRBBP7 was positively correlated with the proliferation and differentiation of myoblasts. Using RNA sequencing, we identified 1633 and 532 differentially expressed genes (DEGs) during myoblast proliferation or differentiation, respectively. The DEGs were found mainly enriched in cell cycle- and skeletal muscle development-related pathways, such as the MDM2/p53 and PI3K-Akt signaling pathways. Further co-IP and IF co-localization analysis revealed that VEGFR-1 is a target of circRBBP7 in myoblasts. qRT-PCR and WB analysis further confirmed the positive correlation between VEGFR-1 and circRBBP7. Moreover, we found that in vivo transfection of circRBBP7 into injured muscle tissues significantly promoted the regeneration and repair of myofibers in mice. Therefore, we speculate that circRBBP7 may affect the activity of MDM2 by targeting VEGFR-1, altering the expression of muscle development-related genes by mediating p53 degradation, and ultimately promoting myoblast development and muscle regeneration. This study provides essential evidence that circRBBP7 can serve as a potential target for myogenesis regulation and a reference for the application of circRBBP7 in cattle genetic breeding and muscle injury treatment.
Collapse
Affiliation(s)
- Yufeng Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- The Animal Husbandry Research Institute of Guangxi Zhuang Autonomous Region, Guangxi Agricultural Vocational University, Nanning, China
| | - Kongwei Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Hancai Jiang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Shuwan Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xiaoxian Xu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yang Liu
- Guangxi Zhuang Autonomous Region Center for Analysis and Test Research, Nanning, China
| | - Qingyou Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Mingsong Wei
- The Animal Husbandry Research Institute of Guangxi Zhuang Autonomous Region, Guangxi Agricultural Vocational University, Nanning, China
| | - Zhipeng Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| |
Collapse
|
22
|
Antony R, Aby K, Montgomery M, Li Y. Skeletal Muscle UCHL1 Negatively Regulates Muscle Development and Recovery after Muscle Injury. Int J Mol Sci 2024; 25:7330. [PMID: 39000437 PMCID: PMC11242864 DOI: 10.3390/ijms25137330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Ubiquitin C-terminal hydrolase L1 (UCHL1) is a deubiquitinating enzyme originally found in the brain. Our previous work revealed that UCHL1 was also expressed in skeletal muscle and affected myoblast differentiation and metabolism. In this study, we further tested the role of UCHL1 in myogenesis and muscle regeneration following muscle ischemia-reperfusion (IR) injury. In the C2C12 myoblast, UCHL1 knockdown upregulated MyoD and myogenin and promoted myotube formation. The skeletal muscle-specific knockout (smKO) of UCHL1 increased muscle fiber sizes in young mice (1 to 2 months old) but not in adult mice (3 months old). In IR-injured hindlimb muscle, UCHL1 was upregulated. UCHL1 smKO ameliorated tissue damage and injury-induced inflammation. UCHL1 smKO also upregulated myogenic factors and promoted functional recovery in IR injury muscle. Moreover, UCHL1 smKO increased Akt and Pink1/Parkin activities. The overall results suggest that skeletal muscle UCHL1 is a negative factor in skeletal muscle development and recovery following IR injury and therefore is a potential therapeutic target to improve muscle regeneration and functional recovery following injuries.
Collapse
Affiliation(s)
| | | | | | - Yifan Li
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (R.A.); (K.A.); (M.M.)
| |
Collapse
|
23
|
Lee SB, Woo TW, Baek DC, Son CG. A standardized herbal combination of Astragalus membranaceus and Paeonia japonica promotes skeletal muscle hypertrophy in a treadmill exercise mouse model. Front Nutr 2024; 11:1362550. [PMID: 38966418 PMCID: PMC11223055 DOI: 10.3389/fnut.2024.1362550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/29/2024] [Indexed: 07/06/2024] Open
Abstract
Background Maintaining a normal range of muscle mass and function is crucial not only for sustaining a healthy life but also for preventing various disorders. Numerous nutritional or natural resources are being explored for their potential muscle hypertrophic properties. Aim We aimed to evaluate the muscle hypertrophic effects of APX, a 1:1 mixture of Astragalus membranaceus and Paeonia japonica. In addition to the myotube differentiation cell assay, we utilized a weighted exercise-based animal model and evaluated changes in muscle hypertrophy using dual-energy X-ray absorptiometry (DXA) and histological analysis. Results The 8-week treadmill exercise led to notable decreases in body weight and fat mass but an increase in muscle mass compared to the control group. Administration of APX significantly accelerated muscle mass gain (p < 0.05) without altering body weight or fat mass compared to the exercise-only group. This muscle hypertrophic effect of APX was consistent with the histologic size of muscle fibers in the gastrocnemius (p > 0.05) and rectus femoris (p < 0.05), as well as the regulation of myogenic transcription factors (MyoD and myogenin), respectively. Furthermore, APX demonstrated a similar action to insulin-like growth factor 1, influencing the proliferation of C2C12 myoblast cells (p < 0.01) and their differentiation into myotubes (p < 0.05) compared to the control group. Conclusion The present study provides experimental evidence that APX has muscle hypertrophic effects, and its underlying mechanisms would involve the modulation of MyoD and myogenin.
Collapse
Affiliation(s)
| | | | | | - Chang-Gue Son
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon, Republic of Korea
| |
Collapse
|
24
|
Chen Y, Wang Z, Qu X, Song B, Tang Y, Li B, Cao G, Yi G. An intronic SNP affects skeletal muscle development by regulating the expression of TP63. Front Vet Sci 2024; 11:1396766. [PMID: 38933706 PMCID: PMC11199888 DOI: 10.3389/fvets.2024.1396766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Background Porcine skeletal muscle development is pivotal for improving meat production. TP63, a transcription factor, regulates vital cellular processes, yet its role in skeletal muscle proliferation is unclear. Methods The effects of TP63 on skeletal muscle cell viability and proliferation were investigated using both mouse and porcine skeletal muscle myoblasts. Selective sweep analysis in Western pigs identified TP63 as a potential candidate gene for skeletal muscle development. The correlation between TP63 overexpression and cell proliferation was assessed using quantitative real-time PCR (RT-qPCR) and 5-ethynyl-2'-deoxyuridine (EDU). Results The study revealed a positive correlation between TP63 overexpression and skeletal muscle cell proliferation. Bioinformatics analysis predicted an interaction between MEF2A, another transcription factor, and the mutation site of TP63. Experimental validation through dual-luciferase assays confirmed that a candidate enhancer SNP could influence MEF2A binding, subsequently regulating TP63 expression and promoting skeletal muscle cell proliferation. Conclusion These findings offer experimental evidence for further exploration of skeletal muscle development mechanisms and the advancement of genetic breeding strategies aimed at improving meat production traits.
