1
|
Ren S, Chai J, Zhang L, Li J, Long X, Zhang T. The role of microRNAs in dexamethasone-induced skeletal muscle atrophy. Exp Gerontol 2025; 205:112749. [PMID: 40250741 DOI: 10.1016/j.exger.2025.112749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
Muscle atrophy is characterized by a decrease in muscle mass, strength, and activity. Recently, it was determined that microRNAs (miRNAs) can regulate muscle atrophy and that dexamethasone (Dex), an allergy and autoimmune disorder treatment that can induce muscle atrophy. Therefore, this study was designed to identify miRNAs expressed in Dex-induced muscle atrophy in mice using small RNA sequencing. A total of 820 miRNAs were identified, with 58 miRNAs expressed explicitly in atrophic muscles. Dex-induced muscle atrophy miRNAs clustered separately from the differential miRNAs in aging, disuse, and cancer-induced muscle atrophy models. The target genes of Dex-induced muscle atrophy miRNAs were independently enriched in inositol phosphate metabolism, hypoxia-inducible factor-1 signaling pathway, etc. Of note, there was a significant increase in the volume of fat cells and adipose weight in the Dex group, suggesting that fat deposition during Dex-induced skeletal muscle atrophy is a unique and typical feature. SIMPLE SUMMARY: Dexamethasone (Dex) is a glucocorticoid used to treat allergic and autoimmune diseases, but excessive use can lead to skeletal muscle atrophy. We used dexamethasone (Dex) to build a muscle atrophy model in mice, and obvious changes had taken place in mouse body weight, muscle tissue morphology and related genes. A large number of microRNAs were found to be differentially expressed, and their functions were enriched in pathways related to muscle development. At the same time, we compared the similarities and differences of microRNAs and their functions between Dex induced muscle atrophy model and other muscle atrophy models. Finally, we were surprised to find that Dex induced muscle atrophy is specifically accompanied by the accumulation of body fat.
Collapse
Affiliation(s)
- Subi Ren
- Chongqing Academy of Animal Science, Rongchang, Chongqing 4024602, China
| | - Jie Chai
- Chongqing Academy of Animal Science, Rongchang, Chongqing 4024602, China; National Center of Technology Innovation for Pigs, Chongqing 4024602, China
| | - Lijuan Zhang
- Chongqing Academy of Animal Science, Rongchang, Chongqing 4024602, China; National Center of Technology Innovation for Pigs, Chongqing 4024602, China
| | - JiGang Li
- Chongqing Academy of Animal Science, Rongchang, Chongqing 4024602, China
| | - Xi Long
- Chongqing Academy of Animal Science, Rongchang, Chongqing 4024602, China; National Center of Technology Innovation for Pigs, Chongqing 4024602, China
| | - Tinghuan Zhang
- Chongqing Academy of Animal Science, Rongchang, Chongqing 4024602, China; National Center of Technology Innovation for Pigs, Chongqing 4024602, China.
| |
Collapse
|
2
|
Li X, Fang L, Zhou R, Yao L, Clayton SW, Muscat S, Kamm DR, Wang C, Liu CJ, Qin L, Tower RJ, Karner CM, Guilak F, Tang SY, Loiselle AE, Meyer GA, Shen J. Current cutting-edge omics techniques on musculoskeletal tissues and diseases. Bone Res 2025; 13:59. [PMID: 40484858 DOI: 10.1038/s41413-025-00442-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/31/2025] [Accepted: 04/27/2025] [Indexed: 06/11/2025] Open
Abstract
Musculoskeletal disorders, including osteoarthritis, rheumatoid arthritis, osteoporosis, bone fracture, intervertebral disc degeneration, tendinopathy, and myopathy, are prevalent conditions that profoundly impact quality of life and place substantial economic burdens on healthcare systems. Traditional bulk transcriptomics, genomics, proteomics, and metabolomics have played a pivotal role in uncovering disease-associated alterations at the population level. However, these approaches are inherently limited in their ability to resolve cellular heterogeneity or to capture the spatial organization of cells within tissues, thus hindering a comprehensive understanding of the complex cellular and molecular mechanisms underlying these diseases. To address these limitations, advanced single-cell and spatial omics techniques have emerged in recent years, offering unparalleled resolution for investigating cellular diversity, tissue microenvironments, and biomolecular interactions within musculoskeletal tissues. These cutting-edge techniques enable the detailed mapping of the molecular landscapes in diseased tissues, providing transformative insights into pathophysiological processes at both the single-cell and spatial levels. This review presents a comprehensive overview of the latest omics technologies as applied to musculoskeletal research, with a particular focus on their potential to revolutionize our understanding of disease mechanisms. Additionally, we explore the power of multi-omics integration in identifying novel therapeutic targets and highlight key challenges that must be overcome to successfully translate these advancements into clinical applications.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Liang Fang
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Renpeng Zhou
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, CT, USA
| | - Lutian Yao
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Sade W Clayton
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Samantha Muscat
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics & Physical Performance, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Dakota R Kamm
- Program in Physical Therapy, Washington University, St. Louis, MO, USA
| | - Cuicui Wang
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, CT, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert J Tower
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Courtney M Karner
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children-St. Louis, St. Louis, MO, USA
| | - Simon Y Tang
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Alayna E Loiselle
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics & Physical Performance, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Gretchen A Meyer
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Program in Physical Therapy, Washington University, St. Louis, MO, USA
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Jie Shen
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA.
| |
Collapse
|
3
|
Qin M, Wang Y, Wang Z, Dong B, Yang P, Liu Y, Xi Q, Ma J. Adipose-derived small extracellular vesicle miR-146a-5p targets Fbx32 to regulate mitochondrial autophagy and delay aging in skeletal muscle. J Nanobiotechnology 2025; 23:287. [PMID: 40211295 PMCID: PMC11983871 DOI: 10.1186/s12951-025-03367-1] [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: 02/26/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
This study investigates how miR-146a-5p, found in adipose tissue-derived small extracellular vesicles (sEV), influences mitochondrial autophagy and its impact on delaying skeletal muscle aging through the targeting of Fbx32. The findings highlight miR-146a-5p as crucial in skeletal muscle development and aging, influencing autophagy, apoptosis, differentiation, and proliferation, collectively impacting muscle atrophy. In C2C12 cells, miR-146a-5p mimics decreased apoptosis, autophagy, and reactive oxygen species (ROS) levels, while enhancing ATP production; conversely, miR-146a-5p inhibitors had the opposite effects. Furthermore, miR-146a-5p-enriched sEV from adipose tissue alleviated skeletal muscle atrophy in aged mice and promoted muscle fiber growth and repair by regulating mitochondrial autophagy and apoptosis. Mechanistically, miR-146a-5p modulated mitochondrial autophagy in myoblasts by targeting Fbx32 and impacting the FoxO3 signaling pathway. This led to a notable decrease in apoptosis-related gene expression, reduced ROS production, and elevated ATP levels. In conclusion, miR-146a-5p derived from WAT-sEV modulates myoblast autophagy, apoptosis, ROS, and differentiation through the Fbx32/FoxO3 signaling axis. This work presents a novel molecular target and theoretical framework for delaying skeletal muscle aging and developing therapies for skeletal muscle-related disorders.
Collapse
Affiliation(s)
- Mengran Qin
- Tianjin Hospital, Tianjin University, Tianjin, 300211, China
- Tianjin Orthopedic Institute, Tianjin, 300050, China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, 510642, Guangdong, China
| | - Yan Wang
- Tianjin Hospital, Tianjin University, Tianjin, 300211, China
- Tianjin Orthopedic Institute, Tianjin, 300050, China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China
| | - Zihan Wang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Benchao Dong
- Tianjin Hospital, Tianjin University, Tianjin, 300211, China
- Tianjin Orthopedic Institute, Tianjin, 300050, China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China
| | - Peichuan Yang
- Tianjin Hospital, Tianjin University, Tianjin, 300211, China
- Tianjin Orthopedic Institute, Tianjin, 300050, China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China
| | - Youyi Liu
- Tianjin Hospital, Tianjin University, Tianjin, 300211, China
- Tianjin Orthopedic Institute, Tianjin, 300050, China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Qianyun Xi
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, 510642, Guangdong, China.
| | - Jianxiong Ma
- Tianjin Hospital, Tianjin University, Tianjin, 300211, China.
- Tianjin Orthopedic Institute, Tianjin, 300050, China.
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China.
| |
Collapse
|
4
|
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] [Download PDF] [Figures] [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
|
5
|
Ko H, Le TT, Nguyen NB, Kang SW, Cha KH, Yang N, Jung SH, Kim M. Poncirus trifoliata Extract and Its Active Coumarins Alleviate Dexamethasone-Induced Skeletal Muscle Atrophy by Regulating Protein Synthesis, Mitochondrial Biogenesis, and Gut Microbiota. Phytother Res 2025. [PMID: 40088055 DOI: 10.1002/ptr.8478] [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: 08/11/2024] [Revised: 01/30/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Sarcopenia, an age-related decline in skeletal muscle mass and function, contributes to frailty and increased morbidity in the elderly. This necessitates the development of effective interventions to combat muscle atrophy. This study investigated the therapeutic potential of Poncirus trifoliata ethanol extract (PT) and its coumarin derivatives against dexamethasone (DEX)-induced muscle atrophy. We employed in vitro and in vivo models of DEX-induced muscle atrophy. C2C12 myotubes were used for mechanistic studies. C57BL/6J mice received DEX injections and oral PT supplementation (50 mg/kg/day) to evaluate effects on muscle mass, function, gene expression, and gut microbiota composition. In vitro, PT enhanced protein synthesis, mitochondrial biogenesis, and myogenic differentiation in DEX-exposed myotubes, with auraptene, ponciol, and triphasiol identified as key bioactive coumarins. In vivo, PT significantly attenuated DEX-induced muscle atrophy, increasing tibialis anterior muscle mass by 36% (p < 0.01), grip strength by 31% (p < 0.001), and maximal running speed by 18% (p < 0.05). Mechanistically, PT upregulated genes associated with muscle function and mitochondrial health. Furthermore, PT modulated gut microbiota composition, notably increasing Phocaeicola vulgatus abundance 2.2-fold, which correlated with improved muscle performance (R = 0.58, p < 0.01). These findings suggest that PT and its coumarin derivatives, particularly auraptene, ponciol, and triphasiol, hold promise as therapeutic agents for combating muscle atrophy. The observed benefits may be mediated through enhanced protein synthesis, improved mitochondrial function, and modulation of the gut-muscle axis.
Collapse
Affiliation(s)
- Hyejin Ko
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
| | - Tam Thi Le
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
| | - Ngoc Bao Nguyen
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
- College of Dentistry, Department of Biochemistry and Molecular Biology, Gangneung Wonju National University, Gangneung, Republic of Korea
| | - Suk Woo Kang
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Daejeon, Republic of Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Nain Yang
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
| | - Sang Hoon Jung
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Myungsuk Kim
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Daejeon, Republic of Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| |
Collapse
|
6
|
Acquarone D, Bertero A, Brancaccio M, Sorge M. Chaperone Proteins: The Rising Players in Muscle Atrophy. J Cachexia Sarcopenia Muscle 2025; 16:e13659. [PMID: 39707668 PMCID: PMC11747685 DOI: 10.1002/jcsm.13659] [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: 07/02/2024] [Revised: 10/18/2024] [Accepted: 10/31/2024] [Indexed: 12/23/2024] Open
Abstract
Despite significant progress in understanding the molecular aetiology of muscle atrophy, there is still a great need for new targets and drugs capable of counteracting muscle wasting. The role of an impaired proteostasis as the underlying causal mechanism of muscle atrophy is a well-established concept. From the earliest work on muscle atrophy and the identification of the first atrogenes, the hyper-activation of the proteolytic systems, such as autophagy and the ubiquitin proteasome system, has been recognized as the major driver of atrophy. However, the role of other key regulators of proteostasis, the chaperone proteins, has been largely overlooked. Chaperone proteins play a pivotal role in protein folding and in preventing the aggregation of misfolded proteins. Indeed, some chaperones, such as αB-crystallin and Hsp25, are involved in compensatory responses aimed at counteracting protein aggregation during sarcopenia. Chaperones also regulate different intracellular signalling pathways crucial for atrogene expression and the control of protein catabolism, such as the AKT and NF-kB pathways, which are regulated by Hsp70 and Hsp90. Furthermore, the downregulation of certain chaperones causes severe muscle wasting per se and experimental strategies aimed at preventing this downregulation have shown promising results in mitigating or reversing muscle atrophy. This highlights the therapeutic potential of targeting chaperones and confirms their crucial anti-atrophic functions. In this review, we summarize the most relevant data showing the modulation and the causative role of chaperone proteins in different types of skeletal muscle atrophies.
