1
|
Wang W, Huang M, Huang X, Ma K, Luo M, Yang N. GsMTx4-blocked PIEZO1 channel promotes myogenic differentiation and alleviates myofiber damage in Duchenne muscular dystrophy. Skelet Muscle 2025; 15:13. [PMID: 40361216 PMCID: PMC12076844 DOI: 10.1186/s13395-025-00383-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a debilitating disease characterized by progressive muscle-wasting and a lack of effective therapy. Although the application of GsMTx4 has been shown to reduce muscle mass loss in dystrophic mice, the mechanism of action remains unclear. METHODS We employed single-nucleus RNA sequencing data to scrutinize the expression of mechanosensitive channels in skeletal muscle. The upregulation of PIEZO1 and its precise localization were corroborated in DMD patients, mdx mice, and activated satellite cells. To delve into the role of the GsMTx4-blocked PIEZO1 channel in the myogenic program, we conducted comprehensive in vitro and in vivo studies encompassing the proliferation of satellite cells, differentiation of myoblasts, and calcium influx into myofibers. Utilizing both a PIEZO1 channel inhibitor, GsMTx4, and a PIEZO1 channel agonist, Yoda1, we explored the PIEZO1 channel's impact on satellite cell proliferation and myogenic differentiation. Additionally, we explored the protective effect of the PIEZO1 channel on myofiber calcium influx using mdx mouse models and isolated single myofibers. RESULTS PIEZO1 was upregulated in the muscle of DMD patients and was predominantly expressed in satellite cells and upregulated during satellite cell proliferation. Treatment with GsMTx4 increased the cross-sectional areas of myofibers and reduced the proportion of centrally nucleated fibers in mdx mice. GsMTx4 inhibited satellite cell proliferation while promoting myogenic differentiation. During myogenic differentiation, the YAP nuclear-cytoplasmic ratio increased in cells treated with GsMTx4 and showed a significant correlation with the nuclear localization of MyoG. In myofibers, GsMTx4 significantly reduced the level of p-CaMKII/CaMKII in muscle and calcium load. CONCLUSIONS PIEZO1 upregulation in DMD could potentially stem from an elevated proportion of proliferating satellite cells triggered by sarcolemma damage and muscle necrosis. The inhibition of the PIEZO1 channel by GsMTx4 plays a beneficial role in fostering myogenic differentiation and mitigating myofiber damage. The PIEZO1 channel emerges as a promising therapeutic target for addressing DMD.
Collapse
MESH Headings
- Animals
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/drug therapy
- Spider Venoms/pharmacology
- Cell Differentiation/drug effects
- Mice, Inbred mdx
- Mice
- Ion Channels/metabolism
- Ion Channels/antagonists & inhibitors
- Ion Channels/genetics
- Muscle Development/drug effects
- Humans
- Satellite Cells, Skeletal Muscle/metabolism
- Male
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle Fibers, Skeletal/drug effects
- Cell Proliferation/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Mice, Inbred C57BL
- Calcium/metabolism
- Intercellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Wengang Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Mingyang Huang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xiusheng Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ke Ma
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ming Luo
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Ningning Yang
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
2
|
Chen P, Jia F, Wang M, Yang S. Analysis of the mechanism of skeletal muscle atrophy from the pathway of decreased protein synthesis. Front Physiol 2025; 16:1533394. [PMID: 40352150 PMCID: PMC12061715 DOI: 10.3389/fphys.2025.1533394] [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: 11/23/2024] [Accepted: 03/18/2025] [Indexed: 05/14/2025] Open
Abstract
Skeletal muscle atrophy is associated with denervation, cancer, diabetes, aging, immobilization, and inflammation, which can significantly impair mobility. It is primarily attributable to increased protein catabolism alongside reduced protein synthesis, although the precise mechanisms underlying this process are not yet fully known. Unlike in the pathway driving increased catabolism, fewer studies have explored the mechanism underpinning muscle atrophy under reduced protein synthesis. Therefore, this study aimed to focus on summarizing relevant studies on the reduction of protein synthesis leading to skeletal muscle atrophy, as driven by alterations in pathways such as the insulin-like growth factor-1-phosphatidylinositol 3-kinase-protein kinase B-rapamycin signaling pathway, glycogen synthase kinase-3, glucocorticoids, 5'-adenosine monophosphate-activated protein kinase, branched-chain amino acid sensors, myostatin, long-term proinflammatory factors, oxidative stress and mitochondrial dysfunction, calciumion concentration, activating transcription factor 4, and glycyl-tRNA synthetase alterations. Consolidating these data will provide a foundation and theoretical basis for further investigation into the mechanisms of muscle atrophy from the perspective of reduced protein synthesis pathways.
Collapse
Affiliation(s)
| | | | | | - Shengbo Yang
- Department of Anatomy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
3
|
Oura M, Son BK, Song Z, Toyoshima K, Nanao-Hamai M, Ogawa S, Akishita M. Testosterone/androgen receptor antagonizes immobility-induced muscle atrophy through Inhibition of myostatin transcription and inflammation in mice. Sci Rep 2025; 15:10568. [PMID: 40148525 PMCID: PMC11950163 DOI: 10.1038/s41598-025-95115-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
Sarcopenia is caused by excessive muscle protein degradation owing to various factors, including disuse. Although testosterone supplementation is an effective treatment, the underlying molecular mechanisms, particularly the role of the androgen receptor (AR), remain unclear. In this study, we examined the preventive actions of testosterone/AR against muscle atrophy in a murine model of immobilization-induced muscle atrophy. The bilateral hindlimbs of 8-week-old male C57BL/6J mice were immobilized using a wire. Testosterone deficiency and supplementation (50 µg/mL) were conducted by castration and intraperitoneal injection (twice a week for a month), respectively. The results showed a remarkable decline in muscle mass and strength after wire-induced immobilization for 14 days. The expression of muscle atrophic factors (Atrogin1 and MuRF1) and inflammatory factors (F4/80 and interleukin-6 (IL-6)) significantly increased (p < 0.001). Notably, muscular AR expression significantly decreased, whereas myostatin and CCAAT/enhancer-binding protein delta (C/EBPδ), a transcriptional activator of myostatin, were significantly elevated (p < 0.05). After castration, AR expression further decreased, and muscular changes with wire-induced immobilization deteriorated. These exacerbations were completely ameliorated by testosterone supplementation and AR upregulation. Our study provides important therapeutic insights into testosterone/AR in muscular atrophy caused by immobilization and shows that muscular AR in a testosterone-dependent manner regulates C/EBPδ/myostatin and inflammation.
Collapse
Affiliation(s)
- Miya Oura
- Department of Geriatric Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Bo-Kyung Son
- Department of Geriatric Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
- Institute for Future Initiatives, Institute of Gerontology, The University of Tokyo, Engineering 8th Building 709, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
- Institute of Gerontology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Zehan Song
- Department of Geriatric Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Koichi Toyoshima
- Department of Geriatric Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Michiko Nanao-Hamai
- Department of Geriatric Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Sumito Ogawa
- Department of Geriatric Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masahiro Akishita
- Department of Geriatric Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
4
|
Dong M, Maturana AD. Effects of aging on calcium channels in skeletal muscle. Front Mol Biosci 2025; 12:1558456. [PMID: 40177518 PMCID: PMC11961898 DOI: 10.3389/fmolb.2025.1558456] [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: 01/10/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
In skeletal muscle, calcium is not only essential to stimulate and sustain their contractions but also for muscle embryogenesis, regeneration, energy production in mitochondria, and fusion. Different ion channels contribute to achieving the various functions of calcium in skeletal muscles. Muscle contraction is initiated by releasing calcium from the sarcoplasmic reticulum through the ryanodine receptor channels gated mechanically by four dihydropyridine receptors of T-tubules. The calcium influx through store-operated calcium channels sustains the contraction and stimulates muscle regeneration. Mitochondrial calcium uniporter allows the calcium entry into mitochondria to stimulate oxidative phosphorylation. Aging alters the expression and activity of these different calcium channels, resulting in a reduction of skeletal muscle force generation and regeneration capacity. Regular physical training and bioactive molecules from nutrients can prevent the effects of aging on calcium channels. This review focuses on the current knowledge of the effects of aging on skeletal muscles' calcium channels.
Collapse
Affiliation(s)
| | - Andrés Daniel Maturana
- Department of Applied Biosciences, Graduate School of Bioagricultural Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
5
|
Zhang ZH, Zhu R, Liu Y, Wang FF, Jiang AY, Dan RC, Liu YH, Tang YX, Liu J, Wang J. IL6-Dependent PIEZO1 Activation Promotes M1-Mediated Orthodontic Root Resorption via CXCL12/CXCR4. J Dent Res 2025:220345251316472. [PMID: 40077814 DOI: 10.1177/00220345251316472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
Orthodontic root resorption (ORR) is a common yet significant complication of orthodontic treatment, largely driven by interactions between periodontal ligament cells (PDLCs) and M1 macrophages. Despite the clinical relevance of ORR, the role of mechanosensitive ion channels in PDLC-mediated ORR and the underlying mechanisms regulating inflammatory cell recruitment remain poorly understood. Here, we identified PIEZO1 as a critical mechanosensitive ion channel that modulates monocyte recruitment and ORR. Using in vivo models treated with the PIEZO1 activator Yoda1 and inhibitor AAV-shPiezo1, we demonstrated that PIEZO1 activation promoted the recruitment of Ly6Chi inflammatory monocytes and exacerbated ORR. In contrast, PIEZO1 inhibition attenuated ORR and the accumulation of M1 macrophages. Mechanistically, PIEZO1 positively regulated the C-X-C motif chemokine 12 (CXCL12) and its receptor, C-X-C chemokine receptor type 4 (CXCR4). Blocking the CXCL12/CXCR4 axis using the CXCR4 antagonist AMD3100 significantly alleviated ORR, reversed M1 macrophage accumulation, and mitigated the recruitment of CD11b+Ly6Chi monocytes. Transwell migration assays with application of the PIEZO1 activator Yoda1 and PIEZO1 inhibitor GsMTX4 consistently confirmed the PIEZO1/CXCL12/CXCR4 axis as a key driver of PDLC-monocyte interactions. Notably, PIEZO1 overactivation was linked to excessive IL-6 production, and IL-6 deficiency inhibited the activation of PIEZO1 induced by Yoda1, leading to attenuation of ORR, M1 macrophage accumulation, and CXCL12/CXCR4 axis activation. Collectively, these findings reveal PIEZO1 in PDLCs as a pivotal modulator of inflammatory monocyte recruitment via the CXCL12/CXCR4 axis in ORR, with IL-6 playing an essential role in PIEZO1 activation. This study provides new insights into the molecular crosstalk between PDLCs and macrophages, offering potential therapeutic targets for mitigating ORR in orthodontic patients.