Collapse
Affiliation(s)
- Yufen Chen
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- College of Animal Science, Shanxi Agricultural University, Jinzhong, China
| | - Zhen Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xiaolu Qu
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Bangmin Song
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yueting Tang
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Bugao Li
- College of Animal Science, Shanxi Agricultural University, Jinzhong, China
| | - Guoqing Cao
- College of Animal Science, Shanxi Agricultural University, Jinzhong, China
| | - Guoqiang Yi
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Kunpeng Institute of Modern Agriculture at Foshan, Chinese Academy of Agricultural Sciences, Foshan, China
- Bama Yao Autonomous County Rural Revitalization Research Institute, Bama, China
| |
Collapse
|
25
|
Chen J, Li Q. Emerging role of HDAC11 in skeletal muscle biology. Front Cell Dev Biol 2024; 12:1368171. [PMID: 38859964 PMCID: PMC11163118 DOI: 10.3389/fcell.2024.1368171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/07/2024] [Indexed: 06/12/2024] Open
Abstract
HDAC11 is an epigenetic repressor of gene transcription, acting through its deacetylase activity to remove functional acetyl groups from the lysine residues of histones at genomic loci. It has been implicated in the regulation of different immune responses, metabolic activities, as well as cell cycle progression. Recent studies have also shed lights on the impact of HDAC11 on myogenic differentiation and muscle development, indicating that HDAC11 is important for histone deacetylation at the promoters to inhibit transcription of cell cycle related genes, thereby permitting myogenic activation at the onset of myoblast differentiation. Interestingly, the upstream networks of HDAC11 target genes are mainly associated with cell cycle regulators and the acetylation of histones at the HDAC11 target promoters appears to be residue specific. As such, selective inhibition, or activation of HDAC11 presents a potential therapeutic approach for targeting distinct epigenetic pathways in clinical applications.
Collapse
Affiliation(s)
- Jihong Chen
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Qiao Li
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
26
|
Rota Graziosi E, François S, Nasser F, Gauthier M, Oger M, Favier AL, Drouet M, Jullien N, Riccobono D. Comparison of Three Antagonists of Hedgehog Pathway to Promote Skeletal Muscle Regeneration after High Dose Irradiation. Radiat Res 2024; 201:429-439. [PMID: 38253061 DOI: 10.1667/rade-23-00140.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/01/2023] [Indexed: 01/24/2024]
Abstract
The current geopolitical context has brought the radiological nuclear risk to the forefront of concerns. High-dose localized radiation exposure leads to the development of a musculocutaneous radiation syndrome affecting the skin and subcutaneous muscles. Despite the implementation of a gold standard treatment based on an invasive surgical procedure coupled with autologous cell therapy, a muscular defect frequently persists. Targeting the modulation of the Hedgehog (Hh) signaling pathway appears to be a promising therapeutic approach. Activation of this pathway enhances cell survival and promotes proliferation after irradiation, while inhibition by Cyclopamine facilitates differentiation. In this study, we compared the effects of three antagonists of Hh, Cyclopamine (CA), Vismodegib (VDG) and Sonidegib (SDG) on differentiation. A stable cell line of murine myoblasts, C2C12, was exposed to X-ray radiation (5 Gy) and treated with CA, VDG or SDG. Analysis of proliferation, survival (apoptosis), morphology, myogenesis genes expression and proteins production were performed. According to the results, VDG does not have a significant impact on C2C12 cells. SDG increases the expression/production of differentiation markers to a similar extent as CA, while morphologically, SDG proves to be more effective than CA. To conclude, SDG can be used in the same way as CA but already has a marketing authorization with an indication against basal cell cancers, facilitating their use in vivo. This proof of concept demonstrates that SDG represents a promising alternative to CA to promotes differentiation of murine myoblasts. Future studies on isolated and cultured satellite cells and in vivo will test this proof of concept.
Collapse
Affiliation(s)
- Emmanuelle Rota Graziosi
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Sabine François
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
- INSERM, UMR1296, Radiations: Defense, Health, Environment, Lyon and Brétigny-sur-Orge, France
| | - Farah Nasser
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Michel Gauthier
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Myriam Oger
- IRBA, French Armed Forces Biomedical Research Institute, Imagery Unit, Department of Platforms and Technology Research, Brétigny-sur-Orge, France
| | - Anne-Laure Favier
- IRBA, French Armed Forces Biomedical Research Institute, Imagery Unit, Department of Platforms and Technology Research, Brétigny-sur-Orge, France
| | - Michel Drouet
- INSERM, UMR1296, Radiations: Defense, Health, Environment, Lyon and Brétigny-sur-Orge, France
- IRBA, French Armed Forces Biomedical Research Institute, Radiations Bioeffects Department, Brétigny-sur-Orge, France
| | - Nicolas Jullien
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Diane Riccobono
- INSERM, UMR1296, Radiations: Defense, Health, Environment, Lyon and Brétigny-sur-Orge, France
- IRBA, French Armed Forces Biomedical Research Institute, Radiations Bioeffects Department, Brétigny-sur-Orge, France
| |
Collapse
|
27
|
Wherley TJ, Thomas S, Millay DP, Saunders T, Roy S. Molecular regulation of myocyte fusion. Curr Top Dev Biol 2024; 158:53-82. [PMID: 38670716 PMCID: PMC11503471 DOI: 10.1016/bs.ctdb.2024.01.016] [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] [Indexed: 04/28/2024]
Abstract
Myocyte fusion is a pivotal process in the development and regeneration of skeletal muscle. Failure during fusion can lead to a range of developmental as well as pathological consequences. This review aims to comprehensively explore the intricate processes underlying myocyte fusion, from the molecular to tissue scale. We shed light on key players, such as the muscle-specific fusogens - Myomaker and Myomixer, in addition to some lesser studied molecules contributing to myocyte fusion. Conserved across vertebrates, Myomaker and Myomixer play a crucial role in driving the merger of plasma membranes of fusing myocytes, ensuring the formation of functional muscle syncytia. Our multiscale approach also delves into broader cell and tissue dynamics that orchestrate the timing and positioning of fusion events. In addition, we explore the relevance of muscle fusogens to human health and disease. Mutations in fusogen genes have been linked to congenital myopathies, providing unique insights into the molecular basis of muscle diseases. We conclude with a discussion on potential therapeutic avenues that may emerge from manipulating the myocyte fusion process to remediate skeletal muscle disorders.
Collapse
Affiliation(s)
- Tanner J Wherley
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Serena Thomas
- Warwick Medical School, University of Warwick, Coventry, United Kingdom; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, Singapore, Singapore
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| | - Timothy Saunders
- Warwick Medical School, University of Warwick, Coventry, United Kingdom; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, Singapore, Singapore.
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore, Singapore; Department of Pediatrics, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
28
|
Gordon T. Brief Electrical Stimulation Promotes Recovery after Surgical Repair of Injured Peripheral Nerves. Int J Mol Sci 2024; 25:665. [PMID: 38203836 PMCID: PMC10779324 DOI: 10.3390/ijms25010665] [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: 10/13/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Injured peripheral nerves regenerate their axons in contrast to those in the central nervous system. Yet, functional recovery after surgical repair is often disappointing. The basis for poor recovery is progressive deterioration with time and distance of the growth capacity of the neurons that lose their contact with targets (chronic axotomy) and the growth support of the chronically denervated Schwann cells (SC) in the distal nerve stumps. Nonetheless, chronically denervated atrophic muscle retains the capacity for reinnervation. Declining electrical activity of motoneurons accompanies the progressive fall in axotomized neuronal and denervated SC expression of regeneration-associated-genes and declining regenerative success. Reduced motoneuronal activity is due to the withdrawal of synaptic contacts from the soma. Exogenous neurotrophic factors that promote nerve regeneration can replace the endogenous factors whose expression declines with time. But the profuse axonal outgrowth they provoke and the difficulties in their delivery hinder their efficacy. Brief (1 h) low-frequency (20 Hz) electrical stimulation (ES) proximal to the injury site promotes the expression of endogenous growth factors and, in turn, dramatically accelerates axon outgrowth and target reinnervation. The latter ES effect has been demonstrated in both rats and humans. A conditioning ES of intact nerve days prior to nerve injury increases axonal outgrowth and regeneration rate. Thereby, this form of ES is amenable for nerve transfer surgeries and end-to-side neurorrhaphies. However, additional surgery for applying the required electrodes may be a hurdle. ES is applicable in all surgeries with excellent outcomes.