Collapse
Affiliation(s)
- Davide Acquarone
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | - Alessandro Bertero
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | - Matteo Sorge
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| |
Collapse
|
7
|
Song C, Zheng W, Liu G, Xu Y, Deng Z, Xiu Y, Zhang R, Yang L, Zhang Y, Yu G, Su Y, Luo J, He B, Xu J, Dai H. Sarcopenic obesity is attenuated by E-syt1 inhibition via improving skeletal muscle mitochondrial function. Redox Biol 2025; 79:103467. [PMID: 39675068 PMCID: PMC11699297 DOI: 10.1016/j.redox.2024.103467] [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: 11/25/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024] Open
Abstract
In aging and metabolic disease, sarcopenic obesity (SO) correlates with intramuscular adipose tissue (IMAT). Using bioinformatics analysis, we found a potential target protein Extended Synaptotagmin 1 (E-syt1) in SO. To investigate the regulatory role of E-syt1 in muscle metabolism, we performed in vivo and in vitro experiments through E-syt1 loss- and gain-of-function on muscle physiology. When E-syt1 is overexpressed in vitro, myoblast proliferation, differentiation, mitochondrial respiration, biogenesis, and mitochondrial dynamics are impaired, which were alleviated by the silence of E-syt1. Furthermore, overexpression of E-syt1 inhibited mitophagic flux. Mechanistically, E-syt1 overexpression leads to mitochondrial calcium overload and mitochondrial ROS burst, inhibits the fusion of mitophagosomes with lysosomes, and impedes the acidification of lysosomes. Animal experiments demonstrated the inhibition of E-syt1 increased the capacity of endurance exercise, muscle mass, mitochondrial function, and oxidative capacity of the muscle fibers in OVX mice. These findings establish E-syt1 as a novel contributor to the pathogenesis of skeletal muscle metabolic disorders in SO. Consequently, targeting E-syt1-induced dysfunction may serve as a viable strategy for attenuating SO.
Collapse
Affiliation(s)
- Chao Song
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China; School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, 350001, China
| | - Wu Zheng
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Guoming Liu
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Yiyang Xu
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Zhibo Deng
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Yu Xiu
- Department of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Rongsheng Zhang
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Linhai Yang
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Yifei Zhang
- Department of Pediatrics, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Guoyu Yu
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Yibin Su
- Department of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Jun Luo
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Bingwei He
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China; School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, 350001, China.
| | - Jie Xu
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China.
| | - Hanhao Dai
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China.
| |
Collapse
|
8
|
Zhang G, Hu F, Huang T, Ma X, Cheng Y, Liu X, Jiang W, Dong B, Fu C. The recent development, application, and future prospects of muscle atrophy animal models. MEDCOMM – FUTURE MEDICINE 2024; 3. [DOI: 10.1002/mef2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/01/2024] [Indexed: 01/06/2025]
Abstract
AbstractMuscle atrophy, characterized by the loss of muscle mass and function, is a hallmark of sarcopenia and cachexia, frequently associated with aging, malignant tumors, chronic heart failure, and malnutrition. Moreover, it poses significant challenges to human health, leading to increased frailty, reduced quality of life, and heightened mortality risks. Despite extensive research on sarcopenia and cachexia, consensus in their assessment remains elusive, with inconsistent conclusions regarding their molecular mechanisms. Muscle atrophy models are crucial tools for advancing research in this field. Currently, animal models of muscle atrophy used for clinical and basic scientific studies are induced through various methods, including aging, genetic editing, nutritional modification, exercise, chronic wasting diseases, and drug administration. Muscle atrophy models also include in vitro and small organism models. Despite their value, each of these models has certain limitations. This review focuses on the limitations and diverse applications of muscle atrophy models to understand sarcopenia and cachexia, and encourage their rational use in future research, therefore deepening the understanding of underlying pathophysiological mechanisms, and ultimately advancing the exploration of therapeutic strategies for sarcopenia and cachexia.
Collapse
Affiliation(s)
- Gongchang Zhang
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Fengjuan Hu
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Tingting Huang
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Xiaoqing Ma
- Longkou People Hospital Longkou Shandong Province China
| | - Ying Cheng
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Xiaolei Liu
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Wenzhou Jiang
- Longkou People Hospital Longkou Shandong Province China
| | - Birong Dong
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Chenying Fu
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
- Department of Laboratory of Aging and Geriatric Medicine National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University Chengdu Sichuan Province China
| |
Collapse
|
9
|
Iki T, Tohda C. Skeletal muscle atrophy induces memory dysfunction via hemopexin action in healthy young mice. Biochem Biophys Res Commun 2024; 733:150606. [PMID: 39208645 DOI: 10.1016/j.bbrc.2024.150606] [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: 08/08/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Age-related morbidity has become an increasingly significant issue worldwide. Sarcopenia, the decline in skeletal muscle mass and strength with age, has been reported to be a risk factor for cognitive impairment. Our previous study revealed that skeletal muscle atrophy shifts the onset of memory dysfunction earlier in young Alzheimer's disease mice and found that hemopexin is a myokine responsible for memory loss. This study aimed to elucidate the occurrence of memory impairment due to skeletal muscle atrophy in non-genetically engineered healthy young mice and the involvement of hemopexin. Closed-colony ddY mice at 12-13 weeks of age were used. Both hind limbs were immobilized by cast attachment for 14 d. Casting for 2 weeks induced a loss of skeletal muscle weight. The memory function of the mice was evaluated using a novel object recognition test. The cast-attached mice exhibited memory impairment. Hemopexin levels in the conditioned medium of the skeletal muscle, plasma, and hippocampus were increased in cast-attached mice. Continuous intracerebroventricular hemopexin infusion induced memory deficits in non-cast mice. To investigate whether hemopexin is the main causative factor of cognitive impairment, cast-attached mice were intracerebroventricularly infused with an anti-hemopexin antibody. Cast-induced memory impairment was reversed by the infusion of an anti-hemopexin antibody. These findings provide new evidence that skeletal muscle atrophy causes memory impairment in healthy young mice through the action of hemopexin in the brain.
Collapse
Affiliation(s)
- Tsukasa Iki
- Section of Neuromedical Science, Division of Bioscience, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Chihiro Tohda
- Section of Neuromedical Science, Division of Bioscience, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| |
Collapse
|
10
|
Tran MP, Ochoa Reyes D, Weitzel AJ, Saxena A, Hiller M, Cooper KL. Gene expression differences associated with intrinsic hindfoot muscle loss in the jerboa, Jaculus jaculus. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2024; 342:453-464. [PMID: 38946691 DOI: 10.1002/jez.b.23268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/16/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024]
Abstract
Vertebrate animals that run or jump across sparsely vegetated habitats, such as horses and jerboas, have reduced the number of distal limb bones, and many have lost most or all distal limb muscle. We previously showed that nascent muscles are present in the jerboa hindfoot at birth and that these myofibers are rapidly and completely lost soon after by a process that shares features with pathological skeletal muscle atrophy. Here, we apply an intra- and interspecies differential RNA-Seq approach, comparing jerboa and mouse muscles, to identify gene expression differences associated with the initiation and progression of jerboa hindfoot muscle loss. We show evidence for reduced hepatocyte growth factor and fibroblast growth factor signaling and an imbalance in nitric oxide signaling; all are pathways that are necessary for skeletal muscle development and regeneration. We also find evidence for phagosome formation, which hints at how myofibers may be removed by autophagy or by nonprofessional phagocytes without evidence for cell death or immune cell activation. Last, we show significant overlap between genes associated with jerboa hindfoot muscle loss and genes that are differentially expressed in a variety of human muscle pathologies and rodent models of muscle loss disorders. All together, these data provide molecular insight into the process of evolutionary and developmental muscle loss in jerboa hindfeet.
Collapse
Affiliation(s)
- Mai P Tran
- Department of Cell and Developmental Biology, University of California San Diego, La Jolla, California, USA
| | - Daniel Ochoa Reyes
- Department of Cell and Developmental Biology, University of California San Diego, La Jolla, California, USA
| | - Alexander J Weitzel
- Department of Cell and Developmental Biology, University of California San Diego, La Jolla, California, USA
| | - Aditya Saxena
- Department of Cell and Developmental Biology, University of California San Diego, La Jolla, California, USA
| | - Michael Hiller
- LOEWE Centre for Translational Biodiversity Genomics, Frankfurt, Germany
- Senckenberg Research Institute, Frankfurt, Germany
- Faculty of Biosciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Kimberly L Cooper
- Department of Cell and Developmental Biology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
11
|
Gutierrez-Monreal MA, Wolff CA, Rijos EE, Viggars MR, Douglas CM, Pagala V, Peng J, Hunt LC, Ding H, Huo Z, Demontis F, Esser KA. Targeted Bmal1 restoration in muscle prolongs lifespan with systemic health effects in aging model. JCI Insight 2024; 9:e174007. [PMID: 39352748 PMCID: PMC11601919 DOI: 10.1172/jci.insight.174007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/26/2024] [Indexed: 10/04/2024] Open
Abstract
Disruption of the circadian clock in skeletal muscle worsens local and systemic health, leading to decreased muscle strength, metabolic dysfunction, and aging-like phenotypes. Whole-body knockout mice that lack Bmal1, a key component of the molecular clock, display premature aging. Here, by using adeno-associated viruses, we rescued Bmal1 expression specifically in the skeletal muscle fibers of Bmal1-KO mice and found that this engaged the circadian clock and clock output gene expression, contributing to extended lifespan. Time course phenotypic analyses found that muscle strength, mobility, and glucose tolerance were improved with no effects on muscle mass or fiber size or type. A multiomics approach at 2 ages further determined that restored muscle Bmal1 improved glucose handling pathways while concomitantly reducing lipid and protein metabolic pathways. The improved glucose tolerance and metabolic flexibility resulted in the systemic reduction of inflammatory signatures across peripheral tissues, including liver, lung, and white adipose fat. Together, these findings highlight the critical role of muscle Bmal1 and downstream target genes for skeletal muscle homeostasis with considerable implications for systemic health.
Collapse
Affiliation(s)
| | | | - Eduardo E. Rijos
- Department of Physiology and Aging
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, USA
| | | | | | - Vishwajeeth Pagala
- Department of Structural Biology, Center for Proteomics and Metabolomics, and
| | - Junmin Peng
- Department of Structural Biology, Center for Proteomics and Metabolomics, and
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Liam C. Hunt
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Biology, Rhodes College, Memphis, Tennessee, USA
| | - Haocheng Ding
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Zhiguang Huo
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Karyn A. Esser
- Department of Physiology and Aging
- Myology Institute, and
| |
Collapse
|
12
|
Li M, Guo J, Qin Y, Lao Y, Kang SG, Huang K, Tong T. Dietary eugenol ameliorates long-term high-fat diet-induced skeletal muscle atrophy: mechanistic insights from integrated multi-omics. Food Funct 2024; 15:10136-10150. [PMID: 39292180 DOI: 10.1039/d4fo03648d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Eugenol (EU), the major constituent of clove oil, possesses a range of bioactivities. Here, the therapeutic potential of oral EU for mitigating skeletal muscle wasting was investigated in a long-term high-fat diet (HFD)-induced obese mice model. Male C57BL/6J mice, aged six weeks, were assigned to either a chow or a HFD for 10 weeks. Subsequently, the weight-matched HFD-fed mice were allocated into two groups, receiving either 0.2% (w/w) EU supplementation or no supplementation for 14 weeks. Our findings revealed that EU supplementation enhanced grip strength, increased hanging duration, and augmented skeletal muscle mass. RNA sequencing analysis demonstrated that EU modified the gastrocnemius muscle transcriptomic profile, and the differentially expressed genes between HFD and EU groups were mainly involved in the HIF-1 signaling pathway, TCR signaling pathway, and cGMP-PKG signaling pathway, which is well-known to be related to skeletal muscle health. Untargeted metabolomics analysis further showed that EU supplementation significantly altered the nucleotide metabolism in the GAS muscle. Analysis of 16S rRNA sequencing demonstrated that EU supplementation ameliorated the gut dysbiosis caused by HFD. The alterations in gut microbiota induced by EU were significantly correlated with indexes related to skeletal muscle atrophy. The multi-omics analysis presented the robust interaction among the skeletal muscle transcriptome, metabolome, and gut microbiome altered by EU supplementation. Our results highlight the potential of EU in skeletal muscle atrophy intervention as a functional dietary supplement.
