1
|
Huot JR, Jamnick NA, Pin F, Livingston PD, Callaway CS, Bonetto A. GL261 glioblastoma induces delayed body weight gain and stunted skeletal muscle growth in young mice. Am J Physiol Regul Integr Comp Physiol 2025; 328:R628-R641. [PMID: 40247678 DOI: 10.1152/ajpregu.00035.2025] [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: 02/12/2025] [Revised: 02/26/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
The survival rate for children and adolescents has increased to over 85%. However, there is limited understanding of the impact of pediatric cancers on muscle development and physiology. Given that brain tumors alone account for 26% of all pediatric cancers, this study aimed to investigate the skeletal muscle consequences of tumor growth in young mice. C2C12 myotubes were cocultured with GL261 murine glioblastoma cells to assess myotube size. GL261 cells were then injected subcutaneously into 4-wk-old male C57BL/6J mice. Animals were euthanized 28 days post-GL261 implantation. Muscle function was tested in vivo and ex vivo. Muscle protein synthesis was estimated via the SUnSET method, and gene/protein expression levels were assessed via Western blotting and qPCR. In vitro, the C2C12 cultures exposed to GL261 exhibited myotube atrophy, consistent with a disrupted anabolic/catabolic balance. In vivo, carcass, heart, and fat mass were significantly reduced in the tumor-bearing mice. Skeletal muscle growth was impeded in the GL261 hosts, along with a smaller muscle cross-sectional area (CSA). Both in vivo muscle torque and the ex vivo Extensor Digitorum Longus (EDL) muscle force were unchanged. At molecular level, the tumor hosts displayed reduced estimations of muscle protein synthesis and increased muscle protein ubiquitination, in disagreement with decreased muscle ubiquitin ligase mRNA expression. Overall, we showed that GL261 tumors impact the growth of pediatric mice by stunting skeletal muscle development, decreasing muscle mass, reducing muscle fiber size, diminishing muscle protein synthesis, and altering protein catabolism signaling.NEW & NOTEWORTHY This study shows that pediatric brain tumors stunt muscle development in young mice. GL261 glioblastoma cells caused myotube atrophy, reduced carcass, heart, and fat mass, and impeded skeletal muscle growth. Tumor-bearing mice had decreased muscle protein synthesis and increased protein ubiquitination. This is the first demonstration that GL261 tumors reduce muscle mass and fiber size, impair muscle function and innervation, and alter muscle protein turnover.
Collapse
Affiliation(s)
- Joshua R Huot
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Department of Kinesiology, School of Health and Human Sciences, Indiana University Purdue University Indianapolis, Indianapolis, Indiana, United States
| | - Nicholas A Jamnick
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Patrick D Livingston
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Chandler S Callaway
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Andrea Bonetto
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Nutrition Obesity Research Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
2
|
Wu J, Tang J, Huang D, Wang Y, Zhou E, Ru Q, Xu G, Chen L, Wu Y. Study on the comorbid mechanisms of sarcopenia and late-life depression. Behav Brain Res 2025; 485:115538. [PMID: 40122287 DOI: 10.1016/j.bbr.2025.115538] [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: 10/15/2024] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
The increasing global aging population has brought greater focus to age-related diseases, particularly muscle-brain comorbidities such as sarcopenia and late-life depression. Sarcopenia, defined by the gradual loss of muscle mass and function, is notably prevalent among older individuals, while late-life depression profoundly affects their mental health and overall well-being. Epidemiological evidence suggests a high co-occurrence of these two conditions, although the precise biological mechanisms linking them remain inadequately understood. This review synthesizes the existing body of literature on sarcopenia and late-life depression, examining their definitions, prevalence, clinical presentations, and available treatments. The goal is to clarify the potential connections between these comorbidities and offer a theoretical framework for the development of future preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Jiale Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Jun Tang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Di Huang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yu Wang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Enyuan Zhou
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
3
|
Yang S, Xiong L, Liao T, Li L, Li Y, Kang L, Yang G, Liang Z. Deubiquitinating Enzyme USP2 Alleviates Muscle Atrophy by Stabilizing PPAR-γ. Diabetes 2025; 74:773-786. [PMID: 39874418 PMCID: PMC12015143 DOI: 10.2337/db24-0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 01/23/2025] [Indexed: 01/30/2025]
Abstract
Insulin resistance, a hallmark of type 2 diabetes, accelerates muscle breakdown and impairs energy metabolism. However, the role of ubiquitin specific peptidase 2 (USP2), a key regulator of insulin resistance, in sarcopenia remains unclear. Peroxisome proliferator-activated receptor γ (PPAR-γ) plays a critical role in regulating muscle atrophy. The role of deubiquitinase USP2 in mitigating muscle atrophy was investigated. Our findings revealed reduced USP2 expression in skeletal muscles of patients with type 2 diabetes. In mouse models of diabetes- and dexamethasone (DEX)-induced muscle atrophy, USP2 expression was downregulated in skeletal muscles. Usp2 knockout exacerbated muscle loss and functional impairment induced by diabetes or DEX. Moreover, skeletal muscle-specific Usp2 knockout further aggravated muscle loss and functional impairment induced by diabetes. Local injection of adeno-associated virus-Usp2 into the gastrocnemius muscles of diabetic mice increased muscle mass and improved skeletal muscle performance and endurance. It enhanced insulin sensitivity in diabetic mice, shown by lower fasting serum glucose and insulin levels and better glucose tolerance. Mechanistic analysis showed USP2 directly interacted with PPAR-γ by deubiquitinating it, stabilizing its protein levels, enhancing insulin signaling and sensitivity, and maintaining muscle mass. Loss of PPAR-γ abolishes the regulatory effects of USP2 on insulin sensitivity and muscle atrophy. MYOD1 activates USP2 transcription by binding to its promoter region. This study demonstrates the protective role of USP2 in mitigating muscle atrophy by stabilizing PPAR-γ through deubiquitination, particularly in models of diabetic and DEX-induced muscle atrophy. Targeting the USP2-PPAR-γ axis may offer promising therapeutic strategies for metabolic disorders and sarcopenia. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Shu Yang
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Lijiao Xiong
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Tingfeng Liao
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Lixing Li
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Yanchun Li
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Lin Kang
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- The Biobank of National Innovation Center for Advanced Medical Devices, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Guangyan Yang
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Zhen Liang
- Department of Geriatrics, Peking University Shenzhen Hospital, Shenzhen, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| |
Collapse
|
4
|
Zhao X, Zhu M, Wang Z, Gao M, Long Y, Zhou S, Wang W. The Alleviative Effect of Sodium Butyrate on Dexamethasone-Induced Skeletal Muscle Atrophy. Cell Biol Int 2025; 49:508-521. [PMID: 39936899 DOI: 10.1002/cbin.70003] [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: 07/03/2024] [Revised: 01/18/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Skeletal muscle mass is significantly negatively regulated by glucocorticoids. Following glucocorticoid administration, the balance between protein synthesis and breakdown in skeletal muscle is disrupted, shifting towards a predominance of catabolic metabolism. Short-chain fatty acids like sodium butyrate have been found to regulate inflammatory reactions and successively activate signaling pathways. The preventive benefits of sodium butyrate against dexamethasone-induced skeletal muscle atrophy and myotube atrophy models were examined in this work, and the underlying mechanism was clarified. A total of 32 6-week-old C57BL/6 inbred male mice were randomly assigned to one of four groups and treated with dexamethasone to induce muscle atrophy and sodium butyrate. We found that sodium succinate alleviated dexamethasone-induced myotube atrophy in the myotube atrophy model by lowering the gene expression of two E3 ubiquitin ligases, Atrogin-1 and MURF1, and activating the AKT/mTOR signaling pathway. Pertussis toxin reversed this effect, indicating that G protein-coupled receptors were involved in sodium butyrate's action as a mediator. Additionally, pre-treatment with sodium butyrate lowered weight and muscle mass loss in a mouse model of skeletal muscle atrophy, dramatically decreased the MURF1 gene expression and decreased the nuclear translocation of the glucocorticoid receptor. In conclusion, this study shows that sodium butyrate inhibits the expression of atrophy genes, thus preventing the breakdown of proteins and the loss of muscle mass, while also inhibiting weight loss, in animal models.
Collapse
Affiliation(s)
- Xingchen Zhao
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Mingqiang Zhu
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Zifan Wang
- College of Animal Science and Veterinary, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Ming Gao
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Yifei Long
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Shuo Zhou
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Wei Wang
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
- College of Animal Science and Veterinary, Shenyang Agricultural University, Shenyang, Liaoning, China
| |
Collapse
|
5
|
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] [Download PDF] [Figures] [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
|
6
|
Cho CH, Chae SH, Thi NHL, Um SH, Lee S, Yu JS, Kang KS, Kim KH. Lambertianic Acid from Platycladus orientalis Inhibits Muscle Atrophy in Dexamethasone-Induced C2C12 Muscle Atrophy Cells. PLANTS (BASEL, SWITZERLAND) 2025; 14:1357. [PMID: 40364384 DOI: 10.3390/plants14091357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
Platycladus orientalis, an evergreen tree belonging to the Cupressaceae family, has been traditionally used to treat various ailments, including fever, cough, diarrhea, diuresis, cold symptoms, and gastrointestinal disorders in folk medicine. As part of our ongoing investigation aimed at discovering bioactive natural products and elucidating their mechanisms of action from various natural sources, we investigated a methanol (MeOH) extract of P. orientalis leaves. This investigation led to the isolation and identification of a labdane-type diterpene, lambertianic acid (LA), via column chromatography and HPLC purification. The structure of LA was elucidated using LC/MS and NMR spectroscopic analyses, including HR-ESIMS, while its absolute configuration was confirmed through electronic circular dichroism (ECD) calculations. Recent studies have reported that labdane-type diterpenes exhibit diverse pharmacological activities, such as anticancer, anti-inflammatory, anti-obesity, and hypolipidemic effects. Notably, LA has been shown to modulate adipocyte metabolism via AMPK signaling; however, its role in skeletal muscle atrophy remains unexplored. Therefore, in this study, we investigated the effects of LA on dexamethasone (Dex)-induced muscle atrophy in C2C12 myotubes. Treatment with LA at concentrations of 25 µM and 50 µM significantly rescued myotube diameter and reduced the expression of atrophy-related proteins, including MuRF-1 and atrogin-1/MAFbx, without compromising cell viability at these moderate concentrations. These findings suggest that LA derived from P. orientalis exerts protective effects against skeletal muscle atrophy, highlighting its potential as a promising natural therapeutic candidate for muscle-wasting disorders.
Collapse
Affiliation(s)
- Chan Hee Cho
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Si Hyeon Chae
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ngoc Han Le Thi
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Sung Hee Um
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seulah Lee
- Department of Oriental Medicine Biotechnology, Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Jae Sik Yu
- Department of Integrative Biological Sciences and Industry, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
7
|
Oyabu M, Ohira Y, Fujita M, Yoshioka K, Kawaguchi R, Kubo A, Hatazawa Y, Yukitoshi H, Ortuste Quiroga HP, Horii N, Miura F, Araki H, Okano M, Hatada I, Gotoh H, Yoshizawa T, Fukada SI, Ogawa Y, Ito T, Ishihara K, Ono Y, Kamei Y. Dnmt3a overexpression disrupts skeletal muscle homeostasis, promotes an aging-like phenotype, and reduces metabolic elasticity. iScience 2025; 28:112144. [PMID: 40151644 PMCID: PMC11937683 DOI: 10.1016/j.isci.2025.112144] [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: 09/27/2023] [Revised: 05/10/2024] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Mammalian aging is reportedly driven by the loss of epigenetic information; however, its impact on skeletal muscle aging remains unclear. This study shows that aging mouse skeletal muscle exhibits increased DNA methylation, and overexpression of DNA methyltransferase 3a (Dnmt3a) induces an aging-like phenotype. Muscle-specific Dnmt3a overexpression leads to an increase in central nucleus-positive myofibers, predominantly in fast-twitch fibers, a shift toward slow-twitch fibers, elevated inflammatory and senescence markers, mitochondrial OXPHOS complex I reduction, and decreased basal autophagy. Dnmt3a overexpression resulted in reduced muscle mass and strength and impaired endurance exercise capacity with age, accompanied by an enhanced inflammatory signature. In addition, Dnmt3a overexpression reduced not only sensitivity to starvation-induced muscle atrophy but also the restorability from muscle atrophy. These findings suggest that increased DNA methylation disrupts skeletal muscle homeostasis, promotes an aging-like phenotype, and reduces muscle metabolic elasticity.
Collapse
Affiliation(s)
- Mamoru Oyabu
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Yuto Ohira
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Mariko Fujita
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Kiyoshi Yoshioka
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
- Institute for Research on Productive Aging (IRPA), Tokyo, Japan
| | - Runa Kawaguchi
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Atsushi Kubo
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yukino Hatazawa
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Hinako Yukitoshi
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Huascar Pedro Ortuste Quiroga
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Naoki Horii
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Fumihito Miura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiromitsu Araki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Masaki Okano
- Department of Pluripotent Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Izuho Hatada
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
- Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Maebashi 371-8511, Japan
| | - Hitoshi Gotoh
- Cell Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 606-0823, Japan
| | - Tatsuya Yoshizawa
- Cell Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 606-0823, Japan
| | - So-ichiro Fukada
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Ito
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Kengo Ishihara
- Department of Food Science and Human Nutrition, Faculty of Agriculture, Ryukoku University, Shiga 520-2194, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Yasutomi Kamei
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| |
Collapse
|
8
|
Ding W, Gong W, Bou T, Shi L, Lin Y, Wu H, Dugarjaviin M, Bai D. Pilot Study on the Profiling and Functional Analysis of mRNA, miRNA, and lncRNA in the Skeletal Muscle of Mongolian Horses, Xilingol Horses, and Grassland-Thoroughbreds. Animals (Basel) 2025; 15:1123. [PMID: 40281957 PMCID: PMC12024394 DOI: 10.3390/ani15081123] [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: 03/01/2025] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Muscle fibers, as the fundamental units of muscle tissue, play a crucial role in determining skeletal muscle function through their growth, development, and composition. To investigate changes in muscle fiber types and their regulatory mechanisms in Mongolian horses (MG), Xilingol horses (XL), and Grassland-Thoroughbreds (CY), we conducted histological and bioinformatic analyses on the gluteus medius muscle of these three horse breeds. Immunofluorescence analysis revealed that Grassland-Thoroughbreds had the highest proportion of fast-twitch muscle fibers at 78.63%, while Mongolian horses had the lowest proportion at 57.54%. Whole-transcriptome analysis identified 105 differentially expressed genes (DEGs) in the CY vs. MG comparison and 104 DEGs in the CY vs. XL comparison. Time-series expression profiling grouped the DEGs into eight gene sets, with three sets showing significantly up-regulated or down-regulated expression patterns (p < 0.05). Additionally, 280 differentially expressed long non-coding RNAs (DELs) were identified in CY vs. MG, and 213 DELs were identified in CY vs. XL. A total of 32 differentially expressed microRNAs (DEMIRs) were identified in CY vs. MG, while 44 DEMIRs were found in CY vs. XL. Functional enrichment analysis indicated that the DEGs were significantly enriched in essential biological processes, such as actin filament organization, muscle contraction, and protein phosphorylation. KEGG pathway analysis showed their involvement in key signaling pathways, including the mTOR signaling pathway, FoxO signaling pathway, and HIF-1 signaling pathway. Furthermore, functional variation-based analyses revealed associations between non-coding RNAs and mRNAs, with some non-coding RNAs targeting genes potentially related to muscle function regulation. These findings provide valuable insights into the molecular basis for the environmental adaptability, athletic performance, and muscle characteristics in horses, offering new perspectives for the breeding of Grassland-Thoroughbreds.
