1
|
Borok MJ, Zaidan L, Relaix F. Transposon expression and repression in skeletal muscle. Mob DNA 2025; 16:18. [PMID: 40217332 PMCID: PMC11992895 DOI: 10.1186/s13100-025-00352-1] [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: 12/31/2024] [Accepted: 03/13/2025] [Indexed: 04/14/2025] Open
Abstract
Transposons and their derivatives make up a major proportion of the human genome, but they are not just relics of ancient genomes. They can still be expressed, potentially affecting the transcription of adjacent genes, and can sometimes even contribute to their coding sequence. Active transposons can integrate into new sites in the genome, potentially modifying the expression of nearby loci and leading to genetic disorders. In this review, we highlight work exploring the expression of transposons in skeletal muscles and transcriptional regulation by the KRAB-ZFP/KAP1/SETDB1 complex. We next focus on specific cases of transposon insertion causing phenotypic variation and distinct muscular dystrophies, as well as the implication of transposon expression in immune myopathies. Finally, we discuss the dysregulation of transposons in facioscapulohumeral dystrophy and aging.
Collapse
Affiliation(s)
- Matthew J Borok
- University Paris-Est Créteil, INSERM U955 IMRB, Créteil, 94010, France.
| | - Louai Zaidan
- University Paris-Est Créteil, INSERM U955 IMRB, Créteil, 94010, France
| | - Frederic Relaix
- University Paris-Est Créteil, INSERM U955 IMRB, Créteil, 94010, France.
- École Nationale Vétérinaire d'Alfort U955 IMRB, Maisons-Alfort, 94700, France.
- EFS IMRB, Créteil, 94010, France.
- Assistance Publique-Hôpitaux de Paris, Hôpital Mondor, Service d'Histologie, Créteil, 94010, France.
| |
Collapse
|
2
|
Shi Y, Qin J, Yin E, Xu J, Chen Y, Tie K, Chen L. Mitochondria Isolated From Bone Mesenchymal Stem Cells Restrain Muscle Disuse Atrophy and Fatty Infiltration After Rotator Cuff Tears. Am J Sports Med 2025; 53:1171-1183. [PMID: 40088076 DOI: 10.1177/03635465251323001] [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] [Indexed: 03/17/2025]
Abstract
BACKGROUND Rotator cuff tears (RCTs) commonly lead to muscle atrophy, fibrosis, and fatty infiltration, complicating treatment. PURPOSE To investigate the use of mitochondria isolated from bone mesenchymal stem cells (BMSC-Mito) for mitigating complications after RCT, focusing on muscle protection. STUDY DESIGN Controlled laboratory study. METHODS RCTs were induced by transecting the tendons of the supraspinatus and infraspinatus in Sprague-Dawley rats. In vivo, 90 rats were randomized into 3 groups: sham (no intervention), RCTs treated with BMSC-Mito, and RCTs treated with phosphate-buffered saline. After 6 weeks of intramuscular injections of BMSC-Mito or phosphate-buffered saline, supraspinatus muscles were harvested for analysis. Evaluations included wet muscle weight, muscle fiber cross-sectional area, fibrosis, fatty infiltration, slow-fast myofiber types and muscle biomechanics, capillary density, mitochondria respiratory chain complex activity, adenosine triphosphate (ATP) concentration, oxidative stress, and mitochondrial ultrastructure. In vitro experiments utilized primary rat skeletal muscle cells pretreated with rhodamine 6G to induce mitochondrial dysfunction, assessing the effects of BMSC-Mito on cell viability, mitochondrial membrane potential, and oxidative stress levels. RESULTS BMSC-Mito can be effectively transplanted into muscles and integrated into the local mitochondrial network. After RCT, the supraspinatus showed significant mass loss, reduced fiber cross-sectional area, fatty infiltration, and a shift from slow to fast myofiber types, which negatively affected muscle biomechanics. These changes were reversed by BMSC-Mito. BMSC-Mito also preserved vascularity (CD31 and α-SMA) impaired by RCT. Additionally, BMSC-Mito notably improved disuse-induced mitochondrial changes, leading to increased mitochondrial number and COX IV expression; furthermore, BMSC-Mito protected mitochondria morphology and enhanced cytosolic superoxide dismutase activity. This treatment also improved mitochondria respiratory chain complex activity and ATP concentration, reducing oxidative stress. In vitro, BMSC-Mito treatment effectively maintained the mitochondrial membrane potential of skeletal muscle cells, improved cell viability, and restored its mitochondrial function and ATP levels. CONCLUSION These findings suggest that BMSC-Mito might play a role in preventing muscle atrophy and fatty infiltration after RCT through the protection of mitochondrial function and the promotion of angiogenesis. CLINICAL RELEVANCE BMSC-Mito present a promising therapeutic approach for addressing rotator cuff muscle degeneration.
Collapse
Affiliation(s)
- Yulong Shi
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Qin
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Enzhi Yin
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Xu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuanyuan Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China
| | - Kai Tie
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Matthews I, Mehra P, Suárez-Calvet X, Piñol-Jurado P, Cox D, Justian V, Carrasco-Rozas A, Laidler Z, Bowey A, Rushton P, López-Fernández S, Díaz-Manera J, Fernández-Simón E. Strategy for drug repurposing in fibroadipogenic replacement during muscle wasting: application to duchenne muscular dystrophy. Front Cell Dev Biol 2025; 13:1505697. [PMID: 40206397 PMCID: PMC11979640 DOI: 10.3389/fcell.2025.1505697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Background Understanding the cell functionality during disease progression or drugs' mechanism are major challenges for precision medicine. Predictive models describing biological phenotypes can be challenging to obtain, particularly in scenarios where sample availability is limited, such as in the case of rare diseases. Here we propose a new method that reproduces the fibroadipogenic expansion that occurs in muscle wasting. Methods We used immortalized fibroadipogenic progenitor cells (FAPs) and differentiated them into fibroblasts or adipocytes. The method successfully identified FAPs cell differentiation fate using accurate measurements of changes in specific proteins, which ultimately constitute a valid cellular in vitro platform for drug screening. Results were confirmed using primary FAPs differentiation as well as comparison with omics data from proteomics and genomic studies. Results Our method allowed us to screen 508 different drugs from 2 compounds libraries. Out of these 508, we identified 4 compounds that reduced fibrogenesis and adipogenesis of ≥30% of fibrogenesis and adipogenesis using immortalized cells. After selecting the optimal dose of each compound, the inhibitory effect on FAP differentiation was confirmed by using primary FAPs from healthy subjects (n = 3) and DMD patients (n = 3). The final 4 selected hits reduced fibrogenic differentiation in healthy and DMD samples. The inhibition of adipogenesis was more evident in DMD samples than healthy samples. After creating an inhibitory map of the tested drugs, we validated the signalling pathways more involved in FAPs differentiation analysing data from proteomic and genomic studies. Conclusion We present a map of molecular targets of approved drugs that helps in predicting which therapeutic option may affect FAP differentiation. This method allows to study the potential effect of signalling circuits on FAP differentiation after drug treatment providing insights into molecular mechanism of action of muscle degeneration. The accuracy of the method is demonstrated by comparing the signal pathway activity obtained after drug treatment with proteomic and genomic data from patient-derived cells.
Collapse
Affiliation(s)
- Izzy Matthews
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, United Kingdom
| | - Priyanka Mehra
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Xavier Suárez-Calvet
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Patricia Piñol-Jurado
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Dan Cox
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Vellia Justian
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ana Carrasco-Rozas
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Zoe Laidler
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew Bowey
- Great North Children’s Hospital, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Paul Rushton
- Great North Children’s Hospital, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Susana López-Fernández
- Plastic Surgery Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jordi Díaz-Manera
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Esther Fernández-Simón
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
4
|
Sun Z, Cheng X, Wang Z, Qiao C, Qian H, Yuan T, Lv Z, Sun W, Zhang H, Liu Y, Lu Z, Lin J, Lai C, Wang Y, Yang X, Wang X, Meng J, Bao N. Single-nucleus transcriptomics reveals subsets of degenerative myonuclei after rotator cuff tear-induced muscle atrophy. Cell Prolif 2025; 58:e13763. [PMID: 39435630 PMCID: PMC11882757 DOI: 10.1111/cpr.13763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Rotator cuff tear (RCT) is the primary cause of shoulder pain and disability and frequently trigger muscle degeneration characterised by muscle atrophy, fatty infiltration and fibrosis. Single-nucleus RNA sequencing (snRNA-seq) was used to reveal the transcriptional changes in the supraspinatus muscle after RCT. Supraspinatus muscles were obtained from patients with habitual shoulder dislocation (n = 3) and RCT (n = 3). In response to the RCT, trajectory analysis showed progression from normal myonuclei to ANKRD1+ myonuclei, which captured atrophy-and fatty infiltration-related regulons (KLF5, KLF10, FOSL1 and BHLHE40). Transcriptomic alterations in fibro/adipogenic progenitors (FAPs) and muscle satellite cells (MuSCs) have also been studied. By predicting cell-cell interactions, we observed communication alterations between myofibers and muscle-resident cells following RCT. Our findings reveal the plasticity of muscle cells in response to RCT and offer valuable insights into the molecular mechanisms and potential therapeutic targets of RCT.
Collapse
Affiliation(s)
- Ziying Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xi Cheng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zheng Wang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Chenfeng Qiao
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| | - Hong Qian
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Tao Yuan
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zhongyang Lv
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Wenshuang Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Hanwen Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Yuan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zhihao Lu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Jintao Lin
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Chengteng Lai
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Yang Wang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xiaojiang Yang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xingyun Wang
- Hongqiao International Institute of Medicine, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Jia Meng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| | - Nirong Bao
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| |
Collapse
|
5
|
Duong VT, Dang TT, Le VP, Le TH, Nguyen CT, Phan HL, Seo J, Lin CC, Back SH, Koo KI. Direct extrusion of multifascicle prevascularized human skeletal muscle for volumetric muscle loss surgery. Biomaterials 2025; 314:122840. [PMID: 39321685 DOI: 10.1016/j.biomaterials.2024.122840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
Skeletal muscle is composed of multiple fascicles, which are parallel bundles of muscle fibers surrounded by connective tissues that contain blood vessels and nerves. Here, we fabricated multifascicle human skeletal muscle scaffolds that mimic the natural structure of human skeletal muscle bundles using a seven-barrel nozzle. For the core material to form the fascicle structure, human skeletal myoblasts were encapsulated in Matrigel with calcium chloride. Meanwhile, the shell that plays a role as the connective tissue, human fibroblasts and human umbilical vein endothelial cells within a mixture of porcine muscle decellularized extracellular matrix and sodium alginate at a 95:5 ratio was used. We assessed four types of extruded scaffolds monolithic-monoculture (Mo-M), monolithic-coculture (Mo-C), multifascicle-monoculture (Mu-M), and multifascicle-coculture (Mu-C) to determine the structural effect of muscle mimicking scaffold. The Mu-C scaffold outperformed other scaffolds in cell proliferation, differentiation, vascularization, mechanical properties, and functionality. In an in vivo mouse model of volumetric muscle loss, the Mu-C scaffold effectively regenerated the tibialis anterior muscle defect, demonstrating its potential for volumetric muscle transplantation. Our nozzle will be further used to produce other volumetric functional tissues, such as tendons and peripheral nerves.