Collapse
Affiliation(s)
- Z H Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Lab of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - R Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Lab of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - F F Wang
- Institute of Hematology, Department of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - A Y Jiang
- Lab of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - R C Dan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Lab of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Y H Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y X Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Lab of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - J Liu
- Lab of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - J Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Tan Y, Ye Y, Huang C, Li J, Huang L, Wei X, Liang T, Qin E, Xiong G, Bin Y. Cigarette Smoking Induces Skeletal Muscle Atrophy in Mice by Activated Macrophage-Mediated Pyroptosis. J Inflamm Res 2025; 18:2447-2464. [PMID: 39991657 PMCID: PMC11847447 DOI: 10.2147/jir.s497631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/07/2025] [Indexed: 02/25/2025] Open
Abstract
Objective Skeletal muscle atrophy is a major comorbidity associated with chronic obstructive pulmonary disease caused by exposure to cigarette smoke (CS). CS-activated macrophages and pyroptosis play an important role in skeletal muscle atrophy, but its specific molecular mechanism remains unclear. This study investigated the role and mechanisms of pyroptosis and activated macrophages in CS-induced skeletal muscle atrophy. Methods In the in vivo model, mice were exposed to either CS or air for 24 weeks, and in the in vitro model, C2C12 murine skeletal muscle cells were co-cultured with macrophages in Transwell chambers. Western blotting, real-time PCR, ELISA, and other methods were used to detect pyroptosis-related markers to investigate the mechanism of CSE-activated macrophages on skeletal muscle atrophy and pyroptosis. Results In vivo, CS-induced atrophy of the mouse gastrocnemius muscle was accompanied by increased expression of pyroptosis-related markers, including NLRP3 inflammasome, cleaved Caspase-1, the GSDMD N-terminal domain, and interleukin (IL)-18. In vitro, CS extract (CSE)-activated macrophages mediates pyroptosis of skeletal muscle cells and induces myotube atrophy. Further studies demonstrated that macrophage-derived TNF-α is the initiating factor of skeletal muscle pyroptosis, and this process appears to be mediated through TNF-α activating the TNFR1/NLRP3/caspase-1/GSDMD signaling pathway. Conclusion TNF-α released by CSE-activated macrophages can promote skeletal muscle pyroptosis by activating the TNFR1/NLRP3/Caspase-1/GSDMD signaling pathway, which likely contributes to skeletal muscle atrophy. These findings provide more insight into the mechanisms underlying skeletal muscle atrophy in COPD.
Collapse
Affiliation(s)
- Yufen Tan
- Department of Respiratory and Critical Care Medicine, the second Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Yuanyuan Ye
- Department of Respiratory and Critical Care Medicine, the second Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Cuibi Huang
- Department of Respiratory and Critical Care Medicine, the second Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Jie Li
- Department of Respiratory and Critical Care Medicine, the second Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Lihua Huang
- Department of Respiratory and Critical Care Medicine, the second Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Xinyan Wei
- Department of Respiratory and Critical Care Medicine, the second Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Tong Liang
- Department of Respiratory and Critical Care Medicine, the second Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Enyuan Qin
- Department of Respiratory and Critical Care Medicine, the second Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Guolin Xiong
- Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Yanfei Bin
- Department of Respiratory and Critical Care Medicine, the second Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| |
Collapse
|
7
|
Li D, Cao S, Chen Y, Liu Y, Huo K, Shi Z, Han S, Wang L. Distribution and functional significance of KLF15 in mouse cerebellum. Mol Brain 2025; 18:3. [PMID: 39838470 PMCID: PMC11749119 DOI: 10.1186/s13041-025-01172-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Kruppel-like factor 15 (KLF15), a member of the KLF family, is closely involved in many biological processes. However, the mechanism by which KLF15 regulates neural development is still unclear. Considering the complexity and importance of neural network development, in this study, we investigated the potent regulatory role of KLF15 in neural network development. KLF15 was detected highly expressed in the cerebellum and enriched in Purkinje cells, with a significant increase in KLF15 expression between 15 and 20 days of neural development. Knockdown of KLF15 led to loss of Purkinje cells and impaired motility in mice. Therefore, our study aims to elucidate the relationship between KLF15 and Purkinje cells in mice, may provide a new research idea for the developmental mechanism of the mouse cerebellum.
Collapse
Affiliation(s)
- Dan Li
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Shuijing Cao
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yanrong Chen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yueyan Liu
- Department of Basic Medicine, Department of Clinical Medicine, West Anhui Health Vocational College, Liuan, Anhui, 237000, China
| | - Kugeng Huo
- Cyagen Biosciences (Guangzhou) Inc., Guangzhou, Guangdong, 510663, China
| | - Zhuangqi Shi
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, 830017, China
| | - Shuxin Han
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, 830017, China.
| | - Liecheng Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, China.
| |
Collapse
|
8
|
Tabuchi A, Poole DC, Kano Y. Intracellular Ca 2+ After Eccentric Muscle Contractions: Key Role for Ryanodine Receptors. Exerc Sport Sci Rev 2025; 53:23-30. [PMID: 39262047 DOI: 10.1249/jes.0000000000000348] [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: 09/13/2024]
Abstract
Eccentric contractions (ECC) induce excessive intracellular calcium ion (Ca 2+ ) accumulation and muscle structural damage in localized regions of the muscle fibers. In this investigation, we present the novel hypothesis that the ryanodine receptor (RyR) plays a central role in evoking a Ca 2+ dynamics profile that is markedly distinguishable from other muscle adaptive responses.
Collapse
Affiliation(s)
| | - David C Poole
- Departments of Anatomy and Physiology and Kinesiology, Kansas State University, Manhattan, KS
| | | |
Collapse
|
9
|
Bo T, Fujii J. Primary Roles of Branched Chain Amino Acids (BCAAs) and Their Metabolism in Physiology and Metabolic Disorders. Molecules 2024; 30:56. [PMID: 39795113 PMCID: PMC11721030 DOI: 10.3390/molecules30010056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/25/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Leucine, isoleucine, and valine are collectively known as branched chain amino acids (BCAAs) and are often discussed in the same physiological and pathological situations. The two consecutive initial reactions of BCAA catabolism are catalyzed by the common enzymes referred to as branched chain aminotransferase (BCAT) and branched chain α-keto acid dehydrogenase (BCKDH). BCAT transfers the amino group of BCAAs to 2-ketoglutarate, which results in corresponding branched chain 2-keto acids (BCKAs) and glutamate. BCKDH performs an oxidative decarboxylation of BCKAs, which produces their coenzyme A-conjugates and NADH. BCAT2 in skeletal muscle dominantly catalyzes the transamination of BCAAs. Low BCAT activity in the liver reduces the metabolization of BCAAs, but the abundant presence of BCKDH promotes the metabolism of muscle-derived BCKAs, which leads to the production of glucose and ketone bodies. While mutations in the genes responsible for BCAA catabolism are involved in rare inherited disorders, an aberrant regulation of their enzymatic activities is associated with major metabolic disorders such as diabetes, cardiovascular disease, and cancer. Therefore, an understanding of the regulatory process of metabolic enzymes, as well as the functions of the BCAAs and their metabolites, make a significant contribution to our health.
Collapse
Affiliation(s)
- Tomoki Bo
- Laboratory Animal Center, Institute for Promotion of Medical Science Research, Faculty of Medicine, Yamagata University, Yamagata 990-9585, Japan
| | - Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| |
Collapse
|
10
|
Li X, Wu C, Lu X, Wang L. Predictive models of sarcopenia based on inflammation and pyroptosis-related genes. Front Genet 2024; 15:1491577. [PMID: 39777262 PMCID: PMC11703911 DOI: 10.3389/fgene.2024.1491577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Background Sarcopenia is a prevalent condition associated with aging. Inflammation and pyroptosis significantly contribute to sarcopenia. Methods Two sarcopenia-related datasets (GSE111016 and GSE167186) were obtained from the Gene Expression Omnibus (GEO), followed by batch effect removal post-merger. The "limma" R package was utilized to identify differentially expressed genes (DEGs). Subsequently, LASSO analysis was conducted on inflammation and pyroptosis-related genes (IPRGs), resulting in the identification of six hub IPRGs. A novel skeletal muscle aging model was developed and validated using an independent dataset. Additionally, Gene Ontology (GO) enrichment analysis was performed on DEGs, along with Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and gene set enrichment analysis (GSEA). ssGSEA was employed to assess differences in immune cell proportions between healthy muscle groups in older versus younger adults. The expression levels of the six core IPRGs were quantified via qRT-PCR. Results A total of 44 elderly samples and 68 young healthy samples were analyzed for DEGs. Compared to young healthy muscle tissue, T cell infiltration levels in aged muscle tissue were significantly reduced, while mast cell and monocyte infiltration levels were relatively elevated. A new diagnostic screening model for sarcopenia based on the six IPRGs demonstrated high predictive efficiency (AUC = 0.871). qRT-PCR results indicated that the expression trends of these six IPRGs aligned with those observed in the database. Conclusion Six biomarkers-BTG2, FOXO3, AQP9, GPC3, CYCS, and SCN1B-were identified alongside a diagnostic model that offers a novel approach for early diagnosis of sarcopenia.
Collapse
Affiliation(s)
- Xiaoqing Li
- Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cheng Wu
- Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiang Lu
- Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Li Wang
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
11
|
Hyodo M, Nomura K, Tsutsumi R, Izumi-Mishima Y, Kawaguchi H, Kawakami A, Hara K, Suzuki Y, Shirakawa T, Osawa K, Matsuo M, Sakaue H. Urinary titin as an early biomarker of skeletal muscle proteolysis and atrophy in various catabolic conditions. Biochem Biophys Res Commun 2024; 737:150918. [PMID: 39488086 DOI: 10.1016/j.bbrc.2024.150918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Skeletal muscle atrophy impairs quality of life and increases the risk of disease, but current methods for assessment of muscle mass have several limitations. We here investigated the urinary concentration of a fragment of the muscle protein titin as a potential biomarker for the early detection of skeletal muscle atrophy. Four mouse models with different atrophy pathways were studied: those of cardiotoxin-induced acute muscle injury, cast-induced muscle immobilization, lipopolysaccharide-induced sepsis, and streptozotocin-induced diabetes. In all four models, urinary titin levels increased early, concurrent with or preceding upregulation of the atrophy-related genes for atrogin-1 and MuRF-1. The increase in the urinary titin concentration was thus associated with initial muscle damage and the onset of proteolysis, rather than with late-stage muscle wasting. Our findings suggest that urinary titin is a promising biomarker for detection of the onset of skeletal muscle catabolism and prediction of the subsequent development of atrophy in different catabolic states. Noninvasive measurement of urinary titin may therefore allow the earlier detection of skeletal muscle proteolysis compared with conventional techniques.