Collapse
Affiliation(s)
- Wenqi Ding
- Key Laboratory of Equus Germplasm Innovation (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (W.D.); (W.G.); (T.B.); (L.S.); (Y.L.); (H.W.); (M.D.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Wendian Gong
- Key Laboratory of Equus Germplasm Innovation (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (W.D.); (W.G.); (T.B.); (L.S.); (Y.L.); (H.W.); (M.D.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Tugeqin Bou
- Key Laboratory of Equus Germplasm Innovation (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (W.D.); (W.G.); (T.B.); (L.S.); (Y.L.); (H.W.); (M.D.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lin Shi
- Key Laboratory of Equus Germplasm Innovation (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (W.D.); (W.G.); (T.B.); (L.S.); (Y.L.); (H.W.); (M.D.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yanan Lin
- Key Laboratory of Equus Germplasm Innovation (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (W.D.); (W.G.); (T.B.); (L.S.); (Y.L.); (H.W.); (M.D.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Huize Wu
- Key Laboratory of Equus Germplasm Innovation (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (W.D.); (W.G.); (T.B.); (L.S.); (Y.L.); (H.W.); (M.D.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Manglai Dugarjaviin
- Key Laboratory of Equus Germplasm Innovation (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (W.D.); (W.G.); (T.B.); (L.S.); (Y.L.); (H.W.); (M.D.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Dongyi Bai
- Key Laboratory of Equus Germplasm Innovation (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; (W.D.); (W.G.); (T.B.); (L.S.); (Y.L.); (H.W.); (M.D.)
- Inner Mongolia Key Laboratory of Equine Science Research and Technology Innovation, Inner Mongolia Agricultural University, Hohhot 010018, China
- Equus Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
9
|
Huang Z, Hu L, Liu Z, Wang S. The Functions and Regulatory Mechanisms of Histone Modifications in Skeletal Muscle Development and Disease. Int J Mol Sci 2025; 26:3644. [PMID: 40332229 PMCID: PMC12027200 DOI: 10.3390/ijms26083644] [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: 03/03/2025] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Skeletal muscle development is a complex biological process regulated by many factors, such as transcription factors, signaling pathways, and epigenetic modifications. Histone modifications are important epigenetic regulatory factors involved in various biological processes, including skeletal muscle development, and play a crucial role in the pathogenesis of skeletal muscle diseases. Histone modification regulators affect the expression of many genes involved in skeletal muscle development and disease by adding or removing certain chemical modifications. In this review, we comprehensively summarize the functions and regulatory activities of the histone modification regulators involved in skeletal muscle development, regeneration, and disease.
Collapse
Affiliation(s)
- Zining Huang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan 430062, China; (Z.H.); (L.H.)
| | - Linqing Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan 430062, China; (Z.H.); (L.H.)
| | - Zhiwei Liu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Shanshan Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan 430062, China; (Z.H.); (L.H.)
| |
Collapse
|
10
|
Burgess J, Arora J, Green A, Singh A, Jarman E, Kang A, Hayashigatani K, Liu YK, Sobel R, Fox PM. Establishing a Murine Model of Muscle Changes in Chronic Nerve Compression. J Hand Surg Am 2025:S0363-5023(25)00130-3. [PMID: 40220004 DOI: 10.1016/j.jhsa.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/16/2025] [Accepted: 03/04/2025] [Indexed: 04/14/2025]
Abstract
PURPOSE We aimed to build upon a previously validated model of early chronic nerve compression (CNC) by evaluating changes in gross muscle weight, muscle gene expression, and muscle function, and correlating the mechanism and timing of muscle- and nerve related changes. METHODS Chronic nerve compression was induced by placing a Silastic tube around the sciatic nerve with the contralateral limb as control. At 6, 8, and 12 weeks of compression, gait analysis, muscle force measurements, and electrodiagnostics (EDX) were performed, and the sciatic nerve, tibialis anterior (TA), extensor digitorum longus (EDL), and gastrocnemius were harvested. Muscle weight (MW), cross-sectional area (CSA), g-ratio, axon area, and axon density were measured. Reverse transcripton polymerase chain reaction of TA+EDL muscle was performed. Genes assayed included atrogenes (Foxo-3, Atrogin-1, and MuRF1), markers of myogenesis (MyoD and MyoG), fatty acid synthase, type-I collagen (Col1a1), and inflammatory markers (tumor necrosis factor-α and interleukin-1β). RESULTS At 6 weeks, we observed a maximum 30.8% decrease in nerve conduction speed. G-ratio was increased 14.4% at 8 weeks, and at all time points, we observed a 25%-26% decrease in axon area. At 12 weeks, we observed a 10.4% decrease in TA+EDL MW, and at 8 weeks, CSA was reduced 13.9%. At 8 weeks, expression of atrogenes was increased 2-3-fold implying ongoing atrophy. MyoD/MyoG expression was reduced 0.3 times, and fatty acid synthase, type-1 collagen, and inflammatory marker expression was increased 1.3-, 1.4-, and >2-fold, respectively. There were no clinically important differences in gait analysis or muscle force measurement between compressed and control limbs at any time-point. CONCLUSIONS The murine model of muscle changes in CNC demonstrates reduced nerve conduction speed, demyelination, and a shift in axon size consistent with early CNC. Changes in MW, CSA, and gene expression occur in the absence of significant differences in muscle function. CLINICAL RELEVANCE These findings establish a mouse model of early muscle changes in CNC that can be used to investigate interventions to reduce or delay muscle changes in compression neuropathies.
Collapse
Affiliation(s)
- Jordan Burgess
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA; Stanford University School of Medicine, Stanford, CA
| | - Jagmeet Arora
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA
| | - Allen Green
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA; Stanford University School of Medicine, Stanford, CA
| | - Amar Singh
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA
| | - Evan Jarman
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA
| | - Augustine Kang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA
| | | | - Yusha Katie Liu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA; Stanford University School of Medicine, Stanford, CA
| | - Raymond Sobel
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA; Stanford University School of Medicine, Stanford, CA
| | - Paige M Fox
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA; Stanford University School of Medicine, Stanford, CA.
| |
Collapse
|
11
|
de Souza ALG, Alves ALR, Martinez CG, de Sousa JC, Kurtenbach E. Biomarkers of Skeletal Muscle Atrophy Based on Atrogenes Evaluation: A Systematic Review and Meta-Analysis Study. Int J Mol Sci 2025; 26:3516. [PMID: 40331994 PMCID: PMC12026492 DOI: 10.3390/ijms26083516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Muscle atrophy leads to decreased muscle mass, weakness, inactivity, and increased mortality. E3 ubiquitin ligases, key regulators of protein degradation via the ubiquitin-proteasome system, play a critical role in atrophic mechanisms. This meta-analysis followed Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines, and its objective was to evaluate the association between E3 ligases Muscle Atrophy F-box (MAFbx)/Atrogin-1 (Fbxo32) and Muscle RING-finger protein 1 (MuRF-1) (TRIM63) E3 ligase mRNA levels, reductions in skeletal muscle CSA measures, and atrophy conditions. We examined papers published on PubMed®, Scopus, and Web of Science that studied E3 ligase gene expression signatures for Fbxo32 (MAFbx/Atrogin-1) and Trim63 (MuRF1) in different types of muscle atrophy and hypertrophy murine models. Twenty-nine studies selected by two independent raters were analyzed. Standardized mean differences (SMDs)/effect sizes (ESs) and 95% confidence intervals (CIs) were calculated for the outcomes using fixed-effects models. We found that 6- and 4.8-fold upregulation, respectively, of Fbxo32 and Trim63 was sufficient to reduce the ES to -3.89 (95% CI: -4.45 to -3.32) for the muscle fiber cross-sectional area and the development of skeletal muscle atrophy. I² and Q test statistics did not indicate heterogeneous data. There was a low probability of bias after both the funnel plot and Egger's test analyses. These results were sustained independently of the atrophic model and muscle type. Therefore, the magnitude of the increase in muscle Fbxo32 and Trim63 mRNA is a feasible, reliable molecular marker for skeletal muscle atrophy in mice. The next step for the Ubiquitin-proteasome system (UPS) field involves elucidating the targets of E3 ligases, paving the way for diagnostic and treatment applications in humans.
Collapse
Affiliation(s)
- André Luiz Gouvêa de Souza
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Anna Luisa Rosa Alves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Camila Guerra Martinez
- Biosciences Applied to Health, Campus Renascença, Universidade Ceuma, São Luis 65075-120, MA, Brazil
| | - Júlia Costa de Sousa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Eleonora Kurtenbach
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
12
|
Fadah K, Mares A, Lange RA. Statin-Associated muscle symptoms and vitamin D supplementation. Curr Opin Cardiol 2025:00001573-990000000-00204. [PMID: 40183368 DOI: 10.1097/hco.0000000000001222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
PURPOSE OF REVIEW Statin medications play a pivotal role in the primary and secondary prevention and management of cardiovascular disease due to their potent lipid-lowering and anti-inflammatory effects. Nevertheless, the clinical application of statins is often hampered by statin-associated muscle symptoms (SAMS), which deter patient adherence and treatment efficacy considerably. SAMS currently affects 10-20% of statin users, thus understanding potential mitigation strategies is crucial. This review focuses on the role of vitamin D in mitigating SAMS, given the growing interest in vitamin D deficiency as a potentially modifiable risk factor. RECENT FINDINGS Despite observational suggestions linking vitamin D deficiency to heightened SAMS risk, recent studies have yielded conflicting results on the role of vitamin D supplementation in preventing SAMS. Some studies report an improvement in statin tolerance following vitamin D repletion. However, recent large-scale clinical trials, particularly the Vitamin D and Omega-3 trial (VITAL) report no significant benefit of vitamin D supplementation in preventing SAMS or reducing statin discontinuation rates, regardless of baseline vitamin D levels. SUMMARY Observational data provides evidence for the use of vitamin D supplementation in SAMS management, however conflicting evidence in recent clinical trials do not support its routine use for preventing or treating SAMS. To explore alternative strategies in improving statin tolerance and adherence, this discourse aims to elucidate the current understanding the complex mechanisms underlying SAMS, the influence of serum vitamin D levels, and the implications for clinical management.
Collapse
Affiliation(s)
- Kahtan Fadah
- Division of Cardiology, Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas
| | - Adriana Mares
- Yale University School of Medicine, New Haven, Connecticut, USA
| | - Richard A Lange
- Division of Cardiology, Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas
| |
Collapse
|
13
|
Yokogawa A, Kido K, Miura I, Oyama E, Takakura D, Tanaka K, Wilkinson D, Smith K, Atherton P, Kawanaka K. Low-Carbohydrate Diet Exacerbates Denervation-Induced Atrophy of Rat Skeletal Muscle Under the Condition of Identical Protein Intake. J Cachexia Sarcopenia Muscle 2025; 16:e13738. [PMID: 40000911 PMCID: PMC11859665 DOI: 10.1002/jcsm.13738] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 12/30/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND While decreased protein intake is associated with muscle mass loss, it is unclear whether a decrease in carbohydrate intake adversely affects muscle atrophy independently of protein intake. Herein, we examined whether a low-carbohydrate (low-CHO) diet exacerbates denervation-induced muscle atrophy under conditions of identical protein intake. METHODS On day one of the experiment, male Wistar rats underwent unilateral denervation. The contralateral leg was used as the control. After denervation, rats were divided into two dietary groups: high-carbohydrate (high-CHO) and low-CHO. Each group was fed a high-CHO (70% carbohydrate) or low-CHO (20% carbohydrate) diet over 7 days. Total protein and energy intakes in both groups were matched by pair feeding. Rats were provided with deuterium oxide (D2O) tracer over the last 3 days of dietary intervention to quantify myofibrillar (muscle) protein synthesis (MPS). RESULTS Denervation reduced wet weight of the gastrocnemius muscle compared to the contralateral control (p < 0.05). Reductions in gastrocnemius muscle weight were greater in the low-CHO group (-34%) than the high-CHO group (-28%) (p < 0.05). Although denervation decreased MPS compared to the contralateral control (p < 0.05), no dietary effect on MPS was observed. Denervation resulted in increased mRNA and protein expression of Atrogin-1, a ubiquitin E3 ligase, compared to that in the contralateral control (p < 0.05). Increases in Atrogin-1 gene and protein expression due to denervation were greater in the low-CHO group than in the high-CHO group (p < 0.05). CONCLUSIONS We conclude that a low-CHO diet may exacerbate denervation-induced atrophy in fast-twitch-dominant muscles compared to a high-CHO diet, even when the same protein intake is maintained. Although blunted MPS contributed to muscle atrophy due to denervation, exacerbation of muscle atrophy by the low-CHO diet was not accompanied by explanatory changes in MPS. The effect of the low-CHO diet might be related to promotion of muscle-specific ubiquitin E3 ligase gene expression.
Collapse
Affiliation(s)
- Aki Yokogawa
- Faculty of Sports and Health SciencesFukuoka UniversityFukuokaJapan
| | - Kohei Kido
- Faculty of Sports and Health SciencesFukuoka UniversityFukuokaJapan
- Institute for Physical ActivityFukuoka UniversityFukuokaJapan
- Health and Medical Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)TakamatsuJapan
| | - Ikuru Miura
- Faculty of Sports and Health SciencesFukuoka UniversityFukuokaJapan
- Institute for Physical ActivityFukuoka UniversityFukuokaJapan
| | - Eisuke Oyama
- Faculty of Sports and Health SciencesFukuoka UniversityFukuokaJapan
| | - Daisuke Takakura
- Faculty of Sports and Health SciencesFukuoka UniversityFukuokaJapan
| | - Keigo Tanaka
- Faculty of Sports and Health SciencesFukuoka UniversityFukuokaJapan
| | - Daniel J. Wilkinson
- MRC‐Versus Arthritis Center for Musculoskeletal Ageing Research, Centre of Metabolism, Ageing & Physiology (COMAP), Royal Derby Hospital CenterUniversity of NottinghamDerbyUK
| | - Kenneth Smith
- MRC‐Versus Arthritis Center for Musculoskeletal Ageing Research, Centre of Metabolism, Ageing & Physiology (COMAP), Royal Derby Hospital CenterUniversity of NottinghamDerbyUK
| | - Philip J. Atherton
- MRC‐Versus Arthritis Center for Musculoskeletal Ageing Research, Centre of Metabolism, Ageing & Physiology (COMAP), Royal Derby Hospital CenterUniversity of NottinghamDerbyUK
| | - Kentaro Kawanaka
- Faculty of Sports and Health SciencesFukuoka UniversityFukuokaJapan
- Institute for Physical ActivityFukuoka UniversityFukuokaJapan
| |
Collapse
|
14
|
Liu H, Wang K, Shang T, Cai Z, Lu C, Shen M, Yu S, Yao X, Shen Y, Chen X, Xu F, Sun H. Astragaloside IV Improves Muscle Atrophy by Modulating the Activity of UPS and ALP via Suppressing Oxidative Stress and Inflammation in Denervated Mice. Mol Neurobiol 2025; 62:4689-4704. [PMID: 39480556 DOI: 10.1007/s12035-024-04590-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: 12/06/2023] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Peripheral nerve injury is common clinically and can lead to neuronal degeneration and atrophy and fibrosis of the target muscle. The molecular mechanisms of muscle atrophy induced by denervation are complex and not fully understood. Inflammation and oxidative stress play an important triggering role in denervated muscle atrophy. Astragaloside IV (ASIV), a monomeric compound purified from astragalus membranaceus, has antioxidant and anti-inflammatory properties. The aim of this study was to investigate the effect of ASIV on denervated muscle atrophy and its molecular mechanism, so as to provide a new potential therapeutic target for the prevention and treatment of denervated muscle atrophy. In this study, an ICR mouse model of muscle atrophy was generated through sciatic nerve dissection. We found that ASIV significantly inhibited the reduction of tibialis anterior muscle mass and muscle fiber cross-sectional area in denervated mice, reducing ROS and oxidative stress-related protein levels. Furthermore, ASIV inhibits the increase in inflammation-associated proteins and infiltration of inflammatory cells, protecting the denervated microvessels in skeletal muscle. We also found that ASIV reduced the expression levels of MAFbx, MuRF1 and FoxO3a, while decreasing the expression levels of autophagy-related proteins, it inhibited the activation of ubiquitin-proteasome and autophagy-lysosome hydrolysis systems and the slow-to-fast myofiber shift. Our results show that ASIV inhibits oxidative stress and inflammatory responses in skeletal muscle due to denervation, inhibits mitophagy and proteolysis, improves microvascular circulation and reverses the transition of muscle fiber types; Therefore, the process of skeletal muscle atrophy caused by denervation can be effectively delayed.