Collapse
Affiliation(s)
- Van Thuy Duong
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Thao Thi Dang
- School of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Van Phu Le
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea.
| | - Thi Huong Le
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea.
| | - Chanh Trung Nguyen
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea.
| | - Huu Lam Phan
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea.
| | - Jongmo Seo
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, 08826, Republic of Korea; Seoul National University Hospital Biomedical Research Institute, Seoul, 03080, Republic of Korea.
| | - Chien-Chi Lin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Sung Hoon Back
- School of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea; Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan, Republic of Korea.
| | - Kyo-In Koo
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea; Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan, Republic of Korea.
| |
Collapse
|
6
|
Jia H, Kaster N, Khan R, Ayari-Akkari A. The Roles of myomiRs in the Pathogenesis of Sarcopenia: From Literature to In Silico Analysis. Mol Biotechnol 2025:10.1007/s12033-025-01373-0. [PMID: 40025274 DOI: 10.1007/s12033-025-01373-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/30/2024] [Indexed: 03/04/2025]
Abstract
Senile sarcopenia is a condition of age-associated muscular disorder and is a significant health issue around the world. In the current review, we curated the information from the NCBI, PubMed, and Google Scholar literature and explored the non-genetic and genetic causes of senile sarcopenia. Interestingly, the myomiRs such as miR-1, miR-206, miR-133a, miR-133b, miR-208b, and miR-499 are skeletal muscle's critical structural and functional regulators. However, very scattered information is available regarding the roles of myomiRs in different skeletal muscle phenotypes through a diverse list of known target genes. Therefore, these pieces of information must be organized to focus on the conserved target genes and comparable effects of the myomiRs in regulating senile sarcopenia. Hence, in the present review, the roles of pathogenetic factors in regulating senile sarcopenia were highlighted. The literature was further curated for the roles of myomiRs such as hsa-miR-1-3p/206, hsa-miR-27-3p, hsa-miR-146-5p, and hsa-miR-499-5p and their target genes. Additionally, we used different bioinformatics tools and predicted target genes of the myomiRs and found the most critical target genes, shared pathways, and their standard functions in regulating muscle structure and functions. The information gathered in the current review will help the researchers to explore their possible therapeutic potential, especially the use of the myomiRs for the treatment of senile sarcopenia.
Collapse
Affiliation(s)
- Huanxia Jia
- Medical College of Xuchang University, No.1389, Xufan Road, Xuchang, 461000, Henan, People's Republic of China
| | - Nurgulsim Kaster
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, People's Republic of China.
- Faculty of Veterinary and Livestock Technology, S. Seifullin Kazakh Agro Technical University, Astana, Kazakhstan.
| | - Rajwali Khan
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, People's Republic of China.
- Department of Livestock Management, Breeding and Genetics, The University of Agriculture, Peshawar, Pakistan.
| | - Amel Ayari-Akkari
- Biology Department, College of Science, King Khalid University, P.O. Box 960, Abha, Saudi Arabia
| |
Collapse
|
7
|
Wang X, Gao X, Deng C, Xu D, Chen Y, Huang J, Li X, Shi Y. Platelet-derived mitochondria attenuate muscle atrophy following rotator cuff tears in a rat model. J Shoulder Elbow Surg 2025:S1058-2746(25)00172-7. [PMID: 39986534 DOI: 10.1016/j.jse.2025.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Rotator cuff tears (RCTs) often result in muscle atrophy, compromising surgical outcomes and recovery. Mitochondrial dysfunction is implicated in this process, suggesting potential for mitochondria-based therapies. This study aimed to investigate the effects of platelet-derived mitochondria (Plt-Mito) administration into the supraspinatus muscle (SSP) following RCTs. METHODS Seventy-two male Sprague-Dawley rats were allocated into 3 distinct groups: (1) a sham surgery group, (2) a group with RCTs treated with Plt-Mito, and (3) a group with RCTs treated with phosphate-buffered saline. Treatments were administered every 2 weeks. After 12 weeks, the SSPs were analyzed for wet muscle weight ratio, muscle fiber cross-sectional area, fibrosis, antioxidant activity, mitochondrial markers, capillary density, and mitochondrial structure. RESULTS Plt-Mito successfully incorporated into SSP, maintaining functional integrity. Compared to the phosphate-buffered saline group, Plt-Mito treatment significantly preserved wet muscle weight, increased mean muscle fiber cross-sectional area, promoted muscle regeneration, reduced fibrosis, enhanced antioxidant activity (increased superoxide dismutase activity and decreased malondialdehyde activity), improved muscle vascularity (increased platelet endothelial cell adhesion molecule-1 and α-smooth muscle actin), increased expression of mitochondrial markers (C oxidase subunit IV and uncoupling protein 1) and maintained mitochondrial density and structure. CONCLUSIONS Our findings demonstrated Plt-Mito administration effectively halted muscle atrophy and fibrosis, while attenuating mitochondrial damage and dysfunction following RCTs.
Collapse
Affiliation(s)
- Xin Wang
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xing Gao
- Animal Experimental Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Xu
- Department of Orthopedic Surgery, Ningbo NO.6 Hospital, Ningbo, China
| | - Yuanyuan Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China
| | - Jiaqi Huang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China
| | - Xiao Li
- Priority Medical Department, General Hospital of Central Theater Command, Wuhan, China.
| | - Yulong Shi
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
8
|
Li Y, Li C, Sun Q, Liu X, Chen F, Cheung Y, Zhao Y, Xie T, Chazaud B, Sun H, Wang H. Skeletal muscle stem cells modulate niche function in Duchenne muscular dystrophy mouse through YY1-CCL5 axis. Nat Commun 2025; 16:1324. [PMID: 39900599 PMCID: PMC11790879 DOI: 10.1038/s41467-025-56474-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 01/15/2025] [Indexed: 02/05/2025] Open
Abstract
Adult skeletal muscle stem cells (MuSCs) are indispensable for muscle regeneration and tightly regulated by macrophages (MPs) and fibro-adipogenic progenitors (FAPs) in their niche. Deregulated MuSC/MP/FAP interactions and the ensuing inflammation and fibrosis are hallmarks of dystrophic muscle. Here we demonstrate intrinsic deletion of transcription factor Yin Yang 1 (YY1) in MuSCs exacerbates dystrophic pathologies by altering composition and heterogeneity of MPs and FAPs. Further analysis reveals YY1 loss induces expression of immune genes in MuSCs, including C-C motif chemokine ligand 5 (Ccl5). Augmented CCL5 secretion promotes MP recruitment via CCL5/C-C chemokine receptor 5 (CCR5) crosstalk, which subsequently hinders FAP clearance through elevated Transforming growth factor-β1 (TGFβ1). Maraviroc-mediated pharmacological blockade of the CCL5/CCR5 axis effectively mitigates muscle dystrophy and improves muscle performance. Lastly, we demonstrate YY1 represses Ccl5 transcription by binding to its enhancer thus facilitating promoter-enhancer looping. Altogether, our study demonstrates the critical role of MuSCs in actively shaping their niche and provides novel insight into the therapeutic intervention of muscle dystrophy.
Collapse
MESH Headings
- Animals
- YY1 Transcription Factor/metabolism
- YY1 Transcription Factor/genetics
- Chemokine CCL5/metabolism
- Chemokine CCL5/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Mice
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Macrophages/metabolism
- Receptors, CCR5/metabolism
- Receptors, CCR5/genetics
- Mice, Inbred mdx
- Stem Cell Niche
- Mice, Knockout
- Maraviroc/pharmacology
- Mice, Inbred C57BL
- Male
- Transforming Growth Factor beta1/metabolism
- Stem Cells/metabolism
- Disease Models, Animal
Collapse
Grants
- 82172436 National Natural Science Foundation of China (National Science Foundation of China)
- 14115319, 14100620, 14106521, 14105823, 14120420, 14103522, 14105123 Research Grants Council, University Grants Committee (RGC, UGC)
- T13-602/21-N Research Grants Council, University Grants Committee (RGC, UGC)
- C6018-19GF Research Grants Council, University Grants Committee (RGC, UGC)
- 10210906, 08190626 Research Grants Council, University Grants Committee (RGC, UGC)
- AoE/M-402/20 Research Grants Council, University Grants Committee (RGC, UGC)
- STG1/E-403/24-N Research Grants Council, University Grants Committee (RGC, UGC)
- National Key R&D Program of China to H.W. (2022YFA0806003) Health and Medical Research Fund (HMRF) from Health Bureau of the Hong Kong Special Administrative Region, China to H.W. (10210906 and 08190626)
Collapse
Affiliation(s)
- Yang Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine Limited, Hong Kong Science Park, Hong Kong SAR, China
| | - Chuhan Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qiang Sun
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine Limited, Hong Kong Science Park, Hong Kong SAR, China
| | - Xingyuan Liu
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fengyuan Chen
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yeelo Cheung
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Zhao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ting Xie
- Center for Tissue Regeneration and Engineering, Division of Life Science, Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Bénédicte Chazaud
- Unité Physiopathologie et Génétique du Neurone et du Muscle, UMR CNRS 5261, Inserm U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Hao Sun
- Warshel Institute for Computational Biology, Faculty of Medicine, Chinese University of Hong Kong (Shenzhen), Guangdong, China.
| | - Huating Wang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China.
- Center for Neuromusculoskeletal Restorative Medicine Limited, Hong Kong Science Park, Hong Kong SAR, China.
| |
Collapse
|
9
|
Hamer MS, Rossi FMV. Multitasking muscle: engineering iPSC-derived myogenic progenitors to do more. Front Cell Dev Biol 2025; 12:1526635. [PMID: 39911186 PMCID: PMC11794491 DOI: 10.3389/fcell.2024.1526635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025] Open
Abstract
The generation of myogenic progenitors from iPSCs (iMPs) with therapeutic potential for in vivo tissue regeneration has long been a goal in the skeletal muscle community. Today, protocols enable the production of potent, albeit immature, iMPs that resemble Pax7+ adult muscle stem cells. While muscular dystrophies are often the primary therapeutic target for these cells, an underexplored application is their use in treating traumatic muscle injuries. Notably absent from recent reviews on iMPs is the concept of engineering these cells to perform functions post-transplantation that non-transgenic cells cannot. Here, we highlight protocols to enhance the generation, purification, and maturation of iMPs, and introduce the idea of engineering these cells to perform functions beyond their normal capacities, envisioning novel therapeutic applications.