Collapse
Affiliation(s)
- Mizusa Hyodo
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kazuhiro Nomura
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan; Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Yuna Izumi-Mishima
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hibiki Kawaguchi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Ayuka Kawakami
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kanako Hara
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yuki Suzuki
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Taku Shirakawa
- Faculty of Health Science, Kobe Tokiwa University, Kobe, Japan
| | - Kayo Osawa
- Faculty of Health Science, Kobe Tokiwa University, Kobe, Japan
| | - Masafumi Matsuo
- Graduate School of Science, Technology, and Innovation, Kobe University, Kobe, Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan; Diabetes Therapeutics and Research Center, University of Tokushima, Tokushima, Japan.
| |
Collapse
|
12
|
Kinsella JA, Debant M, Parsonage G, Morley LC, Bajarwan M, Revill C, Foster R, Beech DJ. Pharmacology of PIEZO1 channels. Br J Pharmacol 2024; 181:4714-4732. [PMID: 39402010 DOI: 10.1111/bph.17351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/15/2024] [Accepted: 08/25/2024] [Indexed: 11/05/2024] Open
Abstract
PIEZO1 is a eukaryotic membrane protein that assembles as trimers to form calcium-permeable, non-selective cation channels with exquisite capabilities for mechanical force sensing and transduction of force into effect in diverse cell types that include blood cells, endothelial cells, epithelial cells, fibroblasts and stem cells and diverse systems that include bone, lymphatics and muscle. The channel has wide-ranging roles and is considered as a target for novel therapeutics in ailments spanning cancers and cardiovascular, dental, gastrointestinal, hepatobiliary, infectious, musculoskeletal, nervous system, ocular, pregnancy, renal, respiratory and urological disorders. The identification of PIEZO1 modulators is in its infancy but useful experimental tools emerged for activating, and to a lesser extent inhibiting, the channels. Elementary structure-activity relationships are known for the Yoda series of small molecule agonists, which show the potential for diverse physicochemical and pharmacological properties. Intriguing effects of Yoda1 include the stimulated removal of excess cerebrospinal fluid. Despite PIEZO1's broad expression, opportunities are suggested for selective positive or negative modulation without intolerable adverse effects. Here we provide a focused, non-systematic, narrative review of progress with this pharmacology and discuss potential future directions for research in the area.
Collapse
Affiliation(s)
- Jacob A Kinsella
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
- School of Chemistry, University of Leeds, Leeds, UK
| | - Marjolaine Debant
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Gregory Parsonage
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Lara C Morley
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Muath Bajarwan
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | | | | | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
13
|
Kitada K. Hypertension research 2024 update and perspectives: basic research. Hypertens Res 2024; 47:3304-3309. [PMID: 39251854 DOI: 10.1038/s41440-024-01878-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024]
Abstract
This review article introduces some basic studies that were recently published in this journal, as a part of Hypertension Research 2024 Update and Perspectives. Including recent basic research trends in other scientific journals, we would like to summarize basic research on keywords such as hypertension and its associated organ damage, new treatments, and others. It is expected that the accumulation of basic studies will lead to breakthroughs in hypertension treatment in the future and lead to the definitive treatment of hypertension beyond blood pressure control with anti-hypertensive drugs.
Collapse
Affiliation(s)
- Kento Kitada
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, 7610793, Japan.
| |
Collapse
|
14
|
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
|
15
|
Xiao B. Mechanisms of mechanotransduction and physiological roles of PIEZO channels. Nat Rev Mol Cell Biol 2024; 25:886-903. [PMID: 39251883 DOI: 10.1038/s41580-024-00773-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/11/2024]
Abstract
Mechanical force is an essential physical element that contributes to the formation and function of life. The discovery of the evolutionarily conserved PIEZO family, including PIEZO1 and PIEZO2 in mammals, as bona fide mechanically activated cation channels has transformed our understanding of how mechanical forces are sensed and transduced into biological activities. In this Review, I discuss recent structure-function studies that have illustrated how PIEZO1 and PIEZO2 adopt their unique structural design and curvature-based gating dynamics, enabling their function as dedicated mechanotransduction channels with high mechanosensitivity and selective cation conductivity. I also discuss our current understanding of the physiological and pathophysiological roles mediated by PIEZO channels, including PIEZO1-dependent regulation of development and functional homeostasis and PIEZO2-dominated mechanosensation of touch, tactile pain, proprioception and interoception of mechanical states of internal organs. Despite the remarkable progress in PIEZO research, this Review also highlights outstanding questions in the field.
Collapse
Affiliation(s)
- Bailong Xiao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
- Beijing Frontier Research Center of Biological Structure, Tsinghua University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
16
|
Ni K, Che B, Gu R, Wang C, Pan Y, Li J, Liu L, Luo M, Deng L. Single-Cell Hypertrophy Promotes Contractile Function of Cultured Human Airway Smooth Muscle Cells via Piezo1 and YAP Auto-Regulation. Cells 2024; 13:1697. [PMID: 39451215 PMCID: PMC11505810 DOI: 10.3390/cells13201697] [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: 08/13/2024] [Revised: 09/30/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
Severe asthma is characterized by increased cell volume (hypertrophy) and enhanced contractile function (hyperresponsiveness) of the airway smooth muscle cells (ASMCs). The causative relationship and underlying regulatory mechanisms between them, however, have remained unclear. Here, we manipulated the single-cell volume of in vitro cultured human ASMCs to increase from 2.7 to 5.2 and 8.2 × 103 μm3 as a simulated ASMC hypertrophy by culturing the cells on micropatterned rectangular substrates with a width of 25 μm and length from 50 to 100 and 200 μm, respectively. We found that as the cell volume increased, ASMCs exhibited a pro-contractile function with increased mRNA expression of contractile proteins, increased cell stiffness and traction force, and enhanced response to contractile stimulation. We also uncovered a concomitant increase in membrane tension and Piezo1 mRNA expression with increasing cell volume. Perhaps more importantly, we found that the enhanced contractile function due to cell volume increase was largely attenuated when membrane tension and Piezo1 mRNA expression were downregulated, and an auto-regulatory loop between Piezo1 and YAP mRNA expression was also involved in perpetuating the contractile function. These findings, thus, provide convincing evidence of a direct link between hypertrophy and enhanced contractile function of ASMCs that was mediated via Piezo1 mRNA expression, which may be specifically targeted as a novel therapeutic strategy to treat pulmonary diseases associated with ASMC hypertrophy such as severe asthma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mingzhi Luo
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| |
Collapse
|
17
|
Isaji M, Horiuchi K, Kondo S, Nakagawa T, Ishizaka T, Amako M, Chiba K. Suppression of TNF-α activity by immobilization rescues Mkx expression and attenuates tendon ossification in a mouse Achilles tenotomy model. J Orthop Res 2024; 42:2140-2148. [PMID: 38806292 DOI: 10.1002/jor.25906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/20/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
Traumatic heterotopic ossification is a condition in which extraskeletal bone formation occurs in soft tissues after injury. It most commonly occurs in patients who had major orthopedic surgery and in those with severe extremity injuries. The lesion causes local pain and can impair motor function of the affected limb, but there is currently no established prophylaxis or treatment for this condition. In this study, we show that immobilization at an early stage of the inflammatory response after injury can attenuate ossification formation in a murine Achilles tenotomy model. Gene expression analysis revealed a decrease in the expression of Tnf and an increase in the expression of Mkx, which encodes one of the master regulators of tendon differentiation, Mohawk. Notably, we found that TNF-α suppressed the expression of Mkx transcripts and accelerated the osteogenic differentiation of tendon-derived mesenchymal stem cells (MSCs), suggesting that TNF-α acts as a negative regulator of Mkx transcription. Consistent with these findings, pharmaceutical inhibition of TNF-α increased the expression of Mkx transcripts and suppressed bone formation in this mouse model. These findings reveal the previously unrecognized involvement of TNF-α in regulating tendon MSC fate through suppression of Mkx expression and suggest that TNF-α is a potential target for preventing traumatic heterotopic ossification.
Collapse
Affiliation(s)
- Masashi Isaji
- Department of Orthopedic Surgery, National Defense Medical College, Saitama, Japan
| | - Keisuke Horiuchi
- Department of Orthopedic Surgery, National Defense Medical College, Saitama, Japan
| | - Shinya Kondo
- Department of Orthopedic Surgery, National Defense Medical College, Saitama, Japan
| | - Takahiro Nakagawa
- Department of Orthopedic Surgery, National Defense Medical College, Saitama, Japan
| | - Takahiro Ishizaka
- Department of Orthopedic Surgery, National Defense Medical College, Saitama, Japan
| | - Masatoshi Amako
- Department of Rehabilitation Medicine, National Defense Medical College Hospital, Saitama, Japan
| | - Kazuhiro Chiba
- Department of Orthopedic Surgery, National Defense Medical College, Saitama, Japan
| |
Collapse
|
18
|
Huang L, Jiao Y, Xia H, Li H, Yu J, Que Y, Zeng Z, Fan C, Wang C, Yang C, Chang J. Strontium zinc silicate simultaneously alleviates osteoporosis and sarcopenia in tail-suspended rats via Piezo1-mediated Ca 2+ signaling. J Orthop Translat 2024; 48:146-155. [PMID: 39229332 PMCID: PMC11369381 DOI: 10.1016/j.jot.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024] Open
Abstract
Background Long-term physical inactivity probably leads to a co-existence of osteoporosis and sarcopenia which result in a high risk of falls, fractures, disability and even mortality. However, universally applicable and feasible approaches are lacking in the concurrent treatment of osteoporosis and sarcopenia. In this study, we evaluated the effect of strontium zinc silicate bioceramic (SZS) extract on osteoporosis and sarcopenia and explored its underlying mechanisms. Methods Hindlimb osteoporosis and sarcopenia were established in a tail-suspended rat model. The bones were conducted μCT scanning, histological examination, and gene expression analysis, and the muscles were conducted histological examination and gene expression analysis. In vitro, the effect of SZS extract on osteoblasts was determined by alizarin red S staining, immunofluorescence and qPCR. Similarly, the effect of SZS extract on myoblasts was determined by immunofluorescence and qPCR.. At last, the role of Piezo1 and the change of intracellular calcium ion (Ca2+) were explored through blockading the Piezo1 by GsMTx4 in MC3T3-E1 and C2C12 cells, respectively. Results We found that SZS extract could concurrently and efficiently prevent bone structure deterioration, muscle atrophy and fibrosis in hind limbs of the tail-suspended rats. The in vivo study also showed that SZS extract could upregulate the mRNA expression of Piezo1, thereby maintaining the homeostasis of bones and muscles. In vitro study demonstrated that SZS extract could promote the proliferation and differentiation of MC3T3-E1 and C2C12 cells by increasing the intracellular Ca2+ in a Piezo1-dependent manner. Conclusion This study demonstrated that SZS extract could increase Piezo1-mediated intracellular Ca2+, and facilitate osteogenic differentiation of osteoblast and myogenic differentiation of myoblasts, contributing to alleviation of osteoporosis and sarcopenia in a tail-suspended rat model. The translational potential of this article The current study might provide a universally applicable and efficient strategy to treat musculoskeletal disorders based on bioactive ceramics. The verification of the role of Piezo1-modulated intracellular Ca2+ during osteogenesis and myogenesis provided a possible therapeutic target against mechanical related diseases.