Collapse
Affiliation(s)
- Hua Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, Nantong, Jiangsu Province, 226600, P. R. China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Tongxin Shang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Zhigang Cai
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, Nantong, Jiangsu Province, 226600, P. R. China
| | - Chunfeng Lu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, Jiangsu Province, 226006, P. R. China
| | - Mi Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Shu Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Xinlei Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Xiaofang Chen
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, Nantong, Jiangsu Province, 226600, P. R. China.
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, Jiangsu Province, 226006, P. R. China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China.
| |
Collapse
|
15
|
Moustafa Mahmoud M, Abdel Hameed NQ, Adel Al Dreny Abd Al Latef B, Samir Kamar S, Ahmed Rashed L, Abdelhameed Gouda SA. High-intensity exercise alongside insulin alleviates muscle atrophy in type 1 diabetes mellitus concomitant with modulation of mitophagy-related proteins in skeletal muscle. Arch Physiol Biochem 2025; 131:293-305. [PMID: 39382178 DOI: 10.1080/13813455.2024.2410791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/08/2024] [Accepted: 09/15/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Diabetes patients' quality of life can be severely impacted by diabetic muscle atrophy. AIM This study aimed to explore the impact of high-intensity exercise (HIE) alongside insulin treatment on muscle atrophy in a rat model of type 1 diabetes mellitus (T1DM). METHODOLOGY Fifty rats were allocated into five groups; Group 1, control sedentary (CS), T1DM was elicited in the rest of the groups by giving them Streptozotocin (STZ) (60 mg/kg), where group 2 (DS) remained sedentary, while groups 3,4,5 were treated with insulin after induction of diabetes. Group 4 (DI+MIE) and 5 (DI+ HIE) underwent moderate and high-intensity exercise, respectively. RESULTS HIE for 14 days combined with insulin treatment significantly restored muscle strength and mass with a significant modification in the mitophagy-related proteins and fibroblast growth factor 21 (FGF 21) compared to other treated groups. CONCLUSION This study concluded that there is a therapeutic role for HIE with insulin against T1DM-induced muscle atrophy.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Muscular Atrophy/therapy
- Muscular Atrophy/etiology
- Muscular Atrophy/metabolism
- Muscular Atrophy/pathology
- Insulin/therapeutic use
- Insulin/pharmacology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/drug effects
- Rats
- Mitophagy/drug effects
- Physical Conditioning, Animal
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/therapy
- Male
- Rats, Wistar
- Hypoglycemic Agents/therapeutic use
- Hypoglycemic Agents/pharmacology
- Streptozocin
- Muscle Strength/drug effects
Collapse
Affiliation(s)
| | | | | | - Samaa Samir Kamar
- Histology department, Faculty of Medicine- Cairo University, Cairo, Egypt
- Histology department, Armed Forces College of Medicine, Cairo, Egypt
| | - Laila Ahmed Rashed
- Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
16
|
Ikeda Y, Kawamura R, Tabara Y, Maruyama K, Shiokawa D, Takakado M, Hadate T, Takata Y, Ohashi J, Saito I, Ogawa Y, Osawa H. Genetic variation in the RETN promoter, accompanied by latent sarcopenic obesity, led to insulin resistance in a Japanese cohort: the Toon Genome Study. Diabetologia 2025; 68:854-865. [PMID: 39670999 PMCID: PMC11950018 DOI: 10.1007/s00125-024-06322-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 09/11/2024] [Indexed: 12/14/2024]
Abstract
AIMS/HYPOTHESIS Resistin, inducing insulin resistance, is elevated in the sera of individuals with the G-A haplotype at c.-420 C>G (rs1862513) and c.-358 G>A (rs3219175). This haplotype is associated with visceral obesity and low grip strength. To elucidate the hidden relationship between the G-A haplotype and insulin resistance, integration of specific phenotypes defined by body composition and 75 g OGTT would be a promising strategy. METHODS The 803 Japanese participants (average age: 62 years), attending annual medical checkups, were evaluated every 5 years. Participants were categorised by skeletal muscle mass, visceral fat score and OGTT results. Hierarchical clustering was performed using body composition and glucose metabolism parameters. Whole blood cells from participants homozygous for the G-A or C-G haplotype (n=25 and 33, respectively), matched for age, sex and BMI, using propensity score matching, were used for RNA-seq, pathway analysis and RT-PCR. RESULTS Multivariate analysis showed that individuals with the G-A haplotype, when accompanied by latent skeletal muscle loss and visceral obesity (latent sarcopenic obesity), presented a pronounced deterioration in insulin resistance over a 5 year period. Cluster 2, identified using hierarchical clustering, was characterised by low skeletal muscle mass, visceral obesity and insulin resistance. This cluster, with the G-A haplotype, demonstrated deterioration in insulin resistance. RNA-seq and RT-PCR revealed altered expression of mitophagy-related genes in whole blood cells of the G-A homozygotes. CONCLUSIONS/INTERPRETATION The G-A haplotype, accompanied by latent low skeletal muscle mass and visceral obesity, led to the deterioration of insulin resistance over a 5 year period in this cohort, possibly through the altered expression of mitophagy-related genes.
Collapse
Affiliation(s)
- Yosuke Ikeda
- Department of Diabetes and Molecular Genetics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ryoichi Kawamura
- Department of Diabetes and Molecular Genetics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yasuharu Tabara
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koutatsu Maruyama
- Department of Bioscience, Graduate School of Agriculture, Ehime University, Ehime, Japan
| | | | - Misaki Takakado
- Department of Diabetes and Molecular Genetics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Toshimi Hadate
- Department of Diabetes and Molecular Genetics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yasunori Takata
- Department of Diabetes and Molecular Genetics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Jun Ohashi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Isao Saito
- Department of Public Health and Epidemiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Haruhiko Osawa
- Department of Diabetes and Molecular Genetics, Ehime University Graduate School of Medicine, Ehime, Japan.
| |
Collapse
|
17
|
Han S, Zhao X, Yu C, Cui C, Zhang Y, Zhu Q, Qiu M, Yang C, Yin H. Nestin Regulates Autophagy-Dependent Ferroptosis Mediated Skeletal Muscle Atrophy by Ubiquitinating MAP 1LC3B. J Cachexia Sarcopenia Muscle 2025; 16:e13779. [PMID: 40183241 PMCID: PMC11969254 DOI: 10.1002/jcsm.13779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Programmed cell death plays a critical role in skeletal muscle atrophy. Ferroptosis, an iron-dependent form of programmed cell death driven by lipid peroxidation, has been implicated in various diseases, but its role in skeletal muscle atrophy remains unclear. METHODS Ferroptosis in skeletal muscle atrophy was investigated using two models: dexamethasone (Dex)-induced atrophy (n = 6 independent cell cultures per group) and simulated microgravity (n = 6 mice per group). Conditional Nestin knockout (KO) mice were generated using CRISPR/Cas9 (n = 6-8 mice per group), with wild-type (WT) controls (n = 6-8). Phenotypic analyses included histopathology (HE staining), functional assessments (muscle strength, weight analysis, treadmill), and dystrophy evaluation (dystrophin staining). Molecular analyses involved flow cytometry, ELISA, transmission electron microscopy, PI staining, and IP/MS to delineate Nestin-regulated ferroptosis pathways in skeletal muscle atrophy. RESULTS Ferroptosis was significantly activated in both atrophy models, with a 2.5-fold increase in lipid peroxidation (p < 0.01), a 2-fold accumulation of Fe2+ (p < 0.01) and a 50% reduction in Nestin expression (p < 0.001). Nestin KO mice exhibited exacerbated muscle atrophy, showing a 40% decrease in muscle weight (p < 0.01) and a 30% reduction in muscle strength (p < 0.05) compared to WT mice. Nestin overexpression mitigated Dex-induced ferroptosis, reducing lipid peroxidation by 40%, decreasing Fe2+ accumulation by 50% (p < 0.01), and improving muscle function by 30% (p < 0.05). Mechanistically, Nestin interacted with MAP 1LC3B (LC3B) to catalyse LC3B polyubiquitination at lysine-51, reducing LC3B availability for autophagy and inhibiting autophagy flux by 60% (p < 0.01), leading to a 50% reduction in ferroptosis (p < 0.001). CONCLUSIONS Our study identifies Nestin as a critical regulator of ferroptosis-autophagy crosstalk in skeletal muscle atrophy. Targeting Nestin-LC3B ubiquitination may offer novel therapeutic strategies for preventing muscle wasting in diseases such as cachexia and sarcopenia.
Collapse
Affiliation(s)
- Shunshun Han
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Xiyu Zhao
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Chunlin Yu
- Animal Breeding and Genetics Key Laboratory of Sichuan ProvinceSichuan Animal Science AcademyChengduChina
| | - Can Cui
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Yao Zhang
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Qing Zhu
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Mohan Qiu
- Animal Breeding and Genetics Key Laboratory of Sichuan ProvinceSichuan Animal Science AcademyChengduChina
| | - Chaowu Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan ProvinceSichuan Animal Science AcademyChengduChina
| | - Huadong Yin
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| |
Collapse
|
18
|
Wang Y, Lu Y, Hou J, Wang Y, Luo L, Lu Z, Xie Y, Cai L, Xiao Z. Upregulation of FAM129B protects against glucocorticoid-induced skeletal muscle atrophy via regulating long non-coding RNA NEAT1. Int J Biol Macromol 2025; 300:140120. [PMID: 39855497 DOI: 10.1016/j.ijbiomac.2025.140120] [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: 08/17/2024] [Revised: 12/29/2024] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Skeletal muscle atrophy, manifested by a reduction in muscle size and quantity, is primarily attributed to excessive protein catabolism. FAM129B, an antioxidant protein, has been previously implicated in muscle growth and development in cattle. Aim of this study is to elucidate the role of FAM129B in muscle atrophy. FAM129B was consistently down-regulated in muscle atrophy models in vitro and in vivo and in human steroid-treated gluteus muscles. FAM129B depletion resulted in myotubes atrophy with reduced diameter, increased MuRF-1 and Atrogin-1. Conversely, FAM129B overexpression ameliorated muscle atrophy by increasing myotube diameter and reducing Atrogin-1 and MuRF-1. Mice overexpressing FAM129B exhibited resistance to muscle atrophy, evidenced by increased grip strength, increased tibial anterior weight, increased myofiber cross-sectional area and decreased MuRF-1 and Atrogin-1. RNA sequencing revealed NEAT1 as a downstream gene of FAM129B. Mechanistically, FAM129B was found to influence the stability of NEAT1 by directly binding to it. The enhanced stability of NEAT1 subsequently led to increased FoxO1 expression and subsequent protein degradation. Our study has provided evidence that the upregulation of FAM129B rescues the glucocorticoid-induced skeletal muscle atrophy, suggesting that FAM129B may be a potential target for alleviating skeletal muscle atrophy.
Collapse
Affiliation(s)
- Yan Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yushen Lu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 430000 Wuhan, China
| | - Jinhui Hou
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 430000 Wuhan, China
| | - Yuyang Wang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 430000 Wuhan, China
| | - Lihuan Luo
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zuneng Lu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuanlong Xie
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 430000 Wuhan, China
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 430000 Wuhan, China.
| | - Zheman Xiao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
19
|
Paul S, Donath L, Hoppstädter J, Hecksteden A. Resistance but not endurance training suppresses glucocorticoid-induced leucine zipper (GILZ) expression in human skeletal muscle. Eur J Appl Physiol 2025; 125:1023-1036. [PMID: 39499305 DOI: 10.1007/s00421-024-05644-7] [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: 06/26/2024] [Accepted: 10/14/2024] [Indexed: 11/07/2024]
Abstract
PURPOSE Within human skeletal muscle, statin treatment leads to elevated levels of the glucocorticoid-induced leucine zipper (GILZ). Further, GILZ mediates the muscle-related side effects of statins. Physical exercise leads to GILZ suppression, in a mechanosensitive manner. Given that statin treatment is rarely tolerated by habitually exercising individuals due to statin-associated muscle symptoms (SAMS), it appears that the opposing regulation of GILZ facilitates this detrimental interaction of two key measures of cardiovascular prevention, specifically for exercise modalities with high muscle strain. Similarly, opposing regulation of atrophy associated genes (atrogenes) may be a further mechanism. If confirmed, these results might have implications for the exercise prescription of statin-users. METHODS A systematic search of the Gene Expression Omnibus (GEO) repository for studies reporting the acute effects of either endurance (END), conventional resistance (RT), or eccentric resistance training (ECC) was conducted. GILZ, as well as the expression of pivotal atrogenes (e.g., muscle atrophy F-box, cathepsin L, etc.) were quantified. RESULTS 15 studies with 204 participants (22 females; 182 males) were included. RT resulted in the highest GILZ suppression, significantly differing from the expressional change after END ( - 0.46 ± 1.11 vs. - 0.07 ± 1.08), but not from ECC ( - 0.46 ± 1.11 vs. - 0.46 ± 0.95). Similar results were seen for various atrogenes. CONCLUSION Our results strengthen the assumption that mechanical loading can be considered a key mediator of exercise-induced changes in GILZ and atrogene expression.
Collapse
Affiliation(s)
- Sebastian Paul
- Department of Training Intervention Research, German Sport University Cologne, 50933, Cologne, Germany.
- Institute of Physiology, Medical University of Innsbruck, 6020, Innsbruck, Austria.
| | - Lars Donath
- Department of Training Intervention Research, German Sport University Cologne, 50933, Cologne, Germany
| | - Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Anne Hecksteden
- Institute of Sport Science, Universität of Innsbruck, 6020, Innsbruck, Austria
- Institute of Physiology, Medical University of Innsbruck, 6020, Innsbruck, Austria
| |
Collapse
|
20
|
SHI J, PAN F, GE H, YANG Z, ZHAN H. Mechanism of Qigu capsule as a treatment for sarcopenia based on network pharmacology and experimental validation. J TRADIT CHIN MED 2025; 45:399-407. [PMID: 40151126 PMCID: PMC11955765 DOI: 10.19852/j.cnki.jtcm.2025.02.001] [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: 11/23/2023] [Accepted: 03/22/2024] [Indexed: 03/29/2025]
Abstract
OBJECTIVE To explore the potential molecular mechanism of Qigu capsule (,QGC) in the treatment of sarcopenia through network pharmacology and to verify it experimentally. METHODS The active compounds of QGC and common targets between QGC and sarcopenia were screened from databases. Then the herbs-compounds-targets network, and protein-protein interaction (PPI) network was constructed. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed by R software. Next, we used a dexamethasone-induced sarcopenia mouse model to evaluate the anti-sarcopenic mechanism of QGC. RESULTS A total of 57 common targets of QGC and sarcopenia were obtained. Based on the enrichment analysis of GO and KEGG, we took the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway as a key target to explore the mechanism of QGC on sarcopenia. Animal experiments showed that QGC could increase muscle strength and inhibit muscle fiber atrophy. In the model group, the expression of muscle ring finger-1 and Atrogin-1 were increased, while myosin heavy chain was decreased, QGC treatment reversed these changes. Moreover, compared with the model group, the expressions of p-PI3K, p-Akt, p-mammalian target of rapamycin and p-Forkhead box O3 in the QGC group were all upregulated. CONCLUSION QGC exerts an anti-sarcopenic effect by activating PI3K/Akt signaling pathway to regulate skeletal muscle protein metabolism.