Collapse
Affiliation(s)
- Mengjie Li
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua Donglu, Beijing 100083, China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
- Beijing Laboratory for Food Quality and Safety, Beijing 100083, China
| | - Jingya Guo
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua Donglu, Beijing 100083, China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
- Beijing Laboratory for Food Quality and Safety, Beijing 100083, China
| | - Yige Qin
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua Donglu, Beijing 100083, China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
- Beijing Laboratory for Food Quality and Safety, Beijing 100083, China
| | - Yujie Lao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua Donglu, Beijing 100083, China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
- Beijing Laboratory for Food Quality and Safety, Beijing 100083, China
| | - Seong-Gook Kang
- Department of Food Engineering and Solar Salt Research Center, Mokpo National University, Muangun 58554, Republic of Korea
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua Donglu, Beijing 100083, China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
- Beijing Laboratory for Food Quality and Safety, Beijing 100083, China
| | - Tao Tong
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua Donglu, Beijing 100083, China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), the Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
- Beijing Laboratory for Food Quality and Safety, Beijing 100083, China
| |
Collapse
|
13
|
Epstein SA, Doles JD, Dasgupta A. KLF10: a point of convergence in cancer cachexia. Curr Opin Support Palliat Care 2024; 18:120-125. [PMID: 39007915 PMCID: PMC11293965 DOI: 10.1097/spc.0000000000000711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
PURPOSE OF THE REVIEW Cancer-associated cachexia is a wasting syndrome entailing loss in body mass and a shortened life expectancy. There is currently no effective treatment to abrogate this syndrome, which leads to 20-30% of deaths in patients with cancer. While there have been advancements in defining signaling factors/pathways in cancer-induced muscle wasting, targeting the same in the clinic has not been as successful. Krüppel-like factor 10 (KLF10), a transcription factor implicated in muscle regulation, is regulated by the transforming growth factor-beta signaling pathway. This review proposes KLF10 as a potential convergence point of diverse signaling pathways involved in muscle wasting. RECENT FINDINGS KLF10 was discovered as a target of transforming growth factor-beta decades ago but more recently it has been shown that deletion of KLF10 rescues cancer-induced muscle wasting. Moreover, KLF10 has also been shown to bind key atrophy genes associated with muscle atrophy in vitro . SUMMARY There is an elevated need to explore targets in cachexia, which will successfully translate into the clinic. Investigating a convergence point downstream of multiple signaling pathways might hold promise in developing effective therapies for cachexia.
Collapse
Affiliation(s)
- Savannah A Epstein
- Department of Anatomy, Cell Biology and Physiology, Indiana School of Medicine, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
14
|
Kok HJ, Fletcher DB, Oster JC, Conover CF, Barton ER, Yarrow JF. Transcriptomics reveals transient and dynamic muscle fibrosis and atrophy differences following spinal cord injury in rats. J Cachexia Sarcopenia Muscle 2024; 15:1309-1323. [PMID: 38764311 PMCID: PMC11294049 DOI: 10.1002/jcsm.13476] [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: 07/05/2023] [Revised: 01/24/2024] [Accepted: 03/10/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND The rate and magnitude of skeletal muscle wasting after severe spinal cord injury (SCI) exceeds most other disuse conditions. Assessing the time course of molecular changes can provide insight into the progression of muscle wasting post-SCI. The goals of this study were (1) to identify potential targets that may prevent the pathologic features of SCI in soleus muscles and (2) to establish therapeutic windows for treating these pathologic changes. METHODS Four-month-old Sprague-Dawley male rats received T9 laminectomy (SHAM surgery) or severe contusion SCI. Hindlimb locomotor function was assessed weekly, with soleus muscles obtained 1 week, 2 weeks, 1 month and 3 months post-surgery (n = 6-7 per group per timepoint). RNA was extracted from muscles for bulk RNA-sequencing analysis (n = 3-5 per group per timepoint). Differentially expressed genes (DEGs) were evaluated between age-matched SHAM and SCI animals. Myofiber size, muscle fibre type and fibrosis were assessed on contralateral muscles. RESULTS SCI produced immediate and persistent hindlimb paralysis, with Basso-Beattie-Bresnahan locomotor scores remaining below 7 throughout the study, contributing to a progressive 25-50% lower soleus mass and myofiber atrophy versus SHAM (P < 0.05 at all timepoints). Transcriptional comparisons of SCI versus SHAM resulted in 184 DEGs (1 week), 436 DEGs (2 weeks), 133 DEGs (1 month) and 1200 DEGs (3 months). Upregulated atrophy-related genes included those associated with cell senescence, nuclear factor kappa B, ubiquitin proteasome and unfolded protein response pathways, along with upregulated genes that negatively influence muscle growth through the transforming growth factor beta pathway and inhibition of insulin-like growth factor-I/Akt/mechanistic target of rapamycin and p38/mitogen-activated protein kinase signalling. Genes associated with extracellular matrix (ECM), including collagens, collagen crosslinkers, proteoglycans and those regulating ECM integrity, were enriched within upregulated DEGs at 1 week but subsequently downregulated at 2 weeks and 3 months and were accompanied by >50% higher ECM areas and hydroxyproline levels in SCI muscles (P < 0.05). Myofiber remodelling genes were enriched in upregulated DEGs at 2 weeks and 1 month and were downregulated at 3 months. Genes that regulate neuromuscular junction remodelling were evident in muscles post-SCI, along with slow-to-fast fibre-type shifts: 1 week and 2 weeks SCI muscles were composed of 90% myosin heavy chain (MHC) type I fibres, which decreased to only 16% at 3 months and were accompanied by 50% fibres containing MHC IIX (P < 0.05). Metabolism genes were enriched in upregulated DEGs at 1 month and were further enriched at 3 months. CONCLUSIONS Our results substantiate many known pathologic features of SCI-induced wasting in rat skeletal muscle and identify a progressive and dynamic transcriptional landscape within the post-SCI soleus. Future studies are warranted to consider these therapeutic treatment windows when countering SCI muscle pathology.
Collapse
Affiliation(s)
- Hui Jean Kok
- Department of Applied Physiology and KinesiologyCollege of Health and Human Performance, University of FloridaGainesvilleFLUSA
- Research Service, Malcolm Randall Department of Veterans Affairs Medical CenterNorth Florida/South Georgia Veterans Health SystemGainesvilleFLUSA
| | - Drew B. Fletcher
- Department of Applied Physiology and KinesiologyCollege of Health and Human Performance, University of FloridaGainesvilleFLUSA
| | - Jacob C. Oster
- Department of Applied Physiology and KinesiologyCollege of Health and Human Performance, University of FloridaGainesvilleFLUSA
| | - Christine F. Conover
- Research Service, Malcolm Randall Department of Veterans Affairs Medical CenterNorth Florida/South Georgia Veterans Health SystemGainesvilleFLUSA
| | - Elisabeth R. Barton
- Department of Applied Physiology and KinesiologyCollege of Health and Human Performance, University of FloridaGainesvilleFLUSA
| | - Joshua F. Yarrow
- Research Service, Malcolm Randall Department of Veterans Affairs Medical CenterNorth Florida/South Georgia Veterans Health SystemGainesvilleFLUSA
- Division of Endocrinology, Diabetes and MetabolismCollege of Medicine, University of FloridaGainesvilleFLUSA
- Brain Rehabilitation Research Center, Malcolm Randall Department of Veterans Affairs Medical CenterNorth Florida/South Georgia Veterans Health SystemGainesvilleFLUSA
- Eastern Colorado Geriatrics Research, Education, and Clinical CenterRocky Mountain Regional Veterans Affairs Medical Center, VA Eastern Colorado Health Care SystemAuroraCOUSA
| |
Collapse
|
15
|
Dowling P, Gargan S, Zweyer M, Henry M, Meleady P, Swandulla D, Ohlendieck K. Proteomic reference map for sarcopenia research: mass spectrometric identification of key muscle proteins of organelles, cellular signaling, bioenergetic metabolism and molecular chaperoning. Eur J Transl Myol 2024; 34:12565. [PMID: 38787292 PMCID: PMC11264233 DOI: 10.4081/ejtm.2024.12565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 05/25/2024] Open
Abstract
During the natural aging process, frailty is often associated with abnormal muscular performance. Although inter-individual differences exit, in most elderly the tissue mass and physiological functionality of voluntary muscles drastically decreases. In order to study age-related contractile decline, animal model research is of central importance in the field of biogerontology. Here we have analyzed wild type mouse muscle to establish a proteomic map of crude tissue extracts. Proteomics is an advanced and large-scale biochemical method that attempts to identify all accessible proteins in a given biological sample. It is a technology-driven approach that uses mass spectrometry for the characterization of individual protein species. Total protein extracts were used in this study in order to minimize the potential introduction of artefacts due to excess subcellular fractionation procedures. In this report, the proteomic survey of aged muscles has focused on organellar marker proteins, as well as proteins that are involved in cellular signaling, the regulation of ion homeostasis, bioenergetic metabolism and molecular chaperoning. Hence, this study has establish a proteomic reference map of a highly suitable model system for future aging research.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| | - Margit Zweyer
- Department of Neonatology and Paediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases, Bonn.
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin.
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin.
| | - Dieter Swandulla
- Institute of Physiology, Medical Faculty, University of Bonn, Bonn.
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| |
Collapse
|
16
|
Dowling P, Gargan S, Zweyer M, Henry M, Meleady P, Swandulla D, Ohlendieck K. Proteomic reference map for sarcopenia research: mass spectrometric identification of key muscle proteins located in the sarcomere, cytoskeleton and the extracellular matrix. Eur J Transl Myol 2024; 34:12564. [PMID: 38787300 PMCID: PMC11264229 DOI: 10.4081/ejtm.2024.12564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 05/25/2024] Open
Abstract
Sarcopenia of old age is characterized by the progressive loss of skeletal muscle mass and concomitant decrease in contractile strength. Age-related skeletal muscle dysfunctions play a key pathophysiological role in the frailty syndrome and can result in a drastically diminished quality of life in the elderly. Here we have used mass spectrometric analysis of the mouse hindlimb musculature to establish the muscle protein constellation at advanced age of a widely used sarcopenic animal model. Proteomic results were further analyzed by systems bioinformatics of voluntary muscles. In this report, the proteomic survey of aged muscles has focused on the expression patterns of proteins involved in the contraction-relaxation cycle, membrane cytoskeletal maintenance and the formation of the extracellular matrix. This includes proteomic markers of the fast versus slow phenotypes of myosin-containing thick filaments and actin-containing thin filaments, as well as proteins that are associated with the non-sarcomeric cytoskeleton and various matrisomal layers. The bioanalytical usefulness of the newly established reference map was demonstrated by the comparative screening of normal versus dystrophic muscles of old age, and findings were verified by immunoblot analysis.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| | - Margit Zweyer
- Department of Neonatology and Paediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases, Bonn.
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin.
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin.
| | - Dieter Swandulla
- Institute of Physiology, Medical Faculty, University of Bonn, Bonn.
