1
|
Shirai T, Uemichi K, Iwai R, Shinkai H, Iwata T, Tanimura R, Sugiyama S, Takemasa T. Systemic effect of combined functional overload and endurance-type swimming exercise on whole body metabolism in mice. Am J Physiol Endocrinol Metab 2025; 328:E695-E710. [PMID: 40248969 DOI: 10.1152/ajpendo.00433.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/06/2025] [Accepted: 03/26/2025] [Indexed: 04/19/2025]
Abstract
In this study, we examined the effects of concurrent functional overload and endurance exercise on muscle hypertrophy, mitochondrial function, and systemic adaptations in male mice. The mice were assigned to three groups: Sham (Sham), overload-induced hypertrophy (OL), and overload with concurrent 60-min free swimming (5 times/wk) (OL + Swim), for 4 wk. Although OL promoted muscle hypertrophy and protein synthesis through the Akt/mammalian/mechanistic target of rapamycin (mTOR) signaling pathway, the addition of swimming (OL + Swim) attenuated these effects, resulting in less pronounced muscle growth and a smaller increase in myofiber cross-sectional area. Notably, the OL + Swim group exhibited enhanced mitochondrial activity and glycogen content compared with the OL group. Both the OL and OL + Swim groups showed elevated rates of protein synthesis, with a significant upregulation of AMP-activated kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) in the OL + Swim group, suggesting enhanced mitochondrial biogenesis and adaptation. Concurrent training also resulted in systemic benefits, including reduced inguinal and epididymal white adipocyte size, improved mitochondrial enzyme activities in adipose and liver tissues, and higher levels of fibronectin type III domain containing protein 5 (FNDC5), fibroblast growth factor 21 (FGF21), and brain-derived neurotrophic factor (BDNF) in serum, which contributed to enhanced muscle protein synthesis in cultured muscle cells. These results highlight the trade-offs between muscle hypertrophy and metabolic health in mice and underscore the importance of balanced training regimens to optimize overall metabolic health and muscle function. Our results provide further insight into how concurrent strength and endurance training can be optimized for health and performance benefits.NEW & NOTEWORTHY This study provides novel insights into the mechanisms underlying the interference effect that occurs in concurrent training, highlighting the potential systemic benefits of combining resistance and endurance exercises. Despite a reduction in muscle hypertrophy, concurrent training enhances metabolic adaptations and systemic health markers and offers a comprehensive approach to improving both muscle and metabolic fitness.
Collapse
Affiliation(s)
- Takanaga Shirai
- Department of Human Sciences, Kanagawa University, Kanagawa, Japan
- Research Fellow of Japan Society for Promotion Science, Tokyo, Japan
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| | - Kazuki Uemichi
- Research Fellow of Japan Society for Promotion Science, Tokyo, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Ryoto Iwai
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Hayato Shinkai
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Tomohiro Iwata
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Riku Tanimura
- Research Fellow of Japan Society for Promotion Science, Tokyo, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Shunsuke Sugiyama
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Tohru Takemasa
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
2
|
Zakyrjanova GF, Matigorova VA, Kuznetsova EA, Dmitrieva SA, Tyapkina OV, Tsentsevitsky AN, Andreyanova SN, Odnoshivkina JG, Shigapova RR, Mukhamedshina YO, Gogolev YV, Petrov AM. Key genes and processes affected by atorvastatin treatment in mouse diaphragm muscle. Arch Toxicol 2025:10.1007/s00204-025-04056-6. [PMID: 40234311 DOI: 10.1007/s00204-025-04056-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Statins are one of the top prescribed medications and are used for preventing or treating cardiovascular diseases. Myalgia, muscle fatigue, weakness, and inflammation are the most common side effects of these drugs collectively named statin-associated muscle symptoms (SAMS). The mechanisms underlying SAMS remain unclear. Given that statins inhibit 3-hydroxy-3-methylglutaryl coenzyme A reductase, the rate-limiting enzyme of mevalonate pathway, responsible for synthesis of cholesterol and other vital molecules, SAMS may be mediated by multiple reasons. Herein, using unbiased whole transcriptome sequencing, we identified statin-affected processes and then assessed them using fluorescent, biochemical, and histological approaches in the mouse diaphragm, the main respiratory muscle. Mice were orally treated for 1 month with atorvastatin, the most prescribed statin, at clinically relevant dose. We found that atorvastatin caused downregulation of genes encoding proteins required for oxidative phosphorylation and anabolic processes, whereas genes of proteins engaged inflammation and muscle atrophy were mainly up-regulated. Furthermore, alterations in gene expression pattern suggest oxidative stress and abnormal lipid accumulation. This transcriptome signature correlated to a decrease in mitochondrial polarization and protein synthesis capacity, as well as an increase in lipid peroxidation and reactive oxygen species production. In addition, atorvastatin treatment caused lipid raft disruption, phospholipidosis, myelin de-compactization, and appearance of greater heterogeneity of muscle fiber cross-section diameter. Thus, atorvastatin treatment can negatively affect diaphragm muscle via oxidative stress accompanied by decrease in mitochondrial activity, protein synthesis, and stability of plasma membrane. As a part of compensatory response can serve enhanced activity of superoxide dismutase and cholesterol uptake capacity.
Collapse
Affiliation(s)
- Guzel F Zakyrjanova
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, 119234, Russia
| | - Valeriya A Matigorova
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
| | - Eva A Kuznetsova
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
| | - Svetlana A Dmitrieva
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
| | - Oksana V Tyapkina
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
| | - Andrei N Tsentsevitsky
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
| | - Sofya N Andreyanova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008, Kazan, Russia
| | - Julia G Odnoshivkina
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
- Department of Normal Physiology, Institute of Neuroscience, Kazan State Medical University, 49 Butlerova Street, Kazan, 420012, Russia
| | - Rezeda R Shigapova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008, Kazan, Russia
| | - Yana O Mukhamedshina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008, Kazan, Russia
| | - Yuri V Gogolev
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008, Kazan, Russia
| | - Alexey M Petrov
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia.