Collapse
Affiliation(s)
- Tessa Gordon
- Division of Reconstructive Surgery, Department of Surgery, University of Toronto, Toronto, ON M4G 1X8, Canada
| |
Collapse
|
29
|
Zhang Y, Lu Y, Yu M, Wang J, Du X, Zhao D, Pian H, He Z, Wu G, Li S, Wang S, Yu D. Transcriptome Profiling Identifies Differentially Expressed Genes in Skeletal Muscle Development in Native Chinese Ducks. Genes (Basel) 2023; 15:52. [PMID: 38254942 PMCID: PMC10815232 DOI: 10.3390/genes15010052] [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: 11/02/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
China boasts a rich diversity of indigenous duck species, some of which exhibit desirable economic traits. Here, we generated transcriptome sequencing datasets of breast muscle tissue samples from 1D of four groups: Pekin duck pure breeding group (P), Jinling White duck breeding group (J), P ♂ × J ♀ orthogonal group (PJ) and J ♂ × P ♀ reciprocal-cross group (JP) (n = 3), chosen based on the distinctive characteristics of duck muscle development during the embryonic period. We identified 5053 differentially expressed genes (DEGs) among the four groups. Network prediction analysis showed that ribosome and oxidative phosphorylation-related genes were the most enriched, and muscular protein-related genes were found in the 14-day-old embryonic group. We found that previously characterized functional genes, such as FN1, AGRN, ADNAMST3, APOB and FGF9, were potentially involved in muscle development in 14-day-old embryos. Functional enrichment analysis suggested that genes that participated in molecular function and cell component and key signaling pathways (e.g., hippo, ribosome, oxidative phosphorylation) were significantly enriched in the development of skeletal muscle at 14 days of embryonic age. These results indicate a possible role of muscle metabolism and myoglobin synthesis in skeletal muscle development in both duck parents and hybrids.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Yinglin Lu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Minli Yu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Jin Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Xubin Du
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Dong Zhao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
- School of Animal Medical, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China
| | - Huifang Pian
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Zongliang He
- Nanjing Academy of Animal Husbandry and Poultry, Nanjing 210095, China
| | - Guansuo Wu
- Nanjing Academy of Animal Husbandry and Poultry, Nanjing 210095, China
| | - Shiwei Li
- College of Animal Science, Xizang Agricultural and Animal Husbandry University, Linzhi 860000, China
| | - Sike Wang
- College of Animal Science, Xizang Agricultural and Animal Husbandry University, Linzhi 860000, China
| | - Debing Yu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| |
Collapse
|
30
|
Ortuste Quiroga HP, Fujimaki S, Ono Y. Pax7 reporter mouse models: a pocket guide for satellite cell research. Eur J Transl Myol 2023; 33:12174. [PMID: 38112596 PMCID: PMC10811643 DOI: 10.4081/ejtm.2023.12174] [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: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Since their discovery, satellite cells have showcased their need as primary contributors to skeletal muscle maintenance and repair. Satellite cells lay dormant, but when needed, activate, differentiate, fuse to fibres and self-renew, that has bestowed satellite cells with the title of muscle stem cells. The satellite cell specific transcription factor Pax7 has enabled researchers to develop animal models against the Pax7 locus in order to isolate and characterise satellite cell-mediated events. This review focuses specifically on describing Pax7 reporter mouse models. Here we describe how each model was generated and the key findings obtained. The strengths and limitations of each model are also discussed. The aim is to provide new and current satellite cell enthusiasts with a basic understanding of the available Pax7 reporter mice and hopefully guide selection of the most appropriate Pax7 model to answer a specific research question.
Collapse
Affiliation(s)
- Huascar Pedro Ortuste Quiroga
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Honjo, Chuo-ku, Kumamoto.
| | - Shin Fujimaki
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Honjo, Chuo-ku, Kumamoto.
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan; Tokyo Metropolitan Institute for Geriatrics and Gerontology (TMIG), Sakae-cho, Itabashi, Tokyo.
| |
Collapse
|
31
|
Jatwani A, Tulsawani R. Ganoderma lucidum Induces Myogenesis Markers to Avert Damage to Skeletal Muscles in Rats Exposed to Hypobaric Hypoxia. High Alt Med Biol 2023; 24:287-295. [PMID: 34142874 DOI: 10.1089/ham.2020.0172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Jatwani, Arti, and Rajkumar Tulsawani. Ganoderma lucidum induces myogenesis markers to avert damage to skeletal muscles in rats exposed to hypobaric hypoxia. High Alt Med Biol. 24:287-295, 2023. Background: Hypobaric hypoxia (HH) has been reported to induce skeletal muscle loss and impair myogenesis. Aqueous extract of G. lucidum (AqGL) contains bioactive metabolites attributed to various pharmacological effects. In this study, protective effect of AqGL in ameliorating muscle mass loss following acute HH has been reported. Materials and Methods: Male Sprague-Dawley rats were divided into following five groups of six rats in each group: unexposed control (Group 1), 6 hours of HH exposure (Group 2), 6 hours of HH exposure+AqGL extract 50 mg/kg body weight (BW) (Group 3), 6 hours of HH exposure+AqGL extract 100 mg/kg BW (Group 4), and 6 hours of HH exposure+AqGL extract 200 mg/kg BW (Group 5). Experimental animals from all groups, except Group, 1 were exposed to HH, simulated altitude of 25,000 ft for 6 hours. After exposure period, gastrocnemius muscle was collected, weighed, and morphological, biochemical, and molecular markers were analyzed. Results: HH-exposed rat muscle showed significant (p < 0.05) increase in oxidative stress markers (reactive oxygen species & malondialdehyde), which was concomitant with decrease in its mass compared to controls. AqGL treatment significantly (p < 0.05) prevented muscle oxidative stress, restored reduced glutathione content, reduced protein carbonyl content and advanced oxidation protein product, and restored muscle mass loss at effective dose of 100 mg/kg BW. Furthermore, AqGL supplementation enhanced Myf5 (p < 0.01), MyoD (p < 0.01), MyoG (p < 0.05), and Mrf4 (nonsignificantly), brain-derived neurotrophic factor (p < 0.01), and interleukin 6 (p < 0.01) expression along with restoration of tumor necrosis factor alpha (p < 0.001) and myostatin (p < 0.05) in hypoxia-exposed muscle, evidencing induction of myogenesis markers. Moreover, histological analysis showed increased myocyte number; nuclei shifted toward the periphery in the treatment group supporting muscle regeneration. Conclusion: AqGL supplementation attenuates muscle mass loss by preventing oxidative stress and inducing modulation in myogenesis markers under HH environment.