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| |
Collapse
|
17
|
Nguyen BN, Hong S, Choi S, Lee CG, Yoo G, Kim M. Dexamethasone-induced muscle atrophy and bone loss in six genetically diverse collaborative cross founder strains demonstrates phenotypic variability by Rg3 treatment. J Ginseng Res 2024; 48:310-322. [PMID: 38707648 PMCID: PMC11069000 DOI: 10.1016/j.jgr.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 11/14/2023] [Accepted: 12/26/2023] [Indexed: 05/07/2024] Open
Abstract
Background Osteosarcopenia is a common condition characterized by the loss of both bone and muscle mass, which can lead to an increased risk of fractures and disability in older adults. The study aimed to elucidate the response of various mouse strains to treatment with Rg3, one of the leading ginsenosides, on musculoskeletal traits and immune function, and their correlation. Methods Six Collaborative Cross (CC) founder strains induced muscle atrophy and bone loss with dexamethasone (15 mg/kg) treatment for 1 month, and half of the mice for each strain were orally administered Rg3 (20 mg/kg). Different responses were observed depending on genetic background and Rg3 treatment. Results Rg3 significantly increased grip strength, running performance, and expression of muscle and bone health-related genes in a two-way analysis of variance considering the genetic backgrounds and Rg3 treatment. Significant improvements in grip strength, running performance, bone area, and muscle mass, and the increased gene expression were observed in specific strains of PWK/PhJ. For traits related to muscle, bone, and immune functions, significant correlations between traits were confirmed following Rg3 administration compared with control mice. The phenotyping analysis was compiled into a public web resource called Rg3-OsteoSarco. Conclusion This highlights the complex interplay between genetic determinants, pathogenesis of muscle atrophy and bone loss, and phytochemical bioactivity and the need to move away from single inbred mouse models to improve their translatability to genetically diverse humans. Rg3-OsteoSarco highlights the use of CC founder strains as a valuable tool in the field of personalized nutrition.
Collapse
Affiliation(s)
- Bao Ngoc Nguyen
- College of Dentistry, Gangneung Wonju National University, Gangneung, Gangwon-do, Republic of Korea
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do, Republic of Korea
| | - Soyeon Hong
- Convergence Research Center for Smart Farm Solution, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do, Republic of Korea
| | - Sowoon Choi
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do, Republic of Korea
| | - Choong-Gu Lee
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju, Gangwon-do, Republic of Korea
| | - GyHye Yoo
- Convergence Research Center for Smart Farm Solution, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
| | - Myungsuk Kim
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon-do, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju, Gangwon-do, Republic of Korea
| |
Collapse
|
18
|
Kurochkina NS, Orlova MA, Vigovskiy MA, Zgoda VG, Vepkhvadze TF, Vavilov NE, Makhnovskii PA, Grigorieva OA, Boroday YR, Philippov VV, Lednev EM, Efimenko AY, Popov DV. Age-related changes in human skeletal muscle transcriptome and proteome are more affected by chronic inflammation and physical inactivity than primary aging. Aging Cell 2024; 23:e14098. [PMID: 38379415 PMCID: PMC11019131 DOI: 10.1111/acel.14098] [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: 10/05/2023] [Revised: 01/13/2024] [Accepted: 01/13/2024] [Indexed: 02/22/2024] Open
Abstract
Evaluation of the influence of primary and secondary aging on the manifestation of molecular and cellular hallmarks of aging is a challenging and currently unresolved issue. Our study represents the first demonstration of the distinct role of primary aging and chronic inflammation/physical inactivity - the most important drivers of secondary aging, in the regulation of transcriptomic and proteomic profiles in human skeletal muscle. To achieve this purpose, young healthy people (n = 15), young (n = 8) and older (n = 37) patients with knee/hip osteoarthritis, a model to study the effect of long-term inactivity and chronic inflammation on the vastus lateralis muscle, were included in the study. It was revealed that widespread and substantial age-related changes in gene expression in older patients relative to young healthy people (~4000 genes regulating mitochondrial function, proteostasis, cell membrane, secretory and immune response) were related to the long-term physical inactivity and chronic inflammation rather than primary aging. Primary aging contributed mainly to the regulation of genes (~200) encoding nuclear proteins (regulators of DNA repair, RNA processing, and transcription), mitochondrial proteins (genes encoding respiratory enzymes, mitochondrial complex assembly factors, regulators of cristae formation and mitochondrial reactive oxygen species production), as well as regulators of proteostasis. It was found that proteins associated with aging were regulated mainly at the post-transcriptional level. The set of putative primary aging genes and their potential transcriptional regulators can be used as a resource for further targeted studies investigating the role of individual genes and related transcription factors in the emergence of a senescent cell phenotype.
Collapse
Affiliation(s)
- Nadia S. Kurochkina
- Institute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
| | - Mira A. Orlova
- Institute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
| | - Maksim A. Vigovskiy
- Medical Research and Educational Center of Lomonosov Moscow State UniversityMoscowRussia
| | | | | | | | | | - Olga A. Grigorieva
- Medical Research and Educational Center of Lomonosov Moscow State UniversityMoscowRussia
| | - Yakov R. Boroday
- Medical Research and Educational Center of Lomonosov Moscow State UniversityMoscowRussia
| | - Vladislav V. Philippov
- Medical Research and Educational Center of Lomonosov Moscow State UniversityMoscowRussia
| | - Egor M. Lednev
- Institute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
| | - Anastasia Yu. Efimenko
- Medical Research and Educational Center of Lomonosov Moscow State UniversityMoscowRussia
| | - Daniil V. Popov
- Institute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
| |
Collapse
|
19
|
Hesketh SJ. Advancing cancer cachexia diagnosis with -omics technology and exercise as molecular medicine. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:1-15. [PMID: 38463663 PMCID: PMC10918365 DOI: 10.1016/j.smhs.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/15/2024] [Accepted: 01/20/2024] [Indexed: 03/12/2024] Open
Abstract
Muscle atrophy exacerbates disease outcomes and increases mortality, whereas the preservation of skeletal muscle mass and function play pivotal roles in ensuring long-term health and overall quality-of-life. Muscle atrophy represents a significant clinical challenge, involving the continued loss of muscle mass and strength, which frequently accompany the development of numerous types of cancer. Cancer cachexia is a highly prevalent multifactorial syndrome, and although cachexia is one of the main causes of cancer-related deaths, there are still no approved management strategies for the disease. The etiology of this condition is based on the upregulation of systemic inflammation factors and catabolic stimuli, resulting in the inhibition of protein synthesis and enhancement of protein degradation. Numerous necessary cellular processes are disrupted by cachectic pathology, which mediate intracellular signalling pathways resulting in the net loss of muscle and organelles. However, the exact underpinning molecular mechanisms of how these changes are orchestrated are incompletely understood. Much work is still required, but structured exercise has the capacity to counteract numerous detrimental effects linked to cancer cachexia. Primarily through the stimulation of muscle protein synthesis, enhancement of mitochondrial function, and the release of myokines. As a result, muscle mass and strength increase, leading to improved mobility, and quality-of-life. This review summarises existing knowledge of the complex molecular networks that regulate cancer cachexia and exercise, highlighting the molecular interplay between the two for potential therapeutic intervention. Finally, the utility of mass spectrometry-based proteomics is considered as a way of establishing early diagnostic biomarkers of cachectic patients.
Collapse
|
20
|
Lord SO, Dawson PW, Chunthorng-Orn J, Ng J, Baehr LM, Hughes DC, Sridhar P, Knowles T, Bodine SC, Lai YC. Uncovering the mechanisms of MuRF1-induced ubiquitylation and revealing similarities with MuRF2 and MuRF3. Biochem Biophys Rep 2024; 37:101636. [PMID: 38283190 PMCID: PMC10818185 DOI: 10.1016/j.bbrep.2023.101636] [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: 11/06/2023] [Revised: 12/12/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
MuRF1 (Muscle-specific RING finger protein 1; gene name TRIM63) is a ubiquitin E3 ligase, associated with the progression of muscle atrophy. As a RING (Really Interesting New Gene) type E3 ligase, its unique activity of ubiquitylation is driven by a specific interaction with a UBE2 (ubiquitin conjugating enzyme). Our understanding of MuRF1 function remains unclear as candidate UBE2s have not been fully elucidated. In the present study, we screened human ubiquitin dependent UBE2s in vitro and found that MuRF1 engages in ubiquitylation with UBE2D, UBE2E, UBE2N/V families and UBE2W. MuRF1 can cause mono-ubiquitylation, K48- and K63-linked polyubiquitin chains in a UBE2 dependent manner. Moreover, we identified a two-step UBE2 dependent mechanism whereby MuRF1 is monoubiquitylated by UBE2W which acts as an anchor for UBE2N/V to generate polyubiquitin chains. With the in vitro ubiquitylation assay, we also found that MuRF2 and MuRF3 not only share the same UBE2 partners as MuRF1 but can also directly ubiquitylate the same substrates: Titin (A168-A170), Desmin, and MYLPF (Myosin Light Chain, Phosphorylatable, Fast Skeletal Muscle; also called Myosin Light Regulatory Chain 2). In summary, our work presents new insights into the mechanisms that underpin MuRF1 activity and reveals overlap in MuRF-induced ubiquitylation which could explain their partial redundancy in vivo.
Collapse
Affiliation(s)
- Samuel O. Lord
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Peter W.J. Dawson
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | | | - Jimi Ng
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Leslie M. Baehr
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - David C. Hughes
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Pooja Sridhar
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - Timothy Knowles
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - Sue C. Bodine
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yu-Chiang Lai
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
21
|
Hughes DC, Goodman CA, Baehr LM, Gregorevic P, Bodine SC. A critical discussion on the relationship between E3 ubiquitin ligases, protein degradation, and skeletal muscle wasting: it's not that simple. Am J Physiol Cell Physiol 2023; 325:C1567-C1582. [PMID: 37955121 PMCID: PMC10861180 DOI: 10.1152/ajpcell.00457.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
Ubiquitination is an important post-translational modification (PTM) for protein substrates, whereby ubiquitin is added to proteins through the coordinated activity of activating (E1), ubiquitin-conjugating (E2), and ubiquitin ligase (E3) enzymes. The E3s provide key functions in the recognition of specific protein substrates to be ubiquitinated and aid in determining their proteolytic or nonproteolytic fates, which has led to their study as indicators of altered cellular processes. MuRF1 and MAFbx/Atrogin-1 were two of the first E3 ubiquitin ligases identified as being upregulated in a range of different skeletal muscle atrophy models. Since their discovery, the expression of these E3 ubiquitin ligases has often been studied as a surrogate measure of changes to bulk protein degradation rates. However, emerging evidence has highlighted the dynamic and complex regulation of the ubiquitin proteasome system (UPS) in skeletal muscle and demonstrated that protein ubiquitination is not necessarily equivalent to protein degradation. These observations highlight the potential challenges of quantifying E3 ubiquitin ligases as markers of protein degradation rates or ubiquitin proteasome system (UPS) activation. This perspective examines the usefulness of monitoring E3 ubiquitin ligases for determining specific or bulk protein degradation rates in the settings of skeletal muscle atrophy. Specific questions that remain unanswered within the skeletal muscle atrophy field are also identified, to encourage the pursuit of new research that will be critical in moving forward our understanding of the molecular mechanisms that govern protein function and degradation in muscle.
Collapse
Affiliation(s)
- David C Hughes
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Craig A Goodman
- Centre for Muscle Research (CMR), Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Leslie M Baehr
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Paul Gregorevic
- Centre for Muscle Research (CMR), Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Neurology, The University of Washington School of Medicine, Seattle, Washington, United States
| | - Sue C Bodine
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| |
Collapse
|
22
|
Marcotte GR, Miller MJ, Kunz HE, Ryan ZC, Strub MD, Vanderboom PM, Heppelmann CJ, Chau S, Von Ruff ZD, Kilroe SP, McKeen AT, Dierdorff JM, Stern JI, Nath KA, Grueter CE, Lira VA, Judge AR, Rasmussen BB, Nair KS, Lanza IR, Ebert SM, Adams CM. GADD45A is a mediator of mitochondrial loss, atrophy, and weakness in skeletal muscle. JCI Insight 2023; 8:e171772. [PMID: 37815864 PMCID: PMC10721312 DOI: 10.1172/jci.insight.171772] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023] Open
Abstract
Aging and many illnesses and injuries impair skeletal muscle mass and function, but the molecular mechanisms are not well understood. To better understand the mechanisms, we generated and studied transgenic mice with skeletal muscle-specific expression of growth arrest and DNA damage inducible α (GADD45A), a signaling protein whose expression in skeletal muscle rises during aging and a wide range of illnesses and injuries. We found that GADD45A induced several cellular changes that are characteristic of skeletal muscle atrophy, including a reduction in skeletal muscle mitochondria and oxidative capacity, selective atrophy of glycolytic muscle fibers, and paradoxical expression of oxidative myosin heavy chains despite mitochondrial loss. These cellular changes were at least partly mediated by MAP kinase kinase kinase 4, a protein kinase that is directly activated by GADD45A. By inducing these changes, GADD45A decreased the mass of muscles that are enriched in glycolytic fibers, and it impaired strength, specific force, and endurance exercise capacity. Furthermore, as predicted by data from mouse models, we found that GADD45A expression in skeletal muscle was associated with muscle weakness in humans. Collectively, these findings identify GADD45A as a mediator of mitochondrial loss, atrophy, and weakness in mouse skeletal muscle and a potential target for muscle weakness in humans.