- Department of Normal Physiology, Institute of Neuroscience, Kazan State Medical University, 49 Butlerova Street, Kazan, 420012, Russia.
| |
Collapse
|
3
|
Wang Y, Xie H, Liu Q, Wang N, Luo X, Sun F, Zhu J, Dong R, Wang Y, Gao J, Gao Z, Huang T, Liu X, Yu Q, Wang T, Li Y, Song D, Liu S, Zhang S, Yin H, Kong W, Wang CY. Kdm2a inhibition in skeletal muscle improves metabolic flexibility in obesity. Nat Metab 2025; 7:383-400. [PMID: 39870955 PMCID: PMC11860252 DOI: 10.1038/s42255-024-01210-9] [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: 07/17/2024] [Accepted: 12/17/2024] [Indexed: 01/29/2025]
Abstract
Skeletal muscle is a critical organ in maintaining homoeostasis against metabolic stress, and histone post-translational modifications are pivotal in those processes. However, the intricate nature of histone methylation in skeletal muscle and its impact on metabolic homoeostasis have yet to be elucidated. Here, we report that mitochondria-rich slow-twitch myofibers are characterized by significantly higher levels of H3K36me2 along with repressed expression of Kdm2a, an enzyme that specifically catalyses H3K36me2 demethylation. Deletion or inhibition of Kdm2a shifts fuel use from glucose under cold challenge to lipids under obese conditions by increasing the proportion of mitochondria-rich slow-twitch myofibers. This protects mice against cold insults and high-fat-diet-induced obesity and insulin resistance. Mechanistically, Kdm2a deficiency leads to a marked increase in H3K36me2 levels, which then promotes the recruitment of Mrg15 to the Esrrg locus to process its precursor messenger RNA splicing, thereby reshaping skeletal muscle metabolic profiles to induce slow-twitch myofiber transition. Collectively, our data support the role of Kdm2a as a viable target against metabolic stress.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Xie
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qianrui Liu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Luo
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Sun
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinghan Zhu
- Department of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruihan Dong
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Gao
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhichao Gao
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Teng Huang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Liu
- Department of Interventional Radiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qilin Yu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Wang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Li
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danni Song
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiwei Liu
- Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Third Hospital of Shanxi Medical University, the Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Taiyuan, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Hao Yin
- Organ Transplant Center, Shanghai Changzheng Hospital (Second Affiliated Hospital of Naval Medical University), Shanghai, China.
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Cong-Yi Wang
- Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Third Hospital of Shanxi Medical University, the Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Taiyuan, China.
- The Center for Biomedical Research, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Kim JT, Jeon DH, Lee HJ. Molecular mechanism of skeletal muscle loss and its prevention by natural resources. Food Sci Biotechnol 2024; 33:3387-3400. [PMID: 39493391 PMCID: PMC11525361 DOI: 10.1007/s10068-024-01678-x] [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: 05/15/2024] [Revised: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 11/05/2024] Open
Abstract
A skeletal muscle disorder has drawn attention due to the global aging issues. The loss of skeletal muscle mass has been suggested to be from the reduced muscle regeneration by dysfunction of muscle satellite cell/fibro-adipogenic progenitor cells and the muscle atrophy by dysfunction of mitochondria, ubiquitin-proteasome system, and autophagy. In this review, we highlighted the underlying mechanisms of skeletal muscle mass loss including Notch signaling, Wnt/β-catenin signaling, Hedgehog signaling, AMP-activated protein kinase (AMPK) signaling, and mammalian target of rapamycin (mTOR) signaling. In addition, we summarized accumulated studies of natural resources investigating their roles in ameliorating the loss of skeletal muscle mass and demonstrating the underlying mechanisms in vitro and in vivo. In conclusion, following the studies of natural resources exerting the preventive activity in muscle mass loss, the signaling-based approaches may accelerate the development of functional foods for sarcopenia prevention.
Collapse
Affiliation(s)
- Jin Tae Kim
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 17546 South Korea
- GreenTech-Based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, 17546 South Korea
| | - Dong Hyeon Jeon
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 17546 South Korea
- GreenTech-Based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, 17546 South Korea
| | - Hong Jin Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 17546 South Korea
- GreenTech-Based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, 17546 South Korea
| |
Collapse
|
5
|
Kusano T, Sotani Y, Takeda R, Hatano A, Kawata K, Kano R, Matsumoto M, Kano Y, Hoshino D. Time-series transcriptomics reveals distinctive mRNA expression dynamics associated with gene ontology specificity and protein expression in skeletal muscle after electrical stimulation-induced resistance exercise. FASEB J 2024; 38:e70153. [PMID: 39545720 PMCID: PMC11698011 DOI: 10.1096/fj.202401420rr] [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: 07/22/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
Resistance exercise upregulates and downregulates the expression of a wide range of genes in skeletal muscle. However, detailed analysis of mRNA dynamics such as response rates and temporal patterns of the transcriptome after resistance exercise has not been performed. We aimed to clarify the dynamics of time-series transcriptomics after resistance exercise. We used electrical stimulation-induced muscle contraction as a resistance exercise model (5 sets × 10 times of 3 s of 100-Hz electrical stimulation) on the tibialis anterior muscle of rats and measured the transcriptome in the muscle before and at 0, 1, 3, 6, and 12 h after muscle contractions by RNA sequencing. We also examined the relationship between the parameters of mRNA dynamics and the increase in protein expression at 12 h after muscle contractions. We found that the function of the upregulated genes differed after muscle contractions depending on their response rate. Genes related to muscle differentiation and response to mechanical stimulus were enriched in the sustainedly upregulated genes. Furthermore, there was a positive correlation between the magnitude of upregulated mRNA expression and the corresponding protein expression level at 12 h after muscle contractions. Although it has been theoretically suggested, this study experimentally demonstrated that the magnitude of the mRNA response after electrical stimulation-induced resistance exercise contributes to skeletal muscle adaptation via increases in protein expression. These findings suggest that mRNA expression dynamics such as response rate, a sustained upregulated expression pattern, and the magnitude of the response contribute to mechanisms underlying adaptation to resistance exercise.