Collapse
Affiliation(s)
- Arti Jatwani
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | | |
Collapse
|
32
|
Cao C, Cai Y, Li Y, Li T, Zhang J, Hu Z, Zhang J. Characterization and comparative transcriptomic analysis of skeletal muscle in female Pekin duck and Hanzhong Ma duck during different growth stages using RNA-seq. Poult Sci 2023; 102:103122. [PMID: 37832186 PMCID: PMC10568565 DOI: 10.1016/j.psj.2023.103122] [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: 06/27/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 10/15/2023] Open
Abstract
Duck is an economically important poultry, and there is currently a major focus on improving its meat quality through breeding. There are wide variations in the growth regulation mechanisms of different duck breeds, that fundamental research on skeletal muscle growth is essential for understanding the regulation of unknown genes. The study aimed to broaden the understanding the duck skeletal muscle development and thereby to improve the performance of domestic ducks. In this study, RNA-seq data from skeletal muscles (breast muscle and leg muscle) of Pekin duck and Hanzhong Ma duck sampled at d 17, 21, and 27 of embryo (E17d, E21d, and E27d), as well as at 6-mo-old following birth (M6), to investigate and compare the mRNA temporal expression profiles and associated pathways that regulate skeletal myogenesis of different duck breeds. There were 331 to 1,440 annotated differentially expressed genes (DEGs) in breast muscle and 380 to 1,790 annotated DEGs in leg muscle from different databases between 2 duck breeds. Gene ontology (GO) enrichment in skeletal muscles indicated that these DEGs were mainly involved in biosynthetic process, developmental process, regulation of protein metabolic process and regulation of gene expression. KEGG analysis in skeletal muscles showed that a total of 41 DEGs were mapped to 7 KEGG pathways, including ECM-receptor interaction, focal adhesion, carbon metabolism, regulation of actin cytoskeleton, calcium signaling pathway, biosynthesis of amino acids and PPAR signaling pathway. The differential expression of 8 selected DEGs was verified by qRT-PCR, and the results were consistent with RNA-seq data. The identified DEGs, such as SDC, SPP1, PAK1, MYL9, PGK1, NOS1, PHGDH, TNNT2, FN1, and AQP4, were specially highlighted, indicating their associations with muscle development in the Pekin duck and Hanzhong Ma duck. This study provides a basis for revealing the differences in skeletal muscle development between Pekin duck and Hanzhong Ma duck.
Collapse
Affiliation(s)
- Chang Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Yingjie Cai
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Yuxiao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Tao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Jiqiao Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Zhigang Hu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Jianqin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China.
| |
Collapse
|
33
|
Maiullari S, Cicirelli A, Picerno A, Giannuzzi F, Gesualdo L, Notarnicola A, Sallustio F, Moretti B. Pulsed Electromagnetic Fields Induce Skeletal Muscle Cell Repair by Sustaining the Expression of Proteins Involved in the Response to Cellular Damage and Oxidative Stress. Int J Mol Sci 2023; 24:16631. [PMID: 38068954 PMCID: PMC10706358 DOI: 10.3390/ijms242316631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Pulsed electromagnetic fields (PEMF) are employed as a non-invasive medicinal therapy, especially in the orthopedic field to stimulate bone regeneration. However, the effect of PEMF on skeletal muscle cells (SkMC) has been understudied. Here, we studied the potentiality of 1.5 mT PEMF to stimulate early regeneration of human SkMC. We showed that human SkMC stimulated with 1.5 mT PEMF for four hours repeated for two days can stimulate cell proliferation without inducing cell apoptosis or significant impairment of the metabolic activity. Interestingly, when we simulated physical damage of the muscle tissue by a scratch, we found that the same PEMF treatment can speed up the regenerative process, inducing a more complete cell migration to close the scratch and wound healing. Moreover, we investigated the molecular pattern induced by PEMF among 26 stress-related cell proteins. We found that the expression of 10 proteins increased after two consecutive days of PEMF stimulation for 4 h, and most of them were involved in response processes to oxidative stress. Among these proteins, we found that heat shock protein 70 (HSP70), which can promote muscle recovery, inhibits apoptosis and decreases inflammation in skeletal muscle, together with thioredoxin, paraoxonase, and superoxide dismutase (SOD2), which can also promote skeletal muscle regeneration following injury. Altogether, these data support the possibility of using PEMF to increase SkMC regeneration and, for the first time, suggest a possible molecular mechanism, which consists of sustaining the expression of antioxidant enzymes to control the important inflammatory and oxidative process occurring following muscle damage.
Collapse
Affiliation(s)
- Silvia Maiullari
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.C.); (A.P.); (F.G.)
| | - Antonella Cicirelli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.C.); (A.P.); (F.G.)
| | - Angela Picerno
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.C.); (A.P.); (F.G.)
| | - Francesca Giannuzzi
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.C.); (A.P.); (F.G.)
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area (DIMEPRE-J), University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Angela Notarnicola
- Orthopaedic and Trauma Unit, Department of Translational Biomedicine and Neuroscience “DiBraiN”, University of Bari “Aldo Moro”, Piazza G. Cesare 11, 70124 Bari, Italy; (A.N.); (B.M.)
| | - Fabio Sallustio
- Nephrology, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area (DIMEPRE-J), University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Biagio Moretti
- Orthopaedic and Trauma Unit, Department of Translational Biomedicine and Neuroscience “DiBraiN”, University of Bari “Aldo Moro”, Piazza G. Cesare 11, 70124 Bari, Italy; (A.N.); (B.M.)
| |
Collapse
|
34
|
Noviello C, Kobon K, Randrianarison-Huetz V, Maire P, Pietri-Rouxel F, Falcone S, Sotiropoulos A. RhoA Is a Crucial Regulator of Myoblast Fusion. Cells 2023; 12:2673. [PMID: 38067102 PMCID: PMC10705320 DOI: 10.3390/cells12232673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
Satellite cells (SCs) are adult muscle stem cells that are mobilized when muscle homeostasis is perturbed. Here we show that RhoA in SCs is indispensable to have correct muscle regeneration and hypertrophy. In particular, the absence of RhoA in SCs prevents a correct SC fusion both to other RhoA-deleted SCs (regeneration context) and to growing control myofibers (hypertrophy context). We demonstrated that RhoA is dispensable for SCs proliferation and differentiation; however, RhoA-deleted SCs have an inefficient movement even if their cytoskeleton assembly is not altered. Proliferative myoblast and differentiated myotubes without RhoA display a decreased expression of Chordin, suggesting a crosstalk between these genes for myoblast fusion regulation. These findings demonstrate the importance of RhoA in SC fusion regulation and its requirement to achieve an efficient skeletal muscle homeostasis restoration.
Collapse
Affiliation(s)
- Chiara Noviello
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
- Centre de Recherche en Myologie, Sorbonne Université, INSERM UMRS 974, Institut de Myologie, F-75013 Paris, France;
| | - Kassandra Kobon
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
| | | | - Pascal Maire
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
| | - France Pietri-Rouxel
- Centre de Recherche en Myologie, Sorbonne Université, INSERM UMRS 974, Institut de Myologie, F-75013 Paris, France;
| | - Sestina Falcone
- Centre de Recherche en Myologie, Sorbonne Université, INSERM UMRS 974, Institut de Myologie, F-75013 Paris, France;
| | - Athanassia Sotiropoulos
- Institut Cochin, Université de Paris, INSERM U1016, CNRS, F-75014 Paris, France (P.M.); (A.S.)
| |
Collapse
|
35
|
Bülow A, Schäfer B, Beier JP. Three-Dimensional Bioprinting in Soft Tissue Engineering for Plastic and Reconstructive Surgery. Bioengineering (Basel) 2023; 10:1232. [PMID: 37892962 PMCID: PMC10604458 DOI: 10.3390/bioengineering10101232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/05/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Skeletal muscle tissue engineering (TE) and adipose tissue engineering have undergone significant progress in recent years. This review focuses on the key findings in these areas, particularly highlighting the integration of 3D bioprinting techniques to overcome challenges and enhance tissue regeneration. In skeletal muscle TE, 3D bioprinting enables the precise replication of muscle architecture. This addresses the need for the parallel alignment of cells and proper innervation. Satellite cells (SCs) and mesenchymal stem cells (MSCs) have been utilized, along with co-cultivation strategies for vascularization and innervation. Therefore, various printing methods and materials, including decellularized extracellular matrix (dECM), have been explored. Similarly, in adipose tissue engineering, 3D bioprinting has been employed to overcome the challenge of vascularization; addressing this challenge is vital for graft survival. Decellularized adipose tissue and biomimetic scaffolds have been used as biological inks, along with adipose-derived stem cells (ADSCs), to enhance graft survival. The integration of dECM and alginate bioinks has demonstrated improved adipocyte maturation and differentiation. These findings highlight the potential of 3D bioprinting techniques in skeletal muscle and adipose tissue engineering. By integrating specific cell types, biomaterials, and printing methods, significant progress has been made in tissue regeneration. However, challenges such as fabricating larger constructs, translating findings to human models, and obtaining regulatory approvals for cellular therapies remain to be addressed. Nonetheless, these advancements underscore the transformative impact of 3D bioprinting in tissue engineering research and its potential for future clinical applications.