Collapse
Affiliation(s)
- George R. Marcotte
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- University of Iowa, Iowa City, Iowa, USA
| | - Matthew J. Miller
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- University of Iowa, Iowa City, Iowa, USA
| | - Hawley E. Kunz
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Zachary C. Ryan
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew D. Strub
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Patrick M. Vanderboom
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Carrie J. Heppelmann
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Sarah Chau
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Sean P. Kilroe
- University of Texas Medical Branch, Galveston, Texas, USA
| | - Andrew T. McKeen
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- University of Iowa, Iowa City, Iowa, USA
| | | | | | - Karl A. Nath
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Andrew R. Judge
- University of Florida, Gainesville, Florida, USA
- Emmyon, Inc., Rochester, Minnesota, USA
| | - Blake B. Rasmussen
- University of Texas Medical Branch, Galveston, Texas, USA
- Emmyon, Inc., Rochester, Minnesota, USA
- University of Texas Health Science Center, San Antonio, Texas, USA
| | - K. Sreekumaran Nair
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ian R. Lanza
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Scott M. Ebert
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Emmyon, Inc., Rochester, Minnesota, USA
| | - Christopher M. Adams
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Emmyon, Inc., Rochester, Minnesota, USA
| |
Collapse
|
23
|
Hunt LC, Pagala V, Stephan A, Xie B, Kodali K, Kavdia K, Wang YD, Shirinifard A, Curley M, Graca FA, Fu Y, Poudel S, Li Y, Wang X, Tan H, Peng J, Demontis F. An adaptive stress response that confers cellular resilience to decreased ubiquitination. Nat Commun 2023; 14:7348. [PMID: 37963875 PMCID: PMC10646096 DOI: 10.1038/s41467-023-43262-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/02/2023] [Indexed: 11/16/2023] Open
Abstract
Ubiquitination is a post-translational modification initiated by the E1 enzyme UBA1, which transfers ubiquitin to ~35 E2 ubiquitin-conjugating enzymes. While UBA1 loss is cell lethal, it remains unknown how partial reduction in UBA1 activity is endured. Here, we utilize deep-coverage mass spectrometry to define the E1-E2 interactome and to determine the proteins that are modulated by knockdown of UBA1 and of each E2 in human cells. These analyses define the UBA1/E2-sensitive proteome and the E2 specificity in protein modulation. Interestingly, profound adaptations in peroxisomes and other organelles are triggered by decreased ubiquitination. While the cargo receptor PEX5 depends on its mono-ubiquitination for binding to peroxisomal proteins and importing them into peroxisomes, we find that UBA1/E2 knockdown induces the compensatory upregulation of other PEX proteins necessary for PEX5 docking to the peroxisomal membrane. Altogether, this study defines a homeostatic mechanism that sustains peroxisomal protein import in cells with decreased ubiquitination capacity.
Collapse
Affiliation(s)
- Liam C Hunt
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
- Department of Biology, Rhodes College, 2000 North Pkwy, Memphis, TN, 38112, USA
| | - Vishwajeeth Pagala
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Anna Stephan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Boer Xie
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Kiran Kodali
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Kanisha Kavdia
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Michelle Curley
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Flavia A Graca
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Yingxue Fu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Suresh Poudel
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yuxin Li
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xusheng Wang
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Haiyan Tan
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Junmin Peng
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
24
|
Kim HJ, Jung DW, Williams DR. Age Is Just a Number: Progress and Obstacles in the Discovery of New Candidate Drugs for Sarcopenia. Cells 2023; 12:2608. [PMID: 37998343 PMCID: PMC10670210 DOI: 10.3390/cells12222608] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
Sarcopenia is a disease characterized by the progressive loss of skeletal muscle mass and function that occurs with aging. The progression of sarcopenia is correlated with the onset of physical disability, the inability to live independently, and increased mortality. Due to global increases in lifespan and demographic aging in developed countries, sarcopenia has become a major socioeconomic burden. Clinical therapies for sarcopenia are based on physical therapy and nutritional support, although these may suffer from low adherence and variable outcomes. There are currently no clinically approved drugs for sarcopenia. Consequently, there is a large amount of pre-clinical research focusing on discovering new candidate drugs and novel targets. In this review, recent progress in this research will be discussed, along with the challenges that may preclude successful translational research in the clinic. The types of drugs examined include mitochondria-targeting compounds, anti-diabetes agents, small molecules that target non-coding RNAs, protein therapeutics, natural products, and repositioning candidates. In light of the large number of drugs and targets being reported, it can be envisioned that clinically approved pharmaceuticals to prevent the progression or even mitigate sarcopenia may be within reach.
Collapse
Affiliation(s)
| | - Da-Woon Jung
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea;
| | - Darren Reece Williams
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea;
| |
Collapse
|
25
|
Golding AE, Li W, Blank PS, Cologna SM, Zimmerberg J. Relative quantification of progressive changes in healthy and dysferlin-deficient mouse skeletal muscle proteomes. Muscle Nerve 2023; 68:805-816. [PMID: 37706611 DOI: 10.1002/mus.27975] [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: 11/09/2022] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023]
Abstract
INTRODUCTION/AIMS Individuals with dysferlinopathies, a group of genetic muscle diseases, experience delay in the onset of muscle weakness. The cause of this delay and subsequent muscle wasting are unknown, and there are currently no clinical interventions to limit or prevent muscle weakness. To better understand molecular drivers of dysferlinopathies, age-dependent changes in the proteomic profile of skeletal muscle (SM) in wild-type (WT) and dysferlin-deficient mice were identified. METHODS Quadriceps were isolated from 6-, 18-, 42-, and 77-wk-old C57BL/6 (WT, Dysf+/+ ) and BLAJ (Dysf-/- ) mice (n = 3, 2 male/1 female or 1 male/2 female, 24 total). Whole-muscle proteomes were characterized using liquid chromatography-mass spectrometry with relative quantification using TMT10plex isobaric labeling. Principle component analysis was utilized to detect age-dependent proteomic differences over the lifespan of, and between, WT and dysferlin-deficient SM. The biological relevance of proteins with significant variation was established using Ingenuity Pathway Analysis. RESULTS Over 3200 proteins were identified between 6-, 18-, 42-, and 77-wk-old mice. In total, 46 proteins varied in aging WT SM (p < .01), while 365 varied in dysferlin-deficient SM. However, 569 proteins varied between aged-matched WT and dysferlin-deficient SM. Proteins with significant variation in expression across all comparisons followed distinct temporal trends. DISCUSSION Proteins involved in sarcolemma repair and regeneration underwent significant changes in SM over the lifespan of WT mice, while those associated with immune infiltration and inflammation were overly represented over the lifespan of dysferlin-deficient mice. The proteins identified herein are likely to contribute to our overall understanding of SM aging and dysferlinopathy disease progression.
Collapse
Affiliation(s)
- Adriana E Golding
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Section on Intracellular Protein Trafficking, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Wenping Li
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois, USA
| | - Paul S Blank
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois, USA
| | - Joshua Zimmerberg
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
26
|
Benz E, Lahousse L, Arinze JT, Wijnant S, de Ridder M, Rivadeneira F, Brusselle G, Stricker BH. Oral corticosteroid use and sarcopenia-related traits in older people with chronic airway disease: a population-based study. ERJ Open Res 2023; 9:00492-2023. [PMID: 37753286 PMCID: PMC10518877 DOI: 10.1183/23120541.00492-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 09/28/2023] Open
Abstract
Background Sarcopenia is characterised by two major phenotypic components: low handgrip strength (HGS) and appendicular skeletal muscle index (ASMI). Oral corticosteroid (OCS) use is an important medication for acute respiratory exacerbations in patients with COPD and asthma. However, the association of OCS and sarcopenia components in older people is largely unexplored. The aim of this study was to examine the association between OCS use and HGS or ASMI in the general population and explore interactions with chronic airway diseases. Methods From the population-based Rotterdam Study, 5054 participants (age 69.0±8.8 years; 56% females) were included in the cross-sectional analysis and 1324 in the longitudinal analysis. Associations between OCS and muscle strength and mass were analysed using linear regression models adjusted for age, sex, fat %, height, kidney function, smoking and comorbidities. Results At baseline, ever-OCS users had lower handgrip strength (β= -0.48, 95% CI -0.84- -0.12) than never-OCS users, with cumulative frequency (≥10 OCS prescriptions)-dependent effects (β= -1.25, 95% CI -2.16- -0.33). COPD ever-OCS users, but not asthma, had lower handgrip strength (β= -0.98, 95% CI -1.91- -0.06) and lower lean mass (β= -0.14, 95% CI -0.27- -0.01) than never-OCS users. After 5.6 years of follow-up in those free of sarcopenia traits at baseline, COPD ever-OCS users developed lower handgrip strength (β= -1.64, 95% CI -2.87- -0.40) with frequency (β= -3.64, 95% CI -6.57- -0.72) and duration (β= -1.51, 95% CI -2.87- -0.15) association compared to never-OCS users. Conclusions OCS use is associated with a decline in handgrip strength in people with COPD in a cumulative frequency and duration-dependent manner. Routine muscle examination may be necessary for patients with COPD.
Collapse
Affiliation(s)
- Elizabeth Benz
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lies Lahousse
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Johnmary T. Arinze
- Department of Respiratory Medicine, Ghent University Hospital, Ghent University, Ghent, Belgium
- Department of Medical Informatics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sara Wijnant
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Maria de Ridder
- Department of Medical Informatics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Guy Brusselle
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Bruno H. Stricker
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
27
|
GrönholdtKlein M, Gorzi A, Wang L, Edström E, Rullman E, Altun M, Ulfhake B. Emergence and Progression of Behavioral Motor Deficits and Skeletal Muscle Atrophy across the Adult Lifespan of the Rat. BIOLOGY 2023; 12:1177. [PMID: 37759577 PMCID: PMC10526071 DOI: 10.3390/biology12091177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023]
Abstract
The facultative loss of muscle mass and function during aging (sarcopenia) poses a serious threat to our independence and health. When activities of daily living are impaired (clinical phase), it appears that the processes leading to sarcopenia have been ongoing in humans for decades (preclinical phase). Here, we examined the natural history of sarcopenia in male outbred rats to compare the occurrence of motor behavioral deficits with the degree of muscle wasting and to explore the muscle-associated processes of the preclinical and clinical phases, respectively. Selected metrics were validated in female rats. We used the soleus muscle because of its long duty cycles and its importance in postural control. Results show that gait and coordination remain intact through middle age (40-60% of median lifespan) when muscle mass is largely preserved relative to body weight. However, the muscle shows numerous signs of remodeling with a shift in myofiber-type composition toward type I. As fiber-type prevalence shifted, fiber-type clustering also increased. The number of hybrid fibers, myofibers with central nuclei, and fibers expressing embryonic myosin increased from being barely detectable to a significant number (5-10%) at late middle age. In parallel, TGFβ1, Smad3, FBXO32, and MuRF1 mRNAs increased. In early (25-month-old) and advanced (30-month-old) aging, gait and coordination deteriorate with the progressive loss of muscle mass. In late middle age and early aging due to type II atrophy (>50%) followed by type I atrophy (>50%), the number of myofibers did not correlate with this process. In advanced age, atrophy is accompanied by a decrease in SCs and βCatenin mRNA, whereas several previously upregulated transcripts were downregulated. The re-expression of embryonic myosin in myofibers and the upregulation of mRNAs encoding the γ-subunit of the nicotinic acetylcholine receptor, the neuronal cell adhesion molecule, and myogenin that begins in late middle age suggest that one mechanism driving sarcopenia is the disruption of neuromuscular connectivity. We conclude that sarcopenia in rats, as in humans, has a long preclinical phase in which muscle undergoes extensive remodeling to maintain muscle mass and function. At later time points, these adaptive mechanisms fail, and sarcopenia becomes clinically manifest.