Collapse
Affiliation(s)
- Mark Stephen Hamer
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Fabio M. V. Rossi
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
10
|
Tomaz da Silva M, Joshi AS, Kumar A. Fibroblast growth factor-inducible 14 regulates satellite cell self-renewal and expansion during skeletal muscle repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.06.616900. [PMID: 39803454 PMCID: PMC11722277 DOI: 10.1101/2024.10.06.616900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Skeletal muscle regeneration in adults is predominantly driven by satellite cells. Loss of satellite cell pool and function leads to skeletal muscle wasting in many conditions and disease states. Here, we demonstrate that the levels of fibroblast growth factor-inducible 14 (Fn14) are increased in satellite cells after muscle injury. Conditional ablation of Fn14 in Pax7-expressing satellite cells drastically reduces their expansion and skeletal muscle regeneration following injury. Fn14 is required for satellite cell self-renewal and proliferation as well as to prevent precocious differentiation. Targeted deletion of Fn14 inhibits Notch signaling but leads to the spurious activation of STAT3 signaling in regenerating skeletal muscle and in cultured muscle progenitor cells. Silencing of STAT3 improves proliferation and inhibits premature differentiation of Fn14-deficient satellite cells. Furthermore, conditional ablation of Fn14 in satellite cells exacerbates myopathy in the mdx mouse model of Duchenne muscular dystrophy (DMD) whereas its overexpression improves the engraftment of exogenous muscle progenitor cells into the dystrophic muscle of mdx mice. Altogether, our study highlights the crucial role of Fn14 in the regulation of satellite cell fate and function and suggests that Fn14 can be a potential molecular target to improve muscle regeneration in muscular disorders.
Collapse
Affiliation(s)
- Meiricris Tomaz da Silva
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Aniket S. Joshi
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Ashok Kumar
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| |
Collapse
|
11
|
Soldado F, López de Jesús M, Beitia M, González-Burguera I, Ocerin G, Elejaga-Jimeno A, Saumell-Esnaola M, Barrondo S, Oraa J, Sallés J, Delgado D, García Del Caño G, Sánchez M. Effects of intramuscular administration of Platelet-Rich Plasma on denervated muscle after peripheral nerve injury. Connect Tissue Res 2025; 66:10-25. [PMID: 39729391 DOI: 10.1080/03008207.2024.2446888] [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: 10/25/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE After peripheral nerve injury (PNI), prolonged denervation of the target muscle prevents adequate reinnervation even if the nerve is repaired. The aim of this work is to analyze the effect of intramuscular Platelet-Rich Plasma (PRP) in a denervated muscle due to PNI.Materials and. METHODS An irreversible PNI was generated in the common peroneal nerve of 80 Wistar rats by nerve resection. Animals were divided into groups: non-treatment (NT), saline (S) and PRP (PRP). 200 uL of saline (S group) and PRP (PRP group) were infiltrated intramuscularly into the tibialis anterior muscle on a weekly basis, from surgery to sacrifice (at 2, 4 and 7 weeks). Muscles were histologically processed for immunofluorescence and Western blotting. Effects on nicotinic acetylcholine receptor (nAChR), satellite cells (SC) and myogenin expression were analyzed. Comparisons were performed by two-way analysis of variance (ANOVA). RESULTS PRP had a platelet concentration 1.5-fold higher than blood, without erythrocytes and leukocytes. The PRP group had a higher percentage weight than the S and NT groups (p < 0.05). The levels of nAChRα1 and nAChRε subunit were lower in the PRP group relative to the NT and S (p < 0.05), while the nAChRγ subunit showed an increase in the PRP group (p < 0.05). The activation of SCs was higher in the PRP group compared to NT and S groups (p < 0.05). CONCLUSION PRP treatment can modulate NMJ configuration as well as key myogenic regulatory factors in denervated muscle, enhancing SC activation while mitigating muscle atrophy.
Collapse
Affiliation(s)
- Francisco Soldado
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, Spain
| | - Maider López de Jesús
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Bioaraba, Cellular and Molecular Neuropharmacology, Vitoria-Gasteiz, Spain
| | - Maider Beitia
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, Spain
| | - Imanol González-Burguera
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Garazi Ocerin
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Ainhoa Elejaga-Jimeno
- Department of Analytical Chemistry, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Miquel Saumell-Esnaola
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Bioaraba, Cellular and Molecular Neuropharmacology, Vitoria-Gasteiz, Spain
| | - Sergio Barrondo
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Jaime Oraa
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, Spain
| | - Joan Sallés
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Diego Delgado
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, Spain
| | - Gontzal García Del Caño
- Bioaraba, Cellular and Molecular Neuropharmacology, Vitoria-Gasteiz, Spain
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Mikel Sánchez
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, Spain
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, Spain
| |
Collapse
|
12
|
Wang T, Zhou D, Hong Z. Sarcopenia and cachexia: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2025; 6:e70030. [PMID: 39764565 PMCID: PMC11702502 DOI: 10.1002/mco2.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/17/2025] Open
Abstract
Sarcopenia is defined as a muscle-wasting syndrome that occurs with accelerated aging, while cachexia is a severe wasting syndrome associated with conditions such as cancer and immunodeficiency disorders, which cannot be fully addressed through conventional nutritional supplementation. Sarcopenia can be considered a component of cachexia, with the bidirectional interplay between adipose tissue and skeletal muscle potentially serving as a molecular mechanism for both conditions. However, the underlying mechanisms differ. Recognizing the interplay and distinctions between these disorders is essential for advancing both basic and translational research in this area, enhancing diagnostic accuracy and ultimately achieving effective therapeutic solutions for affected patients. This review discusses the muscle microenvironment's changes contributing to these conditions, recent therapeutic approaches like lifestyle modifications, small molecules, and nutritional interventions, and emerging strategies such as gene editing, stem cell therapy, and gut microbiome modulation. We also address the challenges and opportunities of multimodal interventions, aiming to provide insights into the pathogenesis and molecular mechanisms of sarcopenia and cachexia, ultimately aiding in innovative strategy development and improved treatments.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Zhen Hong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| |
Collapse
|
13
|
Darfallah L, Sifeddine N, Amalou G, El Cadi CA, Lakhiari H, Barakat A, Rouba H. Identification of a de-novo variant of the MEGF10 gene associated with EMARDD. REVISTA ESPANOLA DE PATOLOGIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ANATOMIA PATOLOGICA Y DE LA SOCIEDAD ESPANOLA DE CITOLOGIA 2025; 58:100795. [PMID: 39827508 DOI: 10.1016/j.patol.2024.100795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/15/2024] [Accepted: 08/14/2024] [Indexed: 01/22/2025]
Abstract
Early-onset Myopathy, Areflexia, Respiratory Distress and Dysphagia (EMARDD) is a congenital neuromuscular disease with a progressive muscle weakness, respiratory failure, joint contractures, and scoliosis without any symptoms of functional brain anomalies caused by variants in the MEGF10 gene. Here, we report the clinical phenotype and genetic features of a Moroccan patient who carries a novel variant associated with EMARDD on the MEGF10 gene. The Whole Exome Sequencing analysis conducted on a 11 year old boy with respiratory and swallowing difficulties revealed the presence of the novel variant c.978T>A (p.Cys326Ter) on exon 9 of the MEGF10 gene; this variant is thought to be associated with EMARDD. Our study reports the first nonsense pathogenic de novo variant in MEGF10 associated with EMARDD worldwide, identified in a Moroccan patient.
Collapse
Affiliation(s)
- Loubna Darfallah
- Laboratory of Genomics and Human Genetics, Pasteur Institute of Morocco, Casablanca, Morocco; Laboratory of Virology, Oncology, Biosciences, Environment and New Energies, Faculty of Science and Technology Mohammedia, University Hassan II Casablanca, Morocco
| | - Najat Sifeddine
- Laboratory of Genomics and Human Genetics, Pasteur Institute of Morocco, Casablanca, Morocco
| | - Ghita Amalou
- Laboratory of Genomics and Human Genetics, Pasteur Institute of Morocco, Casablanca, Morocco
| | - Chaimaa Ait El Cadi
- Laboratory of Genomics and Human Genetics, Pasteur Institute of Morocco, Casablanca, Morocco; Laboratory of Virology, Oncology, Biosciences, Environment and New Energies, Faculty of Science and Technology Mohammedia, University Hassan II Casablanca, Morocco
| | - Hamid Lakhiari
- Laboratory of Virology, Oncology, Biosciences, Environment and New Energies, Faculty of Science and Technology Mohammedia, University Hassan II Casablanca, Morocco
| | - Abdelhamid Barakat
- Laboratory of Genomics and Human Genetics, Pasteur Institute of Morocco, Casablanca, Morocco
| | - Hassan Rouba
- Laboratory of Genomics and Human Genetics, Pasteur Institute of Morocco, Casablanca, Morocco.
| |
Collapse
|
14
|
Liu Y, Li S, Robertson R, Granet JA, Aubry I, Filippelli RL, Tremblay ML, Chang NC. PTPN1/2 inhibition promotes muscle stem cell differentiation in Duchenne muscular dystrophy. Life Sci Alliance 2025; 8:e202402831. [PMID: 39477543 PMCID: PMC11527974 DOI: 10.26508/lsa.202402831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal disease caused by mutations in the DMD gene that encodes dystrophin. Dystrophin deficiency also impacts muscle stem cells (MuSCs), resulting in impaired asymmetric stem cell division and myogenic commitment. Using MuSCs from DMD patients and the DMD mouse model mdx, we found that PTPN1 phosphatase expression is up-regulated and STAT3 phosphorylation is concomitantly down-regulated in DMD MuSCs. To restore STAT3-mediated myogenic signaling, we examined the effect of K884, a novel PTPN1/2 inhibitor, on DMD MuSCs. Treatment with K884 enhanced STAT3 phosphorylation and promoted myogenic differentiation of DMD patient-derived MuSCs. In MuSCs from mdx mice, K884 treatment increased the number of asymmetric cell divisions, correlating with enhanced myogenic differentiation. Interestingly, the pro-myogenic effect of K884 is specific to human and murine DMD MuSCs and is absent from control MuSCs. Moreover, PTPN1/2 loss-of-function experiments indicate that the pro-myogenic impact of K884 is mediated mainly through PTPN1. We propose that PTPN1/2 inhibition may serve as a therapeutic strategy to restore the myogenic function of MuSCs in DMD.
Collapse
MESH Headings
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Animals
- Cell Differentiation/drug effects
- Humans
- Mice
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics
- Mice, Inbred mdx
- STAT3 Transcription Factor/metabolism
- Stem Cells/metabolism
- Stem Cells/cytology
- Muscle Development/genetics
- Muscle Development/drug effects
- Disease Models, Animal
- Phosphorylation
- Signal Transduction/drug effects
- Muscle, Skeletal/metabolism
Collapse
Affiliation(s)
- Yiyang Liu
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Shulei Li
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Rebecca Robertson
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Jules A Granet
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Isabelle Aubry
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Romina L Filippelli
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Michel L Tremblay
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Natasha C Chang
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- Goodman Cancer Institute, McGill University, Montréal, Canada
| |
Collapse
|
15
|
Yoshida Y, Shimizu K, Nakamura H, Fujii Y, Fritsch T, Abdelhameed A, Calabrese V, Osakabe N. An immunohistochemical evaluation of fast twitch muscle formation induced by repeated administration of flavan 3-ols in mice. FASEB J 2024; 38:e70193. [PMID: 39714246 DOI: 10.1096/fj.202401865r] [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: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 12/24/2024]
Abstract
Flavan-3-ols (FL) are poorly bioavailable astringent polyphenols that induce hyperactivation of the sympathetic nervous system. The aim of this study was to investigate the effects of repeated oral administration of FL on mice hindlimb skeletal muscle using immunohistochemical techniques. C57BL/6J male mice were orally administered 50 mg/kg of FL for a period of 2 weeks, and bromideoxyuridine (BrdU) was administered intraperitoneally 3 days prior to the dissection. The soleus and extensor digitorum longus (EDL) were excised and prepared for frozen sections. Myosin heavy chain (MHC) antibodies were used to classify muscle types, in addition, muscle cross-sectional areas (CSA) were measured. We observed a shift in the peak of CSA in the soleus muscle and to a larger extent in the EDL. In addition, a distinct shift toward fast muscle was detected, documented by a reduction in type I and an increase in type IIb in the soleus muscle, whereas in the EDL, we observed a decline in type IIa and an expansion in type IIb. Incorporation of BrdU into cells was significantly increased in all skeletal muscles, with a significant increase in cells co-expressing pair box 7 (Pax7), a marker of differentiation, as observed in the EDL. Given the evidence that β2-adrenergic receptors in skeletal muscles regulate differentiation and size, we measured plasma catecholamine (CA) concentrations following a single differentiation of FL. A single oral dose of FL was observed to significantly increase plasma CA. These findings indicate that catecholamines secreted into the bloodstream from the adrenal gland following oral administration of FL may influence skeletal muscle size and type via β2-receptors.