Collapse
Affiliation(s)
- Jinyu SHI
- 1 Shi's Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
- 2 Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai 200120, China
| | - Fuwei PAN
- 1 Shi's Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
- 2 Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai 200120, China
| | - Haiya GE
- 1 Shi's Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
- 2 Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai 200120, China
| | - Zongrui YANG
- 1 Shi's Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
- 2 Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai 200120, China
| | - Hongsheng ZHAN
- 1 Shi's Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
- 2 Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai 200120, China
| |
Collapse
|
21
|
Schiavo L, Santella B, Mingo M, Rossetti G, Orio M, Cobellis L, Maurano A, Iannelli A, Pilone V. Preliminary Evidence Suggests That a 12-Week Treatment with Tirzepatide Plus Low-Energy Ketogenic Therapy Is More Effective than Its Combination with a Low-Calorie Diet in Preserving Fat-Free Mass, Muscle Strength, and Resting Metabolic Rate in Patients with Obesity. Nutrients 2025; 17:1216. [PMID: 40218974 PMCID: PMC11990520 DOI: 10.3390/nu17071216] [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: 03/01/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Tirzepatide (TZP), a unimolecular dual agonist targeting glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1 receptors, is a promising weight loss agent in obesity. The preservation of metabolically active fat-free mass (FFM), muscle strength (MS), and resting metabolic rate (RMR) is essential for optimizing fat mass (FM) reduction. Although TZP is typically combined with a low-calorie diet (LCD), its impact on FFM is uncertain, and studies on MS and RMR are lacking. Evidence suggests that Low-Energy Ketogenic Therapy (LEKT) may reduce FM while preserving FFM, MS, and RMR. Therefore, this study aimed to compare the effects of an LEKT and an LCD, both combined with TZP, on body weight (BW), FM, FFM, MS, and RMR in patients with obesity. Methods: We prospectively compared the effects of TZP combined with either an LCD or LEKT in 60 patients with obesity (n = 30 per group) over 12 weeks. BW, FM, FFM, MS, and RMR were measured at baseline and after 12 weeks. Clinical parameters, an assessment of dietary compliance, and side effects were also evaluated. Results: At 12-week follow-up, both groups showed a significant BW reduction from baseline (TZP+LEKT, p = 0.0289; TZP+LCD, p = 0.0278), with no significant intergroup difference (p = 0.665). Similarly, FM decreased significantly in both cohorts (TZP+LEKT, p < 0.001; TZP+LCD, p = 0.0185), with the TZP+LEKT group achieving a greater FM loss (p = 0.042). However, the TZP+LCD group exhibited significant declines from baseline in FFM (p = 0.0284), MS (p = 0.0341), and RMR (p < 0.001), whereas we did not observe any significant changes in FFM (p = 0.487), MS (p = 0.691), and RMR (p = 0.263) in the TZP+LEKT group. Intergroup direct comparisons confirmed that the TZP+LCD group experienced significantly greater reductions in FFM (p = 0.0388), MS (p = 0.046), and RMR (p = 0.019). Conclusions: Based on the findings of these preliminary data, we are able to support the hypothesis that TZP+LEKT seems to be superior to TZP+LCD in promoting FM reduction while preserving FFM, MS, and RMR in patients with obesity.
Collapse
Affiliation(s)
- Luigi Schiavo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (B.S.); (M.M.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Biagio Santella
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (B.S.); (M.M.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Monica Mingo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (B.S.); (M.M.)
| | - Gianluca Rossetti
- General and Bariatric Surgery Unit, Abano Terme Policlinic, 35031 Padova, Italy;
| | - Marcello Orio
- Medical and Diabetological Center CMSO, 84123 Salerno, Italy;
| | - Luigi Cobellis
- Unit of General Surgery, Casa Di Cura “Prof. Dott. Luigi Cobellis”, 84078 Vallo della Lucania, Italy;
| | - Attilio Maurano
- Digestive Endoscopic Unit, Ruggiero Clinic, 84013 Cava de Tirreni, Italy;
| | - Antonio Iannelli
- Digestive Surgery and Liver Transplantation Unit, Archet 2 Hospital, Centre Hospitalier Universitaire de Nice, 06001 Nice, France;
- Université Côte d’Azur, 06001 Nice, France
- Team 8 “Hepatic Complications of Obesity and Alcohol”, Inserm, U1065, 06204 Nice, France
| | - Vincenzo Pilone
- Public Health Department, University of Naples Federico II, 80131 Naples, Italy;
| |
Collapse
|
22
|
Menard R, Morin E, Morse D, Halluin C, Pende M, Baanannou A, Grendler J, Fuqua H, Li J, Lancelot L, Drent J, Bonnet F, Graber JH, Murawala P, Dray C, Pradère JP, Coffman JA, Madelaine R. Zebrafish genetic model of neuromuscular degeneration associated with Atrogin-1 expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.07.642048. [PMID: 40196478 PMCID: PMC11975044 DOI: 10.1101/2025.03.07.642048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The degenerative loss of muscle associated with aging leading to muscular atrophy is called sarcopenia. Currently, practicing regular physical exercise is the only efficient way to delay sarcopenia onset. Identification of therapeutic targets to alleviate the symptoms of aging requires in vivo model organisms of accelerated muscle degeneration and atrophy. The zebrafish undergoes aging, with hallmarks including mitochondrial dysfunction, telomere shortening, and accumulation of senescent cells. However, zebrafish age slowly, and no specific zebrafish models of accelerated muscle atrophy associated with molecular events of aging are currently available. We have developed a new genetic tool to efficiently accelerate muscle-fiber degeneration and muscle-tissue atrophy in zebrafish larvae and adults. We used a gain-of-function strategy with a molecule that has been shown to be necessary and sufficient to induce muscle atrophy and a sarcopenia phenotype in mammals: Atrogin-1 (also named Fbxo32). We report the generation, validation, and characterization of a zebrafish genetic model of accelerated neuromuscular atrophy, the atrofish. We demonstrated that Atrogin-1 expression specifically in skeletal muscle tissue induces a muscle atrophic phenotype associated with locomotion dysfunction in both larvae and adult fish. We identified degradation of the myosin light chain as an event occurring prior to muscle-fiber degeneration. Biological processes associated with muscle aging such as proteolysis, inflammation, stress response, extracellular matrix (ECM) remodeling, and apoptosis are upregulated in the atrofish. Surprisingly, we observed a strong correlation between muscle-fiber degeneration and reduced numbers of neuromuscular junctions in the peripheral nervous system, as well as neuronal cell bodies in the spinal cord, suggesting that muscle atrophy could underly a neurodegenerative phenotype in the central nervous system. Finally, while atrofish larvae can recover locomotive functions, adult atrofish have impaired regenerative capacities, as is observed in mammals during muscle aging. In the future, the atrofish could serve as a platform for testing molecules aimed at treating or alleviating the symptoms of muscle aging, thereby opening new therapeutic avenues in the fight against sarcopenia.
Collapse
Affiliation(s)
- Romain Menard
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Elena Morin
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
- RESTORE Research Center, INSERM 1301, CNRS 5070, EFS, ENVT, Université Paul Sabatier, Toulouse, France
- IHU HealthAge, Toulouse, France
| | - Dexter Morse
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Caroline Halluin
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Marko Pende
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Aissette Baanannou
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Janelle Grendler
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Heath Fuqua
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Jijia Li
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
- CARe Graduate School, Université Paul Sabatier, Toulouse, France
| | - Laetitia Lancelot
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
- CARe Graduate School, Université Paul Sabatier, Toulouse, France
| | - Jessica Drent
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Frédéric Bonnet
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Joel H. Graber
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Prayag Murawala
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
- Department of nephrology and hypertension, Hannover Medical School, Hannover, Germany
| | - Cédric Dray
- RESTORE Research Center, INSERM 1301, CNRS 5070, EFS, ENVT, Université Paul Sabatier, Toulouse, France
- IHU HealthAge, Toulouse, France
| | - Jean-Philippe Pradère
- RESTORE Research Center, INSERM 1301, CNRS 5070, EFS, ENVT, Université Paul Sabatier, Toulouse, France
- IHU HealthAge, Toulouse, France
| | - James A. Coffman
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| | - Romain Madelaine
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, Maine, United States of America
| |
Collapse
|
23
|
Nicol M, Deniau B, Rahli R, Genest M, Polidano E, Assad N, Samuel JL, Mebazaa A, Solal AC, Azibani F. Streptozotocin-induced hyperglycemia unmasks cardiotoxicity induced by doxorubicin. Sci Rep 2025; 15:8104. [PMID: 40057546 PMCID: PMC11890863 DOI: 10.1038/s41598-025-91824-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
Late-onset cardiotoxicity induced by anthracyclines occurs years to decades after completion of anti-cancer therapy and is associated with increased morbi-mortality of cancer survivors. Chemotherapy at the time of treatment probably causes cardiac damages for which the juvenile heart compensate. Co-morbidities happening in the adulthood such as type 1 diabetes (DT1), affect the heart and thus can unmask chemotherapy induced cardiotoxicity. To prove our hypothesis, we induced hyperglycemia [Streptozotocin treatment (STZ), 50 mg/kg/day for 5 days] in 11 weeks old mice who previously received doxorubicin treatment (Dox, 3 mg/kg) when they were six-weeks old. Interestingly, streptozotocin-induced hyperglycemia in Dox-pretreated mice (Dox-STZ) induced a higher mortality (p < 0.05) and more severe cardiac dysfunction (p < 0.0001) when compared with mice receiving Dox or STZ alone. Apoptosis evaluated by caspase 3 protein expression and Bax/Bcl2 genes expression was higher in Dox-STZ mice compared to STZ or Dox alone. While Dox and STZ independently induced capillary rarefaction, cardiomyocytes atrophy was only induced by STZ. Furthermore, Sirius-red staining of cardiac sections showed higher fibrosis levels (p < 0.0001) in Dox-STZ compared to Dox or STZ alone. All together, these results demonstrate that STZ precipitates and unmask cardiac dysfunction in previously treated Dox animals.
Collapse
Affiliation(s)
- Martin Nicol
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Cardiology Department, Lariboisière Hospital, University of Paris, Paris, France
| | - Benjamin Deniau
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Department of Anesthesiology and Intensive Care, Lariboisière - Saint Louis Hospitals, APHP, Paris, France
| | - Roza Rahli
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Magali Genest
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Evelyne Polidano
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Noma Assad
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Jane-Lise Samuel
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Alexandre Mebazaa
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Department of Anesthesiology and Intensive Care, Lariboisière - Saint Louis Hospitals, APHP, Paris, France
| | - Alain Cohen Solal
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Cardiology Department, Lariboisière Hospital, University of Paris, Paris, France
| | - Feriel Azibani
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France.
| |
Collapse
|
24
|
Jin H, Seo HS, Shin J, Lee KJ, Kim SJ, Jung SH, Kim B. Dehydroandrographolide succinate attenuates dexamethasone-induced skeletal muscle atrophy by regulating Akt/GSK3β and MuRF-1 pathways. Eur J Pharmacol 2025; 990:177265. [PMID: 39800251 DOI: 10.1016/j.ejphar.2025.177265] [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: 07/10/2024] [Revised: 12/03/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Andrographis paniculata (AGPA) is known for its wide-ranging biological activities, including antiviral, antipyretic, and anticancer properties. However, its effects on muscle atrophy have not been well understood. This study investigates the impact of andrographolide (AD) and dehydroandrographolide succinate (DAS), key components of AGPA, on skeletal muscle atrophy using in vitro and in vivo models. We employed dexamethasone (DEX)-treated mice and C2C12 myotubes as models of skeletal muscle atrophy. While DAS and AD did not reverse the DEX-induced reduction in body weight, both compounds significantly restored grip strength in DEX-treated mice. Notably, DAS treatment, but not AD, markedly improved running speed, endurance time, and distance. Both DAS and AD enhanced lean muscle mass in the whole body, tibialis anterior (TA), and gastrocnemius (GS) muscles, as well as increased TA thickness, with DAS demonstrating superior efficacy compared to AD. In C2C12 myotubes treated with DEX, DAS and AD increased ATP production and myotube diameter. Mechanistically, both compounds upregulated phosphorylation of Akt and GSK3β and downregulated MuRF-1 expression. These results indicate that DAS and AD mitigate muscle atrophy via the Akt/GSK3β and MuRF-1 pathways, with DAS showing greater anti-atrophy efficiency. Thus, DAS emerges as a promising therapeutic candidate for the prevention of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Hengzhe Jin
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea
| | - Hyoung Sig Seo
- Research Institute of KSB TUGEN Inc., Gwangjin-gu, Seoul, 05029, South Korea
| | - Jinyoung Shin
- Department of Family Medicine, Konkuk University School of Medicine, 120-1, Neungdong-ro, Gwangjin-gu, Seoul, 05030, South Korea
| | - Kyung-Jin Lee
- Research Institute of KSB TUGEN Inc., Gwangjin-gu, Seoul, 05029, South Korea
| | - Su Jung Kim
- Research Institute of KSB TUGEN Inc., Gwangjin-gu, Seoul, 05029, South Korea
| | - Seung Hyo Jung
- Research Institute of KSB TUGEN Inc., Gwangjin-gu, Seoul, 05029, South Korea
| | - Bokyung Kim
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea.
| |
Collapse
|
25
|
Zhong P, Li X, Li J. Mechanisms, assessment, and exercise interventions for skeletal muscle dysfunction post-chemotherapy in breast cancer: from inflammation factors to clinical practice. Front Oncol 2025; 15:1551561. [PMID: 40104495 PMCID: PMC11913840 DOI: 10.3389/fonc.2025.1551561] [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: 12/25/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025] Open
Abstract
Chemotherapy remains a central component of breast cancer treatment, significantly improving patient survival rates. However, its toxic side effects, along with cancer-related paraneoplastic syndromes, can lead to the loss of skeletal muscle mass and function, impairing physical abilities and increasing the risk of complications during treatment. Chemotherapeutic agents directly impact skeletal muscle cells by promoting protein degradation, inhibiting protein synthesis, and triggering systemic inflammation, all of which contribute to muscle atrophy. Additionally, these drugs can interfere with the proliferation and differentiation of stem cells, such as satellite cells, disrupting muscle regeneration and repair while inducing abnormal differentiation of intermuscular tissue, thereby worsening muscle wasting. These effects not only reduce the effectiveness of chemotherapy but also negatively affect patients' quality of life and disease prognosis. Recent studies have emphasized the role of exercise as an effective non-pharmacological strategy for preventing muscle loss and preserving muscle mass in cancer patients. This review examines the clinical manifestations of muscle dysfunction following breast cancer chemotherapy, the potential mechanisms underlying these changes, and the evidence supporting exercise as a therapeutic approach for improving muscle function.