Collapse
Affiliation(s)
- Max GrönholdtKlein
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Ali Gorzi
- Department of Sport Sciences, University of Zanjan, Zanjan 45371-38791, Iran;
| | - Lingzhan Wang
- Department of Human Anatomy, Histology and Embryology, Inner Mongolia Minzu University, Tongliao 028000, China;
| | - Erik Edström
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Eric Rullman
- Department of Laboratory Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (E.R.); (M.A.)
| | - Mikael Altun
- Department of Laboratory Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (E.R.); (M.A.)
| | - Brun Ulfhake
- Department of Laboratory Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (E.R.); (M.A.)
| |
Collapse
|
28
|
Dasgupta A, Gibbard DF, Schmitt RE, Arneson-Wissink PC, Ducharme AM, Bruinsma ES, Hawse JR, Jatoi A, Doles JD. A TGF-β/KLF10 signaling axis regulates atrophy-associated genes to induce muscle wasting in pancreatic cancer. Proc Natl Acad Sci U S A 2023; 120:e2215095120. [PMID: 37585460 PMCID: PMC10462925 DOI: 10.1073/pnas.2215095120] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/14/2023] [Indexed: 08/18/2023] Open
Abstract
Cancer cachexia, and its associated complications, represent a large and currently untreatable roadblock to effective cancer management. Many potential therapies have been proposed and tested-including appetite stimulants, targeted cytokine blockers, and nutritional supplementation-yet highly effective therapies are lacking. Innovative approaches to treating cancer cachexia are needed. Members of the Kruppel-like factor (KLF) family play wide-ranging and important roles in the development, maintenance, and metabolism of skeletal muscle. Within the KLF family, we identified KLF10 upregulation in a multitude of wasting contexts-including in pancreatic, lung, and colon cancer mouse models as well as in human patients. We subsequently interrogated loss-of-function of KLF10 as a potential strategy to mitigate cancer associated muscle wasting. In vivo studies leveraging orthotopic implantation of pancreas cancer cells into wild-type and KLF10 KO mice revealed significant preservation of lean mass and robust suppression of pro-atrophy muscle-specific ubiquitin ligases Trim63 and Fbxo32, as well as other factors implicated in atrophy, calcium signaling, and autophagy. Bioinformatics analyses identified Transforming growth factor beta (TGF-β), a known inducer of KLF10 and cachexia promoting factor, as a key upstream regulator of KLF10. We provide direct in vivo evidence that KLF10 KO mice are resistant to the atrophic effects of TGF-β. ChIP-based binding studies demonstrated direct binding to Trim63, a known wasting-associated atrogene. Taken together, we report a critical role for the TGF-β/KLF10 axis in the etiology of pancreatic cancer-associated muscle wasting and highlight the utility of targeting KLF10 as a strategy to prevent muscle wasting and limit cancer-associated cachexia.
Collapse
Affiliation(s)
- Aneesha Dasgupta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN55905
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN46202
- Indiana Center for Musculoskeletal Health, Indianapolis, IN46202
- Tumor Microenvironment & Metastasis Program, Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN46202
| | - Daniel F. Gibbard
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN55905
| | - Rebecca E. Schmitt
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN55905
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN46202
- Indiana Center for Musculoskeletal Health, Indianapolis, IN46202
- Tumor Microenvironment & Metastasis Program, Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN46202
| | - Paige C. Arneson-Wissink
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN55905
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN55905
| | | | | | - John R. Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN55905
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN55905
| | - Aminah Jatoi
- Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - Jason D. Doles
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN55905
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN46202
- Indiana Center for Musculoskeletal Health, Indianapolis, IN46202
- Tumor Microenvironment & Metastasis Program, Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN46202
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN55905
| |
Collapse
|
29
|
Dowling P, Swandulla D, Ohlendieck K. Biochemical and proteomic insights into sarcoplasmic reticulum Ca 2+-ATPase complexes in skeletal muscles. Expert Rev Proteomics 2023; 20:125-142. [PMID: 37668143 DOI: 10.1080/14789450.2023.2255743] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/07/2023] [Accepted: 08/14/2023] [Indexed: 09/06/2023]
Abstract
INTRODUCTION Skeletal muscles contain large numbers of high-molecular-mass protein complexes in elaborate membrane systems. Integral membrane proteins are involved in diverse cellular functions including the regulation of ion handling, membrane homeostasis, energy metabolism and force transmission. AREAS COVERED The proteomic profiling of membrane proteins and large protein assemblies in skeletal muscles are outlined in this article. This includes a critical overview of the main biochemical separation techniques and the mass spectrometric approaches taken to study membrane proteins. As an illustrative example of an analytically challenging large protein complex, the proteomic detection and characterization of the Ca2+-ATPase of the sarcoplasmic reticulum is discussed. The biological role of this large protein complex during normal muscle functioning, in the context of fiber type diversity and in relation to mechanisms of physiological adaptations and pathophysiological abnormalities is evaluated from a proteomics perspective. EXPERT OPINION Mass spectrometry-based muscle proteomics has decisively advanced the field of basic and applied myology. Although it is technically challenging to study membrane proteins, innovations in protein separation methodology in combination with sensitive mass spectrometry and improved systems bioinformatics has allowed the detailed proteomic detection and characterization of skeletal muscle membrane protein complexes, such as Ca2+-pump proteins of the sarcoplasmic reticulum.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth Kildare, Ireland
| | - Dieter Swandulla
- Institute of Physiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth Kildare, Ireland
| |
Collapse
|
30
|
Graca FA, Stephan A, Minden-Birkenmaier BA, Shirinifard A, Wang YD, Demontis F, Labelle M. Platelet-derived chemokines promote skeletal muscle regeneration by guiding neutrophil recruitment to injured muscles. Nat Commun 2023; 14:2900. [PMID: 37217480 DOI: 10.1038/s41467-023-38624-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 05/09/2023] [Indexed: 05/24/2023] Open
Abstract
Skeletal muscle regeneration involves coordinated interactions between different cell types. Injection of platelet-rich plasma is circumstantially considered an aid to muscle repair but whether platelets promote regeneration beyond their role in hemostasis remains unexplored. Here, we find that signaling via platelet-released chemokines is an early event necessary for muscle repair in mice. Platelet depletion reduces the levels of the platelet-secreted neutrophil chemoattractants CXCL5 and CXCL7/PPBP. Consequently, early-phase neutrophil infiltration to injured muscles is impaired whereas later inflammation is exacerbated. Consistent with this model, neutrophil infiltration to injured muscles is compromised in male mice with Cxcl7-knockout platelets. Moreover, neo-angiogenesis and the re-establishment of myofiber size and muscle strength occurs optimally in control mice post-injury but not in Cxcl7ko mice and in neutrophil-depleted mice. Altogether, these findings indicate that platelet-secreted CXCL7 promotes regeneration by recruiting neutrophils to injured muscles, and that this signaling axis could be utilized therapeutically to boost muscle regeneration.
Collapse
Affiliation(s)
- Flavia A Graca
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Anna Stephan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Benjamin A Minden-Birkenmaier
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Oncology, Division of Molecular Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Myriam Labelle
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
- Department of Oncology, Division of Molecular Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
31
|
Gilmore LA, Parry TL, Thomas GA, Khamoui AV. Skeletal muscle omics signatures in cancer cachexia: perspectives and opportunities. J Natl Cancer Inst Monogr 2023; 2023:30-42. [PMID: 37139970 PMCID: PMC10157770 DOI: 10.1093/jncimonographs/lgad006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/13/2023] [Accepted: 02/06/2023] [Indexed: 05/05/2023] Open
Abstract
Cachexia is a life-threatening complication of cancer that occurs in up to 80% of patients with advanced cancer. Cachexia reflects the systemic consequences of cancer and prominently features unintended weight loss and skeletal muscle wasting. Cachexia impairs cancer treatment tolerance, lowers quality of life, and contributes to cancer-related mortality. Effective treatments for cancer cachexia are lacking despite decades of research. High-throughput omics technologies are increasingly implemented in many fields including cancer cachexia to stimulate discovery of disease biology and inform therapy choice. In this paper, we present selected applications of omics technologies as tools to study skeletal muscle alterations in cancer cachexia. We discuss how comprehensive, omics-derived molecular profiles were used to discern muscle loss in cancer cachexia compared with other muscle-wasting conditions, to distinguish cancer cachexia from treatment-related muscle alterations, and to reveal severity-specific mechanisms during the progression of cancer cachexia from early toward severe disease.
Collapse
Affiliation(s)
- L Anne Gilmore
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Traci L Parry
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, NC, USA
| | - Gwendolyn A Thomas
- Department of Kinesiology, Pennsylvania State University, University Park, PA, USA
| | - Andy V Khamoui
- Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, USA
- Institute for Human Health and Disease Intervention, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
32
|
Liu Q, Yuan W, Yan Y, Jin B, You M, Liu T, Gao M, Li J, Gokulnath P, Vulugundam G, Li G, Xu B, Xiao J. Identification of a novel small-molecule inhibitor of miR-29b attenuates muscle atrophy. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:527-540. [PMID: 36891498 PMCID: PMC9988425 DOI: 10.1016/j.omtn.2023.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Muscle atrophy is debilitating and can be induced by several stressors. Unfortunately, there are no effective pharmacological treatment until now. MicroRNA (miR)-29b is an important target that we identified to be commonly involved in multiple types of muscle atrophy. Although sequence-specific inhibition of miR-29b has been developed, in this study, we report a novel small-molecule miR-29b inhibitor that targets miR-29b hairpin precursor (pre-miR-29b) (Targapremir-29b-066 [TGP-29b-066]) considering both its three-dimensional structure and the thermodynamics of interaction between pre-miR-29b and the small molecule. This novel small-molecule inhibitor has been demonstrated to attenuate muscle atrophy induced by angiotensin II (Ang II), dexamethasone (Dex), and tumor necrosis factor α (TNF-α) in C2C12 myotubes, as evidenced by increase in the diameter of myotube and decrease in the expression of Atrogin-1 and MuRF-1. Moreover, it can also attenuate Ang II-induced muscle atrophy in mice, as evidenced by a similar increase in the diameter of myotube, reduced Atrogin-1 and MuRF-1 expression, AKT-FOXO3A-mTOR signaling activation, and decreased apoptosis and autophagy. In summary, we experimentally identified and demonstrated a novel small-molecule inhibitor of miR-29b that could act as a potential therapeutic agent for muscle atrophy.
Collapse
Affiliation(s)
- Qi Liu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Weilin Yuan
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yuwei Yan
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Bing Jin
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Mengke You
- Department of Chemistry, Innovative Drug Research Center, Shanghai University, Shanghai 200444, China
| | - Tianqi Liu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
- Department of Chemistry, Innovative Drug Research Center, Shanghai University, Shanghai 200444, China
| | - Mingchun Gao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Jin Li
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Priyanka Gokulnath
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Bin Xu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai 200444, China
- Department of Chemistry, Innovative Drug Research Center, Shanghai University, Shanghai 200444, China
- Corresponding author Bin Xu, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai 200444, China
- Corresponding author Junjie Xiao, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.
| |
Collapse
|
33
|
Bakhtina AA, Pharaoh GA, Campbell MD, Keller A, Stuppard RS, Marcinek DJ, Bruce JE. Skeletal muscle mitochondrial interactome remodeling is linked to functional decline in aged female mice. NATURE AGING 2023; 3:313-326. [PMID: 37118428 PMCID: PMC10154043 DOI: 10.1038/s43587-023-00366-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 01/10/2023] [Indexed: 04/30/2023]
Abstract
Genomic, transcriptomic and proteomic approaches have been used to gain insight into molecular underpinnings of aging in laboratory animals and in humans. However, protein function in biological systems is under complex regulation and includes factors besides abundance levels, such as modifications, localization, conformation and protein-protein interactions. By making use of quantitative chemical cross-linking technologies, we show that changes in the muscle mitochondrial interactome contribute to mitochondrial functional decline in aging in female mice. Specifically, we identify age-related changes in protein cross-links relating to assembly of electron transport system complexes I and IV, activity of glutamate dehydrogenase, and coenzyme-A binding in fatty acid β-oxidation and tricarboxylic acid cycle enzymes. These changes show a remarkable correlation with complex I respiration differences within the same young-old animal pairs. Each observed cross-link can serve as a protein conformational or protein-protein interaction probe in future studies, which will provide further molecular insights into commonly observed age-related phenotypic differences. Therefore, this data set could become a valuable resource for additional in-depth molecular studies that are needed to better understand complex age-related molecular changes.