Collapse
Affiliation(s)
- Yamato Yoshida
- Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
| | - Kenta Shimizu
- Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
| | - Hitomi Nakamura
- Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
| | - Yasuyuki Fujii
- Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
| | | | - Ali Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Naomi Osakabe
- Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
- Department of Bioscience and Engineering, Faculty of System Science and Engineering, Shibaura Institute of Technology, Saitama, Japan
| |
Collapse
|
16
|
Osiak-Wicha C, Kras K, Arciszewski MB. Comparative Analysis of Muscle Fibers in Selected Muscles of Working and Companion Dog Breeds. Animals (Basel) 2024; 14:3576. [PMID: 39765480 PMCID: PMC11672843 DOI: 10.3390/ani14243576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
The structural and functional characteristics of skeletal muscle fibers play a crucial role in understanding the physical capabilities of dogs, particularly in relation to their breed-specific roles. This study aimed to compare the muscle fiber composition of working and companion dog breeds by analyzing the triceps brachii and biceps femoris muscles, focusing on fiber morphology, myosin heavy chain (MYH) isoform distribution, and nuclei per fiber. A total of 12 dogs, divided equally into working and companion breed groups, were used in this study. Muscle samples were collected post-mortem and prepared for histological analysis using cryosectioning. Immunohistochemical staining was employed to identify the expression of MYH isoforms, including MYH2, MYH4, and MYH7, which correspond to type IIa, IIb, and type I fibers, respectively. The results demonstrated significant differences between the two breed groups. Working dogs exhibited larger muscle fibers, a higher proportion of type IIa (MYH2) and type I (MYH7) fibers, and a greater number of nuclei per fiber, suggesting adaptations for endurance and strength. In contrast, companion dogs showed a higher proportion of type IIb (MYH4) fibers, indicative of their capacity for short bursts of activity rather than sustained exertion. Companion breeds also displayed a higher fiber density but fewer nuclei per fiber, which may contribute to slower muscle regeneration. These findings may provide insights into the muscle adaptations of dogs based on their breed-specific functional demands and highlight the importance of considering these differences in veterinary care and rehabilitation. The study underscores the influence of selective breeding on muscle structure and function in dogs and suggests further research into breed-specific muscle recovery mechanisms.
Collapse
Affiliation(s)
- Cezary Osiak-Wicha
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-950 Lublin, Poland; (K.K.); (M.B.A.)
| | | | | |
Collapse
|
17
|
Li X, Cao Y, Liu Y, Fang W, Xiao C, Cao Y, Zhao Y. Effect of IGF1 on Myogenic Proliferation and Differentiation of Bovine Skeletal Muscle Satellite Cells Through PI3K/AKT Signaling Pathway. Genes (Basel) 2024; 15:1494. [PMID: 39766763 PMCID: PMC11675145 DOI: 10.3390/genes15121494] [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: 11/01/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Cultivated meat, an alternative to conventional meat, has substantial potential for alleviating environmental and ethical concerns. This method of manufacturing meat involves the isolation of skeletal muscle satellite cells (SMSCs) from donor animals, after which they proliferate in vitro and differentiate into primitive muscle fibers. The aim of this research was to evaluate how the insulin-like growth factor 1 (IGF1) gene regulates the myogenic differentiation of bovine skeletal muscle satellite cells (bSMSCs). Methods: bSMSCs isolated from newborn calves were cultured to the third generation in vitro and differentiated into myoblasts via the serum withdrawal method. An overexpression lentivirus and siRNA targeting the IGF1 gene were constructed and transduced into bSMSCs, which were subsequently analyzed via real-time fluorescence quantitative PCR(qRT-PCR) and Western blots. The mRNA and protein levels of the myogenic differentiation markers myosin heavy chain (MyHC) and myogenin (MyoG) were determined. Results: The results revealed that the lentivirus overexpressing the IGF1 gene significantly increased the expression of MyHC and MyoG, whereas the expression of both the MyHC and MyoG mRNAs and proteins was strongly reduced by si-IGF1. Conclusions: IGF1 positively regulates the myogenic differentiation of bSMSCs. This study provides a reference for further elucidating the molecular mechanism by which the IGF1 gene regulates the myogenic differentiation of bSMSCs via the PI3K/Akt signaling pathway and lays a foundation for establishing a regulatory network of bovine muscle growth and development.
Collapse
Affiliation(s)
- Xin Li
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| | - Yang Cao
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| | - Yu Liu
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| | - Wenwen Fang
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| | - Cheng Xiao
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| | - Yang Cao
- Institute of Animal Biotechnology, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China
| | - Yumin Zhao
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| |
Collapse
|
18
|
Di Pietro E, Burla R, La Torre M, González-García MP, Dello Ioio R, Saggio I. Telomeres: an organized string linking plants and mammals. Biol Direct 2024; 19:119. [PMID: 39568075 PMCID: PMC11577926 DOI: 10.1186/s13062-024-00558-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/03/2024] [Indexed: 11/22/2024] Open
Abstract
Telomeres are pivotal determinants of cell stemness, organismal aging, and lifespan. Herein, we examined similarities in telomeres of Arabidopsis thaliana, mice, and humans. We report the common traits, which include their composition in multimers of TTAGGG sequences and their protection by specialized proteins. Moreover, given the link between telomeres, on the one hand, and cell proliferation and stemness on the other, we discuss the counterintuitive convergence between plants and mammals in this regard, focusing on the impact of niches on cell stemness. Finally, we suggest that tackling the study of telomere function and cell stemness by taking into consideration both plants and mammals can aid in the understanding of interconnections and contribute to research focusing on aging and organismal lifespan determinants.
Collapse
Affiliation(s)
- Edison Di Pietro
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza, University of Rome, Rome, Italy
| | - Romina Burla
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza, University of Rome, Rome, Italy
- CNR Institute of Biology and Pathology, Rome, Italy
| | - Mattia La Torre
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza, University of Rome, Rome, Italy
| | - Mary-Paz González-García
- Centro de Biotecnología y Genómica de Plantas (Universidad Politécnica de Madrid - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria), UPM-INIA/CSIC. Campus de Montegancedo, Pozuelo de Alarcón, 28223, Madrid, Spain
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Raffaele Dello Ioio
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza, University of Rome, Rome, Italy.
| | - Isabella Saggio
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza, University of Rome, Rome, Italy.
| |
Collapse
|
19
|
Lee DH, Lee HJ, Yang G, Kim DY, Kim JU, Yook TH, Lee JH, Kim HJ. A novel treatment strategy targeting cellular pathways with natural products to alleviate sarcopenia. Phytother Res 2024; 38:5033-5051. [PMID: 39099170 DOI: 10.1002/ptr.8301] [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: 12/26/2023] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/06/2024]
Abstract
Sarcopenia is a condition marked by a significant reduction in muscle mass and strength, primarily due to the aging process, which critically impacts muscle protein dynamics, metabolic functions, and overall physical functionality. This condition leads to increased body fat and reduced daily activity, contributing to severe health issues and a lower quality of life among the elderly. Recognized in the ICD-10-CM only in 2016, sarcopenia lacks definitive treatment options despite its growing prevalence and substantial social and economic implications. Given the aging global population, addressing sarcopenia has become increasingly relevant and necessary. The primary causes include aging, cachexia, diabetes, and nutritional deficiencies, leading to imbalances in protein synthesis and degradation, mitochondrial dysfunction, and hormonal changes. Exercise remains the most effective intervention, but it is often impractical for individuals with limited mobility, and pharmacological options such as anabolic steroids and myostatin inhibitors are not FDA-approved and are still under investigation. This review is crucial as it examines the potential of natural products as a novel treatment strategy for sarcopenia, targeting multiple mechanisms involved in its pathogenesis. By exploring natural products' multi-targeted effects, this study aims to provide innovative and practical solutions for sarcopenia management. Therefore, this review indicates significant improvements in muscle mass and function with the use of specific natural compounds, suggesting promising alternatives for those unable to engage in regular physical activity.
Collapse
Affiliation(s)
- Da Hee Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Hye Jin Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Gabsik Yang
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Dae Yong Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Jong Uk Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Tae Han Yook
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Jun Ho Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
- Da Capo Co., Ltd., Jeonju-si, Republic of Korea
| | - Hong Jun Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| |
Collapse
|
20
|
Haliloğlu G, Donkervoort S, Yıldız SÖ, Potticary A, Hu Y, Pais L, Carlier RY, Amthor H, Aydıngöz Ü, Bönnemann CG. Distinct whole-body muscle MRI imaging patterns in PAX7-congenital myopathy: A case report. J Neuromuscul Dis 2024; 11:1276-1282. [PMID: 39967430 DOI: 10.1177/22143602241289705] [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: 02/20/2025]
Abstract
PAX7 is a myogenesis transcription factor important for satellite cell specification and function and thus involved in muscle growth, maintenance, repair and regeneration. Recently, a new autosomal recessive congenital myopathy was described that is caused by biallelic variants in PAX7. Our aim is to describe phenotype and whole-body muscle MRI with follow-up imaging findings in a patient with a novel homozygous missense variant in PAX7. We also compare our patients' imaging features with a patient reported in the initial study, to identify a possible emerging pattern for PAX7-congenital myopathy. Generalized muscle hypotrophy and selective sternocleidomastoid, paraspinal and thigh muscle involvement emerge as suggestive findings and could serve as a recognizable fingerprint to differentiate early-onset myopathies within the emerging group of 'primary satellite cell-opathies'.