Collapse
Affiliation(s)
- Lingwei Huang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
- College of Materials Science and Opto-electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiren Jiao
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Hangbin Xia
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, China
| | - Huili Li
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jing Yu
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Yumei Que
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Zhen Zeng
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
- College of Materials Science and Opto-electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chen Fan
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Chen Wang
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Chen Yang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Jiang Chang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| |
Collapse
|
19
|
Zhaoyu L, Xiaomeng Y, Na L, Jiamin S, Guanhua D, Xiuying Y. Roles of natural products on myokine expression and secretion in skeletal muscle atrophy. Gen Comp Endocrinol 2024; 355:114550. [PMID: 38768928 DOI: 10.1016/j.ygcen.2024.114550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Skeletal muscles serve both in movement and as endocrine organs. Myokines secreted by skeletal muscles activate biological functions within muscles and throughout the body via autocrine, paracrine, and/or endocrine pathways. Skeletal muscle atrophy can influence myokine expression and secretion, while myokines can impact the structure and function of skeletal muscles. Regulating the expression and secretion of myokines through the pharmacological approach is a strategy for alleviating skeletal muscle atrophy. Natural products possess complex structures and chemical properties. Previous studies have demonstrated that various natural products exert beneficial effects on skeletal muscle atrophy. This article reviewed the regulatory effects of natural products on myokines and summarized the research progress on skeletal muscle atrophy associated with myokine regulation. The focus is on how small-molecule natural products affect the regulation of interleukin 6 (IL-6), irisin, myostatin, IGF-1, and FGF-21 expression. We contend that the development of small-molecule natural products targeting the regulation of myokines holds promise in combating skeletal muscle atrophy.
Collapse
Affiliation(s)
- Liu Zhaoyu
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Ye Xiaomeng
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Li Na
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Shang Jiamin
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Du Guanhua
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| | - Yang Xiuying
- Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
20
|
Kominami K, Akino M, Kanai M. Efficacy of Neuro-muscular Electrical Stimulation for Orthostatic Hypotension Associated with Long-term Disuse and Diabetic Autonomic Neuropathy: A Case Report. Phys Ther Res 2024; 27:180-185. [PMID: 39866392 PMCID: PMC11756561 DOI: 10.1298/ptr.e10298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/16/2024] [Indexed: 01/28/2025]
Abstract
Patient Background: A 75-year-old man had difficulty moving around at home because of loss of appetite and neglect of medication for several days. He was brought to the emergency room and admitted on the same day with a diagnosis of dehydration and diabetic ketoacidosis. He started physical therapy (PT), had frequent fainting and presyncope due to hypotension, and had difficulty leaving bed. The patient was transferred to our hospital to continue PT. Test results on admission were as follows: short physical performance battery (SPPB) [points], 1/12 points; chair stand 5 times (CS5) [sec], not possible; functional independent measure (FIM) [points], 66/126; standing test: blood pressure (BP) [mmHg], 130/60/HR [beats per minute], 76 in supine, 90/56/79 in sitting, 70/-/79 in standing. PROCESS After transfer, BP continued to fall markedly and he frequently fainted and required assistance with nearly all activities of daily living (ADL). Neuromuscular electrical stimulation (NMES) of the thigh and lower leg was performed five times a week for 30 min. After approximately 3 days of NMES, BP decreased slowly, presyncopic symptoms disappeared, and he could leave bed more frequently and for longer periods. The patient became independent in ADL and was discharged on Day 142. Results at discharge were as follows: SPPB, 11/12; CS5, 13.5; FIM, 114/126. DISCUSSION Although NMES is not effective for orthostatic hypotension (OH) associated with diabetic autonomic neuropathy (DAN), stabilization of BP early after the introduction of NMES may have been due to its peripheral sympathetic nerve-stimulating effect. CONCLUSION The combination of exercise therapy and NMES for OH caused by DAN can alleviate hypotension.
Collapse
Affiliation(s)
| | - Masatoshi Akino
- Department of Internal Medicine, Sapporo Kiyota Hospital, Japan
| | - Motoshi Kanai
- Department of Internal Medicine, Sanseikai Kitano Hospital, Japan
| |
Collapse
|
21
|
Franco-Obregón A, Tai YK. Are Aminoglycoside Antibiotics TRPing Your Metabolic Switches? Cells 2024; 13:1273. [PMID: 39120305 PMCID: PMC11311832 DOI: 10.3390/cells13151273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024] Open
Abstract
Transient receptor potential (TRP) channels are broadly implicated in the developmental programs of most tissues. Amongst these tissues, skeletal muscle and adipose are noteworthy for being essential in establishing systemic metabolic balance. TRP channels respond to environmental stimuli by supplying intracellular calcium that instigates enzymatic cascades of developmental consequence and often impinge on mitochondrial function and biogenesis. Critically, aminoglycoside antibiotics (AGAs) have been shown to block the capacity of TRP channels to conduct calcium entry into the cell in response to a wide range of developmental stimuli of a biophysical nature, including mechanical, electromagnetic, thermal, and chemical. Paradoxically, in vitro paradigms commonly used to understand organismal muscle and adipose development may have been led astray by the conventional use of streptomycin, an AGA, to help prevent bacterial contamination. Accordingly, streptomycin has been shown to disrupt both in vitro and in vivo myogenesis, as well as the phenotypic switch of white adipose into beige thermogenic status. In vivo, streptomycin has been shown to disrupt TRP-mediated calcium-dependent exercise adaptations of importance to systemic metabolism. Alternatively, streptomycin has also been used to curb detrimental levels of calcium leakage into dystrophic skeletal muscle through aberrantly gated TRPC1 channels that have been shown to be involved in the etiology of X-linked muscular dystrophies. TRP channels susceptible to AGA antagonism are critically involved in modulating the development of muscle and adipose tissues that, if administered to behaving animals, may translate to systemwide metabolic disruption. Regenerative medicine and clinical communities need to be made aware of this caveat of AGA usage and seek viable alternatives, to prevent contamination or infection in in vitro and in vivo paradigms, respectively.
Collapse
Affiliation(s)
- Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zürich, 8057 Zürich, Switzerland
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
22
|
Chen ZT, Weng ZX, Lin JD, Meng ZX. Myokines: metabolic regulation in obesity and type 2 diabetes. LIFE METABOLISM 2024; 3:loae006. [PMID: 39872377 PMCID: PMC11749576 DOI: 10.1093/lifemeta/loae006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 01/30/2025]
Abstract
Skeletal muscle plays a vital role in the regulation of systemic metabolism, partly through its secretion of endocrine factors which are collectively known as myokines. Altered myokine levels are associated with metabolic diseases, such as type 2 diabetes (T2D). The significance of interorgan crosstalk, particularly through myokines, has emerged as a fundamental aspect of nutrient and energy homeostasis. However, a comprehensive understanding of myokine biology in the setting of obesity and T2D remains a major challenge. In this review, we discuss the regulation and biological functions of key myokines that have been extensively studied during the past two decades, namely interleukin 6 (IL-6), irisin, myostatin (MSTN), growth differentiation factor 11 (GDF11), fibroblast growth factor 21 (FGF21), apelin, brain-derived neurotrophic factor (BDNF), meteorin-like (Metrnl), secreted protein acidic and rich in cysteine (SPARC), β-aminoisobutyric acid (BAIBA), Musclin, and Dickkopf 3 (Dkk3). Related to these, we detail the role of exercise in myokine expression and secretion together with their contributions to metabolic physiology and disease. Despite significant advancements in myokine research, many myokines remain challenging to measure accurately and investigate thoroughly. Hence, new research techniques and detection methods should be developed and rigorously tested. Therefore, developing a comprehensive perspective on myokine biology is crucial, as this will likely offer new insights into the pathophysiological mechanisms underlying obesity and T2D and may reveal novel targets for therapeutic interventions.
Collapse
Affiliation(s)
- Zhi-Tian Chen
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute (ZJE), School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China
| | - Zhi-Xuan Weng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of Geriatrics, Affiliated Hangzhou First People’s Hospital, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
23
|
Akieda-Asai S, Ma H, Han W, Nagata J, Yamaguchi F, Date Y. Mechanism of muscle atrophy in a normal-weight rat model of type 2 diabetes established by using a soft-pellet diet. Sci Rep 2024; 14:7670. [PMID: 38561446 PMCID: PMC10984920 DOI: 10.1038/s41598-024-57727-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Dietary factors such as food texture affect feeding behavior and energy metabolism, potentially causing obesity and type 2 diabetes. We previously found that rats fed soft pellets (SPs) were neither hyperphagic nor overweight but demonstrated glucose intolerance, insulin resistance, and hyperplasia of pancreatic β-cells. In the present study, we investigated the mechanism of muscle atrophy in rats that had been fed SPs on a 3-h time-restricted feeding schedule for 24 weeks. As expected, the SP rats were normal weight; however, they developed insulin resistance, glucose intolerance, and fat accumulation. In addition, skeletal muscles of SP rats were histologically atrophic and demonstrated disrupted insulin signaling. Furthermore, we learned that the muscle atrophy of the SP rats developed via the IL-6-STAT3-SOCS3 and ubiquitin-proteasome pathways. Our data show that the dietary habit of consuming soft foods can lead to not only glucose intolerance or insulin resistance but also muscle atrophy.
Collapse
Affiliation(s)
- Sayaka Akieda-Asai
- Frontier Science Research Center, University of Miyazaki, Miyazaki, 889-1692, Japan.
| | - Hao Ma
- Frontier Science Research Center, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Wanxin Han
- Frontier Science Research Center, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Junko Nagata
- Department of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Fumitake Yamaguchi
- Frontier Science Research Center, University of Miyazaki, Miyazaki, 889-1692, Japan
- Department of Nursing, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Yukari Date
- Frontier Science Research Center, University of Miyazaki, Miyazaki, 889-1692, Japan.
| |
Collapse
|
24
|
Wood N, Straw S, Cheng CW, Hirata Y, Pereira MG, Gallagher H, Egginton S, Ogawa W, Wheatcroft SB, Witte KK, Roberts LD, Bowen TS. Sodium-glucose cotransporter 2 inhibitors influence skeletal muscle pathology in patients with heart failure and reduced ejection fraction. Eur J Heart Fail 2024; 26:925-935. [PMID: 38468429 DOI: 10.1002/ejhf.3192] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/12/2024] [Accepted: 02/23/2024] [Indexed: 03/13/2024] Open
Abstract
AIMS Patients with heart failure and reduced ejection fraction (HFrEF) exhibit skeletal muscle pathology, which contributes to symptoms and decreased quality of life. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) improve clinical outcomes in HFrEF but their mechanism of action remains poorly understood. We aimed, therefore, to determine whether SGLT2i influence skeletal muscle pathology in patients with HFrEF. METHODS AND RESULTS Muscle biopsies from 28 male patients with HFrEF (New York Heart association class I-III) treated with SGLT2i (>12 months) or without SGLT2i were compared. Comprehensive analyses of muscle structure (immunohistochemistry), transcriptome (RNA sequencing), and metabolome (liquid chromatography-mass spectrometry) were performed, and serum inflammatory profiling (ELISA). Experiments in mice (n = 16) treated with SGLT2i were also performed. Myofiber atrophy was ~20% less in patients taking SGLT2i (p = 0.07). Transcriptomics and follow-up measures identified a unique signature in patients taking SGLT2i related to beneficial effects on atrophy, metabolism, and inflammation. Metabolomics identified influenced tryptophan metabolism in patients taking SGLT2i: kynurenic acid was 24% higher and kynurenine was 32% lower (p < 0.001). Serum profiling identified that SGLT2i treatment was associated with lower (p < 0.05) pro-inflammatory cytokines by 26-64% alongside downstream muscle interleukin (IL)-6-JAK/STAT3 signalling (p = 008 and 0.09). Serum IL-6 and muscle kynurenine were correlated (R = 0.65; p < 0.05). Muscle pathology was lower in mice treated with SGLT2i indicative of a conserved mammalian response to treatment. CONCLUSIONS Treatment with SGLT2i influenced skeletal muscle pathology in patients with HFrEF and was associated with anti-atrophic, anti-inflammatory, and pro-metabolic effects. These changes may be regulated via IL-6-kynurenine signalling. Together, clinical improvements following SGLT2i treatment in patients with HFrEF may be partly explained by their positive effects on skeletal muscle pathology.