Collapse
Affiliation(s)
- Pei Zhong
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xizhuang Li
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiehua Li
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
26
|
Sato T, Morita A, Watanabe Y, Naito Y, Kawaji H, Nakagawa T, Hamaguchi H, Manabe Y, Fujii NL, Ogo N, Asai A, Kamei Y, Miura S. Rebastinib inhibits FoxO1 activity and reduces dexamethasone-induced atrophy and its-related gene expression in cultured myotubes. J Physiol Sci 2025; 75:100012. [PMID: 39985917 PMCID: PMC11905838 DOI: 10.1016/j.jphyss.2025.100012] [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: 10/27/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
FoxO1, a transcription factor, is upregulated in skeletal muscle during atrophy and inactivation of FoxO1 is a potential strategy to prevent muscle loss. This study identified Rebastinib as a potent suppressor of FoxO1 activity among protein kinase inhibitors. To determine whether Rebastinib inhibits atrophy-related ubiquitin ligases gene expression and mitigates atrophy in mouse skeletal muscle-derived cells, we investigated its protective effects of the compound against dexamethasone (DEX)-induced muscle atrophy using C2C12 myotubes. Rebastinib inhibited the DEX-induced upregulation of atrogin-1 and MuRF-1 mRNA, and atrogin-1 protein. Rebastinib also suppressed protein degradation and increased myotube diameter in DEX-treated C2C12 myotubes. Additionally, Rebastinib ameliorated the DEX- and cachexia-induced reduction in contractile force generation. Although the precise mechanisms underlying the action of Rebastinib against muscle atrophy and its efficacy in vivo remains to be elucidated, this compound shows great potential as a therapeutic agent for muscle atrophy.
Collapse
Affiliation(s)
- Tomoki Sato
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Akihito Morita
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yui Watanabe
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yumi Naito
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Haruka Kawaji
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Takumi Nakagawa
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Hiroki Hamaguchi
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Yasuko Manabe
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Nobuharu L Fujii
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Naohisa Ogo
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yasutomi Kamei
- Laboratory of Molecular Nutrition, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Shinji Miura
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan.
| |
Collapse
|
27
|
Zhang F, Hou X. The role of Forkhead box O in diabetes mellitus. Minerva Endocrinol (Torino) 2025; 50:105-112. [PMID: 35708174 DOI: 10.23736/s2724-6507.22.03750-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
Forkhead box O (FOXO) proteins are transcription factors that are involved in many physiological processes, including diabetes mellitus, which is a complex, multifactorial metabolic disorder. FOXO proteins are emerging as pivotal regulators in the progression of diabetes mellitus, mainly by inhibiting insulin or insulin-like growth factor, but little is known about their roles in diabetes mellitus. Although no targeted therapy exists to slow the development of diabetes and diabetes-related complications, several recent advances have clarified the molecular mechanisms underlying the disease. This review summarizes findings about FOXO proteins and diabetes mellitus, and sheds new light on the roles of FOXO proteins in diabetes mellitus.
Collapse
Affiliation(s)
- Fudan Zhang
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xu Hou
- Department of Endocrinology and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China -
| |
Collapse
|
28
|
Liang D, Wang D, Zheng X, Xiang H, Liu S, Yu C, Tian J, Ma J, Niu Y. Aerobic plus resistance exercise attenuates skeletal muscle atrophy induced by dexamethasone through the HDAC4/FoxO3a pathway. Cell Signal 2025; 127:111581. [PMID: 39732306 DOI: 10.1016/j.cellsig.2024.111581] [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: 09/03/2024] [Revised: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
This study aimed to investigate the underlying mechanisms by which physical exercise mitigates muscle atrophy induced by Dexamethasone (Dex). A muscle atrophy model was established in the mouse C2C12 cell line and 8-week-old mice treated with Dex, with subsequent verification of phenotype and atrogene expression. The potential benefits of combined aerobic and resistance exercise in mitigating muscle atrophy were then examined. To elucidate the involvement of Histone deacetylase 4 (HDAC4) in the protective effects of exercise against muscle loss, a combination of RT-PCR, Western blotting, immunoprecipitation, and immunofluorescence staining techniques were employed. The upregulation of HDAC4 was observed following Dex-induced muscle atrophy in vitro and in vivo. Inhibition of HDAC4 in C2C12 cells resulted in an increase in myotube diameter and fusion index, along with a decrease in the expression of Atrogin-1 and MuRF1. Treatment with Tasquinimod, an HDAC4 inhibitor, effectively prevented muscle wasting and dysfunction in mice induced by Dex. After a 6-week exercise intervention, the Dex-Exercise group exhibited significant improvements in body fat level, hyperinsulinemia, muscle mass and function in comparison to the Dex-Sedentary group. Mechanistically, we discovered that HDAC4 bound to and deacetylated Forkhead box protein O 3a (FoxO3a) within the nucleus, leading to decreased phosphorylation of FoxO3a at Ser 253. This interaction subsequently facilitated the expression of downstream atrogene Atrogin-1 and MuRF1, resulting in muscle atrophy. Conversely, exercise was found to potentially mitigate muscle atrophy by inhibiting the HDAC4/FoxO3a pathway. These findings suggest that HDAC4 may be a potential therapeutic target for exercise to combat Dex-induced muscle atrophy.
Collapse
Affiliation(s)
- Dehuan Liang
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Danni Wang
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Xinyue Zheng
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Heng Xiang
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Sujuan Liu
- Department of Anatomy and Histology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
| | - Chunxia Yu
- School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Jiatong Tian
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China
| | - Jianxiong Ma
- Tianjin Hospital, Tianjin University, No. 406 Jiefang South Road, Tianjin 300211, China
| | - Yanmei Niu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
29
|
Yu Y, Jiang X, Yu T, Chen F, Huang R, Xun Z, Wang X, Liu X, Xie X, Sun C, Xu Y, Liu X, Sun H, Yuan X, Ma C, Li Y, Song X, Wang D, Shao D, Shi X, Cao L. Maintaining myoprotein and redox homeostasis via an orally recharged nanoparticulate supplement potentiates sarcopenia treatment. Biomaterials 2025; 314:122863. [PMID: 39366185 DOI: 10.1016/j.biomaterials.2024.122863] [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: 04/10/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Sarcopenia is a progressive skeletal muscle disorder characterized by the accelerated loss of muscle mass and function, with no promising pharmacotherapies. Understanding the imbalance of myoprotein homeostasis within myotubes, which causes sarcopenia, may facilitate the development of novel treatments for clinical use. In this study, we found a strong correlation between low serum selenium levels and muscle function in elderly patients with sarcopenia. We hypothesized that supplementation with selenium might be beneficial for the management of sarcopenia. To verify this hypothesis, we developed diselenide-bridged mesoporous silica nanoparticles (Se-Se-MSNs) with ROS-responsive degradation and release to supplement selenium. Se-Se-MSNs outperformed free selenocysteine in alleviating sarcopenia in both dexamethasone (Dex)- and denervation-induced mouse models. Subsequently, Se-Se-MSNs were loaded with leucine (Leu@Se-Se-MSNs), another nutritional supplement used in sarcopenia management. Oral administration of Leu@Se-Se-MSNs restored myoprotein homeostasis by enhancing mTOR/S6K signaling and inactivating Akt/FoxO3a/MuRF1 signaling, thus exerting optimal therapeutic effects against sarcopenia and exhibiting a more favorable in vivo safety profile. This study provides a proof of concept for treating sarcopenia by maintaining myoprotein and redox homeostasis simultaneously and offers valuable insights into the development of multifunctional nanoparticle-based supplements for sarcopenia management.
Collapse
Affiliation(s)
- Yang Yu
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Xuehan Jiang
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Tianhao Yu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110002, Liaoning, China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, Guangzhou, China
| | - Runnian Huang
- Department of Gerontology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China; Department of Epidemiology and Health Statistics, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Zhe Xun
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiaoxun Wang
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xu Liu
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiaochun Xie
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Chen Sun
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Yingxi Xu
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiyan Liu
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Huayi Sun
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiaoyue Yuan
- Department of Gerontology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China; Department of Epidemiology and Health Statistics, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Chunhua Ma
- Department of Gerontology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China; Department of Epidemiology and Health Statistics, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yibai Li
- Health Sciences Institute, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiaoyu Song
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| | - Difei Wang
- Department of Gerontology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China.
| | - Dan Shao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, Guangzhou, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Xuetao Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, Guangzhou, China.
| | - Liu Cao
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
30
|
Conn MO, DeJong EN, Marko DM, Fayyazi R, Kukje Zada D, Foley KP, Barra NG, Bowdish DME, Schertzer JD. Microbiota protect against frailty and loss of skeletal muscle, and maintain inflammatory tone during aging in mice. Am J Physiol Cell Physiol 2025; 328:C887-C894. [PMID: 39925101 DOI: 10.1152/ajpcell.00869.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 11/26/2024] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
Chronic low-level inflammation or "inflammaging" is hypothesized to contribute to sarcopenia and frailty. Resident microbiota are thought to promote inflammaging, frailty, and loss of skeletal muscle mass. We tested immunity and frailty in male C57BL6/N germ-free (GF), specific pathogen-free (SPF) mice, and mice that were born germ-free and colonized (COL) with an SPF microbiota. Male and female GF mice had lower systemic cellular inflammation indicated by lower blood Ly6Chigh monocytes across their lifespan. Male GF mice had lower body mass, but relative to body mass, GF mice had smaller hindlimb muscles and smaller muscle fibers compared with SPF mice across the lifespan. Male and female GF mice had increased frailty at 18 mo or older. Colonization of female GF mice increased blood Ly6Chigh monocytes but did not affect frailty at 18 mo or older. Colonization of male GF mice increased blood Ly6Chigh monocytes, skeletal muscle size, myofiber fiber size, and decreased frailty at 18 mo or older. Transcriptomic analysis of the tibialis anterior muscle revealed a microbiota-muscle axis with over 550 differentially expressed genes in COL male mice at 18 mo or older. Colonized male mice had transcripts indicative of lower tumor necrosis factor (TNF)-α signaling via nuclear factor κB (NF-κB). Our findings show that microbiota can increase systemic cellular immunity while decreasing muscle inflammation, thereby protecting against muscle loss and frailty. We also found sex differences in the role of microbiota regulating frailty. We propose that microbiota components protect against lower muscle mass and frailty across the lifespan in mice.NEW & NOTEWORTHY Germ-free mice had increased frailty, lower muscle mass, and lower circulating inflammatory monocytes. Therefore, lower systemic inflammation coincided with worse frailty and muscle loss. Microbial colonization decreased frailty, restored muscle mass, and increased circulating inflammatory monocytes while lowering transcripts in inflammatory TNF and NF-κB pathways within muscle. Hence, microbiota can increase circulating inflammation but decrease muscle inflammation to protect against frailty. This microbiota-muscle axis should be investigated for therapeutic potential in muscle wasting and sarcopenia.
Collapse
Affiliation(s)
- Meghan O Conn
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Erica N DeJong
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Daniel M Marko
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Russta Fayyazi
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dana Kukje Zada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Kevin P Foley
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Dawn M E Bowdish
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
31
|
Zhuang M, Gu Y, Wang Z, He X, Chen N. Effects of 12-week whole-body vibration training versus resistance training in older people with sarcopenia. Sci Rep 2025; 15:6981. [PMID: 40011687 PMCID: PMC11865505 DOI: 10.1038/s41598-025-91644-2] [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: 10/28/2024] [Accepted: 02/21/2025] [Indexed: 02/28/2025] Open
Abstract
Sarcopenia is a syndrome commonly found in older people. The aim of this study was to evaluate the effects of whole-body vibration training (WBVT) and resistance training (RT) on body composition, muscle strength, physical performance and blood biomarkers in older people with sarcopenia. We conducted a 12-week, 3-times-weekly assessor-blinded, randomized controlled trial of 27 older people with sarcopenia aged ≥ 65 years. Subjects were randomized into WBVT group (n = 14) and RT group (n = 13). The primary outcome was knee extension strength (KES). Secondary outcomes were body composition [body weight, body mass index (BMI), percentage of body fat (PBF), and appendicular skeletal muscle mass index (ASMI)], muscle strength [handgrip strength (HS)], physical performance [gait speed (GS), 5-time chair stand test (5CST), and short physical performance battery (SPPB)], blood biomarkers (inflammatory factors, hormones, growth factors, and muscle injury biomarker), and quality of life questionnaire [medical outcomes study short-form 36 (SF-36)]. After 12-week intervention, in the WBVT group, we observed significant improvements in body composition (weight, BMI, PBF and ASMI), muscle strength (KES), physical performance (GS, SPPB and 5CST), blood biomarkers [insulin-like growth factor 1 (IGF-1), growth hormone, follistatin (FST) and creatine kinase (CK)] and quality of life. In the RT group, we observed significant improvements in body composition (weight, BMI and PBF), muscle strength (KES), physical performance (GS and SPPB), blood biomarkers (growth hormone, FST and CK) and quality of life. Between-group comparisons were only significant for KES (P = 0.007) and the role-physical (RP) dimension of the SF-36 (P = 0.007). WBVT and RT both improved the physical condition of older people with sarcopenia. RT excelled in muscle strength, but WBVT offered an alternative for those with restrictions. WBVT's low risk and flexibility suited diverse conditions, providing a new rehabilitation option for patients with sarcopenia.
Collapse
Affiliation(s)
- Min Zhuang
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
- The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yifan Gu
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Zhou Wang
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xiangfeng He
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Nan Chen
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China.
- Department of Rehabilitation, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
32
|
Von Ruff ZD, Miller MJ, Moro T, Reidy PT, Ebert SM, Volpi E, Adams CM, Rasmussen BB. Resistance exercise training in older men reduces ATF4-activated and senescence-associated mRNAs in skeletal muscle. GeroScience 2025:10.1007/s11357-025-01564-2. [PMID: 40011348 DOI: 10.1007/s11357-025-01564-2] [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: 10/22/2024] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
Sarcopenia increases the risk of frailty, morbidity, and mortality in older adults. Resistance exercise training improves muscle size and function; however, the response to exercise training is variable in older adults. The objective of our study was to determine both the age-independent and age-dependent changes to the transcriptome following progressive resistance exercise training. Skeletal muscle biopsies were obtained before and after 12 weeks of resistance exercise training in 8 young (24 ± 3.3 years) and 10 older (72 ± 4.9 years) men. RNA was extracted from each biopsy and prepared for analysis via RNA sequencing. We performed differential mRNA expression, gene ontology, and gene set enrichment analyses. We report that when comparing post-training vs pre-training 226 mRNAs and 959 mRNAs were differentially expressed in the skeletal muscle of young and older men, respectively. Additionally, 94 mRNAs increased, and 17 mRNAs decreased in both young and old, indicating limited overlap in response to resistance exercise training. Furthermore, the differential gene expression was larger in older skeletal muscle. Finally, we report three novel findings: 1) resistance exercise training decreased the abundance of ATF4-activated and senescence-associated skeletal muscle mRNAs in older men; 2) resistance exercise-induced increases in lean mass correlate with increased mRNAs encoding mitochondrial proteins; and 3) increases in muscle strength following resistance exercise positively correlate with increased mRNAs involved in translation, rRNA processing, and polyamine metabolism. We conclude that resistance exercise training elicits a differential gene expression response in young and old skeletal muscle, including reduced ATF-4 activated and senescence-associated gene expression.
Collapse
Affiliation(s)
| | - Matthew J Miller
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- University of Iowa, Iowa City, IA, USA
| | - Tatiana Moro
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Paul T Reidy
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, OH, USA
| | - Scott M Ebert
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Elena Volpi
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA
| | - Christopher M Adams
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Blake B Rasmussen
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA.