Collapse
Affiliation(s)
- Astrid Bülow
- Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, 52074 Aachen, Germany; (B.S.); (J.P.B.)
| | | | | |
Collapse
|
36
|
Jara TC, Park K, Vahmani P, Van Eenennaam AL, Smith LR, Denicol AC. Stem cell-based strategies and challenges for production of cultivated meat. NATURE FOOD 2023; 4:841-853. [PMID: 37845547 DOI: 10.1038/s43016-023-00857-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 09/05/2023] [Indexed: 10/18/2023]
Abstract
Cultivated meat scale-up and industrial production will require multiple stable cell lines from different species to recreate the organoleptic and nutritional properties of meat from livestock. In this Review, we explore the potential of stem cells to create the major cellular components of cultivated meat. By using developments in the fields of tissue engineering and biomedicine, we explore the advantages and disadvantages of strategies involving primary adult and pluripotent stem cells for generating cell sources that can be grown at scale. These myogenic, adipogenic or extracellular matrix-producing adult stem cells as well as embryonic or inducible pluripotent stem cells are discussed for their proliferative and differentiation capacity, necessary for cultivated meat. We examine the challenges for industrial scale-up, including differentiation and culture protocols, as well as genetic modification options for stem cell immortalization and controlled differentiation. Finally, we discuss stem cell-related safety and regulatory challenges for bringing cultivated meat to the marketplace.
Collapse
Affiliation(s)
- T C Jara
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - K Park
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - P Vahmani
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - A L Van Eenennaam
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - L R Smith
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, USA.
| | - A C Denicol
- Department of Animal Science, University of California Davis, Davis, CA, USA
| |
Collapse
|
37
|
Shen X, Zhao X, He H, Zhao J, Wei Y, Chen Y, Han S, Zhu Y, Zhang Y, Zhu Q, Yin H. Evolutionary conserved circular MEF2A RNAs regulate myogenic differentiation and skeletal muscle development. PLoS Genet 2023; 19:e1010923. [PMID: 37676887 PMCID: PMC10508632 DOI: 10.1371/journal.pgen.1010923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/19/2023] [Accepted: 08/16/2023] [Indexed: 09/09/2023] Open
Abstract
Circular RNAs (circRNAs) have been recognized as critical regulators of skeletal muscle development. Myocyte enhancer factor 2A (MEF2A) is an evolutionarily conserved transcriptional factor that regulates myogenesis. However, it remains unclear whether MEF2A produces functional circRNAs. In this study, we identified two evolutionarily conserved circular MEF2A RNAs (circMEF2As), namely circMEF2A1 and circMEF2A2, in chicken and mouse muscle stem cells. Our findings revealed that circMEF2A1 promotes myogenesis by regulating the miR-30a-3p/PPP3CA/NFATC1 axis, whereas circMEF2A2 facilitates myogenic differentiation by targeting the miR-148a-5p/SLIT3/ROBO2/β-catenin signaling pathway. Furthermore, in vivo experiments demonstrated that circMEF2As both promote skeletal muscle growth. We also discovered that the linear MEF2A mRNA-derived MEF2A protein binds to its own promoter region, accelerating the transcription of MEF2A and upregulating the expression of both linear MEF2A and circMEF2As, forming a MEF2A autoregulated positive feedback loop. Moreover, circMEF2As positively regulate the expression of linear MEF2A by adsorbing miR-30a-3p and miR-148a-5p, which directly contribute to the MEF2A autoregulated feedback loop. Importantly, we found that mouse circMEF2As are essential for the myogenic differentiation of C2C12 cells. Collectively, our results demonstrated the evolution, function, and underlying mechanisms of circMEF2As in animal myogenesis, which may provide novel insight for both the farm animal meat industry and human medicine.
Collapse
Affiliation(s)
- Xiaoxu Shen
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiyu Zhao
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Haorong He
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jing Zhao
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yuanhang Wei
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yuqi Chen
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shunshun Han
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yifeng Zhu
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yao Zhang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qing Zhu
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Huadong Yin
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
38
|
Szabó L, Telek A, Fodor J, Dobrosi N, Dócs K, Hegyi Z, Gönczi M, Csernoch L, Dienes B. Reduced Expression of Septin7 Hinders Skeletal Muscle Regeneration. Int J Mol Sci 2023; 24:13536. [PMID: 37686339 PMCID: PMC10487768 DOI: 10.3390/ijms241713536] [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: 06/05/2023] [Revised: 07/28/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Septins are considered the fourth component of the cytoskeleton with the septin7 isoform playing a critical role in the formation of diffusion barriers in phospholipid bilayers and intra- and extracellular scaffolds. While its importance has already been confirmed in different intracellular processes, very little is known about its role in skeletal muscle. Muscle regeneration was studied in a Sept7 conditional knock-down mouse model to prove the possible role of septin7 in this process. Sterile inflammation in skeletal muscle was induced which was followed by regeneration resulting in the upregulation of septin7 expression. Partial knock-down of Sept7 resulted in an increased number of inflammatory cells and myofibers containing central nuclei. Taken together, our data suggest that partial knock-down of Sept7 hinders the kinetics of muscle regeneration, indicating its crucial role in skeletal muscle functions.
Collapse
Affiliation(s)
- László Szabó
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Andrea Telek
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Nóra Dobrosi
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Klaudia Dócs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Zoltán Hegyi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Mónika Gönczi
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
39
|
Molinaro C, Scalise M, Leo I, Salerno L, Sabatino J, Salerno N, De Rosa S, Torella D, Cianflone E, Marino F. Polarizing Macrophage Functional Phenotype to Foster Cardiac Regeneration. Int J Mol Sci 2023; 24:10747. [PMID: 37445929 DOI: 10.3390/ijms241310747] [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] [Received: 05/24/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
There is an increasing interest in understanding the connection between the immune and cardiovascular systems, which are highly integrated and communicate through finely regulated cross-talking mechanisms. Recent evidence has demonstrated that the immune system does indeed have a key role in the response to cardiac injury and in cardiac regeneration. Among the immune cells, macrophages appear to have a prominent role in this context, with different subtypes described so far that each have a specific influence on cardiac remodeling and repair. Similarly, there are significant differences in how the innate and adaptive immune systems affect the response to cardiac damage. Understanding all these mechanisms may have relevant clinical implications. Several studies have already demonstrated that stem cell-based therapies support myocardial repair. However, the exact role that cardiac macrophages and their modulation may have in this setting is still unclear. The current need to decipher the dual role of immunity in boosting both heart injury and repair is due, at least for a significant part, to unresolved questions related to the complexity of cardiac macrophage phenotypes. The aim of this review is to provide an overview on the role of the immune system, and of macrophages in particular, in the response to cardiac injury and to outline, through the modulation of the immune response, potential novel therapeutic strategies for cardiac regeneration.