Collapse
Affiliation(s)
- Anna A Bakhtina
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Gavin A Pharaoh
- Department of Radiology, University of Washington, Seattle, WA, USA
| | | | - Andrew Keller
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, WA, USA.
| | - James E Bruce
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
| |
Collapse
|
34
|
Wang BYH, Hsiao AWT, Wong N, Chen YF, Lee CW, Lee WYW. Is dexamethasone-induced muscle atrophy an alternative model for naturally aged sarcopenia model? J Orthop Translat 2023; 39:12-20. [PMID: 36605620 PMCID: PMC9793312 DOI: 10.1016/j.jot.2022.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Background Primary sarcopenia is usually known as age-related skeletal muscle loss; however, other factors like endocrine, lifestyle and inflammation can also cause muscle loss, known as secondary sarcopenia. Although many studies have used different sarcopenia animal models for exploring the underlying mechanism and therapeutic approaches for sarcopenia, limited study has provided evidence of the relevance of these animal models. This study aims to investigate the similarity and difference in muscle qualities between primary and secondary sarcopenia mice models, using naturally aged mice and dexamethasone-induced mice. Methods 21-month-old mice were used as naturally aged primary sarcopenia mice and 3-month-old mice received daily intraperitoneal injection of dexamethasone (20 mg/ kg body weight) for 10 days were used as secondary sarcopenia model. This study provided measurements for muscle mass and functions, including Dual-energy X-ray absorptiometry (DXA) scanning, handgrip strength test and treadmill running to exhaustion test. Besides, muscle contraction, muscle fibre type measurements and gene expression were also performed to provide additional information on muscle qualities. Results The results suggest two sarcopenia animal models shared a comparable decrease in forelimb lean mass, muscle fibre size, grip strength and muscle contraction ability. Besides, the upregulation of protein degradation genes was also observed in two sarcopenia animal models. However, only primary sarcopenia mice were identified with an early stage of mtDNA deletion. Conclusion Collectively, this study evaluated that the dexamethasone-induced mouse model could be served as an alternative model for primary sarcopenia, according to the comparable muscle mass and functional changes. However, whether dexamethasone-induced mice can be used as an animal model when studying the molecular mechanisms of sarcopenia needs to be carefully evaluated. The translational potential of this article The purpose of sarcopenia research is to investigate appropriate treatments for reversing the loss of skeletal muscle mass and functions. Using animal models for the preclinical study could predict the safety and efficacy of the treatments. This study compared the typical age-related sarcopenia mice model and dexamethasone-induced secondary sarcopenia mice to provide evidence of the pathological and functional changes in the mice models.
Collapse
Affiliation(s)
- Belle Yu-Hsuan Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong
| | - Allen Wei-Ting Hsiao
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Nicodemus Wong
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong
| | - Yi-Fan Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, 11031 Taipei, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, 11031 Taipei, Taiwan
| | - Chien-Wei Lee
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| | - Wayne Yuk Wai Lee
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
35
|
Progressive development of melanoma-induced cachexia differentially impacts organ systems in mice. Cell Rep 2023; 42:111934. [PMID: 36640353 PMCID: PMC9983329 DOI: 10.1016/j.celrep.2022.111934] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/12/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022] Open
Abstract
Cachexia is a systemic wasting syndrome that increases cancer-associated mortality. How cachexia progressively and differentially impacts distinct tissues is largely unknown. Here, we find that the heart and skeletal muscle undergo wasting at early stages and are the tissues transcriptionally most impacted by cachexia. We also identify general and organ-specific transcriptional changes that indicate functional derangement by cachexia even in tissues that do not undergo wasting, such as the brain. Secreted factors constitute a top category of cancer-regulated genes in host tissues, and these changes include upregulation of the angiotensin-converting enzyme (ACE). ACE inhibition with the drug lisinopril improves muscle force and partially impedes cachexia-induced transcriptional changes, although wasting is not prevented, suggesting that cancer-induced host-secreted factors can regulate tissue function during cachexia. Altogether, by defining prevalent and temporal and tissue-specific responses to cachexia, this resource highlights biomarkers and possible targets for general and tissue-tailored anti-cachexia therapies.
Collapse
|
36
|
Jiao J, Curley M, Graca FA, Robles-Murguia M, Shirinifard A, Finkelstein D, Xu B, Fan Y, Demontis F. Modulation of protease expression by the transcription factor Ptx1/PITX regulates protein quality control during aging. Cell Rep 2023; 42:111970. [PMID: 36640359 PMCID: PMC9933915 DOI: 10.1016/j.celrep.2022.111970] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 10/31/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Protein quality control is important for healthy aging and is dysregulated in age-related diseases. The autophagy-lysosome and ubiquitin-proteasome are key for proteostasis, but it remains largely unknown whether other proteolytic systems also contribute to maintain proteostasis during aging. Here, we find that expression of proteolytic enzymes (proteases/peptidases) distinct from the autophagy-lysosome and ubiquitin-proteasome systems declines during skeletal muscle aging in Drosophila. Age-dependent protease downregulation undermines proteostasis, as demonstrated by the increase in detergent-insoluble poly-ubiquitinated proteins and pathogenic huntingtin-polyQ levels in response to protease knockdown. Computational analyses identify the transcription factor Ptx1 (homologous to human PITX1/2/3) as a regulator of protease expression. Consistent with this model, Ptx1 protein levels increase with aging, and Ptx1 RNAi counteracts the age-associated downregulation of protease expression. Moreover, Ptx1 RNAi improves muscle protein quality control in a protease-dependent manner and extends lifespan. These findings indicate that proteases and their transcriptional modulator Ptx1 ensure proteostasis during aging.
Collapse
Affiliation(s)
- Jianqin Jiao
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Michelle Curley
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Flavia A. Graca
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Maricela Robles-Murguia
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Beisi Xu
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA,Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Yiping Fan
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
37
|
Dowling P, Gargan S, Swandulla D, Ohlendieck K. Fiber-Type Shifting in Sarcopenia of Old Age: Proteomic Profiling of the Contractile Apparatus of Skeletal Muscles. Int J Mol Sci 2023; 24:2415. [PMID: 36768735 PMCID: PMC9916839 DOI: 10.3390/ijms24032415] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
The progressive loss of skeletal muscle mass and concomitant reduction in contractile strength plays a central role in frailty syndrome. Age-related neuronal impairments are closely associated with sarcopenia in the elderly, which is characterized by severe muscular atrophy that can considerably lessen the overall quality of life at old age. Mass-spectrometry-based proteomic surveys of senescent human skeletal muscles, as well as animal models of sarcopenia, have decisively improved our understanding of the molecular and cellular consequences of muscular atrophy and associated fiber-type shifting during aging. This review outlines the mass spectrometric identification of proteome-wide changes in atrophying skeletal muscles, with a focus on contractile proteins as potential markers of changes in fiber-type distribution patterns. The observed trend of fast-to-slow transitions in individual human skeletal muscles during the aging process is most likely linked to a preferential susceptibility of fast-twitching muscle fibers to muscular atrophy. Studies with senescent animal models, including mostly aged rodent skeletal muscles, have confirmed fiber-type shifting. The proteomic analysis of fast versus slow isoforms of key contractile proteins, such as myosin heavy chains, myosin light chains, actins, troponins and tropomyosins, suggests them as suitable bioanalytical tools of fiber-type transitions during aging.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Dieter Swandulla
- Institute of Physiology, University of Bonn, D53115 Bonn, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| |
Collapse
|
38
|
Zhang M, Chen M, Li Y, Rao M, Wang D, Wang Z, Zhang L, Yin P, Tang P. Delayed denervation-induced muscle atrophy in Opg knockout mice. Front Physiol 2023; 14:1127474. [PMID: 36909232 PMCID: PMC9992212 DOI: 10.3389/fphys.2023.1127474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Recent evidence has shown a crucial role for the osteoprotegerin/receptor activator of nuclear factor κ-B ligand/RANK (OPG/RANKL/RANK) signaling axis not only in bone but also in muscle tissue; however, there is still a lack of understanding of its effects on muscle atrophy. Here, we found that denervated Opg knockout mice displayed better functional recovery and delayed muscle atrophy, especially in a specific type IIB fiber. Moreover, OPG deficiency promoted milder activation of the ubiquitin-proteasome pathway, which further verified the protective role of Opg knockout in denervated muscle damage. Furthermore, transcriptome sequencing indicated that Opg knockout upregulated the expression of Inpp5k, Rbm3, and Tet2 and downregulated that of Deptor in denervated muscle. In vitro experiments revealed that satellite cells derived from Opg knockout mice displayed a better differentiation ability than those acquired from wild-type littermates. Higher expression levels of Tet2 were also observed in satellite cells derived from Opg knockout mice, which provided a possible mechanistic basis for the protective effects of Opg knockout on muscle atrophy. Taken together, our findings uncover the novel role of Opg in muscle atrophy process and extend the current understanding in the OPG/RANKL/RANK signaling axis.
Collapse
Affiliation(s)
- Mingming Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Ming Chen
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Yi Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Man Rao
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Duanyang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhongqi Wang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| |
Collapse
|
39
|
Keeble AR, Brightwell CR, Latham CM, Thomas NT, Mobley CB, Murach KA, Johnson DL, Noehren B, Fry CS. Depressed Protein Synthesis and Anabolic Signaling Potentiate ACL Tear-Resultant Quadriceps Atrophy. Am J Sports Med 2023; 51:81-96. [PMID: 36475881 PMCID: PMC9813974 DOI: 10.1177/03635465221135769] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Anterior cruciate ligament (ACL) tear (ACLT) leads to protracted quadriceps muscle atrophy. Protein turnover largely dictates muscle size and is highly responsive to injury and loading. Regulation of quadriceps molecular protein synthetic machinery after ACLT has largely been unexplored, limiting development of targeted therapies. PURPOSE To define the effect of ACLT on (1) the activation of protein synthetic and catabolic signaling within quadriceps biopsy specimens from human participants and (2) the time course of alterations to protein synthesis and its molecular regulation in a mouse ACL injury model. STUDY DESIGN Descriptive laboratory study. METHODS Muscle biopsy specimens were obtained from the ACL-injured and noninjured vastus lateralis of young adult humans after an overnight fast (N = 21; mean ± SD, 19 ± 5 years). Mice had their limbs assigned to ACLT or control, and whole quadriceps were collected 6 hours or 1, 3, or 7 days after injury with puromycin injected before tissue collection for assessment of relative protein synthesis. Muscle fiber size and expression and phosphorylation of protein anabolic and catabolic signaling proteins were assessed at the protein and transcript levels (RNA sequencing). RESULTS Human quadriceps showed reduced phosphorylation of ribosomal protein S6 (-41%) in the ACL-injured limb (P = .008), in addition to elevated phosphorylation of eukaryotic initiation factor 2α (+98%; P = .006), indicative of depressed protein anabolic signaling in the injured limb. No differences in E3 ubiquitin ligase expression were noted. Protein synthesis was lower at 1 day (P = .01 vs control limb) and 3 days (P = .002 vs control limb) after ACLT in mice. Pathway analyses revealed shared molecular alterations between human and mouse quadriceps after ACLT. CONCLUSION (1) Global protein synthesis and anabolic signaling deficits occur in the quadriceps in response to ACL injury, without notable changes in measured markers of muscle protein catabolism. (2) Importantly, these deficits occur before the onset of significant atrophy, underscoring the need for early intervention. CLINICAL RELEVANCE These findings suggest that blunted protein anabolism as opposed to increased catabolism likely mediates quadriceps atrophy after ACL injury. Thus, future interventions should aim to restore muscle protein anabolism rapidly after ACLT.