Collapse
Affiliation(s)
- Göknur Haliloğlu
- Department of Pediatrics, Division of Pediatric Neurology, Hacettepe University Faculty of Medicine, Ankara, Turkey
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Sibel Öz Yıldız
- Department of Pediatrics, Division of Pediatric Neurology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Abigail Potticary
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Lynn Pais
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert Y Carlier
- Department of Radiology, Assistance Publique-Hôpitaux de Paris (AP-HP), DMU Smart Imaging, Raymond Poincaré Teaching Hospital, Garches, France
| | - Helge Amthor
- Service de Neurologie et Ré animation Pédiatriques, AP-HP. Université Paris-Saclay, Hôpital Raymond Poincaré, Garches, France
| | - Üstün Aydıngöz
- Department of Radiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
21
|
Wang T, Zhou D, Hong Z. Adipose tissue in older individuals: a contributing factor to sarcopenia. Metabolism 2024; 160:155998. [PMID: 39128607 DOI: 10.1016/j.metabol.2024.155998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
Sarcopenia is a geriatric syndrome characterized by a functional decline in muscle. The prevalence of sarcopenia increases with natural aging, becoming a serious health problem among elderly individuals. Therefore, understanding the pathology of sarcopenia is critical for inhibiting age-related alterations and promoting health and longevity in elderly individuals. The development of sarcopenia may be influenced by interactions between visceral and subcutaneous adipose tissue and skeletal muscle, particularly under conditions of chronic low-grade inflammation and metabolic dysfunction. This hypothesis is supported by the following observations: (i) accumulation of senescent cells in both adipose tissue and skeletal muscle with age; (ii) gut dysbiosis, characterized by an imbalance in gut microbial communities as the main trigger for inflammation, sarcopenia, and aged adipose tissue; and (iii) microbial dysbiosis, which could impact the onset or progression of a senescent state. Moreover, adipose tissue acts as an endocrine organ, releasing molecules that participate in intricate communication networks between organs. Our discussion focuses on novel adipokines and their role in regulating adipose tissue and muscle, particularly those influenced by aging and obesity, emphasizing their contributions to disease development. On the basis of these findings, we propose that age-related adipose tissue and sarcopenia are disorders characterized by chronic inflammation and metabolic dysregulation. Finally, we explore new potential therapeutic strategies involving specialized proresolving mediator (SPM) G protein-coupled receptor (GPCR) agonists, non-SPM GPCR agonists, transient receptor potential (TRP) channels, antidiabetic drugs in conjunction with probiotics and prebiotics, and compounds designed to target senescent cells and mitigate their pro-inflammatory activity.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China.
| | - Dong Zhou
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China
| | - Zhen Hong
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
22
|
Sun C, Swoboda CO, Morales FM, Calvo C, Petrany MJ, Parameswaran S, VonHandorf A, Weirauch MT, Lepper C, Millay DP. Lineage tracing of nuclei in skeletal myofibers uncovers distinct transcripts and interplay between myonuclear populations. Nat Commun 2024; 15:9372. [PMID: 39477931 PMCID: PMC11526147 DOI: 10.1038/s41467-024-53510-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Multinucleated skeletal muscle cells need to acquire additional nuclei through fusion with activated skeletal muscle stem cells when responding to both developmental and adaptive growth stimuli. A fundamental question in skeletal muscle biology has been the reason underlying this need for new nuclei in cells that already harbor hundreds of nuclei. Here we utilize nuclear RNA-sequencing approaches and develop a lineage tracing strategy capable of defining the transcriptional state of recently fused nuclei and distinguishing this state from that of pre-existing nuclei. Our findings reveal the presence of conserved markers of newly fused nuclei both during development and after a hypertrophic stimulus in the adult. However, newly fused nuclei also exhibit divergent gene expression that is determined by the myogenic environment to which they fuse. Moreover, accrual of new nuclei through fusion is required for nuclei already resident in adult myofibers to mount a normal transcriptional response to a load-inducing stimulus. We propose a model of mutual regulation in the control of skeletal muscle development and adaptations, where newly fused and pre-existing myonuclear populations influence each other to maintain optimal functional growth.
Collapse
Affiliation(s)
- Chengyi Sun
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Casey O Swoboda
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Fabian Montecino Morales
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Cristofer Calvo
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael J Petrany
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sreeja Parameswaran
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew VonHandorf
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Christoph Lepper
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
23
|
Han J, Zhang J, Zhang X, Luo W, Liu L, Zhu Y, Liu Q, Zhang XA. Emerging role and function of Hippo-YAP/TAZ signaling pathway in musculoskeletal disorders. Stem Cell Res Ther 2024; 15:386. [PMID: 39468616 PMCID: PMC11520482 DOI: 10.1186/s13287-024-04011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024] Open
Abstract
Hippo pathway is an evolutionarily conservative key pathway that regulates organ size and tissue regeneration by regulating cell proliferation, differentiation and apoptosis. Yes-associated protein 1 (YAP)/ WW domain-containing transcription regulator 1 (TAZ) serves as a pivotal transcription factor within the Hippo signaling pathway, which undergoes negative regulation by the Hippo pathway. The expression of YAP/TAZ affects various biological processes, including differentiation of osteoblasts (OB) and osteoclasts (OC), cartilage homeostasis, skeletal muscle development, regeneration and quality maintenance. At the same time, the dysregulation of the Hippo pathway can concurrently contribute to the development of various musculoskeletal disorders, including bone tumors, osteoporosis (OP), osteoarthritis (OA), intervertebral disc degeneration (IDD), muscular dystrophy, and rhabdomyosarcoma (RMS). Therefore, targeting the Hippo pathway has emerged as a promising therapeutic strategy for the treatment of musculoskeletal disorders. The focus of this review is to elucidate the mechanisms by which the Hippo pathway maintains homeostasis in bone, cartilage, and skeletal muscle, while also providing a comprehensive summary of the pivotal role played by core components of this pathway in musculoskeletal diseases. The efficacy and feasibility of Hippo pathway-related drugs for targeted therapy of musculoskeletal diseases are also discussed in our study. These endeavors offer novel insights into the application of Hippo signaling in musculoskeletal disorders, providing effective therapeutic targets and potential drug candidates for treating such conditions.
Collapse
Affiliation(s)
- Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Jiale Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Xiaoyi Zhang
- College of Second Clinical Medical, China Medical University, Shenyang, 110122, China
| | - Wenxin Luo
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Lifei Liu
- Department of Rehabilitation, The People's Hospital of Liaoning Province, Shenyang, 110016, China
| | - Yuqing Zhu
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Qingfeng Liu
- Department of General Surgery, Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China
| | - Xin-An Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China.
| |
Collapse
|
24
|
Banik O, Salve AL, Kumar P, Kumar S, Banoth E. Electrically conductive nanomaterials: transformative applications in biomedical engineering-a review. NANOTECHNOLOGY 2024; 36:022001. [PMID: 39389095 DOI: 10.1088/1361-6528/ad857d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/10/2024] [Indexed: 10/12/2024]
Abstract
In recent years, significant advancements in nanotechnology have improved the various disciplines of scientific fields. Nanomaterials, like, carbon-based (carbon nanotubes, graphene), metallic, metal oxides, conductive polymers, and 2D materials (MXenes) exhibit exceptional electrical conductivity, mechanical strength, flexibility, thermal property and chemical stability. These materials hold significant capability in transforming material science and biomedical engineering by enabling the creation of more efficient, miniaturized, and versatile devices. The indulgence of nanotechnology with conductive materials in biological fields promises a transformative innovation across various industries, from bioelectronics to environmental regulations. The conductivity of nanomaterials with a suitable size and shape exhibits unique characteristics, which provides a platform for realization in bioelectronics as biosensors, tissue engineering, wound healing, and drug delivery systems. It can be explored for state-of-the-art cardiac, skeletal, nerve, and bone scaffold fabrication while highlighting their proof-of-concept in the development of biosensing probes and medical imaging. This review paper highlights the significance and application of the conductive nanomaterials associated with conductivity and their contribution towards a new perspective in improving the healthcare system globally.
Collapse
Affiliation(s)
- Oindrila Banik
- Opto-Biomedical Microsystems Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, India
| | - Amol Lalchand Salve
- Opto-Biomedical Microsystems Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, India
| | - Prasoon Kumar
- BioDesign and Medical Devices, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, India
| | - Santosh Kumar
- Department of Electronics and Communication Engineering, Centre of Excellence for Nanotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Andhra Pradesh 522302, India
| | - Earu Banoth
- Opto-Biomedical Microsystems Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, India
| |
Collapse
|
25
|
Lu Y, Walji T, Pandey P, Zhou C, Habela CW, Snapper SB, Li R, Chen EH. Branched actin polymerization drives invasive protrusion formation to promote myoblast fusion during skeletal muscle regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.30.615960. [PMID: 39416162 PMCID: PMC11482830 DOI: 10.1101/2024.09.30.615960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Skeletal muscle regeneration is a multistep process involving the activation, proliferation, differentiation, and fusion of muscle stem cells, known as satellite cells. The fusion of satellite cell-derived mononucleated muscle cells (SCMs) is indispensable for the generation of multinucleated, contractile myofibers during muscle repair. However, the molecular and cellular mechanisms underlying SCM fusion during muscle regeneration remain poorly understood. In this study, we uncovered an essential role for branched actin polymerization in SCM fusion. Using conditional knockouts of the Arp2/3 complex and its actin nucleation-promoting factors, N-WASP and WAVE, we demonstrated that branched actin polymerization is required for the SCM fusion, but not for satellite cell proliferation, differentiation, and migration. We showed that the N-WASP and WAVE complexes have partially redundant functions in regulating SCM fusion. Furthermore, we showed that branched actin polymerization is essential for generating invasive protrusions at the fusogenic synapses in SCMs. Taken together, our study has identified new components of the myoblast fusion machinery in skeletal muscle regeneration and demonstrated a critical role for branched actin-propelled invasive protrusions in this process.
Collapse
Affiliation(s)
- Yue Lu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tezin Walji
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pratima Pandey
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chuanli Zhou
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christa W. Habela
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Scott B. Snapper
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
| | - Rong Li
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Elizabeth H. Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
26
|
Chen X, Müller A, Pishnamaz M, Hildebrand F, Bollheimer LC, Nourbakhsh M. Differential Fatty Acid Response of Resident Macrophages in Human Skeletal Muscle Fiber and Intermuscular Adipose Tissue. Int J Mol Sci 2024; 25:10722. [PMID: 39409051 PMCID: PMC11477279 DOI: 10.3390/ijms251910722] [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: 09/12/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Human skeletal muscle contains different types of tissues with skeletal muscle fibers (SMFs) and intermuscular adipose tissues (IMATs) as the main components. We maintained human skeletal muscle tissues from 12 study participants under native conditions in vitro for 11 days to investigate the dynamics of macrophages that reside in adjacent IMATs and SMFs simultaneously. The samples were subjected to immunohistochemical analysis for macrophage phenotyping and mitochondrial mass assessment before and after maintenance in vitro. Multiplex protein analysis was used to determine cytokine/chemokine expression in tissue extracts. The results revealed significant correlations between donor age or body mass index (BMI) and distinct phenotypes of resident macrophages in SMFs and IMATs. The dynamics of SMF- and IMAT-resident macrophages differed significantly in vitro and exhibited inverse correlations with chemokine/cytokine expression levels and mitochondrial activity. Moreover, the responses of macrophages to saturated and unsaturated fatty acids (FAs) differed substantially between SMFs and IMATs. These findings showed the functional diversity of phenotypically identical macrophages in adjacent niches. Thus, the currently available macrophage markers cannot capture the functional diversity of human tissue-resident macrophages. The model used in the present study may help elucidate how macrophages affect muscle homeostasis and disease in humans.