Collapse
Affiliation(s)
- Tatsuya Kusano
- Bioscience and Technology Program, Department of Engineering ScienceThe University of Electro‐CommunicationsChofuTokyoJapan
| | - Yuta Sotani
- Bioscience and Technology Program, Department of Engineering ScienceThe University of Electro‐CommunicationsChofuTokyoJapan
| | - Reo Takeda
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)TsukubaIbarakiJapan
| | - Atsushi Hatano
- Department of Omics and Systems Biology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataNiigataJapan
| | - Kentaro Kawata
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)TsukubaIbarakiJapan
| | - Ryotaro Kano
- Bioscience and Technology Program, Department of Engineering ScienceThe University of Electro‐CommunicationsChofuTokyoJapan
| | - Masaki Matsumoto
- Department of Omics and Systems Biology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataNiigataJapan
| | - Yutaka Kano
- Bioscience and Technology Program, Department of Engineering ScienceThe University of Electro‐CommunicationsChofuTokyoJapan
| | - Daisuke Hoshino
- Bioscience and Technology Program, Department of Engineering ScienceThe University of Electro‐CommunicationsChofuTokyoJapan
| |
Collapse
|
6
|
Yang S, Xiong L, Yang G, Xiang J, Li L, Kang L, Liang Z. KLF13 restrains Dll4-muscular Notch2 axis to improve the muscle atrophy. J Cachexia Sarcopenia Muscle 2024; 15:1869-1882. [PMID: 38973459 PMCID: PMC11446702 DOI: 10.1002/jcsm.13538] [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: 10/30/2023] [Revised: 05/25/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Muscle atrophy can cause muscle dysfunction and weakness. Krüppel-like factor 13 (KLF13), a central regulator of cellular energy metabolism, is highly expressed in skeletal muscles and implicated in the pathogenesis of several diseases. This study investigated the role of KLF13 in muscle atrophy, which could be a novel therapeutic target. METHODS The effects of gene knockdown and pharmacological targeting of KLF13 on skeletal muscle atrophy were investigated using cell-based and animal models. Clofoctol, an antibiotic and KLF13 agonist, was also investigated as a candidate for repurposing. The mechanisms related to skeletal muscle atrophy were assessed by measuring the expression levels and activation statuses of key regulatory pathways and validated using gene knockdown and RNA sequencing. RESULTS In a dexamethasone-induced muscle atrophy mouse model, the KLF13 knockout group had decreased muscle strength (N) (1.77 ± 0.10 vs. 1.48 ± 0.16, P < 0.01), muscle weight (%) [gastrocnemius (Gas): 76.0 ± 5.69 vs. 60.7 ± 7.23, P < 0.001; tibialis anterior (TA): 75.8 ± 6.21 vs. 67.5 ± 5.01, P < 0.05], and exhaustive running distance (m) (495.5 ± 64.8 vs. 315.5 ± 60.9, P < 0.05) compared with the control group. KLF13 overexpression preserved muscle mass (Gas: 100 ± 6.38 vs. 120 ± 14.4, P < 0.01) and the exhaustive running distance (423.8 ± 59.04 vs. 530.2 ± 77.45, P < 0.05) in an in vivo diabetes-induced skeletal muscle atrophy model. Clofoctol treatment protected against dexamethasone-induced muscle atrophy. Myotubes treated with dexamethasone, an atrophy-inducing glucocorticoid, were aggravated by KLF13 knockout, but anti-atrophic effects were achieved by inducing KLF13 overexpression. We performed a transcriptome analysis and luciferase reporter assays to further explore this mechanism, finding that delta-like 4 (Dll4) was a novel target gene of KLF13. The KLF13 transcript repressed Dll4, inhibiting the Dll4-Notch2 axis and preventing muscle atrophy. Dexamethasone inhibited KLF13 expression by inhibiting myogenic differentiation 1 (i.e., MYOD1)-mediated KLF13 transcriptional activation and promoting F-Box and WD repeat domain containing 7 (i.e., FBXW7)-mediated KLF13 ubiquitination. CONCLUSIONS This study sheds new light on the mechanisms underlying skeletal muscle atrophy and potential drug targets. KLF13 regulates muscle atrophy and is a potential therapeutic target. Clofoctol is an attractive compound for repurposing studies to treat skeletal muscle atrophy.
Collapse
Affiliation(s)
- Shu Yang
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhenChina
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)ShenzhenChina
| | - Lijiao Xiong
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhenChina
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)ShenzhenChina
| | - Guangyan Yang
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhenChina
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)ShenzhenChina
| | - Jiaqing Xiang
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhenChina
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)ShenzhenChina
| | - Lixing Li
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhenChina
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)ShenzhenChina
| | - Lin Kang
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhenChina
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)ShenzhenChina
- The Biobank of National Innovation Center for Advanced Medical DevicesShenzhen People's HospitalShenzhenChina
| | - Zhen Liang
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhenChina
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)ShenzhenChina
| |
Collapse
|
7
|
Hotta K, Shimotsu R, Behnke BJ, Masamoto K, Yagishita K, Poole DC, Kano Y. Effect of diabetes on microvascular morphology and permeability of rat skeletal muscle: in vivo imaging using two-photon laser scanning microscopy. J Appl Physiol (1985) 2024; 137:963-974. [PMID: 39143906 DOI: 10.1152/japplphysiol.00222.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/29/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024] Open
Abstract
This investigation evaluated the microvascular permeability and ultrastructure of skeletal muscle capillaries in the skeletal muscle of diabetic (DIA) rats using two-photon laser scanning microscopy (TPLSM) and transmission electron microscopy (TEM). Microvascular permeability was assessed in the tibialis anterior muscle of control (CON) and DIA (streptozocin) male Wistar rats (n = 20, 10-14 wk) by in vivo imaging using TPLSM after fluorescent dye intravenous infusion. Fluorescent dye leakage was quantified to determine microvascular permeability. The ultrastructure was imaged by TEM ex vivo to calculate the size and number of intercellular clefts between capillary endothelial cells and also intracellular vesicles. Compared with control, the volumetrically determined interstitial fluorescent dye leakage, the endothelial cell thickness, and the number of intercellular clefts per capillary perimeter were significantly higher, and the cleft width was significantly narrower in tibialis anterior (TA) of DIA (interstitial fluorescent dye leakage, 2.88 ± 1.40 vs. 10.95 ± 1.41 µm3 × min × 106; endothelial thickness, 0.28 ± 0.02 vs. 0.45 ± 0.03 µm; number of intercellular clefts per capillary perimeter, 6.3 ± 0.80 vs. 13.6 ± 1.7/100 µm; cleft width, 11.92 ± 0.95 vs. 8.40 ± 1.03 nm, CON vs. DIA, respectively, all P < 0.05). The size of intracellular vesicles in the vascular endothelium showed an increased proportion of large vesicles in the DIA group compared with the CON group (P < 0.05). Diabetes mellitus enhances the microvascular permeability of skeletal muscle microvessels due, in part, to a higher density and narrowing of the endothelial intercellular clefts, and larger intracellular vesicles.NEW & NOTEWORTHY Microvascular permeability in diabetic muscle was investigated using our original two-photon scanning laser microscopy method. Compared with controls, the leakage volume was increased in diabetic muscle, which was atrophic with smaller capillary diameter, endothelial cell thickening, and the appearance of more endothelial intercellular gaps or clefts, and large vesicles. Hyperpermeability was closely related to ultrafine structural changes of the capillary endothelial cell junctions.