Collapse
Affiliation(s)
- Nathanael Wood
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Sam Straw
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Chew W Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Yu Hirata
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Marcelo G Pereira
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Harrison Gallagher
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Stuart Egginton
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Klaus K Witte
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Clinic for Cardiology, Angiology and Internal Intensive Care Medicine, RWTH Aachen University, Aachen, Germany
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
25
|
Nguyen J, Gilbert PM. Decoding the forces that shape muscle stem cell function. Curr Top Dev Biol 2024; 158:279-306. [PMID: 38670710 DOI: 10.1016/bs.ctdb.2024.02.009] [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] [Indexed: 04/28/2024]
Abstract
Skeletal muscle is a force-producing organ composed of muscle tissues, connective tissues, blood vessels, and nerves, all working in synergy to enable movement and provide support to the body. While robust biomechanical descriptions of skeletal muscle force production at the body or tissue level exist, little is known about force application on microstructures within the muscles, such as cells. Among various cell types, skeletal muscle stem cells reside in the muscle tissue environment and play a crucial role in driving the self-repair process when muscle damage occurs. Early evidence indicates that the fate and function of skeletal muscle stem cells are controlled by both biophysical and biochemical factors in their microenvironments, but much remains to accomplish in quantitatively describing the biophysical muscle stem cell microenvironment. This book chapter aims to review current knowledge on the influence of biophysical stresses and landscape properties on muscle stem cells in heath, aging, and diseases.
Collapse
Affiliation(s)
- Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
26
|
Zhang X, Liu J, Deng X, Bo L. Understanding COVID-19-associated endothelial dysfunction: role of PIEZO1 as a potential therapeutic target. Front Immunol 2024; 15:1281263. [PMID: 38487535 PMCID: PMC10937424 DOI: 10.3389/fimmu.2024.1281263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Due to its high infectivity, the pandemic has rapidly spread and become a global health crisis. Emerging evidence indicates that endothelial dysfunction may play a central role in the multiorgan injuries associated with COVID-19. Therefore, there is an urgent need to discover and validate novel therapeutic strategies targeting endothelial cells. PIEZO1, a mechanosensitive (MS) ion channel highly expressed in the blood vessels of various tissues, has garnered increasing attention for its potential involvement in the regulation of inflammation, thrombosis, and endothelial integrity. This review aims to provide a novel perspective on the potential role of PIEZO1 as a promising target for mitigating COVID-19-associated endothelial dysfunction.
Collapse
Affiliation(s)
| | | | - Xiaoming Deng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lulong Bo
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
27
|
Xu X, Guo Y, Liu P, Zhang H, Wang Y, Li Z, Mei Y, Niu L, Liu R. Piezo Mediates the Mechanosensation and Injury-Repair of Pulpo-Dentinal Complex. Int Dent J 2024; 74:71-80. [PMID: 37833209 PMCID: PMC10829354 DOI: 10.1016/j.identj.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 10/15/2023] Open
Abstract
OBJECTIVES The aim of this research was to investigate the functions of Piezo channels in dentin defect, including mechanical signalling and odontoblast responses. METHODS Rat dentin-defect models were constructed, and spatiotemporal expression of Piezo proteins was detected in the pulpo-dentinal complex. Real-time polymerase chain reaction (rtPCR) was used to investigate the functional expression pattern of Piezo channels in odontoblasts. Moreover, RNA interference technology was employed to uncover the underlying mechanisms of the Piezo-driven inflammatory response and repair under fluid shear stress (FSS) conditions in vitro. RESULTS Piezo1 and Piezo2 were found to be widely expressed in the odontoblast layer and dental pulp in the rat dentin-defect model during the end stage of reparative dentin formation. The expression levels of the Piezo1 and Piezo2 genes in MDPC-23 cells were high in the initial stage under FSS loading and then decreased over time. Moreover, the expression trends of inflammatory, odontogenic, and mineralisation genes were generally contrary to those of Piezo1 and Piezo2 over time. After silencing of Piezo1/Piezo2, FSS stimulation resulted in significantly higher expression of inflammatory, odontogenesis, and mineralisation genes in MDPC-23 cells. Finally, the expression of genes involved in the integrin β1/ERK1 and Wnt5b/β-catenin signalling pathways was changed in response to RNA silencing of Piezo1 and Piezo2. CONCLUSIONS Piezo1 and Piezo2 may be involved in regulating the expression of inflammatory and odontogenic genes in odontoblasts stimulated by FSS.
Collapse
Affiliation(s)
- Xiaoqiao Xu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Yi Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Peiqi Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Hui Zhang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, China
| | - Yijie Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Zhen Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Yukun Mei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Lin Niu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China; Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
| | - Ruirui Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China; Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
28
|
He J, Cheng X, Fang B, Shan S, Li Q. Mechanical stiffness promotes skin fibrosis via Piezo1-Wnt2/Wnt11-CCL24 positive feedback loop. Cell Death Dis 2024; 15:84. [PMID: 38267432 PMCID: PMC10808102 DOI: 10.1038/s41419-024-06466-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 01/26/2024]
Abstract
Skin fibrosis is characterized by the excessive accumulation of extracellular matrix (ECM) caused by fibrotic disorders of the skin. In recent years, ECM stiffness has emerged as a prominent mechanical cue that precedes skin fibrosis and drives its progression by promoting fibroblasts activation. However, how stiffness influences fibroblasts activation for skin fibrosis progression remains unknown. Here, we report a positive feedback loop mediated by the mechanosensitive ion channel Piezo1 and aberrant tissue mechanics in driving skin fibrosis. Piezo1 is upregulated in fibrotic skin in both humans and mice. Piezo1 knockdown dermal fibroblasts lose their fibroproliferative phenotypes despite being grown on a stiffer substrate. We show that Piezo1 acts through the Wnt2/Wnt11 pathway to mechanically induce secretion of C-C motif chemokine ligand 24 (CCL24, also known as eotaxin-2), a potent cytokine associated with fibrotic disorders. Importantly, adeno-associated virus (AAV)-mediated Piezo1 knockdown ameliorated the progression of skin fibrosis and skin stiffness in mice. Overall, increased matrix stiffness promotes skin fibrosis through the inflammatory Piezo1-Wnt2/Wnt11-CCL24 pathway. In turn, a stiffer skin microenvironment increases Piezo1 expression to exacerbate skin fibrosis aggression. Therefore, targeting Piezo1 represents a strategy to break the positive feedback loop between fibroblasts mechanotransduction and aberrant tissue mechanics in skin fibrosis.
Collapse
Affiliation(s)
- Jiahao He
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
| | - Xinwei Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
| | - Bin Fang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China.
| | - Shengzhou Shan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China.
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China.
| |
Collapse
|
29
|
Tan R, Zhang K, Si Y, Zhang S, Yang J, Hu J. Implantable Epigallocatechin Gallate Sustained-Release Nanofibers for the Prevention of Immobilization-Induced Muscle Atrophy. ACS NANO 2024; 18:919-930. [PMID: 38142426 DOI: 10.1021/acsnano.3c09634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
Long-term immobilization of joints can lead to disuse atrophy of the muscles in the joints. Oral nutrients are used clinically for rehabilitation and therapeutic purposes, but bioavailability and targeting are limited. Here, we report tea polyphenols (dietary polyphenols), sustained-release nanofilms that release tea polyphenols through slow local degradation of core-shell nanofibers in muscles. This dietary polyphenol does not require gastrointestinal consumption and multiple doses and can directly remove inflammatory factors and superoxide generated in muscle tissue during joint fixation. The quality of muscles is increased by 30%, and muscle movement function is effectively improved. Although nanofibers need to be implanted into muscles, they can improve bacterial infections after joint surgery. To investigate the biological mechanism of this core-shell nanomembrane prevention, we conducted further transcriptomic studies on muscle, confirming that in addition to achieving antioxidation and anti-inflammation by inhibiting TNF-α and NF-κB signaling pathways, tea polyphenol core-shell nanofibers can also promote muscle formation by activating the p-Akt signaling pathway.
Collapse
Affiliation(s)
- Renjie Tan
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Ke Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Yifan Si
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Shuai Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Jieqiong Yang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Jinlian Hu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| |
Collapse
|
30
|
Rutledge CA. Molecular mechanisms underlying sarcopenia in heart failure. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:7. [PMID: 38455513 PMCID: PMC10919908 DOI: 10.20517/jca.2023.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
The loss of skeletal muscle, also known as sarcopenia, is an aging-associated muscle disorder that is disproportionately present in heart failure (HF) patients. HF patients with sarcopenia have poor outcomes compared to the overall HF patient population. The prevalence of sarcopenia in HF is only expected to grow as the global population ages, and novel treatment strategies are needed to improve outcomes in this cohort. Multiple mechanistic pathways have emerged that may explain the increased prevalence of sarcopenia in the HF population, and a better understanding of these pathways may lead to the development of therapies to prevent muscle loss. This review article aims to explore the molecular mechanisms linking sarcopenia and HF, and to discuss treatment strategies aimed at addressing such molecular signals.