Collapse
Affiliation(s)
- Xiaoying Chen
- Clinic for Geriatric Medicine, RWTH Aachen University Hospital, 52074 Aachen, Germany; (X.C.); (A.M.); (L.C.B.)
| | - Aline Müller
- Clinic for Geriatric Medicine, RWTH Aachen University Hospital, 52074 Aachen, Germany; (X.C.); (A.M.); (L.C.B.)
| | - Miguel Pishnamaz
- Clinic for Orthopedics, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (M.P.); (F.H.)
| | - Frank Hildebrand
- Clinic for Orthopedics, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (M.P.); (F.H.)
| | - Leo Cornelius Bollheimer
- Clinic for Geriatric Medicine, RWTH Aachen University Hospital, 52074 Aachen, Germany; (X.C.); (A.M.); (L.C.B.)
| | - Mahtab Nourbakhsh
- Clinic for Geriatric Medicine, RWTH Aachen University Hospital, 52074 Aachen, Germany; (X.C.); (A.M.); (L.C.B.)
| |
Collapse
|
27
|
Tollance A, Prola A, Michel D, Bouche A, Turzi A, Hannouche D, Berndt S, Laumonier T. Platelet-Rich Plasma Promotes the Expansion of Human Myoblasts and Favors the In Vitro Generation of Human Muscle Reserve Cells in a Deeper State of Quiescence. Stem Cell Rev Rep 2024; 20:1981-1994. [PMID: 39001964 PMCID: PMC11445347 DOI: 10.1007/s12015-024-10760-0] [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] [Accepted: 06/30/2024] [Indexed: 07/15/2024]
Abstract
Stem cell therapy holds significant potential for skeletal muscle repair, with in vitro-generated human muscle reserve cells (MuRCs) emerging as a source of quiescent myogenic stem cells that can be injected to enhance muscle regeneration. However, the clinical translation of such therapies is hampered by the need for fetal bovine serum (FBS) during the in vitro generation of human MuRCs. This study aimed to determine whether fresh allogeneic human platelet-rich plasma (PRP) combined or not with hyaluronic acid (PRP-HA) could effectively replace xenogeneic FBS for the ex vivo expansion and differentiation of human primary myoblasts. Cells were cultured in media supplemented with either PRP or PRP-HA and their proliferation rate, cytotoxicity and myogenic differentiation potential were compared with those cultured in media supplemented with FBS. The results showed similar proliferation rates among human myoblasts cultured in PRP, PRP-HA or FBS supplemented media, with no cytotoxic effects. Human myoblasts cultured in PRP or PRP-HA showed reduced fusion ability upon differentiation. Nevertheless, we also observed that human MuRCs generated from PRP or PRP-HA myogenic cultures, exhibited increased Pax7 expression and delayed re-entry into the cell cycle upon reactivation, indicating a deeper quiescent state of human MuRCs. These results suggest that allogeneic human PRP effectively replaces FBS for the ex vivo expansion and differentiation of human myoblasts and favors the in vitro generation of Pax7High human MuRCs, with important implications for the advancement of stem cell-based muscle repair strategies.
Collapse
Affiliation(s)
- Axel Tollance
- Department of Orthopedic Surgery, Geneva University Hospitals & Faculty of Medicine, Geneva, Switzerland
- Regen Lab SA, 1052, Le Mont-Sur-Lausanne, Switzerland
| | - Alexandre Prola
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland
| | - Diego Michel
- Department of Orthopedic Surgery, Geneva University Hospitals & Faculty of Medicine, Geneva, Switzerland
| | - Axelle Bouche
- Department of Orthopedic Surgery, Geneva University Hospitals & Faculty of Medicine, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland
| | - Antoine Turzi
- Regen Lab SA, 1052, Le Mont-Sur-Lausanne, Switzerland
| | - Didier Hannouche
- Department of Orthopedic Surgery, Geneva University Hospitals & Faculty of Medicine, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland
| | - Sarah Berndt
- Regen Lab SA, 1052, Le Mont-Sur-Lausanne, Switzerland
| | - Thomas Laumonier
- Department of Orthopedic Surgery, Geneva University Hospitals & Faculty of Medicine, Geneva, Switzerland.
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland.
| |
Collapse
|
28
|
Zheng H, Yang Z, Zhou L, Zhang B, Cheng R, Zhang Q. Bioactive Nb 2C MXene-Functionalized Hydrogel with Microenvironment Remodeling and Enhanced Neurogenesis to Promote Skeletal Muscle Regeneration and Functional Restoration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310483. [PMID: 39254284 DOI: 10.1002/smll.202310483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/18/2024] [Indexed: 09/11/2024]
Abstract
The complete structure-functional repair of volumetric muscle loss (VML) remains a giant challenge and biomedical hydrogels to remodel microenvironment and enhance neurogenesis have appeared to be a promising direction. However, the current hydrogels for VML repair hardly achieve these two goals simultaneously due to their insufficient functionality and the challenge in high-cost of bioactive factors. In this study, a facile strategy using Nb2C MXene-functionalized hydrogel (OPTN) as a bioactive scaffold is proposed to promote VML repair with skeletal muscle regeneration and functional restoration. In vitro experiments show that OPTN scaffold can effectively scavenge reactive oxygen species (ROS), guide macrophages polarization toward M2 phenotype, and resist bacterial infection, providing a favorable microenvironment for myoblasts proliferation as well as the endothelial cells proliferation, migration, and tube formation. More importantly, OPTN scaffold with electroactive feature remarkably boosts myoblasts differentiation and mesenchymal stem cells neural differentiation. Animal experiments further confirm that OPTN scaffold can achieve a prominent structure-functional VML repair by attenuating ROS levels, alleviating inflammation, reducing fibrosis, and facilitating angiogenesis, newborn myotube formation, and neurogenesis. Collectively, this study provides a highly promising and effective strategy for the structure-functional VML repair through designing bioactive multifunctional hydrogel with microenvironment remodeling and enhanced neurogenesis.
Collapse
Affiliation(s)
- Hua Zheng
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710129, P. R. China
- Xi'an Key Laboratory of Functional Organic Porous Materials, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Zuoting Yang
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710129, P. R. China
- Xi'an Key Laboratory of Functional Organic Porous Materials, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Li Zhou
- Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Baoliang Zhang
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710129, P. R. China
- Xi'an Key Laboratory of Functional Organic Porous Materials, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Ruidong Cheng
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710129, P. R. China
- Xi'an Key Laboratory of Functional Organic Porous Materials, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Qiuyu Zhang
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710129, P. R. China
- Xi'an Key Laboratory of Functional Organic Porous Materials, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| |
Collapse
|
29
|
Sevegnani M, Lama A, Girardi F, Hess MW, Castelo MP, Pichler I, Biressi S, Piccoli G. Parkin R274W mutation affects muscle and mitochondrial physiology. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167302. [PMID: 38878834 DOI: 10.1016/j.bbadis.2024.167302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024]
Abstract
Recessive mutations in the Parkin gene (PRKN) are the most common cause of young-onset inherited parkinsonism. Parkin is a multifunctional E3 ubiquitin ligase that plays a variety of roles in the cell including the degradation of proteins and the maintenance of mitochondrial homeostasis, integrity, and biogenesis. In 2001, the R275W mutation in the PRKN gene was identified in two unrelated families with a multigenerational history of postural tremor, dystonia and parkinsonism. Drosophila models of Parkin R275W showed selective and progressive degeneration of dopaminergic neuronal clusters, mitochondrial abnormalities, and prominent climbing defects. In the Prkn mouse orthologue, the amino acid R274 corresponds to human R275. Here we described an age-related motor impairment and a muscle phenotype in R274W +/+ mice. In vitro, Parkin R274W mutation correlates with abnormal myoblast differentiation, mitochondrial defects, and alteration in mitochondrial mRNA and protein levels. Our data suggest that the Parkin R274W mutation may impact mitochondrial physiology and eventually myoblast proliferation and differentiation.
Collapse
Affiliation(s)
| | - Adriano Lama
- Department CIBIO, University of Trento, Trento, Italy
| | | | - Michael W Hess
- Innsbruck Medical University, Institute of Histology and Embryology, Medical University of Innsbruck, Austria
| | - Maria Paulina Castelo
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Irene Pichler
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | | | | |
Collapse
|
30
|
Saliu TP, Goh J, Kang G, Burke BI, Ismaeel A, McCarthy JJ. Satellite cell dynamics during skeletal muscle hypertrophy. Biochem Soc Trans 2024; 52:1921-1926. [PMID: 39136196 PMCID: PMC11660404 DOI: 10.1042/bst20240201] [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: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/26/2024]
Abstract
Skeletal muscle stem cells (MuSCs) display distinct behavior crucial for tissue maintenance and repair. Upon activation, MuSCs exhibit distinct modes of division: symmetric division, facilitating either self-renewal or differentiation, and asymmetric division, which dictates divergent cellular fates. This review explores the nuanced dynamics of MuSC division and the molecular mechanisms governing this behavior. Furthermore, it introduces a novel phenomenon observed in a subset of MuSCs under hypertrophic stimuli termed division-independent differentiation. Insights into the underlying mechanisms driving this process are discussed, alongside its broader implications for muscle physiology.
Collapse
Affiliation(s)
- Tolulope P. Saliu
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
| | - Jensen Goh
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
| | - Gyumin Kang
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
- Division of Biomedical Informatics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
| | - Benjamin I. Burke
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
| | - Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
| | - John J. McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, U.S.A
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, U.S.A
| |
Collapse
|
31
|
Song Z, Lin Q, Liang J, Zhang W. Inhibition of Sesn2 has negative regulatory effects on the myogenic differentiation of C2C12 myoblasts. MOLECULAR BIOMEDICINE 2024; 5:31. [PMID: 39117956 PMCID: PMC11310181 DOI: 10.1186/s43556-024-00193-z] [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: 03/26/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
Sestrin2 (Sesn2) has been previously confirmed to be a stress-response molecule. However, the influence of Sesn2 on myogenic differentiation remains elusive. This study was conducted to analyze the role of Sesn2 in the myogenic differentiation of C2C12 myoblasts and related aspects in mdx mice, an animal model of Duchenne muscular dystrophy (DMD). Our results showed that knockdown of Sesn2 reduced the myogenic differentiation capacity of C2C12 myoblasts. Predictive analysis from two databases suggested that miR-182-5p is a potential regulator of Sesn2. Further experimental validation revealed that overexpression of miR-182-5p decreased both the protein and mRNA levels of Sesn2 and inhibited myogenesis of C2C12 myoblasts. These findings suggest that miR-182-5p negatively regulates myogenesis by repressing Sesn2 expression. Extending to an in vivo model of DMD, knockdown of Sesn2 led to decreased Myogenin (Myog) expression and increased Pax7 expression, while its overexpression upregulated Myog levels and enhanced the proportion of slow-switch myofibers. These findings indicate the crucial role of Sesn2 in promoting myogenic differentiation and skeletal muscle regeneration, providing potential therapeutic targets for muscular dystrophy.