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA.
| |
Collapse
|
33
|
Yeh SL, Chen PY, Liao JW, Huang RL, Yu SH, Chen LN, Lee MH, Chen LW, Chen HW, Yang YC, Wu YL, Liu KL. The Protective Effects of Perch Essence Against Muscle Atrophy in Cancer Cachexia and Cisplatin Treatment. Curr Issues Mol Biol 2025; 47:152. [PMID: 40136406 PMCID: PMC11941385 DOI: 10.3390/cimb47030152] [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: 01/22/2025] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Muscle atrophy, through several pathways including increased protein catabolism, leads to adverse effects in cachexia induced by cancer and chemotherapy. Perch essence (PE) is a perch extract rich in branched-chain amino acids and peptides. The present study initially investigated the effects of PE supplementation on muscle atrophy in a mouse model of cancer cachexia induced by C26 cancer cells and compared these effects with those of tryptone. Compared with the tumor-only group, we found that PE supplementation significantly improved body weight, muscle mass, maximum limb grip strength (MLGS), and myosin heavy chain expression in the muscles of tumor-bearing mice. PE also significantly inhibited the expression of factors related to protein degradation, oxidative stress, and inflammation, while enhancing the expression of antioxidant enzymes in tumor-bearing mice. These effects of PE were associated with an increased expression of phosphorylated Akt and forkhead box protein O1, along with a reduced expression of phosphorylated nuclear factor-κB p65 in the muscles of tumor-bearing mice. Furthermore, PE similarly increased MLGS and attenuated muscle atrophy in mice exposed to cisplatin by inhibiting protein degradation. All the therapeutic effects of PE supplementation mentioned above were generally greater than those of tryptone supplementation. These results suggest the potential of PE in protecting against muscle atrophy induced by tumors or chemotherapy.
Collapse
Affiliation(s)
- Shu-Lan Yeh
- Department of Nutrition, Chung Shan Medical University, Taichung 40203, Taiwan; (S.-L.Y.); (R.-L.H.); (S.-H.Y.)
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung 40203, Taiwan
| | - Pei-Yin Chen
- Department of Senior Citizen Welfare and Long-Term Care Business (Master Program), Hungkuang University, Taichung 433304, Taiwan;
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, College of Veterinary Medicine, National Chung Hsing University, Taichung 402202, Taiwan;
| | - Ruo-Li Huang
- Department of Nutrition, Chung Shan Medical University, Taichung 40203, Taiwan; (S.-L.Y.); (R.-L.H.); (S.-H.Y.)
| | - Shu-Han Yu
- Department of Nutrition, Chung Shan Medical University, Taichung 40203, Taiwan; (S.-L.Y.); (R.-L.H.); (S.-H.Y.)
| | - Ling-Ni Chen
- Anyong Biotechnology Inc., Kaohsiung 827012, Taiwan; (L.-N.C.); (M.-H.L.)
| | - Mao-Hsiang Lee
- Anyong Biotechnology Inc., Kaohsiung 827012, Taiwan; (L.-N.C.); (M.-H.L.)
| | - Li-Wen Chen
- Division of Nutrition Therapy, Jen-Ai Hospital, Taichung 412224, Taiwan;
| | - Haw-Wen Chen
- Department of Nutrition, China Medical University, Taichung 40402, Taiwan;
| | - Ya-Chen Yang
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 413, Taiwan;
| | - Yu-Ling Wu
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung 40203, Taiwan; (S.-L.Y.); (R.-L.H.); (S.-H.Y.)
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung 40203, Taiwan
| |
Collapse
|
34
|
Ru Q, Li Y, Zhang X, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in muscle diseases and disorders: mechanisms and therapeutic prospects. Bone Res 2025; 13:27. [PMID: 40000618 PMCID: PMC11861620 DOI: 10.1038/s41413-024-00398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
The muscular system plays a critical role in the human body by governing skeletal movement, cardiovascular function, and the activities of digestive organs. Additionally, muscle tissues serve an endocrine function by secreting myogenic cytokines, thereby regulating metabolism throughout the entire body. Maintaining muscle function requires iron homeostasis. Recent studies suggest that disruptions in iron metabolism and ferroptosis, a form of iron-dependent cell death, are essential contributors to the progression of a wide range of muscle diseases and disorders, including sarcopenia, cardiomyopathy, and amyotrophic lateral sclerosis. Thus, a comprehensive overview of the mechanisms regulating iron metabolism and ferroptosis in these conditions is crucial for identifying potential therapeutic targets and developing new strategies for disease treatment and/or prevention. This review aims to summarize recent advances in understanding the molecular mechanisms underlying ferroptosis in the context of muscle injury, as well as associated muscle diseases and disorders. Moreover, we discuss potential targets within the ferroptosis pathway and possible strategies for managing muscle disorders. Finally, we shed new light on current limitations and future prospects for therapeutic interventions targeting ferroptosis.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
35
|
Wu C, Tong Y, Huang J, Wang S, Kobori H, Zhang Z, Suzuki K. Atrophic C2C12 Myotubes Activate Inflammatory Response of Macrophages In Vitro. Cells 2025; 14:317. [PMID: 40072046 PMCID: PMC11899532 DOI: 10.3390/cells14050317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Skeletal muscle wasting is commonly observed in aging, immobility, and chronic diseases. In pathological conditions, the impairment of skeletal muscle and immune system often occurs simultaneously. Recent studies have highlighted the initiative role of skeletal muscle in interactions with immune cells. However, the impact of skeletal muscle wasting on macrophage inflammatory responses remains poorly understood. METHODS To investigate the effect of atrophic myotubes on the inflammatory response of macrophages, we established two in vitro models to induce myotube atrophy: one induced by D-galactose and the other by starvation. Conditioned medium (CM) from normal and atrophic myotubes were collected and administered to bone marrow-derived macrophages (BMDMs) from mice. Subsequently, lipopolysaccharide (LPS) stimulation was applied, and the expression of inflammatory cytokines was measured via RT-qPCR. RESULTS Both D-galactose and starvation treatments reduced myotube diameter and upregulated muscle atrophy-related gene expression. CM from both atrophic myotubes models augmented the gene expression of pro-inflammatory factors in BMDMs following LPS stimulation, including Il6, Il1b, and Nfkb1. Notably, CM from starvation-induced atrophic myotubes also enhanced Il12b, Tnf, and Nos2 expression in BMDMs after stimulation, a response not observed in D-galactose-induced atrophic myotubes. CONCLUSIONS These findings suggest that CM from atrophic myotubes enhanced the expression of LPS-induced pro-inflammatory mediators in macrophages.
Collapse
Affiliation(s)
- Cong Wu
- Graduate School of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan; (C.W.); (Y.T.); (J.H.); (S.W.); (H.K.); (Z.Z.)
| | - Yishan Tong
- Graduate School of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan; (C.W.); (Y.T.); (J.H.); (S.W.); (H.K.); (Z.Z.)
| | - Jiapeng Huang
- Graduate School of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan; (C.W.); (Y.T.); (J.H.); (S.W.); (H.K.); (Z.Z.)
| | - Shuo Wang
- Graduate School of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan; (C.W.); (Y.T.); (J.H.); (S.W.); (H.K.); (Z.Z.)
| | - Haruki Kobori
- Graduate School of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan; (C.W.); (Y.T.); (J.H.); (S.W.); (H.K.); (Z.Z.)
| | - Ziwei Zhang
- Graduate School of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan; (C.W.); (Y.T.); (J.H.); (S.W.); (H.K.); (Z.Z.)
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
| |
Collapse
|
36
|
Cefis M, Marcangeli V, Hammad R, Granet J, Leduc-Gaudet JP, Gaudreau P, Trumpff C, Huang Q, Picard M, Aubertin-Leheudre M, Bélanger M, Robitaille R, Morais JA, Gouspillou G. Impact of physical activity on physical function, mitochondrial energetics, ROS production, and Ca 2+ handling across the adult lifespan in men. Cell Rep Med 2025; 6:101968. [PMID: 39933528 PMCID: PMC11866497 DOI: 10.1016/j.xcrm.2025.101968] [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: 07/16/2024] [Revised: 11/05/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
Aging-related muscle atrophy and weakness contribute to loss of mobility, falls, and disability. Mitochondrial dysfunction is widely considered a key contributing mechanism to muscle aging. However, mounting evidence positions physical activity as a confounding factor, making unclear whether muscle mitochondria accumulate bona fide defects with aging. To disentangle aging from physical activity-related mitochondrial adaptations, we functionally profiled skeletal muscle mitochondria in 51 inactive and 88 active men aged 20-93. Physical activity status confers partial protection against age-related decline in physical performance. Mitochondrial respiration remains unaltered in active participants, indicating that aging per se does not alter mitochondrial respiratory capacity. Mitochondrial reactive oxygen species (ROS) production is unaffected by aging and higher in active participants. In contrast, mitochondrial calcium retention capacity decreases with aging regardless of physical activity and correlates with muscle mass, performance, and the stress-responsive metabokine/mitokine growth differentiation factor 15 (GDF15). Targeting mitochondrial calcium handling may hold promise for treating aging-related muscle impairments.
Collapse
Affiliation(s)
- Marina Cefis
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; INSERM UMR1093-CAPS, UFR des Sciences de santé, Université de Bourgogne, Dijon, France
| | - Vincent Marcangeli
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Département des sciences biologiques, Université du Québec À Montréal, Montreal, QC, Canada
| | - Rami Hammad
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Département des sciences biologiques, Université du Québec À Montréal, Montreal, QC, Canada; Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, QC, Canada; Al-Ahliyya Amman university, Faculty of educational sciences, Department of physical and health education, Amman, Jordan
| | - Jordan Granet
- Département des sciences biologiques, Université du Québec À Montréal, Montreal, QC, Canada; Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, QC, Canada
| | - Jean-Philippe Leduc-Gaudet
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Research Group in Cellular Signaling, Department of Medical Biology, Université du Québec À Trois-Rivières, Trois-Rivières, Canada
| | - Pierrette Gaudreau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Département de médecine, Université de Montréal, Montreal, QC, Canada
| | - Caroline Trumpff
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, and Robert N Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Qiuhan Huang
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, and Robert N Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Martin Picard
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, and Robert N Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Mylène Aubertin-Leheudre
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, QC, Canada
| | - Marc Bélanger
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada
| | - Richard Robitaille
- Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Département de neurosciences, Université de Montréal, Montreal, QC, Canada; Centre interdisciplinaire de recherche sur le cerveau et l'apprentissage, Montreal, QC, Canada
| | - José A Morais
- Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Research Institute of the McGill University Health Centre, Montreal, QC, Canada; Division of Geriatric Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Gilles Gouspillou
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, QC, Canada; Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
37
|
Bogucka D, Wajda A, Stypińska B, Radkowski MJ, Targowski T, Modzelewska E, Kmiołek T, Ejma-Multański A, Filipowicz G, Kaliberda Y, Dudek E, Paradowska-Gorycka A. Epigenetic factors and inflammaging: FOXO3A as a potential biomarker of sarcopenia and upregulation of DNMT3A and SIRT3 in older adults. Front Immunol 2025; 16:1467308. [PMID: 40034697 PMCID: PMC11872893 DOI: 10.3389/fimmu.2025.1467308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Background Epigenetic factors influence inflammaging and geriatric disorders such as sarcopenia and frailty. It is necessary to develop a biomarker/panel of biomarkers for fast and easy diagnostics. Currently, hard-to-access equipment is required to diagnose sarcopenia. The development of a biomarker/panel of biomarkers will prevent many older adults from being excluded from the diagnostic process. Methods In this study, we analyzed selected gene expression profiles, namely, SIRT1, SIRT3, SIRT6, DNMT3A, FOXO1, FOXO3A, and ELAVL1, in whole blood. The study included 168 subjects divided into five groups: patients hospitalized at the Geriatrics Clinic and Polyclinic with sarcopenia, frailty syndrome, or without those disorders (geriatric control), and non-hospitalized healthy controls (HC) aged 25 to 30 years and over 50 years. Results We revealed a lower mRNA level of FOXO3A (p<0.001) in sarcopenic patients compared to the geriatric controls. Furthermore, we detected upregulation of DNMT3A (p=0.003) and SIRT3 (p=0.015) in HC over 50 years old compared to HC aged 25 to 30 years. Interestingly, we observed 2 cluster formations during the gene expression correlation analysis (SIRT1, SIRT3, DNMT3A, and FOXO1, ELAVL1). We also noted correlations of clinical parameters with mRNA levels in the sarcopenic patients group, such as vitamin D level with SIRT1 (r=0.64, p=0.010), creatine kinase with SIRT3 (r=-0.58, p=0.032) and DNMT3A (r=-0.59, p=0.026), creatinine with DNMT3A (r=0.57, p=0.026), erythrocyte sedimentation rate (ESR) with FOXO3A (r=0.69, p=0.004), and lactate dehydrogenase (LDH) with FOXO3A (r=-0.86, p=0.007). In the frailty syndrome group, we noted a correlation of appendicular skeletal muscle mass (ASMM) with ELAVL1 (r=0.59, p=0.026) mRNA level. In the geriatric controls, we observed a correlation of serum iron with FOXO3A mRNA level (r=-0.79, p=0.036). Conclusions Our study revealed FOXO3A as a potential biomarker of sarcopenia. Furthermore, we observed a high expression of epigenetic factors (DNMT3A and SIRT3) in older adults.
Collapse
Affiliation(s)
- Diana Bogucka
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Anna Wajda
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Barbara Stypińska
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Marcin Jerzy Radkowski
- Department of Geriatrics, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Tomasz Targowski
- Department of Geriatrics, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Ewa Modzelewska
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Tomasz Kmiołek
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Adam Ejma-Multański
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Gabriela Filipowicz
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Yana Kaliberda
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Ewa Dudek
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Agnieszka Paradowska-Gorycka
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| |
Collapse
|
38
|
Huot JR, Jamnick NA, Pin F, Livingston PD, Callaway CS, Bonetto A. GL261 glioblastoma induces delayed body weight gain and stunted skeletal muscle growth in young mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.635159. [PMID: 39990490 PMCID: PMC11844426 DOI: 10.1101/2025.02.10.635159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Introduction The survival rate for children and adolescents has increased to over 85%. However, there is limited understanding of the impact of pediatric cancers on muscle development and physiology. Given that brain tumors alone account for 26% of all pediatric cancers, this study aimed to investigate the skeletal muscle consequences of tumor growth in young mice. Methods C2C12 myotubes were co-cultured with GL261 murine glioblastoma cells to assess myotube size. GL261 cells were then injected subcutaneously into 4-week-old male C57BL/6J mice. Animals were euthanized 28 days post-GL261 implantation. Muscle function was tested in vivo and ex vivo . Muscle protein synthesis was measured via the SUnSET method, and gene/protein expression levels were assessed via Western blotting and qPCR. Results In vitro , the C2C12 cultures exposed to GL261 exhibited myotube atrophy, consistent with a disrupted anabolic/catabolic balance. In vivo , carcass, heart, and fat mass were significantly reduced in the tumor-bearing mice. Skeletal muscle growth was impeded in the GL261 hosts, along with smaller muscle CSA. Both in vivo muscle torque and the ex vivo EDL muscle force were unchanged. At molecular level, the tumor hosts displayed reduced muscle protein synthesis and increased muscle protein ubiquitination, in disagreement with decreased muscle ubiquitin ligase mRNA expression. Conclusions Overall, we showed that GL261 tumors impact the growth of pediatric mice by stunting skeletal muscle development, decreasing muscle mass, reducing muscle fiber size, diminishing muscle protein synthesis, and altering protein catabolism signaling.