Collapse
Affiliation(s)
- Cody A. Rutledge
- Acute Medicine Section, Division of Medicine, Louis Stokes Cleveland Veteran Affairs Medical Center, Cleveland, OH 44106, USA
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
31
|
Gan D, Jin X, Wang X, Tao C, Yan Q, Jia Q, Huo S, Chen D, Yao Q, Xiao G. Pathological progress and remission strategies of osteoarthritic lesions caused by long-term joint immobilization. Arthritis Res Ther 2023; 25:237. [PMID: 38062473 PMCID: PMC10702075 DOI: 10.1186/s13075-023-03223-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
OBJECTIVE While joint immobilization is a useful repair method for intra-articular ligament injury and periarticular fracture, prolonged joint immobilization can cause multiple complications. A better understanding how joint immobilization and remobilization impact joint function and homeostasis will help clinicians develop novel strategies to reduce complications. DESIGN We first determined the effects of long-term immobilization on joint pain and osteophyte formation in patients after an extraarticular fracture or ligament injury. We then developed a mouse model of joint immobilization and harvested the knee joint samples at 2, 4, and 8 weeks. We further determined the effects of remobilization on recovery of the osteoarthritis (OA) lesions induced by immobilization in mice. RESULTS We found that the long-term (6 weeks) joint immobilization caused significant joint pain and osteophytes in patients. In mice, 2-week immobilization already induced moderate sensory innervation and increased pain sensitivity and infiltration in synovium without inducing marked osteophyte formation and cartilage loss. Long-term immobilization (4 and 8 weeks) induced more severe sensory innervation and inflammatory infiltration in synovium, massive osteophyte formation on both sides of the femoral condyle, and the edge of the tibial plateau and significant loss of the articular cartilage in mice. Remobilization, which ameliorates normal joint load and activity, restored to certain extent some of the OA lesions and joint function in mice. CONCLUSIONS Joint immobilization caused multiple OA-like lesions in both mice and humans. Joint immobilization induced progressive sensory innervation, synovitis, osteophyte formation, and cartilage loss in mice, which can be partially ameliorated by remobilization.
Collapse
Affiliation(s)
- Donghao Gan
- Department of Biochemistry, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xiaowan Jin
- Department of Biochemistry, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xiangpeng Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chu Tao
- Department of Biochemistry, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Qinnan Yan
- Department of Biochemistry, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Qingyun Jia
- Department of Orthopedics, Linyi People's Hospital, Linyi, China
| | - Shaochuan Huo
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Di Chen
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qing Yao
- Department of Biochemistry, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| | - Guozhi Xiao
- Department of Biochemistry, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
32
|
Ieko T, Fujiki J, Hasegawa Y, Iwasaki T, Iwano H, Maeda N. Mechanism of skeletal muscle atrophy by muscle fiber types in male rats under long-term fasting stress. Steroids 2023; 200:109328. [PMID: 37863411 DOI: 10.1016/j.steroids.2023.109328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/04/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
Fasting induces metabolic changes in muscles, which are differentiated by muscle fiber type. In this study, the mechanism of fasting-induced muscle atrophy in rats was examined to determine the differences between muscle fiber types in energy production. Fasting for 96 h did not alter the weight of the soleus (SOL), a fiber type I muscle, but did significantly reduce the weight of gastrocnemius (GM), a fiber type II muscle. GM, SOL and blood pregnenolone and testosterone levels decreased under fasting, which induced energy deprivation, whereas corticosterone (CORT) levels significantly increased. However, the expression of 3β-HSD and P45011β in GM was unaffected by fasting. The decrease in GM weight may be due to decreased levels of testosterone and reduced synthesis of mammalian target of rapamycin (mTOR). Significant increases in CORT both GM and SOL were associated with increases in the amount of branched-chain amino acids available for energy production. However, decreased levels of mTOR and IGF1 and increased levels of CORT and IL-6 in SOL suggest that GM proteolysis was followed by SOL proteolysis for additional energy production. In conclusion, IGF1 levels decreased significantly in SOL, whereas those of IL-6 significantly increased in SOL and blood but decreased in GM. Blood branched-chain amino acids (BCAA) levels were unaffected due to fasting, whereas an increase was noted in the levels of BCAA in GM and SOL. These results show that fasting for 96 h restricts energy supply, producing fast-twitch muscle atrophy followed by slow-twitch muscle atrophy.
Collapse
Affiliation(s)
- Takahiro Ieko
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido 069-8501, Japan
| | - Jumpei Fujiki
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido 069-8501, Japan
| | - Yasuhiro Hasegawa
- Laboratory of Meat Science, Department of Food Science and Human Wellness, Rakuno Gakuen University, Hokkaido 069-8501, Japan
| | - Tomohito Iwasaki
- Laboratory of Meat Science, Department of Food Science and Human Wellness, Rakuno Gakuen University, Hokkaido 069-8501, Japan
| | - Hidetomo Iwano
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido 069-8501, Japan
| | - Naoyuki Maeda
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido 069-8501, Japan; Laboratory of Meat Science, Department of Food Science and Human Wellness, Rakuno Gakuen University, Hokkaido 069-8501, Japan.
| |
Collapse
|
33
|
Zeineddine Y, Friedman MA, Buettmann EG, Abraham LB, Hoppock GA, Donahue HJ. Genetic diversity modulates the physical and transcriptomic response of skeletal muscle to simulated microgravity in male mice. NPJ Microgravity 2023; 9:86. [PMID: 38040743 PMCID: PMC10692100 DOI: 10.1038/s41526-023-00334-8] [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: 05/17/2023] [Accepted: 11/09/2023] [Indexed: 12/03/2023] Open
Abstract
Developments in long-term space exploration necessitate advancements in countermeasures against microgravity-induced skeletal muscle loss. Astronaut data shows considerable variation in muscle loss in response to microgravity. Previous experiments suggest that genetic background influences the skeletal muscle response to unloading, but no in-depth analysis of genetic expression has been performed. Here, we placed eight, male, inbred founder strains of the diversity outbred mice (129S1/SvImJ, A/J, C57BL/6J, CAST/EiJ, NOD/ShiLtJ, NZO/HILtJ, PWK/PhJ, and WSB/EiJ) in simulated microgravity (SM) via hindlimb unloading for three weeks. Body weight, muscle morphology, muscle strength, protein synthesis marker expression, and RNA expression were collected. A/J and CAST/EiJ mice were most susceptible to SM-induced muscle loss, whereas NOD/ShiLtJ mice were the most protected. In response to SM, A/J and CAST/EiJ mice experienced reductions in body weight, muscle mass, muscle volume, and muscle cross-sectional area. A/J mice had the highest number of differentially expressed genes (68) and associated gene ontologies (328). Downregulation of immunological gene ontologies and genes encoding anabolic immune factors suggest that immune dysregulation contributes to the response of A/J mice to SM. Several muscle properties showed significant interactions between SM and mouse strain and a high degree of heritability. These data imply that genetic background plays a role in the degree of muscle loss in SM and that more individualized programs should be developed for astronauts to protect their skeletal muscles against microgravity on long-term missions.
Collapse
Affiliation(s)
- Yasmina Zeineddine
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael A Friedman
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Evan G Buettmann
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Lovell B Abraham
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Gabriel A Hoppock
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Henry J Donahue
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
34
|
Gao S, Huang S, Zhang Y, Fang G, Liu Y, Zhang C, Li Y, Du J. The transcriptional regulator KLF15 is necessary for myoblast differentiation and muscle regeneration by activating FKBP5. J Biol Chem 2023; 299:105226. [PMID: 37673339 PMCID: PMC10622842 DOI: 10.1016/j.jbc.2023.105226] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/18/2023] [Accepted: 08/27/2023] [Indexed: 09/08/2023] Open
Abstract
Successful muscle regeneration following injury is essential for functional homeostasis of skeletal muscles. Krüppel-like factor 15 (KLF15) is a metabolic transcriptional regulator in the muscles. However, little is known regarding its function in muscle regeneration. Here, we examined microarray datasets from the Gene Expression Omnibus database, which indicated downregulated KLF15 in muscles from patients with various muscle diseases. Additionally, we found that Klf15 knockout (Klf15KO) impaired muscle regeneration following injury in mice. Furthermore, KLF15 expression was robustly induced during myoblast differentiation. Myoblasts with KLF15 deficiency showed a marked reduction in their fusion capacity. Unbiased transcriptome analysis of muscles on day 7 postinjury revealed downregulated genes involved in cell differentiation and metabolic processes in Klf15KO muscles. The FK506-binding protein 51 (FKBP5), a positive regulator of myoblast differentiation, was ranked as one of the most strongly downregulated genes in the Klf15KO group. A mechanistic search revealed that KLF15 binds directly to the promoter region of FKBP5 and activates FKBP5 expression. Local delivery of FKBP5 rescued the impaired muscle regeneration in Klf15KO mice. Our findings reveal a positive regulatory role of KLF15 in myoblast differentiation and muscle regeneration by activating FKBP5 expression. KLF15 signaling may be a novel therapeutic target for muscle disorders associated with injuries or diseases.
Collapse
Affiliation(s)
- Shijuan Gao
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shan Huang
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yanhong Zhang
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guangming Fang
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yan Liu
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Congcong Zhang
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Yulin Li
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Jie Du
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
35
|
Mirzoev TM. The emerging role of Piezo1 channels in skeletal muscle physiology. Biophys Rev 2023; 15:1171-1184. [PMID: 37975010 PMCID: PMC10643716 DOI: 10.1007/s12551-023-01154-6] [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: 06/15/2023] [Accepted: 09/25/2023] [Indexed: 11/19/2023] Open
Abstract
Piezo1 channels are mechanically activated (MA) cation channels that are involved in sensing of various mechanical perturbations, such as membrane stretch and shear stress, and play a crucial role in cell mechanotransduction. In response to mechanical stimuli, these channels open up and allow cations to travel into the cell and induce biochemical reactions that can change the cell's metabolism and function. Skeletal muscle cells/fibers inherently depend upon mechanical cues in the form of fluid shear stress and contractions (physical exercise). For example, an exposure of skeletal muscles to chronic mechanical loading leads to increased anabolism and fiber hypertrophy, while prolonged mechanical unloading results in muscle atrophy. MA Piezo1 channels have recently emerged as key mechanosensors that are capable of linking mechanical signals and intramuscular signaling in skeletal muscle cells/fibers. This review will summarize the emerging role of Piezo1 channels in the development and regeneration of skeletal muscle tissue as well as in the regulation of skeletal muscle atrophy. In addition, an overview of potential Piezo1-related signaling pathways underlying anabolic and catabolic processes will be provided. A better understanding of Piezo1's role in skeletal muscle mechanotransduction may represent an important basis for the development of therapeutic strategies for maintaining muscle functions under disuse conditions and in some disease states.
Collapse
Affiliation(s)
- Timur M. Mirzoev
- Myology Laboratory, Institute of Biomedical Problems RAS, Moscow, Russia
| |
Collapse
|
36
|
Zong B, Yu F, Zhang X, Pang Y, Zhao W, Sun P, Li L. Mechanosensitive Piezo1 channel in physiology and pathophysiology of the central nervous system. Ageing Res Rev 2023; 90:102026. [PMID: 37532007 DOI: 10.1016/j.arr.2023.102026] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/29/2023] [Accepted: 07/29/2023] [Indexed: 08/04/2023]
Abstract
Since the discovery of the mechanosensitive Piezo1 channel in 2010, there has been a significant amount of research conducted to explore its regulatory role in the physiology and pathology of various organ systems. Recently, a growing body of compelling evidence has emerged linking the activity of the mechanosensitive Piezo1 channel to health and disease of the central nervous system. However, the exact mechanisms underlying these associations remain inadequately comprehended. This review systematically summarizes the current research on the mechanosensitive Piezo1 channel and its implications for central nervous system mechanobiology, retrospects the results demonstrating the regulatory role of the mechanosensitive Piezo1 channel on various cell types within the central nervous system, including neural stem cells, neurons, oligodendrocytes, microglia, astrocytes, and brain endothelial cells. Furthermore, the review discusses the current understanding of the involvement of the Piezo1 channel in central nervous system disorders, such as Alzheimer's disease, multiple sclerosis, glaucoma, stroke, and glioma.