Collapse
MESH Headings
- Animals
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/pathology
- Muscle, Skeletal/diagnostic imaging
- Male
- Rats, Wistar
- Capillary Permeability/physiology
- Rats
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/diagnostic imaging
- Microscopy, Confocal/methods
- Capillaries/diagnostic imaging
- Capillaries/pathology
- Capillaries/ultrastructure
- Endothelial Cells/pathology
- Endothelial Cells/ultrastructure
- Endothelial Cells/metabolism
- Fluorescent Dyes
- Microvessels/diagnostic imaging
- Microvessels/pathology
- Microscopy, Electron, Transmission/methods
Collapse
Affiliation(s)
- Kazuki Hotta
- Department of Rehabilitation Sciences, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
- Department of Rehabilitation, Kitasato University School of Allied Health Sciences, Sagamihara, Japan
| | - Rie Shimotsu
- Department of Engineering Science, Bioscience and Technology Program, University of Electro-Communications, Chofu, Japan
- Center for Neuroscience and Biomedical Engineering, University of Electro-Communications, Chofu, Japan
| | - Bradley J Behnke
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, United States
- Department of Kinesiology, Kansas State University, Manhattan, Kansas, United States
| | - Kazuto Masamoto
- Department of Engineering Science, Bioscience and Technology Program, University of Electro-Communications, Chofu, Japan
- Center for Neuroscience and Biomedical Engineering, University of Electro-Communications, Chofu, Japan
| | - Kazuyoshi Yagishita
- Clinical Center for Sports Medicine and Sports Dentistry, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - David C Poole
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, United States
- Department of Kinesiology, Kansas State University, Manhattan, Kansas, United States
| | - Yutaka Kano
- Department of Engineering Science, Bioscience and Technology Program, University of Electro-Communications, Chofu, Japan
- Center for Neuroscience and Biomedical Engineering, University of Electro-Communications, Chofu, Japan
| |
Collapse
|
8
|
Hayashi T, Sadaki S, Tsuji R, Okada R, Fuseya S, Kanai M, Nakamura A, Okamura Y, Muratani M, Wenchao G, Sugasawa T, Mizuno S, Warabi E, Kudo T, Takahashi S, Fujita R. Dual-specificity phosphatases 13 and 27 as key switches in muscle stem cell transition from proliferation to differentiation. Stem Cells 2024; 42:830-847. [PMID: 38975693 PMCID: PMC11384902 DOI: 10.1093/stmcls/sxae045] [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: 01/09/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024]
Abstract
Muscle regeneration depends on muscle stem cell (MuSC) activity. Myogenic regulatory factors, including myoblast determination protein 1 (MyoD), regulate the fate transition of MuSCs. However, the direct target of MYOD in the process is not completely clear. Using previously established MyoD knock-in (MyoD-KI) mice, we revealed that MyoD targets dual-specificity phosphatase (Dusp) 13 and Dusp27. In Dusp13:Dusp27 double knock-out mice, the ability for muscle regeneration after injury was reduced. Moreover, single-cell RNA sequencing of MyoD-high expressing MuSCs from MyoD-KI mice revealed that Dusp13 and Dusp27 are expressed only in specific populations within MyoD-high MuSCs, which also express Myogenin. Overexpressing Dusp13 in MuSCs causes premature muscle differentiation. Thus, we propose a model where DUSP13 and DUSP27 contribute to the fate transition of MuSCs from proliferation to differentiation during myogenesis.
Collapse
Affiliation(s)
- Takuto Hayashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Shunya Sadaki
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
- PhD Program in Humanics, School of Integrative and Global Majors, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Ryosuke Tsuji
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
- PhD Program in Humanics, School of Integrative and Global Majors, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Risa Okada
- JEM Utilization Center, Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency (JAXA), Ibaraki 305-8505, Japan
| | - Sayaka Fuseya
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Ibaraki 305-8565, Japan
| | - Maho Kanai
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Ayano Nakamura
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
- College of Medicine, School of Medicine and Health Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yui Okamura
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
- College of Medicine, School of Medicine and Health Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Masafumi Muratani
- Department of Genome Biology, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Gu Wenchao
- Department of Diagnostic and Interventional Radiology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takehito Sugasawa
- Laboratory of Clinical Examination and Sports Medicine, Department of Clinical Medicine, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Laboratory Animal Science, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Eiji Warabi
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takashi Kudo
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Ryo Fujita
- Division of Regenerative Medicine, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
9
|
Jin Z, Mao Y, Guo Q, Yin Y, Kiram A, Zhou D, Yang J, Zhou Z, Xue J, Feng Z, Liu Z, Qiu Y, Fu T, Gan Z, Zhu Z. Imbalanced Skeletal Muscle Mitochondrial Proteostasis Causes Bone Loss. RESEARCH (WASHINGTON, D.C.) 2024; 7:0465. [PMID: 39221030 PMCID: PMC11362843 DOI: 10.34133/research.0465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
Although microgravity has been implicated in osteoporosis, the precise molecular mechanism remains elusive. Here, we found that microgravity might induce mitochondrial protein buildup in skeletal muscle, alongside reduced levels of LONP1 protein. We revealed that disruptions in mitochondrial proteolysis, induced by the targeted skeletal muscle-specific deletion of the essential mitochondrial protease LONP1 or by the acute inducible deletion of muscle LONP1 in adult mice, cause reduced bone mass and compromised mechanical function. Moreover, the bone loss and weakness phenotypes were recapitulated in skeletal muscle-specific overexpressing ΔOTC mice, a known protein degraded by LONP1. Mechanistically, mitochondrial proteostasis imbalance triggered the mitochondrial unfolded protein response (UPRmt) in muscle, leading to an up-regulation of multiple myokines, including FGF21, which acts as a pro-osteoclastogenic factor. Surprisingly, this mitochondrial proteostasis stress influenced muscle-bone crosstalk independently of ATF4 in skeletal muscle. Furthermore, we established a marked association between serum FGF21 levels and bone health in humans. These findings emphasize the pivotal role of skeletal muscle mitochondrial proteostasis in responding to alterations in loading conditions and in coordinating UPRmt to modulate bone metabolism.