Collapse
Affiliation(s)
- Claudia Molinaro
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Isabella Leo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Jolanda Sabatino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Nadia Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
40
|
Galasso L, Cappella A, Mulè A, Castelli L, Ciorciari A, Stacchiotti A, Montaruli A. Polyamines and Physical Activity in Musculoskeletal Diseases: A Potential Therapeutic Challenge. Int J Mol Sci 2023; 24:9798. [PMID: 37372945 DOI: 10.3390/ijms24129798] [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: 05/10/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Autophagy dysregulation is commonplace in the pathogenesis of several invalidating diseases, such as musculoskeletal diseases. Polyamines, as spermidine and spermine, are small aliphatic cations essential for cell growth and differentiation, with multiple antioxidant, anti-inflammatory, and anti-apoptotic effects. Remarkably, they are emerging as natural autophagy regulators with strong anti-aging effects. Polyamine levels were significantly altered in the skeletal muscles of aged animals. Therefore, supplementation of spermine and spermidine may be important to prevent or treat muscle atrophy. Recent in vitro and in vivo experimental studies indicate that spermidine reverses dysfunctional autophagy and stimulates mitophagy in muscles and heart, preventing senescence. Physical exercise, as polyamines, regulates skeletal muscle mass inducing proper autophagy and mitophagy. This narrative review focuses on the latest evidence regarding the efficacy of polyamines and exercise as autophagy inducers, alone or coupled, in alleviating sarcopenia and aging-dependent musculoskeletal diseases. A comprehensive description of overall autophagic steps in muscle, polyamine metabolic pathways, and effects of the role of autophagy inducers played by both polyamines and exercise has been presented. Although literature shows few data in regard to this controversial topic, interesting effects on muscle atrophy in murine models have emerged when the two "autophagy-inducers" were combined. We hope these findings, with caution, can encourage researchers to continue investigating in this direction. In particular, if these novel insights could be confirmed in further in vivo and clinical studies, and the two synergic treatments could be optimized in terms of dose and duration, then polyamine supplementation and physical exercise might have a clinical potential in sarcopenia, and more importantly, implications for a healthy lifestyle in the elderly population.
Collapse
Affiliation(s)
- Letizia Galasso
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Annalisa Cappella
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
- U.O. Laboratorio di Morfologia Umana Applicata, I.R.C.C.S. Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Antonino Mulè
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Lucia Castelli
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Andrea Ciorciari
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Alessandra Stacchiotti
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
- U.O. Laboratorio di Morfologia Umana Applicata, I.R.C.C.S. Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Angela Montaruli
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
- I.R.C.C.S. Ospedale Galeazzi-Sant'Ambrogio, 20157 Milan, Italy
| |
Collapse
|
41
|
Odame E, Li L, Nabilla JA, Cai H, Xiao M, Ye J, Chen Y, Kyei B, Dai D, Zhan S, Cao J, Guo J, Zhong T, Wang L, Zhang H. miR-145-3p Inhibits MuSCs Proliferation and Mitochondria Mass via Targeting MYBL1 in Jianzhou Big-Eared Goats. Int J Mol Sci 2023; 24:ijms24098341. [PMID: 37176056 PMCID: PMC10179409 DOI: 10.3390/ijms24098341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/30/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Muscle growth and injury-induced regeneration are controlled by skeletal muscle satellite cells (MuSCs) through myogenesis in postnatal animals. Meanwhile, myogenesis is accompanied by mitochondrial function and enzyme activity. Nevertheless, the underlying molecular mechanisms involving non-coding RNAs including circular RNAs (circRNAs) and microRNAs (miRNAs) remain largely unsolved. Here, we explored the myogenic roles of miR-145-3p and MYBL1 on muscle development and mitochondrial mass. We noticed that overexpression of miR-145-3p inhibited MuSCs proliferation and reduced the number of viable cells. Meanwhile, deficiency of miR-145-3p caused by LNAantimiR-145-3p or an inhibitor retarded the differentiation of MuSCs. miR-145-3p altered the mitochondrial mass in MuSCs. Moreover, miR-145-3p targeted and negatively regulated the expression of CDR1as and MYBL1. The knockdown of the MYBL1 using ASO-2'MOE modification simulated the inhibitory function of miR-145-3p on cell proliferation. Additionally, MYBL1 mediated the regulation of miR-145-3p on Vexin, VCPIP1, COX1, COX2, and Pax7. These imply that CDR1as/miR-145-3p/MYBL1/COX1, COX2, VCPIP1/Vexin expression at least partly results in a reduction in mitochondrial mass and MuSCs proliferation. These novel findings confirm the importance of mitochondrial mass during myogenesis and the boosting of muscle/meat development in mammals.
Collapse
Affiliation(s)
- Emmanuel Odame
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Joshua Abdulai Nabilla
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - He Cai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Miao Xiao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiangfeng Ye
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuan Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Bismark Kyei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Dinghui Dai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Siyuan Zhan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiaxue Cao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiazhong Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Tao Zhong
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Linjie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
42
|
Park J, Lee J, Shim K. Effects of heat stress exposure on porcine muscle satellite cells. J Therm Biol 2023; 114:103569. [PMID: 37344027 DOI: 10.1016/j.jtherbio.2023.103569] [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: 07/15/2022] [Revised: 04/06/2023] [Accepted: 04/08/2023] [Indexed: 06/23/2023]
Abstract
Heat stress (HS) affects cell culture as well as animal production. Although there have been many reports on the disparate effects of heat stress, its effects on mammalian muscle stem cells are still unclear. In this study, we isolated porcine muscle satellite cells (PMSCs) from the femurs of 1-day-old piglets, and cultured them under three temperature conditions: 37 °C, 39 °C, and 41 °C. Exposure to HS not only decreased the viability and proliferation rates of PMSCs, but also regulated the cell cycle and induced apoptosis. High-temperature culture conditions decreased both protein and gene expression of Pax7, a proliferation and maintenance marker of muscle satellite cells, whereas it increased both protein and gene expression of MyoG, a differentiation marker, and promoted myotube formation in the early stage of differentiation induction. In addition, the protein and gene expression of several heat shock proteins (HSPs) in PMSCs increased due to heat treatment. In conclusion, HS induced the cell cycle arrest of PMSCs, thereby reducing the proliferation rate. In addition, high-temperature culture conditions promoted the formation of myotubes at the early stage of differentiation of PMSCs without additives.
Collapse
Affiliation(s)
- Jinryong Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, South Korea; 3D Tissue Culture Research Center, Konkuk University, Seoul, 05029, South Korea
| | - Jeongeun Lee
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Kwanseob Shim
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, 54896, South Korea; Department of Animal Biotechnology, Jeonbuk National University, Jeonju, 54896, South Korea.
| |
Collapse
|
43
|
Suroto H, Wardana GR, Sugianto JA, Aprilya D, Samijo S. Time to surgery and myo-d expression in biceps muscle of adult brachial plexus injury: a preliminary study. BMC Res Notes 2023; 16:51. [PMID: 37055794 PMCID: PMC10103435 DOI: 10.1186/s13104-023-06317-y] [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] [Received: 08/22/2022] [Accepted: 03/27/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Brachial Plexus Injury (BPI) is one of the peripheral nerve injuries which causes severe functional impairment and disability. Without prompt treatment, prolonged denervation will cause severe muscle atrophy. MyoD, which is expressed by satellite cells, is one of the parameters that relate to the regeneration process in post-injury muscle and it is presumed to determine the clinical outcome following neurotization procedure. This study aims to understand the correlation between time to surgery (TTS) and MyoD expression in satellite cells in the biceps muscle of adult brachial plexus injury patients. METHODS Analytic observational study with a cross-sectional design was conducted at Dr. Soetomo General Hospital. All patients with BPI who underwent surgery between May 2013 and December 2015 were included. Muscle biopsy was taken and stained using immunohistochemistry for MyoD expression. Pearson correlation test was used to assess the correlation between MyoD expression with TTS and with age. RESULTS Twenty-two biceps muscle samples were examined. Most patients are males (81.8%) with an average age of 25.5 years. MyoD expression was found to be highest at TTS of 4 months and then dropped significantly (and plateau) from 9 to 36 months. MyoD expression is significantly correlated with TTS (r=-0.895; p = 0.00) but not with age (r=-0.294; p = 0.184). CONCLUSION Our study found, from the cellular point of view, that treatment of BPI needs to be done as early as possible before the regenerative potential - as indicated by MyoD expression - declined.