Collapse
Affiliation(s)
- Boyi Zong
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Fengzhi Yu
- School of Exercise and Health, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaoyou Zhang
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Yige Pang
- Department of Neurosurgery, Zibo Central Hospital, Zibo 255000, Shandong, China
| | - Wenrui Zhao
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua 321004, Zhejiang, China
| | - Peng Sun
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Lin Li
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
37
|
Wang WG, Chen ZSD, Sun J, Yang CM, He HB, Lu XK, Wang WY. Bioinformatic analysis of biological changes involved in pelvic organ prolapse. Medicine (Baltimore) 2023; 102:e33823. [PMID: 37266648 PMCID: PMC10237711 DOI: 10.1097/md.0000000000033823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/21/2023] [Accepted: 05/01/2023] [Indexed: 06/03/2023] Open
Abstract
The molecular mechanisms involved in the pathogenesis of pelvic organ prolapse (POP) remain unclear. This study aimed to identify key molecules involved in the pathogenesis and progression of POP. Differentially expressed genes (DEGs) were identified based on gene expression data extracted from the GSE53868, GSE28660, and GSE12852 datasets in the gene expression omnibus database. The R software was used for data mining, and gene ontology functional annotation and Kyoto encyclopedia of genes and genomes enrichment analyses were performed to explore the biological functions of DEGs. A protein-protein interaction network (PPI) was constructed using the Search Tool for the Retrieval of Interacting Genes database, and hub genes were identified by the Cytoscape plug-in cytoHubba. In addition, the CIBERSORT algorithm was used to analyze and evaluate immune cell infiltration in POP tissues. A total of 92 upregulated DEGs were identified and subjected to enrichment analysis. Gene ontology analysis revealed that these DEGs were associated with response to hormones, positive regulation of cell death, collagen-containing extracellular matrix, and extracellular matrix. Kyoto encyclopedia of genes and genomes pathway analysis showed that the upregulated genes were mainly enriched in the phosphatidylinositol 3-kinase-AKT signaling pathway. The PPI network was structured. Nodes in the PPI network were associated with structural molecular activity and collagen-containing extracellular matrix. A total of 10 hub genes were identified, namely, CDKN1A, IL-6, PPARG, ADAMTS4, ADIPOQ, AREG, activating transcription factor 3, CCL2, CD36, and Cell death-inducing DNA fragmentation factor-like effector A. Furthermore, patients with POP were found to have a higher abundance of CD8-positive T cells in the 3 gene expression omnibus datasets. The abundance of CD8-positive T cells was negatively correlated with that of follicular helper T cells (Pearson correlation coefficient = -0.34, P < .01) or gamma delta T cells (Pearson correlation coefficient = -0.33, P < .01). But was positively correlated with that of M2 macrophages (Pearson correlation coefficient = 0.35, P < .01) and activated memory CD4 T cells (Pearson correlation coefficient = 0.34, P < .01). Altogether, PPARG, ADAMTS4, ADIPOQ, AREG, CD36, and Cell death-inducing DNA fragmentation factor-like effector A genes were discovered in the POP process for the first time, which should be intensively investigated.
Collapse
Affiliation(s)
- Wei Guo Wang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhang Sen Di Chen
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ji Sun
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chun Mei Yang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Bo He
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xian Kun Lu
- Science and Technology Department of Sichuan Province, Chengdu, Sichuan, China
| | - Wei Yuan Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
38
|
Zhang Y, Wang L, Kang H, Lin CY, Fan Y. Unlocking the Therapeutic Potential of Irisin: Harnessing Its Function in Degenerative Disorders and Tissue Regeneration. Int J Mol Sci 2023; 24:ijms24076551. [PMID: 37047523 PMCID: PMC10095399 DOI: 10.3390/ijms24076551] [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: 03/03/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Physical activity is well-established as an important protective factor against degenerative conditions and a promoter of tissue growth and renewal. The discovery of Fibronectin domain-containing protein 5 (FNDC5) as the precursor of Irisin in 2012 sparked significant interest in its potential as a diagnostic biomarker and a therapeutic agent for various diseases. Clinical studies have examined the correlation between plasma Irisin levels and pathological conditions using a range of assays, but the lack of reliable measurements for endogenous Irisin has led to uncertainty about its prognostic/diagnostic potential as an exercise surrogate. Animal and tissue-engineering models have shown the protective effects of Irisin treatment in reversing functional impairment and potentially permanent damage, but dosage ambiguities remain unresolved. This review provides a comprehensive examination of the clinical and basic studies of Irisin in the context of degenerative conditions and explores its potential as a therapeutic approach in the physiological processes involved in tissue repair/regeneration.
Collapse
Affiliation(s)
- Yuwei Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Lizhen Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Correspondence:
| | - Hongyan Kang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Chia-Ying Lin
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Department of Biomedical, Chemical & Environmental Engineering, University of Cincinnati, Cincinnati, OH 45267, USA
- Department of Orthopaedic Surgery, University of Cincinnati, Cincinnati, OH 45267, USA
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- School of Engineering Medicine, Beihang University, Beijing 100083, China
| |
Collapse
|
39
|
Dienes B, Bazsó T, Szabó L, Csernoch L. The Role of the Piezo1 Mechanosensitive Channel in the Musculoskeletal System. Int J Mol Sci 2023; 24:ijms24076513. [PMID: 37047487 PMCID: PMC10095409 DOI: 10.3390/ijms24076513] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
Since the recent discovery of the mechanosensitive Piezo1 channels, many studies have addressed the role of the channel in various physiological or even pathological processes of different organs. Although the number of studies on their effects on the musculoskeletal system is constantly increasing, we are still far from a precise understanding. In this review, the knowledge available so far regarding the musculoskeletal system is summarized, reviewing the results achieved in the field of skeletal muscles, bones, joints and cartilage, tendons and ligaments, as well as intervertebral discs.
Collapse
|
40
|
Savadipour A, Palmer D, Ely EV, Collins KH, Garcia-Castorena JM, Harissa Z, Kim YS, Oestrich A, Qu F, Rashidi N, Guilak F. The role of PIEZO ion channels in the musculoskeletal system. Am J Physiol Cell Physiol 2023; 324:C728-C740. [PMID: 36717101 PMCID: PMC10027092 DOI: 10.1152/ajpcell.00544.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 02/01/2023]
Abstract
PIEZO1 and PIEZO2 are mechanosensitive cation channels that are highly expressed in numerous tissues throughout the body and exhibit diverse, cell-specific functions in multiple organ systems. Within the musculoskeletal system, PIEZO1 functions to maintain muscle and bone mass, sense tendon stretch, and regulate senescence and apoptosis in response to mechanical stimuli within cartilage and the intervertebral disc. PIEZO2 is essential for transducing pain and touch sensations as well as proprioception in the nervous system, which can affect musculoskeletal health. PIEZO1 and PIEZO2 have been shown to act both independently as well as synergistically in different cell types. Conditions that alter PIEZO channel mechanosensitivity, such as inflammation or genetic mutations, can have drastic effects on these functions. For this reason, therapeutic approaches for PIEZO-related disease focus on altering PIEZO1 and/or PIEZO2 activity in a controlled manner, either through inhibition with small molecules, or through dietary control and supplementation to maintain a healthy cell membrane composition. Although many opportunities to better understand PIEZO1 and PIEZO2 remain, the studies summarized in this review highlight how crucial PIEZO channels are to musculoskeletal health and point to promising possible avenues for their modulation as a therapeutic target.
Collapse
Affiliation(s)
- Alireza Savadipour
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Daniel Palmer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Erica V Ely
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Kelsey H Collins
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jaquelin M Garcia-Castorena
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Zainab Harissa
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Yu Seon Kim
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Arin Oestrich
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Feini Qu
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Neda Rashidi
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| |
Collapse
|
41
|
Mechanotransduction for Muscle Protein Synthesis via Mechanically Activated Ion Channels. Life (Basel) 2023; 13:life13020341. [PMID: 36836698 PMCID: PMC9962945 DOI: 10.3390/life13020341] [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: 01/03/2023] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Cell mechanotransduction, the ability to detect physical forces and convert them into a series of biochemical events, is important for a wide range of physiological processes. Cells express an array of mechanosensors transducing physical forces into intracellular signaling cascades, including ion channels. Ion channels that can be directly activated by mechanical cues are known as mechanically activated (MA), or stretch-activated (SA), channels. In response to repeated exposures to mechanical stimulation in the form of resistance training, enhanced protein synthesis and fiber hypertrophy are elicited in skeletal muscle, whereas a lack of mechanical stimuli due to inactivity/mechanical unloading leads to reduced muscle protein synthesis and fiber atrophy. To date, the role of MA channels in the transduction of mechanical load to intracellular signaling pathways regulating muscle protein synthesis is poorly described. This review article will discuss MA channels in striated muscle, their regulation, and putative roles in the anabolic processes in muscle cells/fibers in response to mechanical stimuli.
Collapse
|
42
|
Jung SC, Zhou T, Ko EA. Age-dependent expression of ion channel genes in rat. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2023; 27:85-94. [PMID: 36575936 PMCID: PMC9806634 DOI: 10.4196/kjpp.2023.27.1.85] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 12/29/2022]
Abstract
Ion channels regulate a large number of cellular functions and their functional role in many diseases makes them potential therapeutic targets. Given their diverse distribution across multiple organs, the roles of ion channels, particularly in age-associated transcriptomic changes in specific organs, are yet to be fully revealed. Using RNA-seq data, we investigated the rat transcriptomic profiles of ion channel genes across 11 organs/tissues and 4 developmental stages in both sexes of Fischer 344 rats and identify tissue-specific and age-dependent changes in ion channel gene expression. Organ-enriched ion channel genes were identified. In particular, the brain showed higher tissue-specificity of ion channel genes, including Gabrd, Gabra6, Gabrg2, Grin2a, and Grin2b. Notably, age-dependent changes in ion channel gene expression were prominently observed in the thymus, including in Aqp1, Clcn4, Hvcn1, Itpr1, Kcng2, Kcnj11, Kcnn3, and Trpm2. Our comprehensive study of ion channel gene expression will serve as a primary resource for biological studies of aging-related diseases caused by abnormal ion channel functions.