Collapse
Affiliation(s)
- Zubiao Song
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Qing Lin
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Jiahui Liang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Weixi Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China.
| |
Collapse
|
32
|
Pimentel Neto J, Batista RD, Rocha-Braga LC, Chacur M, Camargo PO, Ciena AP. The telocytes relationship with satellite cells: Extracellular vesicles mediate the myotendinous junction remodeling. Microsc Res Tech 2024; 87:1733-1741. [PMID: 38501548 DOI: 10.1002/jemt.24549] [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: 08/24/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
The peripheral nerve injury (PNI) affects the morphology of the whole locomotor apparatus, which can reach the myotendinous junction (MTJ) interface. In the injury condition, the skeletal muscle satellite cells (SC) are triggered, activated, and proliferated to repair their structure, and in the MTJ, the telocytes (TC) are associated to support the interface with the need for remodeling; in that way, these cells can be associated with SC. The study aimed to describe the SC and TC relationship after PNI at the MTJ. Sixteen adult Wistar rats were divided into Control Group (C, n = 8) and PNI Group (PNI, n = 8), PNI was performed by the constriction of the sciatic nerve. The samples were processed for transmission electron microscopy and immunostaining analysis. In the C group was evidenced the arrangement of sarcoplasmic evaginations and invaginations, the support collagen layer with a TC inside it, and an SC through vesicles internally and externally to then. In the PNI group were observed the disarrangement of invaginations and evaginations and sarcomeres degradation at MTJ, as the disposition of telopodes adjacent and in contact to the SC with extracellular vesicles and exosomes in a characterized paracrine activity. These findings can determine a link between the TCs and the SCs at the MTJ remodeling. RESEARCH HIGHLIGHTS: Peripheral nerve injury promotes the myotendinous junction (MTJ) remodeling. The telocytes (TC) and the satellite cells (SC) are present at the myotendinous interface. TC mediated the SC activity at MTJ.
Collapse
Affiliation(s)
- Jurandyr Pimentel Neto
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | - Rodrigo Daniel Batista
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | - Lara Caetano Rocha-Braga
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | - Marucia Chacur
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Paula Oliveira Camargo
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | - Adriano Polican Ciena
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| |
Collapse
|
33
|
Fujii I, Kinoshita R, Akiyama H, Nakamura A, Iwamori K, Fukada SI, Honda H, Shimizu K. Discovery of fibroblast growth factor 2-derived peptides for enhancing mice skeletal muscle satellite cell proliferation. Biotechnol J 2024; 19:e2400278. [PMID: 39212202 DOI: 10.1002/biot.202400278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/20/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Skeletal muscle satellite cells (SCs) are essential for muscle regeneration. Their proliferation and differentiation are influenced by fibroblast growth factor (FGF)-2. In this study, we screened for FGF-2-derived peptides that promote SC proliferation. Utilizing photocleavable peptide array technology, a library of 7-residue peptides was synthesized, and its effect on SC proliferation was examined using a mixture of five peptides. The results showed that peptides 1-5 (136%), 21-25 (136%), 26-30 (141%), 31-35 (159%), 71-75 (135%), 76-80 (144%), and 126-130 (137%) significantly increased SC proliferation. Further experiments revealed that peptide 33, CKNGGFF, enhanced SC proliferation. Furthermore, its extended form, peptide 33-13, CKNGGFFLRIHPD, promoted SC proliferation and increased the percentage of Pax7-positive cells, indicating that SCs were maintained in an undifferentiated state. The addition of FGF-2 and peptide 33-13 further induced cell proliferation but did not increase the percentage of Pax7-positive cells. A proliferation assay using an FGF receptor (FGFR) inhibitor suggested that peptide 33-13 acts through the FGFR-mediated and other pathways. Although further research is necessary to explore the mechanisms of action of these peptides and their potential for in vivo and in vitro use, the high sequence conservation of peptides 33 and 33-13 in FGF-2 across multiple species suggests their broad application prospects in biomedical engineering and biotechnology.
Collapse
Affiliation(s)
- Itsuki Fujii
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Remi Kinoshita
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Hirokazu Akiyama
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Ayasa Nakamura
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kanako Iwamori
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - So-Ichiro Fukada
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hiroyuki Honda
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| |
Collapse
|
34
|
Kitajima Y, Yoshioka K, Mikumo Y, Ohki S, Maehara K, Ohkawa Y, Ono Y. Loss of Tob1 promotes muscle regeneration through muscle stem cell expansion. J Cell Sci 2024; 137:jcs261886. [PMID: 39037211 DOI: 10.1242/jcs.261886] [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: 12/11/2023] [Accepted: 06/17/2024] [Indexed: 07/23/2024] Open
Abstract
Muscle stem cells (MuSCs) play an indispensable role in postnatal muscle growth and hypertrophy in adults. MuSCs also retain a highly regenerative capacity and are therefore considered a promising stem cell source for regenerative therapy for muscle diseases. In this study, we identify tumor-suppressor protein Tob1 as a Pax7 target protein that negatively controls the population expansion of MuSCs. Tob1 protein is undetectable in the quiescent state but is upregulated during activation in MuSCs. Tob1 ablation in mice accelerates MuSC population expansion and boosts muscle regeneration. Moreover, inactivation of Tob1 in MuSCs ameliorates the efficiency of MuSC transplantation in a murine muscular dystrophy model. Collectively, selective targeting of Tob1 might be a therapeutic option for the treatment of muscular diseases, including muscular dystrophy and age-related sarcopenia.
Collapse
Affiliation(s)
- Yasuo Kitajima
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811, Japan
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Kiyoshi Yoshioka
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811, Japan
| | - Yoko Mikumo
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811, Japan
| | - Shun Ohki
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Kazumitsu Maehara
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811, Japan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
- Division of Biological Regulation, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
- Muscle Biology Laboratory, Research Team for Aging Science, Tokyo Metropolitan Institute for Geriatrics and Gerontology (TMIG), Tokyo, 173-0015, Japan
| |
Collapse
|
35
|
Kawashima M, Nagata I, Terada E, Tamari A, Kurauchi M, Sakuraya T, Sonomura T, Oyanagi E, Yano H, Peake JM, Arakawa T. Frequent Icing Stimulates Skeletal Muscle Regeneration Following Injury With Necrosis in a Small Fraction of Myofibers in Rats. J Histochem Cytochem 2024; 72:569-584. [PMID: 39240248 PMCID: PMC11456165 DOI: 10.1369/00221554241274882] [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: 05/16/2024] [Accepted: 07/19/2024] [Indexed: 09/07/2024] Open
Abstract
Icing interventions on the injured skeletal muscle affect the macrophage-related regenerative events and muscle repair. However, despite its importance for the practice in sport medicine, the influence of different icing protocols on muscle regeneration remains unclear. Here, using a rodent model of mild muscle injury with necrosis in a small fraction of myofibers, the injured animals were allocated to four groups: non-icing control (Con) and a single treatment (Ice-1), three treatments (Ice-3), or nine treatments (Ice-9) with a 30-min icing each time within two days following injury. Muscle regeneration was compared between the groups on post-injury days 1, 3, 5, and 7. The results showed that compared with the Con group, muscle regeneration was faster in the Ice-9 group (but not in the Ice-1 and Ice-3 groups), as indicated by more rapid accumulation of satellite cells within the regenerating area and enlarged size of regenerating myofibers (p<0.05, respectively). There was also less macrophage accumulation (p<0.05) and a trend toward early removal of necrotic myofibers in the damaged/regenerating area in the Ice-9 group (p=0.0535). These results demonstrate that in the case of mild muscle damage, more frequent icing treatment is more effective to stimulate muscle regeneration.
Collapse
Affiliation(s)
- Masato Kawashima
- Department of Health and Sports Science, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, Kurashiki, Japan
| | - Itsuki Nagata
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Erika Terada
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Asano Tamari
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Mami Kurauchi
- Faculty of Health Sciences, Kobe University School of Medicine, Kobe, Japan
- Dynamic Sports Medicine Institute, Osaka, Japan
| | - Tohma Sakuraya
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
- Department of Oral Anatomy, School of Dentistry, Asahi University, Gifu, Japan
| | - Takahiro Sonomura
- Department of Oral Anatomy, School of Dentistry, Asahi University, Gifu, Japan
| | - Eri Oyanagi
- Department of Health and Sports Science, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, Kurashiki, Japan
| | - Hiromi Yano
- Department of Health and Sports Science, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, Kurashiki, Japan
- Graduate School of Health Science and Technology, Kawasaki University of Medical Welfare, Kurashiki, Japan
| | - Jonathan M. Peake
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- Sport Performance Innovation and Knowledge Excellence, Queensland Academy of Sport, Brisbane, QLD, Australia
| | - Takamitsu Arakawa
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
36
|
Cao M, Sheng R, Sun Y, Cao Y, Wang H, Zhang M, Pu Y, Gao Y, Zhang Y, Lu P, Teng G, Wang Q, Rui Y. Delivering Microrobots in the Musculoskeletal System. NANO-MICRO LETTERS 2024; 16:251. [PMID: 39037551 PMCID: PMC11263536 DOI: 10.1007/s40820-024-01464-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/16/2024] [Indexed: 07/23/2024]
Abstract
Disorders of the musculoskeletal system are the major contributors to the global burden of disease and current treatments show limited efficacy. Patients often suffer chronic pain and might eventually have to undergo end-stage surgery. Therefore, future treatments should focus on early detection and intervention of regional lesions. Microrobots have been gradually used in organisms due to their advantages of intelligent, precise and minimally invasive targeted delivery. Through the combination of control and imaging systems, microrobots with good biosafety can be delivered to the desired area for treatment. In the musculoskeletal system, microrobots are mainly utilized to transport stem cells/drugs or to remove hazardous substances from the body. Compared to traditional biomaterial and tissue engineering strategies, active motion improves the efficiency and penetration of local targeting of cells/drugs. This review discusses the frontier applications of microrobotic systems in different tissues of the musculoskeletal system. We summarize the challenges and barriers that hinder clinical translation by evaluating the characteristics of different microrobots and finally point out the future direction of microrobots in the musculoskeletal system.
Collapse
Affiliation(s)
- Mumin Cao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Renwang Sheng
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yimin Sun
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
| | - Ying Cao
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
| | - Hao Wang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Ming Zhang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yunmeng Pu
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
| | - Yucheng Gao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yuanwei Zhang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Panpan Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Gaojun Teng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China.
| | - Qianqian Wang
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China.
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China.
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China.
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
37
|
Majchrzak K, Hentschel E, Hönzke K, Geithe C, von Maltzahn J. We need to talk-how muscle stem cells communicate. Front Cell Dev Biol 2024; 12:1378548. [PMID: 39050890 PMCID: PMC11266305 DOI: 10.3389/fcell.2024.1378548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Skeletal muscle is one of the tissues with the highest ability to regenerate, a finely controlled process which is critically depending on muscle stem cells. Muscle stem cell functionality depends on intrinsic signaling pathways and interaction with their immediate niche. Upon injury quiescent muscle stem cells get activated, proliferate and fuse to form new myofibers, a process involving the interaction of multiple cell types in regenerating skeletal muscle. Receptors in muscle stem cells receive the respective signals through direct cell-cell interaction, signaling via secreted factors or cell-matrix interactions thereby regulating responses of muscle stem cells to external stimuli. Here, we discuss how muscle stem cells interact with their immediate niche focusing on how this controls their quiescence, activation and self-renewal and how these processes are altered in age and disease.