Collapse
Affiliation(s)
- Zhen Jin
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
- Division of Spine Surgery, Department of Orthopedic Surgery,
Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yan Mao
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Qiqi Guo
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Yujing Yin
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Abdukahar Kiram
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Danxia Zhou
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Jing Yang
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zheng Zhou
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Jiachen Xue
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zhenhua Feng
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zhen Liu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
- Division of Spine Surgery, Department of Orthopedic Surgery,
Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yong Qiu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Tingting Fu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zhenji Gan
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zezhang Zhu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing University, Nanjing, China
- Division of Spine Surgery, Department of Orthopedic Surgery,
Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Bolado-Carrancio A, Tapia O, Rodríguez-Rey JC. Ubiquitination Insight from Spinal Muscular Atrophy-From Pathogenesis to Therapy: A Muscle Perspective. Int J Mol Sci 2024; 25:8800. [PMID: 39201486 PMCID: PMC11354275 DOI: 10.3390/ijms25168800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/03/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Spinal muscular atrophy (SMA) is one of the most frequent causes of death in childhood. The disease's molecular basis is deletion or mutations in the SMN1 gene, which produces reduced survival motor neuron protein (SMN) levels. As a result, there is spinal motor neuron degeneration and a large increase in muscle atrophy, in which the ubiquitin-proteasome system (UPS) plays a significant role. In humans, a paralogue of SMN1, SMN2 encodes the truncated protein SMNΔ7. Structural differences between SMN and SMNΔ7 affect the interaction of the proteins with UPS and decrease the stability of the truncated protein. SMN loss affects the general ubiquitination process by lowering the levels of UBA1, one of the main enzymes in the ubiquitination process. We discuss how SMN loss affects both SMN stability and the general ubiquitination process, and how the proteins involved in ubiquitination could be used as future targets for SMA treatment.
Collapse
Affiliation(s)
- Alfonso Bolado-Carrancio
- Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria-and Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain;
| | - Olga Tapia
- Departamento de Ciencias Médicas Básicas, Instituto de Tecnologías Biomédicas, Universidad de la Laguna, 38200 La Laguna, Spain
| | - José C. Rodríguez-Rey
- Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria-and Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain;
| |
Collapse
|
11
|
Madigan LA, Jaime D, Chen I, Fallon JR. MuSK-BMP signaling in adult muscle stem cells maintains quiescence and regulates myofiber size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.17.541238. [PMID: 37292636 PMCID: PMC10245747 DOI: 10.1101/2023.05.17.541238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A central question in adult stem cell biology is elucidating the signaling pathways regulating their dynamics and function in diverse physiological and age-related contexts. Muscle stem cells in adults (Satellite Cells; SCs) are generally quiescent but can activate and contribute to muscle repair and growth. Here we tested the role of the MuSK-BMP pathway in regulating adult SC quiescence by deletion of the BMP-binding MuSK Ig3 domain ('ΔIg3-MuSK'). At 3 months of age SC and myonuclei numbers and myofiber size were comparable to WT. However, at 5 months of age SC density was decreased while myofiber size, myonuclear number and grip strength were increased - indicating that SCs had activated and productively fused into the myofibers over this interval. Transcriptomic analysis showed that SCs from uninjured ΔIg3-MuSK mice exhibit signatures of activation. Regeneration experiments showed that ΔIg3-MuSK SCs maintain full stem cell function. Expression of ΔIg3-MuSK in adult SCs was sufficient to break quiescence and increase myofiber size. We conclude that the MuSK-BMP pathway regulates SC quiescence and myofiber size in a cell autonomous, age-dependent manner. Targeting MuSK-BMP signaling in muscle stem cells thus emerges a therapeutic strategy for promoting muscle growth and function in the settings of injury, disease, and aging. Highlights MuSK, in its role as a BMP co-receptor, regulates adult muscle stem cell quiescenceThe MuSK-BMP pathway acts cell autonomouslyIncreased muscle size and function with preservation of myonuclear density and stemness in mice with attenuated MuSK-BMP signaling.
Collapse
|
12
|
Rodríguez C, Timóteo-Ferreira F, Minchiotti G, Brunelli S, Guardiola O. Cellular interactions and microenvironment dynamics in skeletal muscle regeneration and disease. Front Cell Dev Biol 2024; 12:1385399. [PMID: 38840849 PMCID: PMC11150574 DOI: 10.3389/fcell.2024.1385399] [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: 02/12/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Skeletal muscle regeneration relies on the intricate interplay of various cell populations within the muscle niche-an environment crucial for regulating the behavior of muscle stem cells (MuSCs) and ensuring postnatal tissue maintenance and regeneration. This review delves into the dynamic interactions among key players of this process, including MuSCs, macrophages (MPs), fibro-adipogenic progenitors (FAPs), endothelial cells (ECs), and pericytes (PCs), each assuming pivotal roles in orchestrating homeostasis and regeneration. Dysfunctions in these interactions can lead not only to pathological conditions but also exacerbate muscular dystrophies. The exploration of cellular and molecular crosstalk among these populations in both physiological and dystrophic conditions provides insights into the multifaceted communication networks governing muscle regeneration. Furthermore, this review discusses emerging strategies to modulate the muscle-regenerating niche, presenting a comprehensive overview of current understanding and innovative approaches.
Collapse
Affiliation(s)
- Cristina Rodríguez
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso”, CNR, Naples, Italy
| | | | - Gabriella Minchiotti
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso”, CNR, Naples, Italy
| | - Silvia Brunelli
- School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Ombretta Guardiola
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso”, CNR, Naples, Italy
| |
Collapse
|
13
|
Karthikeyan S, Asakura A. Imaging analysis for muscle stem cells and regeneration. Front Cell Dev Biol 2024; 12:1411401. [PMID: 38774645 PMCID: PMC11106391 DOI: 10.3389/fcell.2024.1411401] [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: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 05/24/2024] Open
Abstract
Composed of a diverse variety of cells, the skeletal muscle is one of the body's tissues with the remarkable ability to regenerate after injury. One of the key players in the regeneration process is the muscle satellite cell (MuSC), a stem cell population for skeletal muscle, as it is the source of new myofibers. Maintaining MuSC quiescence during homeostasis involves complex interactions between MuSCs and other cells in their corresponding niche in adult skeletal muscle. After the injury, MuSCs are activated to enter the cell cycle for cell proliferation and differentiate into myotubes, followed by mature myofibers to regenerate muscle. Despite decades of research, the exact mechanisms underlying MuSC maintenance and activation remain elusive. Traditional methods of analyzing MuSCs, including cell cultures, animal models, and gene expression analyses, provide some insight into MuSC biology but lack the ability to replicate the 3-dimensional (3-D) in vivo muscle environment and capture dynamic processes comprehensively. Recent advancements in imaging technology, including confocal, intra-vital, and multi-photon microscopies, provide promising avenues for dynamic MuSC morphology and behavior to be observed and characterized. This chapter aims to review 3-D and live-imaging methods that have contributed to uncovering insights into MuSC behavior, morphology changes, interactions within the muscle niche, and internal signaling pathways during the quiescence to activation (Q-A) transition. Integrating advanced imaging modalities and computational tools provides a new avenue for studying complex biological processes in skeletal muscle regeneration and muscle degenerative diseases such as sarcopenia and Duchenne muscular dystrophy (DMD).