Collapse
Affiliation(s)
- Heri Suroto
- Department of Orthopaedic & Traumatology, Faculty of Medicine, Universitas Airlangga/Dr. Soetomo General Hospital, Surabaya, 60132, Indonesia.
- Cell and Tissue Bank-Regenerative Medicine, Faculty of Medicine, Dr Soetomo General Academic Hospital, Universitas Airlangga, Surabaya, 60132, Indonesia.
| | - Gestana Retaha Wardana
- Department of Orthopaedic & Traumatology, Faculty of Medicine, Universitas Airlangga/Dr. Soetomo General Hospital, Surabaya, 60132, Indonesia
| | - Julius Albert Sugianto
- Department of Orthopaedic & Traumatology, Faculty of Medicine, Universitas Airlangga/Dr. Soetomo General Hospital, Surabaya, 60132, Indonesia
| | - Dina Aprilya
- Orthopedic and Traumatology Department, Siloam Agora Hospital, Jakarta, Indonesia
| | | |
Collapse
|
44
|
Su Y, Gao X, Wang Y, Li X, Zhang W, Zhao J. Astragalus polysaccharide promotes sheep satellite cell differentiation by regulating miR-133a through the MAPK/ERK signaling pathway. Int J Biol Macromol 2023; 239:124351. [PMID: 37023880 DOI: 10.1016/j.ijbiomac.2023.124351] [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: 12/12/2022] [Revised: 03/21/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Astragalus polysaccharide (APS) possesses extensive biological activities, pharmacological effects, and anti-fatigue function. MiR-133a is a specifically expressed miRNA in skeletal muscle that participates in the regulation of myoblast proliferation and differentiation. However, little is known about the role of APS in the development of sheep skeletal muscle. In this study, we aimed to investigate the underlying mechanism of APS and miR-133a on the differentiation of sheep skeletal muscle satellite cells (SMSCs) and the regulatory relationship between APS and miR-133a. The results suggested that APS plays a positive regulatory role in the proliferation and differentiation of sheep SMSCs. Moreover, miR-133a significantly promotes SMSC differentiation and the activity of the MAPK/ERK signaling pathway. Importantly, we found that APS function requires the mediation of miR-133a in the differentiation of sheep SMSCs. Taken together, our results indicate that APS accelerates SMSC differentiation by regulating miR-133a via the MAPK/ERK signaling pathway in sheep.
Collapse
Affiliation(s)
- Yuan Su
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China
| | - Xuyang Gao
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China
| | - Yu Wang
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China
| | - Xuying Li
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China
| | - Weipeng Zhang
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China
| | - Junxing Zhao
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China.
| |
Collapse
|
45
|
Hirota J, Hasegawa T, Inui A, Takeda D, Amano-Iga R, Yatagai N, Saito I, Arimoto S, Akashi M. Local application of a transcutaneous carbon dioxide paste prevents excessive scarring and promotes muscle regeneration in a bupivacaine-induced rat model of muscle injury. Int Wound J 2023; 20:1151-1159. [PMID: 36250918 PMCID: PMC10031219 DOI: 10.1111/iwj.13974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 11/29/2022] Open
Abstract
In postoperative patients with head and neck cancer, scar tissue formation may interfere with the healing process, resulting in incomplete functional recovery and a reduced quality of life. Percutaneous application of carbon dioxide (CO2 ) has been reported to improve hypoxia, stimulate angiogenesis, and promote fracture repair and muscle damage. However, gaseous CO2 cannot be applied to the head and neck regions. Previously, we developed a paste that holds non-gaseous CO2 in a carrier and can be administered transdermally. Here, we investigated whether this paste could prevent excessive scarring and promote muscle regeneration using a bupivacaine-induced rat model of muscle injury. Forty-eight Sprague Dawley rats were randomly assigned to either a control group or a CO2 group. Both groups underwent surgery to induce muscle injury, but the control group received no treatment, whereas the CO2 group received the CO2 paste daily after surgery. Then, samples of the experimental sites were taken on days 3, 7, 14, and 21 post-surgery to examine the following: (1) inflammatory (interleukin [IL]-1β, IL-6), and transforming growth factor (TGF)-β and myogenic (MyoD and myogenin) gene expression by polymerase chain reaction, (2) muscle regeneration with haematoxylin and eosin staining, and (3) MyoD and myogenin protein expression using immunohistochemical staining. Rats in the CO2 group showed higher MyoD and myogenin expression and lower IL-1β, IL-6, and TGF-β expression than the control rats. In addition, treated rats showed evidence of accelerated muscle regeneration. Our study demonstrated that the CO2 paste prevents excessive scarring and accelerates muscle regeneration. This action may be exerted through the induction of an artificial Bohr effect, which leads to the upregulation of MyoD and myogenin, and the downregulation of IL-1β, IL-6, and TGF-β. The paste is inexpensive and non-invasive. Thus, it may be the treatment of choice for patients with muscle damage.
Collapse
Affiliation(s)
- Junya Hirota
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takumi Hasegawa
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Atsuyuki Inui
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Daisuke Takeda
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Rika Amano-Iga
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Nanae Yatagai
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Izumi Saito
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satomi Arimoto
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masaya Akashi
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
46
|
Nicholson TA, Sagmeister M, Wijesinghe SN, Farah H, Hardy RS, Jones SW. Oligonucleotide Therapeutics for Age-Related Musculoskeletal Disorders: Successes and Challenges. Pharmaceutics 2023; 15:237. [PMID: 36678864 PMCID: PMC9866666 DOI: 10.3390/pharmaceutics15010237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
Age-related disorders of the musculoskeletal system including sarcopenia, osteoporosis and arthritis represent some of the most common chronic conditions worldwide, for which there remains a great clinical need to develop safer and more efficacious pharmacological treatments. Collectively, these conditions involve multiple tissues, including skeletal muscle, bone, articular cartilage and the synovium within the joint lining. In this review, we discuss the potential for oligonucleotide therapies to combat the unmet clinical need in musculoskeletal disorders by evaluating the successes of oligonucleotides to modify candidate pathological gene targets and cellular processes in relevant tissues and cells of the musculoskeletal system. Further, we discuss the challenges that remain for the clinical development of oligonucleotides therapies for musculoskeletal disorders and evaluate some of the current approaches to overcome these.