Collapse
|
39
|
Guo Y, Han S, Yu W, Xu Y, Ying Y, Xu H, Feng H, Wang X, Wu W, Wang D, Liu L, Han X, Lou W. Deciphering molecular crosstalk mechanisms between skeletal muscle atrophy and KRAS-mutant pancreatic cancer: a literature review. Hepatobiliary Surg Nutr 2025; 14:78-95. [PMID: 39925900 PMCID: PMC11806137 DOI: 10.21037/hbsn-24-282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 09/30/2024] [Indexed: 02/11/2025]
Abstract
Background and Objective Cachexia-induced skeletal muscle atrophy is a critical manifestation in Kirsten rat sarcoma viral oncogene homologue (KRAS)-mutant pancreatic cancer (PC) patients, predominantly characterized by a shift in metabolic equilibrium towards catabolism that accelerates protein degradation in myofibers and leads to muscle atrophy. This metabolic reprogramming not only supports tumor growth but also precipitates energy depletion in skeletal muscle tissues. Exploring these mechanisms reveals potential therapeutic targets in the metabolic and proteolytic pathways associated with KRAS-mutant PC. Methods A comprehensive search for literature was conducted in PubMed, Web of Science, Google Scholar and other search engines up to May 21st, 2024. Studies on PC models and patients were included. Key Content and Findings The crosstalk between KRAS-mutant PC and skeletal muscle atrophy can be categorized into four principal domains: (I) KRAS-driven metabolic reprogramming in cancer cells leads to the depletion of muscle energy reserves, thereby influencing the reallocation of myofiber energy towards fueling cancer cell; (II) KRAS-mutant cancer cells rely on nutrient-scavenging pathways, resulting in altered cytokine profiles, increased ubiquitin mRNA expression and autophagy-lysosome pathway, which facilitate myotube degradation and inhibit muscle regeneration, thereby disrupting muscular homeostasis and causing a one-way nutrient flux; (III) tumor-induced oxidative stress inflicts damage on myotubes, highlighting the detrimental effects of reactive oxygen species on muscle structure; (IV) KRAS-mutant cancer cells remodulate immune cell dynamics within the tumor environment, thereby reshaping host immunity. Together, these findings illuminate the intricate interplay between KRAS-mutant PC and skeletal muscle atrophy, mapping the pathophysiological framework that is crucial for understanding sarcopenia and related disorders. Conclusions This comprehensive analysis advances our understanding of the complex etiology of cancer cachexia and stimulates the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Yuquan Guo
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Siyang Han
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weisheng Yu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yaolin Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Ying
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huaxiang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haokang Feng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xu’an Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenchuan Wu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dansong Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xu Han
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenhui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, The Shanghai Geriatrics Medical Center, Fudan University, Shanghai, China
| |
Collapse
|
40
|
Flaherty S, Song L, Albuquerque B, Rinaldi A, Piper M, Shanthappa D, Chen X, Stansfield J, Asano S, Pashos E, Ross T, Jagarlapudi S, Sheikh A, Zhang B, Wu Z. GDF15 Neutralization Ameliorates Muscle Atrophy and Exercise Intolerance in a Mouse Model of Mitochondrial Myopathy. J Cachexia Sarcopenia Muscle 2025; 16:e13715. [PMID: 39976232 PMCID: PMC11840706 DOI: 10.1002/jcsm.13715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 11/27/2024] [Accepted: 12/25/2024] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Primary mitochondrial myopathies (PMMs) are disorders caused by mutations in genes encoding mitochondrial proteins and proteins involved in mitochondrial function. PMMs are characterized by loss of muscle mass and strength as well as impaired exercise capacity. Growth/Differentiation Factor 15 (GDF15) was reported to be highly elevated in PMMs and cancer cachexia. Previous studies have shown that GDF15 neutralization is effective in improving skeletal muscle mass and function in cancer cachexia. It remains to be determined if the inhibition of GDF15 could be beneficial for PMMs. The purpose of the present study is to assess whether treatment with a GDF15 neutralizing antibody can alleviate muscle atrophy and physical performance impairment in a mouse model of PMM. METHODS The effects of GDF15 neutralization on PMM were assessed using PolgD257A/D257A (POLG) mice. These mice express a proofreading-deficient version of the mitochondrial DNA polymerase gamma, leading to an increased rate of mutations in mitochondrial DNA (mtDNA). These animals display increased circulating GDF15 levels, reduced muscle mass and function, exercise intolerance, and premature aging. Starting at 9 months of age, the mice were treated with an anti-GDF15 antibody (mAB2) once per week for 12 weeks. Body weight, food intake, body composition, and muscle mass were assessed. Muscle function and exercise capacity were evaluated using in vivo concentric max force stimulation assays, forced treadmill running and voluntary home-cage wheel running. Mechanistic investigations were performed via muscle histology, bulk transcriptomic analysis, RT-qPCR and western blotting. RESULTS Anti-GDF15 antibody treatment ameliorated the metabolic phenotypes of the POLG animals, improving body weight (+13% ± 8%, p < 0.0001), lean mass (+13% ± 15%, p < 0.001) and muscle mass (+35% ± 24%, p < 0.001). Additionally, the treatment improved skeletal muscle max force production (+35% ± 43%, p < 0.001) and exercise performance, including treadmill (+40% ± 29%, p < 0.05) and voluntary wheel running (+320% ± 19%, p < 0.05). Mechanistically, the beneficial effects of GDF15 neutralization are linked to the reversal of the transcriptional dysregulation in genes involved in autophagy and proteasome signalling. The treatment also appears to dampen glucocorticoid signalling by suppressing circulating corticosterone levels in the POLG animals. CONCLUSIONS Our findings highlight the potential of GDF15 neutralization with a monoclonal antibody as a therapeutic avenue to enhance physical performance and mitigate adverse clinical outcomes in patients with PMM.
Collapse
Affiliation(s)
- Stephen E. Flaherty
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Obesity and ComplicationsEli LillyBostonMassachusettsUSA
| | - LouJin Song
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Bina Albuquerque
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Anthony Rinaldi
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Program Mamager, Preclinical Sciences, ToxicologyVertex PharmaceuticalsBostonMassachusettsUSA
| | - Mary Piper
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | | | - Xian Chen
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - John Stansfield
- Biostatistics, Early Clinical DevelopmentPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Shoh Asano
- Inflammation and Immunology Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Evanthia Pashos
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Trenton Thomas Ross
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Srinath Jagarlapudi
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Abdul Sheikh
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Bei Zhang
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Zhidan Wu
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| |
Collapse
|
41
|
Brown JL, Xu H, Duggan E, Rosenfeld CS, Remmen HV. Pharmacological reduction of lipid hydroperoxides as a potential modulator of sarcopenia. J Physiol 2025; 603:837-854. [PMID: 39777675 PMCID: PMC12042244 DOI: 10.1113/jp287090] [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: 06/13/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
We previously reported that elevated expression of phospholipid hydroperoxide glutathione peroxidase 4, an enzyme that regulates membrane lipid hydroperoxides, can mitigate sarcopenia in mice. However, it is still unknown whether a pharmacological intervention designed to modulate lipid hydroperoxides might be an effective strategy to reduce sarcopenia in aged mice. Here we asked whether a newly developed compound, CMD-35647 (CMD), can reduce muscle atrophy induced by sciatic nerve transection. We treated mice daily with vehicle or CMD (15 mg/kg, i.p. injection) starting 1 day prior to denervation. CMD treatment reduced hydroperoxide generation and blunted muscle atrophy by over 17% in denervated muscle. To test whether CMD can reduce ageing-induced muscle atrophy and weakness, we treated mice with either vehicle or CMD (15 mg/kg, i.p. injection) 3 days per week for 8 months, starting at 18 months of age until 26 months of age. We measured muscle mass, functional status of neuromuscular junctions, muscle contractile function and mitochondrial function in control and CMD-treated 26-month-old female mice. Treatment with CMD conferred protection against muscle atrophy in both tibialis anterior and extensor digitorum longus that was associated with maintenance of fibre size of MHC 2b and 2x fibres. Mitochondrial respiration was also protected in CMD-treated mice. We also found that muscle force generation was protected with CMD treatment despite denervation in ∼25% of the muscle fibres. Overall, this study shows that pharmacological interventions designed to reduce lipid hydroperoxides might be effective for preventing sarcopenia. KEY POINTS: Sarcopenia in aged mice is associated with muscle loss, contractile dysfunction, denervation, and reduced mitochondrial respiration. CMD-35647 is a pharmocological compound that can neutralize lipid hydroperoxides. 8 month treatment of CMD-35647 mitigated muscle atrophy in tibialis anterior and extensor digitorum longus. 8 month treatment of CMD-35647 improved muscle function in aged mice independent of the neuromuscular junction. Aged mice treated with CMD-35647 had greater respiration in red gastrocnemius muscle when compared to vehicle treated mice.
Collapse
Affiliation(s)
- Jacob L. Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
- Oklahoma City VA Medical Center, Oklahoma City, OK 73104, United States
| | - Hongyang Xu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
| | - Elizabeth Duggan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
- Oklahoma City VA Medical Center, Oklahoma City, OK 73104, United States
| | | | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, United States
- Oklahoma City VA Medical Center, Oklahoma City, OK 73104, United States
| |
Collapse
|
42
|
Gopal Krishnan PD, Lee WX, Goh KY, Choy SM, Turqueza LRR, Lim ZH, Tang HW. Transcriptional regulation of autophagy in skeletal muscle stem cells. Dis Model Mech 2025; 18:DMM052007. [PMID: 39925192 PMCID: PMC11849978 DOI: 10.1242/dmm.052007] [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: 02/11/2025] Open
Abstract
Muscle stem cells (MuSCs) are essential for the regenerative capabilities of skeletal muscles. MuSCs are maintained in a quiescent state, but, when activated, can undergo proliferation and differentiation into myocytes, which fuse and mature to generate muscle fibers. The maintenance of MuSC quiescence and MuSC activation are processes that are tightly regulated by autophagy, a conserved degradation system that removes unessential or dysfunctional cellular components via lysosomes. Both the upregulation and downregulation of autophagy have been linked to impaired muscle regeneration, causing myopathies such as cancer cachexia, sarcopenia and Duchenne muscular dystrophy. In this Review, we highlight the importance of autophagy in regulating MuSC activity during muscle regeneration. Additionally, we summarize recent studies that link the transcriptional dysregulation of autophagy to muscle atrophy, emphasizing the dominant roles that transcription factors play in myogenic programs. Deciphering and understanding the roles of these transcription factors in the regulation of autophagy during myogenesis could advance the development of regenerative medicine.
Collapse
Affiliation(s)
- Priya D. Gopal Krishnan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Wen Xing Lee
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Kah Yong Goh
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Sze Mun Choy
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | | | - Zhuo Han Lim
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Hong-Wen Tang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore
| |
Collapse
|
43
|
Yang A, Lv Q, Han Z, Dai S, Li Y, Hao M, Yu R, Zhu J, Yang C, Shi Z, Zhou J. The Effects of Vitamin D on Muscle Strength Are Influenced by Testosterone Levels. J Cachexia Sarcopenia Muscle 2025; 16:e13733. [PMID: 39957010 PMCID: PMC11830628 DOI: 10.1002/jcsm.13733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Although the role of vitamin D receptor (VDR) in muscle mass and strength is well established, the effects of vitamin D (VD) on muscle remain controversial due to various factors. Herein, the influence of sex on the effects of VD on muscle function and the underlying reasons was explored. METHODS Male and female Sod1 gene knockout (SKO) mice, serving as a model for skeletal muscle atrophy, were treated with the VD active analogue calcipotriol, and RNA sequencing was employed to investigate this potential signalling pathway. The National Health and Nutrition Examination Survey (NHANES) database was utilized to explore whether testosterone affects the correlation between VD and grip strength in human participants. Experiments involving C2C12 cells and castrated male mice subjected to immobilization were conducted to demonstrate the enhancing effects of testosterone on VD function. RESULTS In male SKO mice, Vdr expression in the gastrocnemius muscle was positively correlated with grip strength (R2 = 0.4689, p < 0.001), whereas no such correlation was identified in female mice. At 28 weeks of age, both male and female SKO mice exhibited significantly reduced grip strength compared to Sod1 wild-type (SWT) mice, and calcipotriol restored grip strength in male SKO mice (SWT-veh: 0.0716 ± 0.0006, SWT-cal: 0.0686 ± 0.0010, SKO-veh: 0.0601 ± 0.0010, SKO-cal: 0.0703 ± 0.0007; p < 0.05). Calcipotriol increased muscle protein synthesis and mitochondrial biogenesis while decreasing inflammation and atrogenes in gastrocnemius muscle of male SKO mice. However, the effect of calcipotriol on muscle was not significant in female SKO mice. Compared to wild-type mice, both male and female SKO mice exhibited reduced levels of 1,25(OH)2D3 due to ROS-induced hepatic CYP3A4 overexpression, thereby excluding the influence of baseline VD levels. The serum 25(OH)D3 and testosterone interactively affect grip strength in adults (p < 0.05). Using C2C12 cells differentiated into myotubes, testosterone significantly enhanced the inducing effects of VD on VDR, androgen receptor (AR), P-AKT, PGC1α, Beclin1 and LC3B. Calcipotriol improved grip strength in sham-operated mice but had a negligible effect on grip strength in castrated mice. However, a significant improvement in grip strength was observed in castrated mice following testosterone restoration (p < 0.05). CONCLUSIONS This study demonstrates the existence of sex heterogeneity in the effects of VD on muscle and that testosterone enhances the strength and molecular responses to VD. These findings underscore the importance of considering testosterone levels when utilizing VD to enhance muscle strength.
Collapse
Affiliation(s)
- Aolin Yang
- School of Public Health (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhenGuangdongChina
- Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| | - Qingqing Lv
- School of Public Health (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Ziyu Han
- School of Public Health (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Shimiao Dai
- School of Public Health (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Yao Li
- School of Public Health (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Mengru Hao
- School of Public Health (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Ruirui Yu
- School of Public Health (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Junying Zhu
- School of Public Health (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Chenggang Yang
- School of Public Health (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Zhan Shi
- School of Public Health (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Ji‐Chang Zhou
- School of Public Health (Shenzhen)Shenzhen Campus of Sun Yat‐sen UniversityShenzhenGuangdongChina
- Guangdong Province Engineering Laboratory for Nutrition TranslationSun Yat‐sen UniversityShenzhenGuangdongChina
- Guangdong Provincial Key Laboratory of Food, Nutrition and HealthSun Yat‐sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
44
|
Li X, Yao X, Zhao W, Wei B, Zhang R, Yan G, Ma M, Wang Z, Liu X, Liu Y, Wang G, Li H, Kong Q, Wang J, Mu L. Muscle fiber types switched during the development of experimental autoimmune myasthenia gravis via the PI3K/Akt signaling pathway. Mol Immunol 2025; 178:41-51. [PMID: 39832429 DOI: 10.1016/j.molimm.2025.01.006] [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: 11/16/2024] [Revised: 12/25/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
As one of the largest organs of our human body, skeletal muscle has good research prospects in myasthenia gravis (MG), the symptoms of which include systemic skeletal muscle weakness. Skeletal muscle is composed of two types of muscle fibers. Different fiber subtypes can be converted into each other; however, the underlying mechanism is not yet clear. In this paper, we firstly established an experimental autoimmune myasthenia gravis (EAMG) rat model and found that the skeletal muscle fibers of the EAMG group were atrophied, with a change in the proportion of fiber subtypes, which switched from type IIa to type I in the EAMG group at the peak stage, as verified by histological and molecular analyses. Second-generation sequencing results predicted that the PI3K-Akt signaling pathway might be involved in the switch, and the mRNA expression levels of the PI3K-Akt pathway-related genesNr4a1, IL2rb, Col1A1 and Ddit4 were significantly different. In conclusion, this study indicates that the switch of muscle fiber subtypes in MG via the PI3K-Akt signaling pathway may be a potential target for the treatment of MG-related skeletal muscle atrophy in the future.