Collapse
Affiliation(s)
- Smrithi Karthikeyan
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, United States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Atsushi Asakura
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, United States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
14
|
Lai Y, Ramírez-Pardo I, Isern J, An J, Perdiguero E, Serrano AL, Li J, García-Domínguez E, Segalés J, Guo P, Lukesova V, Andrés E, Zuo J, Yuan Y, Liu C, Viña J, Doménech-Fernández J, Gómez-Cabrera MC, Song Y, Liu L, Xu X, Muñoz-Cánoves P, Esteban MA. Multimodal cell atlas of the ageing human skeletal muscle. Nature 2024; 629:154-164. [PMID: 38649488 PMCID: PMC11062927 DOI: 10.1038/s41586-024-07348-6] [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: 08/24/2022] [Accepted: 03/25/2024] [Indexed: 04/25/2024]
Abstract
Muscle atrophy and functional decline (sarcopenia) are common manifestations of frailty and are critical contributors to morbidity and mortality in older people1. Deciphering the molecular mechanisms underlying sarcopenia has major implications for understanding human ageing2. Yet, progress has been slow, partly due to the difficulties of characterizing skeletal muscle niche heterogeneity (whereby myofibres are the most abundant) and obtaining well-characterized human samples3,4. Here we generate a single-cell/single-nucleus transcriptomic and chromatin accessibility map of human limb skeletal muscles encompassing over 387,000 cells/nuclei from individuals aged 15 to 99 years with distinct fitness and frailty levels. We describe how cell populations change during ageing, including the emergence of new populations in older people, and the cell-specific and multicellular network features (at the transcriptomic and epigenetic levels) associated with these changes. On the basis of cross-comparison with genetic data, we also identify key elements of chromatin architecture that mark susceptibility to sarcopenia. Our study provides a basis for identifying targets in the skeletal muscle that are amenable to medical, pharmacological and lifestyle interventions in late life.
Collapse
Affiliation(s)
- Yiwei Lai
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Ignacio Ramírez-Pardo
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA
| | - Joan Isern
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA
| | - Juan An
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Eusebio Perdiguero
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA
| | - Antonio L Serrano
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA
| | - Jinxiu Li
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Esther García-Domínguez
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia and CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Jessica Segalés
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Pengcheng Guo
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Jilin, China
| | - Vera Lukesova
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Eva Andrés
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jing Zuo
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Yue Yuan
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Chuanyu Liu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - José Viña
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia and CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Julio Doménech-Fernández
- Servicio de Cirugía Ortopédica y Traumatología, Hospital Arnau de Vilanova y Hospital de Liria and Health Care Department Arnau-Lliria, Valencia, Spain
- Department of Orthopedic Surgery, Clinica Universidad de Navarra, Pamplona, Spain
| | - Mari Carmen Gómez-Cabrera
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia and CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Yancheng Song
- Department of Orthopedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Longqi Liu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xun Xu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Pura Muñoz-Cánoves
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA.
- ICREA, Barcelona, Spain.
| | - Miguel A Esteban
- BGI Research, Hangzhou, China.
- BGI Research, Shenzhen, China.
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Jilin, China.
- The Fifth Affiliated Hospital of Guangzhou Medical University-BGI Research Center for Integrative Biology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
15
|
Kim HM, Bathe M. Force-free activation of Notch with DNA origami. Trends Genet 2024; 40:293-295. [PMID: 38493041 DOI: 10.1016/j.tig.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 03/18/2024]
Abstract
The Notch signaling pathway is a highly conserved, fundamental process to embryogenesis and neurogenesis. While force-induced conformational change is known to activate Notch receptors, Smyrlaki et al. recently used DNA origami to reveal an additional, force-independent mode of Notch activation via soluble presentation of spatially controlled ligand nanopatterns.
Collapse
Affiliation(s)
- Hyun Min Kim
- Department of Biological Engineering, MIT, Cambridge, MA, USA
| | - Mark Bathe
- Department of Biological Engineering, MIT, Cambridge, MA, USA; HMS Initiative for RNA Medicine, BIDMC Cancer Center, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT & Harvard, Cambridge, MA, USA.
| |
Collapse
|
16
|
Fischer A, Alsina-Sanchis E. Disturbed endothelial cell signaling in tumor progression and therapy resistance. Curr Opin Cell Biol 2024; 86:102287. [PMID: 38029706 DOI: 10.1016/j.ceb.2023.102287] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023]
Abstract
Growth of new blood vessels is considered requisite to cancer progression. Recent findings revealed that in addition to inducing angiogenesis, tumor-derived factors alter endothelial cell gene transcription within the tumor mass but also systemically throughout the body. This subsequently contributes to immunosuppression, altered metabolism, therapy resistance and metastasis. Clinical studies demonstrated that targeting the endothelium can increase the success rate of immunotherapy. Single-cell technologies revealed remarkable organ-specific endothelial heterogeneity that becomes altered by the presence of a tumor. In metastases, endothelial transcription differs remarkably between newly formed and co-opted vessels which may provide a basis for developing new therapies to target endothelial cells and overcome therapy resistance more effectively. This review addresses how cancers impact the endothelium to facilitate tumor progression.
Collapse
Affiliation(s)
- Andreas Fischer
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen University, 37075 Göttingen, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Germany.
| | - Elisenda Alsina-Sanchis
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen University, 37075 Göttingen, Germany
| |
Collapse
|
17
|
Smyrlaki I, Fördős F, Rocamonde-Lago I, Wang Y, Shen B, Lentini A, Luca VC, Reinius B, Teixeira AI, Högberg B. Soluble and multivalent Jag1 DNA origami nanopatterns activate Notch without pulling force. Nat Commun 2024; 15:465. [PMID: 38238313 PMCID: PMC10796381 DOI: 10.1038/s41467-023-44059-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 11/28/2023] [Indexed: 01/22/2024] Open
Abstract
The Notch signaling pathway has fundamental roles in embryonic development and in the nervous system. The current model of receptor activation involves initiation via a force-induced conformational change. Here, we define conditions that reveal pulling force-independent Notch activation using soluble multivalent constructs. We treat neuroepithelial stem-like cells with molecularly precise ligand nanopatterns displayed from solution using DNA origami. Notch signaling follows with clusters of Jag1, and with chimeric structures where most Jag1 proteins are replaced by other binders not targeting Notch. Our data rule out several confounding factors and suggest a model where Jag1 activates Notch upon prolonged binding without appearing to need a pulling force. These findings reveal a distinct mode of activation of Notch and lay the foundation for the development of soluble agonists.