Collapse
Affiliation(s)
- Alexander R. Keeble
- Department of Physiology, College of Medicine, University of Kentucky
- Center for Muscle Biology, University of Kentucky
| | - Camille R. Brightwell
- Center for Muscle Biology, University of Kentucky
- Department of Athletic Training and Clinical Nutrition, University of Kentucky
| | - Christine M. Latham
- Center for Muscle Biology, University of Kentucky
- Department of Athletic Training and Clinical Nutrition, University of Kentucky
| | - Nicholas T. Thomas
- Center for Muscle Biology, University of Kentucky
- Department of Athletic Training and Clinical Nutrition, University of Kentucky
| | - C. Brooks Mobley
- Department of Physiology, College of Medicine, University of Kentucky
- Center for Muscle Biology, University of Kentucky
| | - Kevin A. Murach
- Center for Muscle Biology, University of Kentucky
- Department of Physical Therapy, University of Kentucky
| | - Darren L. Johnson
- Department of Orthopaedic Surgery & Sports Medicine, University of Kentucky
| | - Brian Noehren
- Center for Muscle Biology, University of Kentucky
- Department of Physical Therapy, University of Kentucky
- Department of Orthopaedic Surgery & Sports Medicine, University of Kentucky
| | - Christopher S. Fry
- Center for Muscle Biology, University of Kentucky
- Department of Athletic Training and Clinical Nutrition, University of Kentucky
| |
Collapse
|
40
|
Rai M, Demontis F. Muscle-to-Brain Signaling Via Myokines and Myometabolites. Brain Plast 2022; 8:43-63. [PMID: 36448045 PMCID: PMC9661353 DOI: 10.3233/bpl-210133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle health and function are important determinants of systemic metabolic homeostasis and organism-wide responses, including disease outcome. While it is well known that exercise protects the central nervous system (CNS) from aging and disease, only recently this has been found to depend on the endocrine capacity of skeletal muscle. Here, we review muscle-secreted growth factors and cytokines (myokines), metabolites (myometabolites), and other unconventional signals (e.g. bioactive lipid species, enzymes, and exosomes) that mediate muscle-brain and muscle-retina communication and neuroprotection in response to exercise and associated processes, such as the muscle unfolded protein response and metabolic stress. In addition to impacting proteostasis, neurogenesis, and cognitive functions, muscle-brain signaling influences complex brain-dependent behaviors, such as depression, sleeping patterns, and biosynthesis of neurotransmitters. Moreover, myokine signaling adapts feeding behavior to meet the energy demands of skeletal muscle. Contrary to protective myokines induced by exercise and associated signaling pathways, inactivity and muscle wasting may derange myokine expression and secretion and in turn compromise CNS function. We propose that tailoring muscle-to-CNS signaling by modulating myokines and myometabolites may combat age-related neurodegeneration and brain diseases that are influenced by systemic signals.
Collapse
Affiliation(s)
- Mamta Rai
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
41
|
Wei W, Zha C, Jiang A, Chao Z, Hou L, Liu H, Huang R, Wu W. A Combined Differential Proteome and Transcriptome Profiling of Fast- and Slow-Twitch Skeletal Muscle in Pigs. Foods 2022; 11:foods11182842. [PMID: 36140968 PMCID: PMC9497725 DOI: 10.3390/foods11182842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Skeletal muscle fiber types can contribute in part to affecting pork quality parameters. Biceps femoris (Bf) (fast muscle or white muscle) and Soleus (Sol) (slow muscle or red muscle) are two typical skeletal muscles characterized by obvious muscle fiber type differences in pigs. However, the critical proteins and potential regulatory mechanisms regulating porcine skeletal muscle fibers have yet to be clearly defined. In this study, the isobaric Tag for Relative and Absolute Quantification (iTRAQ)-based proteome was used to identify the key proteins affecting the skeletal muscle fiber types with Bf and Sol, by integrating the previous transcriptome data, while function enrichment analysis and a protein–protein interaction (PPI) network were utilized to explore the potential regulatory mechanisms of skeletal muscle fibers. A total of 126 differentially abundant proteins (DAPs) between the Bf and Sol were identified, and 12 genes were found to be overlapping between differentially expressed genes (DEGs) and DAPs, which are the critical proteins regulating the formation of skeletal muscle fibers. Functional enrichment and PPI analysis showed that the DAPs were mainly involved in the skeletal-muscle-associated structural proteins, mitochondria and energy metabolism, tricarboxylic acid cycle, fatty acid metabolism, and kinase activity, suggesting that PPI networks including DAPs are the main regulatory network affecting muscle fiber formation. Overall, these data provide valuable information for understanding the molecular mechanism underlying the formation and conversion of muscle fiber types, and provide potential markers for the evaluation of meat quality.
Collapse
Affiliation(s)
- Wei Wei
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chengwan Zha
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Aiwen Jiang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhe Chao
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou 571100, China
| | - Liming Hou
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Honglin Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruihua Huang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wangjun Wu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- Correspondence: ; Tel.: +86-25-84399762
| |
Collapse
|
42
|
E L D, T J W, A R F, A B P, K M R, C P D, C R G, J E W, K V S, R J U, M SM. Cancer and Associated Therapies Impact the Skeletal Muscle Proteome. Front Physiol 2022; 13:879263. [PMID: 35694399 PMCID: PMC9184684 DOI: 10.3389/fphys.2022.879263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Both cancer and cancer associated therapies (CAT; including chemotherapy or concurrent chemoradiation) disrupt cellular metabolism throughout the body, including the regulation of skeletal muscle mass and function. Adjunct testosterone therapy during standard of care chemotherapy and chemoradiation modulates CAT-induced dysregulation of skeletal muscle metabolism and protects lean body mass during CAT. However, the extent to which the skeletal muscle proteome is altered under these therapeutic conditions is unknown. Objective: We probed the skeletal muscle proteome of cancer patients as an ancillary analysis following a randomized, double-blind, placebo-controlled phase II trial investigating the effect of adjunct testosterone on body composition in men and women with advanced cancers undergoing CAT. Methods: Men and women diagnosed with late stage (≥IIB) or recurrent head and neck or cervical cancer who were scheduled to receive standard of care CAT were administered an adjunct 7 weeks treatment of weekly intramuscular injections of either 100 mg testosterone (CAT+T, n = 7; 2M/5F) or placebo/saline (CAT+P, n = 6; 4M/2F). Biopsies were performed on the vastus lateralis before (PRE) and after (POST) the 7 weeks treatment. Extracted proteins were separated with 2-dimensional gel electrophoresis (2DE), and subjected to analyses of total protein abundance, phosphorylation and S-nitrosylation. Proteoforms showing significant 1.5 fold differences (t-test p ≤ 0.05) between PRE and POST timepoints were identified by mass spectroscopy (MS), and lists of altered proteins were subjected to Gene Set Enrichment Analysis (GSEA) to identify affected pathways. Results: A total of 756 distinct protein spots were identified. Of those spots, 102 were found to be altered in terms of abundance, phosphorylation, or S-nitrosylation, and identified by mass spectroscopy analysis to represent 58 unique proteins. Among the biological processes and pathways identified, CAT+P predominantly impacted metabolic processes, cell assembly, oxygen transport, and apoptotic signaling, while CAT+T impacted transcription regulation, muscle differentiation, muscle development, and contraction. Conclusion: Cancer and CAT significantly altered the skeletal muscle proteome in a manner suggestive of loss of structural integrity, reduced contractile function, and disrupted metabolism. Proteomic analysis suggests that the addition of adjunct testosterone minimized the structural and contractile influence of cancer and its associated therapies.
Collapse
Affiliation(s)
- Dillon E L
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Wright T J
- Department of Health and Kinesiology, Texas A&M University, College Station, TX, United States
| | - Filley A R
- Department of Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Pulliam A B
- Department of Health and Kinesiology, Texas A&M University, College Station, TX, United States
| | - Randolph K M
- Department of Health and Kinesiology, Texas A&M University, College Station, TX, United States
| | - Danesi C P
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Gilkison C R
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Wiktorowicz J E
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Soman K V
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Urban R J
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Sheffield-Moore M
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
43
|
Stephan A, Graca FA, Hunt LC, Demontis F. Electroporation of Small Interfering RNAs into Tibialis Anterior Muscles of Mice. Bio Protoc 2022; 12:e4428. [PMID: 35799907 PMCID: PMC9244496 DOI: 10.21769/bioprotoc.4428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/08/2022] [Accepted: 04/06/2022] [Indexed: 12/29/2022] Open
Abstract
Aging and wasting of skeletal muscle reduce organismal fitness. Regrettably, only limited interventions are currently available to address this unmet medical need. Many methods have been developed to study this condition, including the intramuscular electroporation of DNA plasmids. However, this technique requires surgery and high electrical fields, which cause tissue damage. Here, we report an optimized protocol for the electroporation of small interfering RNAs (siRNAs) into the tibialis anterior muscle of mice. This protocol does not require surgery and, because of the small siRNA size, mild electroporation conditions are utilized. By inducing target mRNA knockdown, this method can be used to interrogate gene function in muscles of mice from different strains, genotypes, and ages. Moreover, a complementary method for siRNA transfection into differentiated myotubes can be used for testing siRNA efficacy before in vivo use. Altogether, this streamlined protocol is instrumental for basic science and translational studies in muscles of mice and other animal models.
Collapse
Affiliation(s)
- Anna Stephan
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Flavia A. Graca
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Liam C. Hunt
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
,
*For correspondence:
| |
Collapse
|
44
|
Hughes DC, Hardee JP, Waddell DS, Goodman CA. CORP: Gene delivery into murine skeletal muscle using in vivo electroporation. J Appl Physiol (1985) 2022; 133:41-59. [PMID: 35511722 DOI: 10.1152/japplphysiol.00088.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The strategy of gene delivery into skeletal muscles has provided exciting avenues in identifying new potential therapeutics towards muscular disorders and addressing basic research questions in muscle physiology through overexpression and knockdown studies. In vivo electroporation methodology offers a simple, rapidly effective technique for the delivery of plasmid DNA into post-mitotic skeletal muscle fibers and the ability to easily explore the molecular mechanisms of skeletal muscle plasticity. The purpose of this review is to describe how to robustly electroporate plasmid DNA into different hindlimb muscles of rodent models. Further, key parameters (e.g., voltage, hyaluronidase, plasmid concentration) which contribute to the successful introduction of plasmid DNA into skeletal muscle fibers will be discussed. In addition, details on processing tissue for immunohistochemistry and fiber cross-sectional area (CSA) analysis will be outlined. The overall goal of this review is to provide the basic and necessary information needed for successful implementation of in vivo electroporation of plasmid DNA and thus open new avenues of discovery research in skeletal muscle physiology.
Collapse
Affiliation(s)
- David C Hughes
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Justin P Hardee
- Centre for Muscle Research (CMR), Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - David S Waddell
- Department of Biology, University of North Florida, Jacksonville, FL, United States
| | - Craig A Goodman
- Centre for Muscle Research (CMR), Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
45
|
Detection of Target Genes for Drug Repurposing to Treat Skeletal Muscle Atrophy in Mice Flown in Spaceflight. Genes (Basel) 2022; 13:genes13030473. [PMID: 35328027 PMCID: PMC8953707 DOI: 10.3390/genes13030473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/25/2022] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle atrophy is a common condition in aging, diabetes, and in long duration spaceflights due to microgravity. This article investigates multi-modal gene disease and disease drug networks via link prediction algorithms to select drugs for repurposing to treat skeletal muscle atrophy. Key target genes that cause muscle atrophy in the left and right extensor digitorum longus muscle tissue, gastrocnemius, quadriceps, and the left and right soleus muscles are detected using graph theoretic network analysis, by mining the transcriptomic datasets collected from mice flown in spaceflight made available by GeneLab. We identified the top muscle atrophy gene regulators by the Pearson correlation and Bayesian Markov blanket method. The gene disease knowledge graph was constructed using the scalable precision medicine knowledge engine. We computed node embeddings, random walk measures from the networks. Graph convolutional networks, graph neural networks, random forest, and gradient boosting methods were trained using the embeddings, network features for predicting links and ranking top gene-disease associations for skeletal muscle atrophy. Drugs were selected and a disease drug knowledge graph was constructed. Link prediction methods were applied to the disease drug networks to identify top ranked drugs for therapeutic treatment of skeletal muscle atrophy. The graph convolution network performs best in link prediction based on receiver operating characteristic curves and prediction accuracies. The key genes involved in skeletal muscle atrophy are associated with metabolic and neurodegenerative diseases. The drugs selected for repurposing using the graph convolution network method were nutrients, corticosteroids, anti-inflammatory medications, and others related to insulin.
Collapse
|