Collapse
Affiliation(s)
- Sung-Cherl Jung
- Department of Physiology, School of Medicine, Jeju National University, Jeju 63243, Korea
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Eun-A Ko
- Department of Physiology, School of Medicine, Jeju National University, Jeju 63243, Korea,Correspondence Eun-A Ko, E-mail:
| |
Collapse
|
43
|
Ono Y, Saito M, Sakamoto K, Maejima Y, Misaka S, Shimomura K, Nakanishi N, Inoue S, Kotani J. C188-9, a specific inhibitor of STAT3 signaling, prevents thermal burn-induced skeletal muscle wasting in mice. Front Pharmacol 2022; 13:1031906. [PMID: 36588738 PMCID: PMC9800842 DOI: 10.3389/fphar.2022.1031906] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Burn injury is the leading cause of death and disability worldwide and places a tremendous economic burden on society. Systemic inflammatory responses induced by thermal burn injury can cause muscle wasting, a severe involuntary loss of skeletal muscle that adversely affects the survival and functional outcomes of these patients. Currently, no pharmacological interventions are available for the treatment of thermal burn-induced skeletal muscle wasting. Elevated levels of inflammatory cytokines, such as interleukin-6 (IL-6), are important hallmarks of severe burn injury. The levels of signal transducer and activator of transcription 3 (STAT3)-a downstream component of IL-6 inflammatory signaling-are elevated with muscle wasting in various pro-catabolic conditions, and STAT3 has been implicated in the regulation of skeletal muscle atrophy. Here, we tested the effects of the STAT3-specific signaling inhibitor C188-9 on thermal burn injury-induced skeletal muscle wasting in vivo and on C2C12 myotube atrophy in vitro after the administration of plasma from burn model mice. In mice, thermal burn injury severity dependently increased IL-6 in the plasma and tibialis anterior muscles and activated the STAT3 (increased ratio of phospho-STAT3/STAT3) and ubiquitin-proteasome proteolytic pathways (increased Atrogin-1/MAFbx and MuRF1). These effects resulted in skeletal muscle atrophy and reduced grip strength. In murine C2C12 myotubes, plasma from burn mice activated the same inflammatory and proteolytic pathways, leading to myotube atrophy. In mice with burn injury, the intraperitoneal injection of C188-9 (50 mg/kg) reduced activation of the STAT3 and ubiquitin-proteasome proteolytic pathways, reversed skeletal muscle atrophy, and increased grip strength. Similarly, pretreatment of murine C2C12 myotubes with C188-9 (10 µM) reduced activation of the same inflammatory and proteolytic pathways, and ameliorated myotube atrophy induced by plasma taken from burn model mice. Collectively, these results indicate that pharmacological inhibition of STAT3 signaling may be a novel therapeutic strategy for thermal burn-induced skeletal muscle wasting.
Collapse
Affiliation(s)
- Yuko Ono
- Department of Disaster and Emergency Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan,Department of Bioregulation and Pharmacological Medicine, School of Medicine, Fukushima Medical University, Fukushima, Japan,*Correspondence: Yuko Ono,
| | - Masafumi Saito
- Department of Disaster and Emergency Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Kazuho Sakamoto
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Shingen Misaka
- Department of Bioregulation and Pharmacological Medicine, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Nobuto Nakanishi
- Department of Disaster and Emergency Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Shigeaki Inoue
- Department of Disaster and Emergency Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Joji Kotani
- Department of Disaster and Emergency Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan
| |
Collapse
|
44
|
Bouredji Z, Argaw A, Frenette J. The inflammatory response, a mixed blessing for muscle homeostasis and plasticity. Front Physiol 2022; 13:1032450. [PMID: 36505042 PMCID: PMC9726740 DOI: 10.3389/fphys.2022.1032450] [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: 08/30/2022] [Accepted: 11/10/2022] [Indexed: 11/24/2022] Open
Abstract
Skeletal muscle makes up almost half the body weight of heathy individuals and is involved in several vital functions, including breathing, thermogenesis, metabolism, and locomotion. Skeletal muscle exhibits enormous plasticity with its capacity to adapt to stimuli such as changes in mechanical loading, nutritional interventions, or environmental factors (oxidative stress, inflammation, and endocrine changes). Satellite cells and timely recruited inflammatory cells are key actors in muscle homeostasis, injury, and repair processes. Conversely, uncontrolled recruitment of inflammatory cells or chronic inflammatory processes leads to muscle atrophy, fibrosis and, ultimately, impairment of muscle function. Muscle atrophy and loss of function are reported to occur either in physiological situations such as aging, cast immobilization, and prolonged bed rest, as well as in many pathological situations, including cancers, muscular dystrophies, and several other chronic illnesses. In this review, we highlight recent discoveries with respect to the molecular mechanisms leading to muscle atrophy caused by modified mechanical loading, aging, and diseases. We also summarize current perspectives suggesting that the inflammatory process in muscle homeostasis and repair is a double-edged sword. Lastly, we review recent therapeutic approaches for treating muscle wasting disorders, with a focus on the RANK/RANKL/OPG pathway and its involvement in muscle inflammation, protection and regeneration processes.
Collapse
Affiliation(s)
- Zineb Bouredji
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CRCHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC, Canada
| | - Anteneh Argaw
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CRCHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC, Canada
| | - Jérôme Frenette
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CRCHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC, Canada,Département de Réadaptation, Faculté de Médecine, Université Laval, Quebec City, QC, Canada,*Correspondence: Jérôme Frenette,
| |
Collapse
|
45
|
Wu M, Zhang Y. Integrated bioinformatics, network pharmacology, and artificial intelligence to predict the mechanism of celastrol against muscle atrophy caused by colorectal cancer. Front Genet 2022; 13:1012932. [PMID: 36419834 PMCID: PMC9676937 DOI: 10.3389/fgene.2022.1012932] [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: 08/08/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
Muscle atrophy due to colorectal cancer severely reduces the quality of life and survival time of patients. However, the underlying causative mechanisms and therapeutic agents are not well understood. The aim of this study was to screen and identify the microRNA (miRNA)–mRNA regulatory network and therapeutic targets of celastrol in colorectal cancer causing muscle atrophy via blood exosomes. Datasets were downloaded from the Gene Expression Omnibus online database. Differential expression analysis was first performed using the blood exosome dataset GSE39833 from colorectal cancer and normal humans to identify differentially expressed (DE) miRNAs, and then, transcriptional enrichment analysis was performed to identify important enriched genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed by FunRich software. Using the muscle atrophy sample GSE34111, the DE mRNAs in the muscle atrophy sample were analyzed, a regulatory network map was established based on miRNA‒mRNA regulatory mechanisms, further GO and KEGG enrichment analyses were performed for the DE genes in muscle atrophy via Cytoscape’s ClueGO plug-in, and the network pharmacology pharmacophore analysis method was used to analyze the celastrol therapeutic targets, taking intersections to find the therapeutic targets of celastrol, using the artificial intelligence AlphaFold2 to predict the protein structures of the key targets, and finally using molecular docking to verify whether celastrol and the target proteins can be successfully docked. A total of 82 DE miRNAs were obtained, and the top 10 enriched target genes were identified. The enrichment of the 82 miRNAs showed a close correlation with muscle atrophy, and 332 DE mRNAs were found by differential expression analysis in muscle atrophy samples, among which 44 mRNA genes were involved in miRNA‒mRNA networks. The DE genes in muscle atrophy were enriched for 30 signaling pathways, and 228 target genes were annotated after pharmacophore target analysis. The NR1D2 gene, the target of treatment, was found by taking intersections, the protein structure of this target was predicted by AlphaFold2, and the structure was successfully docked and validated using molecular docking. In our present study, colorectal cancer likely enters the muscle from blood exosomes and regulates skeletal muscle atrophy through miRNA‒mRNA regulatory network mechanisms, and celastrol treats muscle through NR1D2 in the miRNA‒mRNA regulatory network.
Collapse
Affiliation(s)
- Ming Wu
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Yan Zhang
- Department of Orthopedics, Gongli Hospital of Pudong New Area, Shanghai, China
- *Correspondence: Yan Zhang,
| |
Collapse
|
46
|
A preliminary study on the role of Piezo1 channels in myokine release from cultured mouse myotubes. Biochem Biophys Res Commun 2022; 623:148-153. [DOI: 10.1016/j.bbrc.2022.07.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/14/2022] [Indexed: 11/20/2022]
|
47
|
Metabolic Pathways and Ion Channels Involved in Skeletal Muscle Atrophy: A Starting Point for Potential Therapeutic Strategies. Cells 2022; 11:cells11162566. [PMID: 36010642 PMCID: PMC9406740 DOI: 10.3390/cells11162566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/08/2022] [Accepted: 08/16/2022] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle tissue has the important function of supporting and defending the organism. It is the largest apparatus in the human body, and its function is important for contraction and movements. In addition, it is involved in the regulation of protein synthesis and degradation. In fact, inhibition of protein synthesis and/or activation of catabolism determines a pathological condition called muscle atrophy. Muscle atrophy is a reduction in muscle mass resulting in a partial or complete loss of function. It has been established that many physiopathological conditions can cause a reduction in muscle mass. Nevertheless, it is not well known that the molecular mechanisms and signaling processes caused this dramatic event. There are multiple concomitant processes involved in muscle atrophy. In fact, the gene transcription of some factors, oxidative stress mechanisms, and the alteration of ion transport through specific ion channels may contribute to muscle function impairment. In this review, we focused on the molecular mechanisms responsible for muscle damage and potential drugs to be used to alleviate this disabling condition.
Collapse
|
48
|
Bernareggi A, Bosutti A, Massaria G, Giniatullin R, Malm T, Sciancalepore M, Lorenzon P. The State of the Art of Piezo1 Channels in Skeletal Muscle Regeneration. Int J Mol Sci 2022; 23:ijms23126616. [PMID: 35743058 PMCID: PMC9224226 DOI: 10.3390/ijms23126616] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 02/07/2023] Open
Abstract
Piezo1 channels are highly mechanically-activated cation channels that can sense and transduce the mechanical stimuli into physiological signals in different tissues including skeletal muscle. In this focused review, we summarize the emerging evidence of Piezo1 channel-mediated effects in the physiology of skeletal muscle, with a particular focus on the role of Piezo1 in controlling myogenic precursor activity and skeletal muscle regeneration and vascularization. The disclosed effects reported by pharmacological activation of Piezo1 channels with the selective agonist Yoda1 indicate a potential impact of Piezo1 channel activity in skeletal muscle regeneration, which is disrupted in various muscular pathological states. All findings reported so far agree with the idea that Piezo1 channels represent a novel, powerful molecular target to develop new therapeutic strategies for preventing or ameliorating skeletal muscle disorders characterized by an impairment of tissue regenerative potential.
Collapse
Affiliation(s)
- Annalisa Bernareggi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (G.M.); (M.S.); (P.L.)
- Correspondence:
| | - Alessandra Bosutti
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (G.M.); (M.S.); (P.L.)
| | - Gabriele Massaria
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (G.M.); (M.S.); (P.L.)
| | - Rashid Giniatullin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (R.G.); (T.M.)
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (R.G.); (T.M.)
| | - Marina Sciancalepore
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (G.M.); (M.S.); (P.L.)
| | - Paola Lorenzon
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (G.M.); (M.S.); (P.L.)
| |
Collapse
|