Collapse
Affiliation(s)
- Ioanna Smyrlaki
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ferenc Fördős
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Iris Rocamonde-Lago
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Yang Wang
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Boxuan Shen
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Alto, Finland
| | - Antonio Lentini
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Vincent C Luca
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| | - Björn Reinius
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ana I Teixeira
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Björn Högberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
18
|
Liu G, Wei J, Xiao W, Xie W, Ru Q, Chen L, Wu Y, Mobasheri A, Li Y. Insights into the Notch signaling pathway in degenerative musculoskeletal disorders: Mechanisms and perspectives. Biomed Pharmacother 2023; 169:115884. [PMID: 37981460 DOI: 10.1016/j.biopha.2023.115884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023] Open
Abstract
Degenerative musculoskeletal disorders are a group of age-related diseases of the locomotive system that severely affects the patient's ability to work and cause adverse sequalae such as fractures and even death. The incidence and prevalence of degenerative musculoskeletal disorders is rising owing to the aging of the world's population. The Notch signaling pathway, which is expressed in almost all organ systems, extensively regulates cell proliferation and differentiation as well as cellular fate. Notch signaling shows increased activity in degenerative musculoskeletal disorders and retards the progression of degeneration to some extent. The review focuses on four major degenerative musculoskeletal disorders (osteoarthritis, intervertebral disc degeneration, osteoporosis, and sarcopenia) and summarizes the pathophysiological functions of Notch signaling in these disorders, especially its role in stem/progenitor cells in each disorder. Finally, a conclusion will be presented to explore the research and application of the perspectives on Notch signaling in degenerative musculoskeletal disorders.
Collapse
Affiliation(s)
- Gaoming Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410011, China
| | - Jun Wei
- Department of Clinical Medical School, Xinjiang Medical University, Urumqi 830054, China
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410011, China; Department of Clinical Medical School, Xinjiang Medical University, Urumqi 830054, China.
| |
Collapse
|
19
|
Ferreira RP, Duarte JA. Protein Turnover in Skeletal Muscle: Looking at Molecular Regulation towards an Active Lifestyle. Int J Sports Med 2023; 44:763-777. [PMID: 36854391 DOI: 10.1055/a-2044-8277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Skeletal muscle is a highly plastic tissue, able to change its mass and functional properties in response to several stimuli. Skeletal muscle mass is influenced by the balance between protein synthesis and breakdown, which is regulated by several signaling pathways. The relative contribution of Akt/mTOR signaling, ubiquitin-proteasome pathway, autophagy among other signaling pathways to protein turnover and, therefore, to skeletal muscle mass, differs depending on the wasting or loading condition and muscle type. By modulating mitochondria biogenesis, PGC-1α has a major role in the cell's bioenergetic status and, thus, on protein turnover. In fact, rates of protein turnover regulate differently the levels of distinct protein classes in response to atrophic or hypertrophic stimuli. Mitochondrial protein turnover rates may be enhanced in wasting conditions, whereas the increased turnover of myofibrillar proteins triggers muscle mass gain. The present review aims to update the knowledge on the molecular pathways implicated in the regulation of protein turnover in skeletal muscle, focusing on how distinct muscle proteins may be modulated by lifestyle interventions with emphasis on exercise training. The comprehensive analysis of the anabolic effects of exercise programs will pave the way to the tailored management of muscle wasting conditions.
Collapse
Affiliation(s)
- Rita Pinho Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Jose Alberto Duarte
- TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
- CIAFEL, Faculty of Sports, University of Porto and Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
| |
Collapse
|
20
|
Zhou Q, Li B, Li J. DLL4-Notch signalling in acute-on-chronic liver failure: State of the art and perspectives. Life Sci 2023; 317:121438. [PMID: 36709913 DOI: 10.1016/j.lfs.2023.121438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/28/2023]
Abstract
Acute-on-chronic liver failure (ACLF) is a syndrome characterized by acute decompensation of chronic liver disease associated with multiple-organ failures and high short-term mortality. Acute insults to patients with chronic liver disease can lead to ACLF, among which, hepatitis B virus-related ACLF is the most common type of liver failure in the Asia-Pacific region. Currently, immune-metabolism disorders and systemic inflammation are proposed to be the main mechanisms of ACLF. The resulting cholestasis and intrahepatic microcirculatory dysfunction accelerate the development of ACLF. Treatments targeting immune regulation, metabolic balance, microcirculation maintenance and bile duct repair can alleviate inflammation and restore the tissue structure. An increasing number of studies have demonstrated that delta-like ligand 4 (DLL4), one of the Notch signalling ligands, plays a vital role in immune regulation, metabolism, angiogenesis, and biliary regeneration, which participate in liver pathological and physiological processes. The detailed mechanism of the DLL4-Notch signalling pathway in ACLF has rarely been investigated. Here, we review the evidence showing that DLL4-Notch signalling is involved in ACLF and analyse the potential role of DLL4 in the treatment of ACLF.
Collapse
Affiliation(s)
- Qian Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Bingqi Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Jun Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China; Precision Medicine Center of Taizhou Central Hospital, Taizhou University Medical School, Taizhou, China.
| |
Collapse
|
21
|
Gao Y, Fu Z, Guan J, Liu X, Zhang Q. The role of Notch signaling pathway in metabolic bone diseases. Biochem Pharmacol 2023; 207:115377. [PMID: 36513140 DOI: 10.1016/j.bcp.2022.115377] [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: 09/29/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Metabolic bone diseases is the third most common endocrine diseases after diabetes and thyroid diseases. More than 500 million people worldwide suffer from metabolic bone diseases. The generation and development of bone metabolic diseases is a complex process regulated by multiple signaling pathways, among which the Notch signaling pathway is one of the most important pathways. The Notch signaling pathway regulates the differentiation and function of osteoblasts and osteoclasts, and affects the process of cartilage formation, bone formation and bone resorption. Genetic mutations in upstream and downstream of Notch signaling genes can lead to a series of metabolic bone diseases, such as Alagille syndrome, Adams-Oliver syndrome and spondylocostal dysostosis. In this review, we analyzed the mechanisms of Notch ligands, Notch receptors and signaling molecules in the process of signal transduction, and summarized the progress on the pathogenesis and clinical manifestations of bone metabolic diseases caused by Notch gene mutation. We hope to draw attention to the role of the Notch signaling pathway in metabolic bone diseases and provide new ideas and approaches for the diagnosis and treatment of metabolic bone diseases.