Collapse
MESH Headings
- Animals
- Signal Transduction
- Proto-Oncogene Proteins c-akt/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Myasthenia Gravis, Autoimmune, Experimental/pathology
- Myasthenia Gravis, Autoimmune, Experimental/metabolism
- Myasthenia Gravis, Autoimmune, Experimental/immunology
- Rats
- Muscle Fibers, Skeletal/pathology
- Muscle Fibers, Skeletal/metabolism
- Female
- Muscle, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Rats, Inbred Lew
- Muscular Atrophy/pathology
Collapse
Affiliation(s)
- Xinrong Li
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Xiuhua Yao
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Wei Zhao
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Bo Wei
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Ran Zhang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Geng Yan
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Mingyu Ma
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Zhenhai Wang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Xijun Liu
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Yumei Liu
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Guangyou Wang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Qingfei Kong
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Jinghua Wang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China; Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Lili Mu
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China.
| |
Collapse
|
45
|
Fung TKH, Cheung KK, Wang X, Lau BWM, Ngai SPC. Transcriptomic Profiling Reveals Differences in Slow-Twitch and Fast-Twitch Muscles of a Cigarette Smoke-Exposed Rat Model. J Cachexia Sarcopenia Muscle 2025; 16:e13633. [PMID: 39611217 DOI: 10.1002/jcsm.13633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 10/03/2024] [Accepted: 10/11/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Cigarette smoking is known to affect muscle function and exercise capacity, including muscle fatigue resistance. Most studies showed diminished cross-sectional area and fibre type shifting in slow-twitch muscles such as the soleus, while effects on fast-twitch muscles were seldom reported and the differential responses between muscle types in response to exposure to cigarette smoke (CS) were largely unknown. This study aimed to elucidate the histomorphological, biochemical and transcriptomic changes induced by CS on both slow-twitch and fast-twitch muscles. METHOD Male Sprague-Dawley rats were randomly divided into two groups: sham air (SA) and CS. The rats were exposed to CS for 8 weeks using an exposure chamber system to mimic smoking conditions. Histomorphological analyses on muscle fibre type and cross-sectional area were determined in soleus and extensor digitorum longus (EDL). Transcriptomic profiles were investigated for identifying differentially expressed genes (DEGs) and potential mechanistic pathways involved. Inflammatory responses in terms of the macrophage population and the level of inflammatory cytokines were measured. Markers for muscle-specific proteolysis were also examined. RESULT Soleus muscle, but not in EDL, exhibited a significant increase in Type IIa fibres (SA: 9.0 ± 3.3%; CS: 19.8 ± 2.4%, p = 0.002) and decrease in Type I fibres (SA: 90.1 ± 3.6%; CS: 77.9 ± 3.3%, p = 0.003) after CS exposure. RNA sequencing revealed 165 identified DEGs in soleus including upregulation of 'Cd68', 'Ccl2' and 'Ucp2' as well as downregulation of 'Ucp3', etc. Pathways enrichment analysis revealed that the upregulated pathways in soleus were related to immune system and cellular response, while the downregulated pathways were related to oxidative metabolism. Only 10 DEGs were identified in EDL with less enriched pathways. The soleus also showed elevated pro-inflammatory cytokines, and the total macrophage marker CD68 was significantly higher in soleus of CS compared to the SA group (CD68+/no. of fibre: SA = 60.3 ± 39.3%; CS = 106.5 ± 27.2%, p = 0.0039), while the two groups in EDL muscle showed no significant difference. The expression of E3 ubiquitin ligase atrogin-1 associated with muscle degradation pathways was 1.63-fold higher in the soleus after CS, while no significant differences were observed in the EDL. CONCLUSION The CS-induced inflammatory responses on soleus muscle are likely mediated via targeting mitochondrial-related signalling, resulting in mitochondrial dysfunction and impaired oxidative capacity. The presumably less active mitochondrial-related signalling in EDL renders it less susceptible to changes towards CS, accounting for differential impacts between muscle types.
Collapse
Affiliation(s)
- Timothy K H Fung
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Kwok Kuen Cheung
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Xia Wang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Benson W M Lau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Shirley P C Ngai
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| |
Collapse
|
46
|
Song J, Yang M, Xia L, Wang L, Wang K, Xiang Y, Cheng J, Chen J, Liu J, Zhao R, Liu F, Sun Z, Hou X, Zang N, Chen L. Aptamer-Conjugated Exosomes Ameliorate Diabetes-Induced Muscle Atrophy by Enhancing SIRT1/FoxO1/3a-Mediated Mitochondrial Function. J Cachexia Sarcopenia Muscle 2025; 16:e13717. [PMID: 39871746 PMCID: PMC11773161 DOI: 10.1002/jcsm.13717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Muscle atrophy is associated with Type 2 diabetes mellitus, which reduces the quality of life and lacks effective treatment strategies. Previously, it was determined that human umbilical cord mesenchymal stromal cell (hucMSC)-derived exosomes (EXOs) ameliorate diabetes-induced muscle atrophy. However, the systemic application of EXOs is less selective for diseased tissues, which reduces their efficacy and safety associated with their nonspecific biological distribution in vivo. Therefore, improving exosomal targeting is imperative. In this study, a skeletal muscle-specific aptamer (Apt) was used to explore the effects of Apt-functionalized EXOs derived from hucMSCs in diabetes-associated muscle atrophy and its specific mechanisms. METHODS Diabetic db/db mice and C2C12 myotubes were used to explore the effects of MSC-EXOs or Apt-EXOs in alleviating muscle atrophy. Grip strength, muscle weight and muscle fibre cross-sectional area (CSA) were used to evaluate skeletal muscle strength and muscle mass. Western blot analysis of muscle atrophy signalling, including MuRF1 and Atrogin 1 and the mitochondrial complex and Seahorse analysis were performed to investigate the underlying mechanisms of MSC-EXOs or Apt-EXOs on muscle atrophy. RESULTS MSC-EXOs increased grip strength (p = 0.0002) and muscle mass (p = 0.0044 for tibialis anterior (TA) muscle, p = 0.002 for soleus (SO) muscle) in db/db mice. It also increased the CSA of muscle fibres (p = 0.0011 for all fibres, p = 0.0036 for slow muscle fibres and p = 0.0089 for fast muscle fibres) and the percentage of slow-to-fast muscle fibres (p = 0.0109). However, Atrogin 1 (p = 0.0455) and MuRF1 expression (p = 0.0168) was reduced. MSC-EXOs activated SIRT1/FoxO1/3a signalling and enhanced mitochondrial function in db/db mice and C2C12 myotubes. SIRT1 knockdown decreased the beneficial antiatrophic effects of MSC-EXOs. Additionally, Apt conjugation increased the effect of MSC-EXOs on muscle atrophy and myofiber-type transition (p = 0.0133 for grip strength, p = 0.0124 for TA muscle weight, p = 0.0008 for SO muscle weight, p < 0.0001 for CSA of all muscle fibres, p = 0.0198 for CSA of slow muscle fibres, p = 0.0213 for CSA of fast muscle fibres, p = 0.011 for percentage of slow-fast muscle fibres, p = 0.0141 for Atrogin 1 expression and p = 0.005 for MuRF1 expression). CONCLUSIONS The results suggest that hucMSC-derived exosomes ameliorate diabetes-associated muscle atrophy by enhancing SIRT1/FoxO1/3a-mediated mitochondrial function and that Apt conjugation strengthens the effects of MSC-EXOs on muscle atrophy. These findings demonstrate the therapeutic potential of muscle-targeted MSC-EXOs for the treatment of muscle atrophy.
Collapse
Affiliation(s)
- Jia Song
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Mengmeng Yang
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Longqing Xia
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Liming Wang
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Kewei Wang
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Yingyue Xiang
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Jun Cheng
- Department of Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker DetectionThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Jun Chen
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Jidong Liu
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Ruxing Zhao
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Fuqiang Liu
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Zheng Sun
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Xinguo Hou
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
- Shandong Provincial Key Laboratory of Spatiotemporal Regulation and Precision Intervention in Endocrine and Metabolic Diseases, Shandong Provincial Engineering Research Center for Advanced Technologies in Prevention and Treatment of Chromic Metabolic Diseases, Institute of Endocrine and Metabolic Diseases of Shandong UniversityJinanShandongChina
| | - Nan Zang
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
- Shandong Provincial Key Laboratory of Spatiotemporal Regulation and Precision Intervention in Endocrine and Metabolic Diseases, Shandong Provincial Engineering Research Center for Advanced Technologies in Prevention and Treatment of Chromic Metabolic Diseases, Institute of Endocrine and Metabolic Diseases of Shandong UniversityJinanShandongChina
| | - Li Chen
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
- Shandong Provincial Key Laboratory of Spatiotemporal Regulation and Precision Intervention in Endocrine and Metabolic Diseases, Shandong Provincial Engineering Research Center for Advanced Technologies in Prevention and Treatment of Chromic Metabolic Diseases, Institute of Endocrine and Metabolic Diseases of Shandong UniversityJinanShandongChina
| |
Collapse
|
47
|
Yang W, Si SC, Wang WH, Li J, Ma YX, Zhao H, Liu J. Gut dysbiosis in primary sarcopenia: potential mechanisms and implications for novel microbiome-based therapeutic strategies. Front Microbiol 2025; 16:1526764. [PMID: 39935646 PMCID: PMC11810907 DOI: 10.3389/fmicb.2025.1526764] [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: 11/12/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
Primary sarcopenia is characterized by a progressive loss of skeletal muscle mass, strength, and physical function that occurs with aging. Despite the related adverse or even serious health outcomes, no medications are currently available for treating primary sarcopenia. Here, we discuss recent advancements in understanding the mechanistic role of gut microbiota-muscle cross-talk in primary sarcopenia, and the therapeutic implications. The mechanistic insights encompass a causal role of gut dysbiosis in primary sarcopenia, potentially mediated through gut microbiota-derived bioactive metabolites, such as short-chain fatty acids (SCFAs), secondary bile acids, and their associated signaling pathways, which may be translated into the development of new microbiome-based treatment and diagnostic approaches. Furthermore, we identify challenges that need addressing in future studies to facilitate the translation into potential novel treatment and differential diagnosis for older individuals with sarcopenia.
Collapse
Affiliation(s)
- Wei Yang
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
48
|
Sedlacek J. Activation of the 26S Proteasome to Reduce Proteotoxic Stress and Improve the Efficacy of PROTACs. ACS Pharmacol Transl Sci 2025; 8:21-35. [PMID: 39816802 PMCID: PMC11729432 DOI: 10.1021/acsptsci.4c00408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
The 26S proteasome degrades the majority of cellular proteins and affects all aspects of cellular life. Therefore, the 26S proteasome abundance, proper assembly, and activity in different life contexts need to be precisely controlled. Impaired proteasome activity is considered a causative factor in several serious disorders. Recent advances in proteasome biology have revealed that the proteasome can be activated by different factors or small molecules. Thus, activated ubiquitin-dependent proteasome degradation has effects such as extending the lifespan in different models, preventing the accumulation of protein aggregates, and reducing their negative impact on cells. Increased 26S proteasome-mediated degradation reduces proteotoxic stress and can potentially improve the efficacy of engineered degraders, such as PROTACs, particularly in situations characterized by proteasome malfunction. Here, emerging ideas and recent insights into the pharmacological activation of the proteasome at the transcriptional and posttranslational levels are summarized.
Collapse
Affiliation(s)
- Jindrich Sedlacek
- Department
of Genetics and Microbiology, Charles University
and Research Center BIOCEV, Pru°myslová 595, Vestec 252 50, Czech Republic
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 16610 Prague, Czech
Republic
| |
Collapse
|
49
|
Balnis J, Jackson EL, Drake LA, Singer DV, Bossardi Ramos R, Singer HA, Jaitovich A. Rapamycin improves satellite cells' autophagy and muscle regeneration during hypercapnia. JCI Insight 2025; 10:e182842. [PMID: 39589836 PMCID: PMC11721297 DOI: 10.1172/jci.insight.182842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
Both CO2 retention, or hypercapnia, and skeletal muscle dysfunction predict higher mortality in critically ill patients. Mechanistically, muscle injury and reduced myogenesis contribute to critical illness myopathy, and while hypercapnia causes muscle wasting, no research has been conducted on hypercapnia-driven dysfunctional myogenesis in vivo. Autophagy flux regulates myogenesis by supporting skeletal muscle stem cell - satellite cell - activation, and previous data suggest that hypercapnia inhibits autophagy. We tested whether hypercapnia worsens satellite cell autophagy flux and myogenic potential and if autophagy induction reverses these deficits. Satellite cell transplantation and lineage-tracing experiments showed that hypercapnia undermined satellite cells' activation, replication, and myogenic capacity. Bulk and single-cell sequencing analyses indicated that hypercapnia disrupts autophagy, senescence, and other satellite cell programs. Autophagy activation was reduced in hypercapnic cultured myoblasts, and autophagy genetic knockdown phenocopied these changes in vitro. Rapamycin stimulation led to AMPK activation and downregulation of the mTOR pathway, which are both associated with accelerated autophagy flux and cell replication. Moreover, hypercapnic mice receiving rapamycin showed improved satellite cell autophagy flux, activation, replication rate, and posttransplantation myogenic capacity. In conclusion, we have shown that hypercapnia interferes with satellite cell activation, autophagy flux, and myogenesis, and systemic rapamycin administration improves these outcomes.
Collapse
Affiliation(s)
- Joseph Balnis
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Emily L. Jackson
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Lisa A. Drake
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Diane V. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Ramon Bossardi Ramos
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Harold A. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| |
Collapse
|
50
|
Liu SY, Chen LK, Chung YT, Chen CW, Wu GL, Chang YC, Chen PR, Chang YI, Lin HF, Wu LY, Juan CC. Glucosamine inhibits myoblast proliferation and differentiation, and stimulates myotube atrophy through distinct signal pathways. J Nutr Biochem 2025; 135:109762. [PMID: 39251145 DOI: 10.1016/j.jnutbio.2024.109762] [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: 05/28/2024] [Revised: 08/01/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
Glucosamine (GlcN) is one of the dietary supplements used in the treatment of osteoarthritis. Endogenously, GlcN is synthesized from glucose through the hexosamine pathway. In addition to ameliorating arthritis, several biological functions of GlcN have been reported, including insulin resistance in skeletal muscle. However, the regulatory role of GlcN in skeletal muscle development is not clear. We therefore investigated the effect of GlcN on myoblast proliferation, differentiation, and myotube development and their underlying mechanisms in C2C12 cells. Myoblast proliferation was measured by MTT assay. The expressions of MyoD, myogenin (MyoG), and myosin heavy chain (MyHC) were identified as determinants of myoblast differentiation. Expressions of atrogin-1 and muscle RING-finger protein-1 (MuRF-1) were identified as markers of myotube atrophy. The results show that treatment with GlcN significantly reduced myoblast proliferation and phosphorylation of Stat3 and S6K. These findings suggest that GlcN can inhibit growth of myoblasts through inhibiting phosphorylation of Stat3 and S6K. In addition, GlcN significantly suppressed the expression of MyoD, MyoG, and MyHC, as well as myotube formation. Pretreatment of C2C12 myoblast cells with ER stress inhibitors significantly blocked GlcN-inhibited MyHC expression and myotube formation. It can be concluded that GlcN suppressed myogenic differentiation via a pathway that involved ER stress. Moreover, GlcN decreased myotube diameter and expression of MyHC, as well as increased MuRF-1 in C2C12 myotubes. Meanwhile, GlcN also reduced the expressions of phosphorylated Akt and mTOR were stimulated after GlcN treatment in C2C12 myotubes. Thus, GlcN induced skeletal muscle atrophy by inhibiting the protein synthesis pathway. Chronic GlcN infusion also caused skeletal muscle atrophy in mice. In conclusion, GlcN regulated important stages of skeletal muscle development through different signaling pathways.
Collapse
Affiliation(s)
- Shui-Yu Liu
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Luen-Kui Chen
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ting Chung
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chien-Wei Chen
- Department of Physical Education, Health, and Recreation, Teachers College, National Chiayi University, Chiayi, Taiwan
| | - Guan-Lin Wu
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Chieh Chang
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pin-Rong Chen
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yuan-I Chang
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Heng-Fu Lin
- Division of Trauma, Department of Surgery, Far-Eastern Memorial Hospital, New Taipei City, Taiwan; Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan.
| | - Liang-Yi Wu
- Department of Bioscience Technology, College of Science, Chung-Yuan Christian University, Chung Li, Taiwan.
| | - Chi-Chang Juan
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|