Collapse
Affiliation(s)
- Thomas A. Nicholson
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Michael Sagmeister
- Institute for Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Susanne N. Wijesinghe
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Hussein Farah
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Rowan S. Hardy
- Institute for Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Simon W. Jones
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
47
|
Park J, Choi H, Shim K. Inhibition of GSK3β Promotes Proliferation and Suppresses Apoptosis of Porcine Muscle Satellite Cells. Animals (Basel) 2022; 12:ani12233328. [PMID: 36496849 PMCID: PMC9738253 DOI: 10.3390/ani12233328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/17/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
As the global population increases, interest in cultured meat (a new research field) is gradually increasing. The main raw material for the production of cultured meat is muscle stem cells called satellite cells isolated from livestock. However, how to mass proliferate and maintain satellite cells in vitro without genetic manipulation remains unclear. In the present study, we isolated and purified porcine muscle satellite cells (PMSCs) from the femur of a 1-day-old piglet and cultured PMSCs by treating them with an inhibitor (XAV939, Tankyrase (TNKS) inhibitor) or an activator (CHIR99021, glycogen synthase kinase 3 beta (GSK3β) inhibitor) of Wnt signaling. The CHIR group treated with 3 μM CHIR99021 showed a significantly increased proliferation rate of PMSCs compared to the SC group (control), whereas the XAV group treated with 1 μM XAV939 showed a significantly decreased proliferation rate of PMSCs. CHIR99021 also inhibited the differentiation of PMSCs by reducing the expression of MyoD while maintaining the expression of Pax7 and suppressed apoptosis by regulating the expression of apoptosis-related proteins and genes. RNA sequencing was performed to obtain gene expression profiles following inhibition or activation of the Wnt signaling pathway and various signaling mechanisms related to the maintenance of satellite cells were identified. Our results suggest that inhibition of GSK3β could dramatically improve the maintenance and mass proliferation ability of PMSCs in vitro by regulating the expression of myogenic markers and the cell cycle.
Collapse
Affiliation(s)
- Jinryong Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
- 3D Tissue Culture Research Center, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyunwoo Choi
- Department of Animal Science, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Kwanseob Shim
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Correspondence: ; Tel.: +82-063-270-2609
| |
Collapse
|
48
|
Lactate Activates AMPK Remodeling of the Cellular Metabolic Profile and Promotes the Proliferation and Differentiation of C2C12 Myoblasts. Int J Mol Sci 2022; 23:ijms232213996. [PMID: 36430479 PMCID: PMC9694550 DOI: 10.3390/ijms232213996] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/02/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Lactate is a general compound fuel serving as the fulcrum of metabolism, which is produced from glycolysis and shuttles between different cells, tissues and organs. Lactate is usually accumulated abundantly in muscles during exercise. It remains unclear whether lactate plays an important role in the metabolism of muscle cells. In this research, we assessed the effects of lactate on myoblasts and clarified the underlying metabolic mechanisms through NMR-based metabonomic profiling. Lactate treatment promoted the proliferation and differentiation of myoblasts, as indicated by significantly enhanced expression levels of the proteins related to cellular proliferation and differentiation, including p-AKT, p-ERK, MyoD and myogenin. Moreover, lactate treatment profoundly regulated metabolisms in myoblasts by promoting the intake and intracellular utilization of lactate, activating the TCA cycle, and thereby increasing energy production. For the first time, we found that lactate treatment evidently promotes AMPK signaling as reflected by the elevated expression levels of p-AMPK and p-ACC. Our results showed that lactate as a metabolic regulator activates AMPK, remodeling the cellular metabolic profile, and thereby promoting the proliferation and differentiation of myoblasts. This study elucidates molecular mechanisms underlying the effects of lactate on skeletal muscle in vitro and may be of benefit to the exploration of lactate acting as a metabolic regulator.
Collapse
|
49
|
Chen W, Chen W, Liu P, Qian S, Tao S, Huang M, Xu W, Li C, Chen X, Lin H, Qin Z, Lu J, Xie S. Role of lncRNA Has2os in Skeletal Muscle Differentiation and Regeneration. Cells 2022; 11:3497. [PMID: 36359891 PMCID: PMC9655701 DOI: 10.3390/cells11213497] [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: 08/19/2022] [Revised: 10/28/2022] [Accepted: 11/03/2022] [Indexed: 09/26/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) regulate a series of physiological processes and play an important role in development, metabolism and disease. Our previous studies showed that lncRNAs involved in skeletal muscle differentiation. Here, we demonstrated that lncRNA Has2os is highly expressed in skeletal muscle and significantly elevated during skeletal cell differentiation. The knockdown of Has2os inhibited myocyte fusion and impeded the expression of the myogenic factors MyHC and Mef2C. Mechanically, Has2os regulates skeletal muscle differentiation by inhibiting the JNK/MAPK signaling pathway. Furthermore, we also revealed that Has2os is involved in the early stage of regeneration after muscle injury, and the JNK/MAPK signaling pathway is activated at both protein and mRNA levels during early repair. Our results demonstrate the new function of lncRNA Has2os, which plays crucial roles during skeletal muscle differentiation and muscle regeneration, providing a basis for the therapy of lncRNA-related muscle diseases.
Collapse
Affiliation(s)
- Wanxin Chen
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Weicai Chen
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Peng Liu
- Laboratory Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Shiyu Qian
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Shuang Tao
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Mengchun Huang
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Wanyi Xu
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Cuiping Li
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Xiaoyan Chen
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Huizhu Lin
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Zhenshu Qin
- Department of Trauma Orthopaedics, Chenzhou First People’s Hospital Affiliated to South China University, Chenzhou 423000, China
| | - Jianxi Lu
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Shujuan Xie
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
- Vaccine Research Institute of Sun Yat-Sen University, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| |
Collapse
|
50
|
Lee J, Park J, Choe H, Shim K. Insect peptide CopA3 promotes proliferation and PAX7 and MYOD expression in porcine muscle satellite cells. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:1132-1143. [PMID: 36812017 PMCID: PMC9890342 DOI: 10.5187/jast.2022.e81] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/27/2022] [Accepted: 10/04/2022] [Indexed: 12/14/2022]
Abstract
Insects are a valuable natural source that can produce a variety of bioactive compounds due to their increasing species diversity. CopA3 is an antimicrobial peptide derived from Copris tripartitus (i.e., the dung beetle). It is known to increase the proliferation of colonic epithelial and neuronal stem cells by regulating cell cycle. This research hypothesized that CopA3 can promote the proliferation of porcine muscle satellite cells (MSCs). The effects of CopA3 on porcine MSCs, which are important for muscle growth and regeneration, remain unclear. Here, we investigated the effects of CopA3 on porcine MSCs. According to viability results, we designed four groups: control (without CopA3) and three treatment groups (treated with 5,10, and 25 μg/mL of CopA3). At a CopA3 concentration of 5 μg/mL and 10 μg/mL, the proliferation of MSCs increased more than that observed in the control group. Furthermore, compared to that in the control, CopA3 treatment increased the S phase but decreased the G0/G1 phase ratio. Additionally, early and late apoptotic cells were found to be decreased in the 5 μg/mL group. The expressions of the myogenesis-related transcription factor PAX7 and MYOD proteins were significantly upregulated in the 5 μg/mL and 10 μg/mL groups, whereas the MYOG protein remained undetected in all group. This study suggested that CopA3 promotes muscle cell proliferation by regulating the cell cycle of MSCs and can regulate the activity of MSCs by increasing the expressions of PAX7 and MYOD.
Collapse
Affiliation(s)
- Jeongeun Lee
- Department of Agricultural Convergence
Technology, Jeonbuk National University, Jeonju 54896,
Korea
| | - Jinryoung Park
- Department of Stem Cell and Regenerative
Biotechnology, Konkuk University, Seoul 06591, Korea,3D Tissue Culture Research Center, Konkuk
University, Seoul 06591, Korea
| | - Hosung Choe
- Department of Animal Biotechnology,
Jeonbuk National University, Jeonju 54896, Korea
| | - Kwanseob Shim
- Department of Agricultural Convergence
Technology, Jeonbuk National University, Jeonju 54896,
Korea,Department of Animal Biotechnology,
Jeonbuk National University, Jeonju 54896, Korea,Corresponding author: Kwanseob Shim,
Department of Agricultural Convergence Technology, Jeonbuk National University,
Jeonju 54896, Korea. Tel: +82-63-270-2609, E-mail:
| |
Collapse
|