Collapse
Affiliation(s)
- Yongguang Gao
- Tangshan Key Laboratory of Green Speciality Chemicals, Department of Chemistry, Tangshan Normal University, Tangshan 063000, China.
| | - Zhanda Fu
- Tangshan Key Laboratory of Green Speciality Chemicals, Department of Chemistry, Tangshan Normal University, Tangshan 063000, China
| | - Junxia Guan
- Tangshan Key Laboratory of Green Speciality Chemicals, Department of Chemistry, Tangshan Normal University, Tangshan 063000, China
| | - Xinhua Liu
- Tangshan Key Laboratory of Green Speciality Chemicals, Department of Chemistry, Tangshan Normal University, Tangshan 063000, China
| | - Qing Zhang
- Tangshan Key Laboratory of Green Speciality Chemicals, Department of Chemistry, Tangshan Normal University, Tangshan 063000, China.
| |
Collapse
|
22
|
Chen K, Gao P, Li Z, Dai A, Yang M, Chen S, Su J, Deng Z, Li L. Forkhead Box O Signaling Pathway in Skeletal Muscle Atrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1648-1657. [PMID: 36174679 DOI: 10.1016/j.ajpath.2022.09.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/01/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Skeletal muscle atrophy is the consequence of protein degradation exceeding protein synthesis because of disease, aging, and physical inactivity. Patients with skeletal muscle atrophy have decreased muscle mass and fiber cross-sectional area, and experience reduced survival quality and motor function. The forkhead box O (FOXO) signaling pathway plays an important role in the pathogenesis of skeletal muscle atrophy by regulating E3 ubiquitin ligases and some autophagy factors. However, the mechanism of FOXO signaling pathway leading to skeletal muscle atrophy is still unclear. The development of treatment strategies for skeletal muscle atrophy has been a thorny clinical problem. FOXO-targeted therapy to treat skeletal muscle atrophy is a promising approach, and an increasing number of relevant studies have been reported. This article reviews the mechanism and therapeutic targets of the FOXO signaling pathway mediating skeletal muscle atrophy, and provides ideas for the clinical treatment of this condition.
Collapse
Affiliation(s)
- Kun Chen
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Peng Gao
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Zongchao Li
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Aonan Dai
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Ming Yang
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Siyu Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Jingyue Su
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China.
| | - Liangjun Li
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China.
| |
Collapse
|
23
|
Den Hartog L, Asakura A. Implications of notch signaling in duchenne muscular dystrophy. Front Physiol 2022; 13:984373. [PMID: 36237531 PMCID: PMC9553129 DOI: 10.3389/fphys.2022.984373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
This review focuses upon the implications of the Notch signaling pathway in muscular dystrophies, particularly Duchenne muscular dystrophy (DMD): a pervasive and catastrophic condition concerned with skeletal muscle degeneration. Prior work has defined the pathogenesis of DMD, and several therapeutic approaches have been undertaken in order to regenerate skeletal muscle tissue and ameliorate the phenotype. There is presently no cure for DMD, but a promising avenue for novel therapies is inducing muscle regeneration via satellite cells (muscle stem cells). One specific target using this approach is the Notch signaling pathway. The canonical Notch signaling pathway has been well-characterized and it ultimately governs cell fate decision, cell proliferation, and induction of differentiation. Additionally, inhibition of the Notch signaling pathway has been directly implicated in the deficits seen with muscular dystrophies. Here, we explore the connection between the Notch signaling pathway and DMD, as well as how Notch signaling may be targeted to improve the muscle degeneration seen in muscular dystrophies.
Collapse
|
24
|
Dungan CM, Brightwell CR, Wen Y, Zdunek CJ, Latham CM, Thomas NT, Zagzoog AM, Brightwell BD, Nolt GL, Keeble AR, Watowich SJ, Murach KA, Fry CS. Muscle-Specific Cellular and Molecular Adaptations to Late-Life Voluntary Concurrent Exercise. FUNCTION 2022; 3:zqac027. [PMID: 35774589 PMCID: PMC9233305 DOI: 10.1093/function/zqac027] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/06/2022] [Accepted: 05/08/2022] [Indexed: 01/07/2023] Open
Abstract
Murine exercise models can provide information on factors that influence muscle adaptability with aging, but few translatable solutions exist. Progressive weighted wheel running (PoWeR) is a simple, voluntary, low-cost, high-volume endurance/resistance exercise approach for training young mice. In the current investigation, aged mice (22-mo-old) underwent a modified version of PoWeR for 8 wk. Muscle functional, cellular, biochemical, transcriptional, and myonuclear DNA methylation analyses provide an encompassing picture of how muscle from aged mice responds to high-volume combined training. Mice run 6-8 km/d, and relative to sedentary mice, PoWeR increases plantarflexor muscle strength. The oxidative soleus of aged mice responds to PoWeR similarly to young mice in every parameter measured in previous work; this includes muscle mass, glycolytic-to-oxidative fiber type transitioning, fiber size, satellite cell frequency, and myonuclear number. The oxidative/glycolytic plantaris adapts according to fiber type, but with modest overall changes in muscle mass. Capillarity increases markedly with PoWeR in both muscles, which may be permissive for adaptability in advanced age. Comparison to published PoWeR RNA-sequencing data in young mice identified conserved regulators of adaptability across age and muscles; this includes Aldh1l1 which associates with muscle vasculature. Agrn and Samd1 gene expression is upregulated after PoWeR simultaneous with a hypomethylated promoter CpG in myonuclear DNA, which could have implications for innervation and capillarization. A promoter CpG in Rbm10 is hypomethylated by late-life exercise in myonuclei, consistent with findings in muscle tissue. PoWeR and the data herein are a resource for uncovering cellular and molecular regulators of muscle adaptation with aging.
Collapse
Affiliation(s)
- Cory M Dungan
- Department of Physical Therapy, University of Kentucky, Lexington 40536, KY, USA
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
| | - Camille R Brightwell
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Yuan Wen
- Department of Physical Therapy, University of Kentucky, Lexington 40536, KY, USA
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
| | | | - Christine M Latham
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Nicholas T Thomas
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Alyaa M Zagzoog
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Benjamin D Brightwell
- Kinesiology and Health Promotion Graduate Program, University of Kentucky, Lexington 40536, KY, USA
| | - Georgia L Nolt
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
| | - Alexander R Keeble
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Stanley J Watowich
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston 77555, TX, USA
| | - Kevin A Murach
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville 72701, AR, USA
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville 72701, AR, USA
| | - Christopher S Fry
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| |
Collapse
|