Collapse
Affiliation(s)
- Karolina Majchrzak
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Erik Hentschel
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Katja Hönzke
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Christiane Geithe
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Julia von Maltzahn
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
- Faculty for Environment and Natural Sciences, Brandenburg University of Technology Cottbus—Senftenberg, Senftenberg, Germany
| |
Collapse
|
38
|
Beaudry K, De Lisio M. Sex-Based Differences in Muscle Stem Cell Regulation Following Exercise. Exerc Sport Sci Rev 2024; 52:87-94. [PMID: 38445901 DOI: 10.1249/jes.0000000000000337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Sexual dimorphism, driven by the sex hormones testosterone and estrogen, influences body composition, muscle fiber type, and inflammation. Research related to muscle stem cell (MuSC) responses to exercise has mainly focused on males. We propose a novel hypothesis that there are sex-based differences in MuSC regulation following exercise, such that males have more MuSCs, whereas females demonstrate a greater capacity for regeneration.
Collapse
Affiliation(s)
- Kayleigh Beaudry
- School of Human Kinetics , Department of Cellular and Molecular Medicine, Regenerative Medicine Program, Centre on Neuromuscular Disease , University of Ottawa, Ottawa, Ontario, Canada
| | | |
Collapse
|
39
|
Engquist EN, Greco A, Joosten LA, van Engelen BG, Banerji CR, Zammit PS. Transcriptomic gene signatures measure satellite cell activity in muscular dystrophies. iScience 2024; 27:109947. [PMID: 38840844 PMCID: PMC11150970 DOI: 10.1016/j.isci.2024.109947] [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: 12/12/2023] [Revised: 03/20/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
The routine need for myonuclear turnover in skeletal muscle, together with more sporadic demands for hypertrophy and repair, are performed by resident muscle stem cells called satellite cells. Muscular dystrophies are characterized by muscle wasting, stimulating chronic repair/regeneration by satellite cells. Here, we derived and validated transcriptomic signatures for satellite cells, myoblasts/myocytes, and myonuclei using publicly available murine single cell RNA-Sequencing data. Our signatures distinguished disease from control in transcriptomic data from several muscular dystrophies including facioscapulohumeral muscular dystrophy (FSHD), Duchenne muscular dystrophy, and myotonic dystrophy type I. For FSHD, the expression of our gene signatures correlated with direct counts of satellite cells on muscle sections, as well as with increasing clinical and pathological severity. Thus, our gene signatures enable the investigation of myogenesis in bulk transcriptomic data from muscle biopsies. They also facilitate study of muscle regeneration in transcriptomic data from human muscle across health and disease.
Collapse
Affiliation(s)
- Elise N. Engquist
- King’s College London, Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, London SE1 1UL, UK
| | - Anna Greco
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Geert Grooteplein Zuid 10, Nijmegen 6525 GA, The Netherlands
| | - Leo A.B. Joosten
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Geert Grooteplein Zuid 10, Nijmegen 6525 GA, The Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University if Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Baziel G.M. van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Christopher R.S. Banerji
- King’s College London, Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, London SE1 1UL, UK
- The Alan Turing Institute, The British Library, 96 Euston Road, London NW1 2DB, UK
- University College London Hospitals, NHS Foundation Trust, London NW1 2BU, UK
| | - Peter S. Zammit
- King’s College London, Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, London SE1 1UL, UK
| |
Collapse
|
40
|
Florczyk-Soluch U, Polak K, Jelinkova S, Bronisz-Budzyńska I, Sabo R, Bolisetty S, Agarwal A, Werner E, Józkowicz A, Stępniewski J, Szade K, Dulak J. Targeted expression of heme oxygenase-1 in satellite cells improves skeletal muscle pathology in dystrophic mice. Skelet Muscle 2024; 14:13. [PMID: 38867250 PMCID: PMC11167827 DOI: 10.1186/s13395-024-00346-2] [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: 12/21/2023] [Accepted: 06/03/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Adult muscle-resident myogenic stem cells, satellite cells (SCs), that play non-redundant role in muscle regeneration, are intrinsically impaired in Duchenne muscular dystrophy (DMD). Previously we revealed that dystrophic SCs express low level of anti-inflammatory and anti-oxidative heme oxygenase-1 (HO-1, HMOX1). Here we assess whether targeted induction of HMOX1 affect SC function and alleviates hallmark symptoms of DMD. METHODS We generated double-transgenic mouse model (mdx;HMOX1Pax7Ind) that allows tamoxifen (TX)-inducible HMOX1 expression in Pax7 positive cells of dystrophic muscles. Mdx;HMOX1Pax7Ind and control mdx mice were subjected to 5-day TX injections (75 mg/kg b.w.) followed by acute exercise protocol with high-speed treadmill (12 m/min, 45 min) and downhill running to worsen skeletal muscle phenotype and reveal immediate effects of HO-1 on muscle pathology and SC function. RESULTS HMOX1 induction caused a drop in SC pool in mdx;HMOX1Pax7Ind mice (vs. mdx counterparts), while not exaggerating the effect of physical exercise. Upon physical exercise, the proliferation of SCs and activated CD34- SC subpopulation, was impaired in mdx mice, an effect that was reversed in mdx;HMOX1Pax7Ind mice, however, both in vehicle- and TX-treated animals. This corresponded to the pattern of HO-1 expression in skeletal muscles. At the tissue level, necrotic events of selective skeletal muscles of mdx mice and associated increase in circulating levels of muscle damage markers were blunted in HO-1 transgenic animals which showed also anti-inflammatory cytokine profile (vs. mdx). CONCLUSIONS Targeted expression of HMOX1 plays protective role in DMD and alleviates dystrophic muscle pathology.
Collapse
MESH Headings
- Animals
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/metabolism
- Satellite Cells, Skeletal Muscle/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Mice, Inbred mdx
- Mice, Transgenic
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Mice
- PAX7 Transcription Factor/genetics
- PAX7 Transcription Factor/metabolism
- Male
- Mice, Inbred C57BL
- Physical Conditioning, Animal
- Membrane Proteins
Collapse
Affiliation(s)
- Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Katarzyna Polak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Sarka Jelinkova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Iwona Bronisz-Budzyńska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Reece Sabo
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ewa Werner
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
- Laboratory of Stem Cells Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
41
|
Espino-Gonzalez E, Dalbram E, Mounier R, Gondin J, Farup J, Jessen N, Treebak JT. Impaired skeletal muscle regeneration in diabetes: From cellular and molecular mechanisms to novel treatments. Cell Metab 2024; 36:1204-1236. [PMID: 38490209 DOI: 10.1016/j.cmet.2024.02.014] [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: 09/07/2023] [Revised: 01/10/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024]
Abstract
Diabetes represents a major public health concern with a considerable impact on human life and healthcare expenditures. It is now well established that diabetes is characterized by a severe skeletal muscle pathology that limits functional capacity and quality of life. Increasing evidence indicates that diabetes is also one of the most prevalent disorders characterized by impaired skeletal muscle regeneration, yet underlying mechanisms and therapeutic treatments remain poorly established. In this review, we describe the cellular and molecular alterations currently known to occur during skeletal muscle regeneration in people with diabetes and animal models of diabetes, including its associated comorbidities, e.g., obesity, hyperinsulinemia, and insulin resistance. We describe the role of myogenic and non-myogenic cell types on muscle regeneration in conditions with or without diabetes. Therapies for skeletal muscle regeneration and gaps in our knowledge are also discussed, while proposing future directions for the field.
Collapse
Affiliation(s)
- Ever Espino-Gonzalez
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Jean Farup
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
42
|
Collins BC, Shapiro JB, Scheib MM, Musci RV, Verma M, Kardon G. Three-dimensional imaging studies in mice identify cellular dynamics of skeletal muscle regeneration. Dev Cell 2024; 59:1457-1474.e5. [PMID: 38569550 PMCID: PMC11153043 DOI: 10.1016/j.devcel.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 12/06/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
The function of many organs, including skeletal muscle, depends on their three-dimensional structure. Muscle regeneration therefore requires not only reestablishment of myofibers but also restoration of tissue architecture. Resident muscle stem cells (SCs) are essential for regeneration, but how SCs regenerate muscle architecture is largely unknown. We address this problem using genetic labeling of mouse SCs and whole-mount imaging to reconstruct, in three dimensions, muscle regeneration. Unexpectedly, we found that myofibers form via two distinct phases of fusion and the residual basement membrane of necrotic myofibers is critical for promoting fusion and orienting regenerated myofibers. Furthermore, the centralized myonuclei characteristic of regenerated myofibers are associated with myofibrillogenesis and endure months post injury. Finally, we elucidate two cellular mechanisms for the formation of branched myofibers, a pathology characteristic of diseased muscle. We provide a synthesis of the cellular events of regeneration and show that these differ from those used during development.
Collapse
Affiliation(s)
- Brittany C Collins
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Jacob B Shapiro
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Mya M Scheib
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Robert V Musci
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Mayank Verma
- Department of Pediatrics, Division of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
43
|
Kuriki M, Korb A, Comai G, Tajbakhsh S. Interplay between Pitx2 and Pax7 temporally governs specification of extraocular muscle stem cells. PLoS Genet 2024; 20:e1010935. [PMID: 38875306 PMCID: PMC11178213 DOI: 10.1371/journal.pgen.1010935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/05/2024] [Indexed: 06/16/2024] Open
Abstract
Gene regulatory networks that act upstream of skeletal muscle fate determinants are distinct in different anatomical locations. Despite recent efforts, a clear understanding of the cascade of events underlying the emergence and maintenance of the stem cell pool in specific muscle groups remains unresolved and debated. Here, we invalidated Pitx2 with multiple Cre-driver mice prenatally, postnatally, and during lineage progression. We showed that this gene becomes progressively dispensable for specification and maintenance of the muscle stem (MuSC) cell pool in extraocular muscles (EOMs) despite being, together with Myf5, a major upstream regulator during early development. Moreover, constitutive inactivation of Pax7 postnatally led to a greater loss of MuSCs in the EOMs compared to the limb. Thus, we propose a relay between Pitx2, Myf5 and Pax7 for EOM stem cell maintenance. We demonstrate also that MuSCs in the EOMs adopt a quiescent state earlier that those in limb muscles and do not spontaneously proliferate in the adult, yet EOMs have a significantly higher content of Pax7+ MuSCs per area pre- and post-natally. Finally, while limb MuSCs proliferate in the mdx mouse model for Duchenne muscular dystrophy, significantly less MuSCs were present in the EOMs of the mdx mouse model compared to controls, and they were not proliferative. Overall, our study provides a comprehensive in vivo characterisation of MuSC heterogeneity along the body axis and brings further insights into the unusual sparing of EOMs during muscular dystrophy.
Collapse
Affiliation(s)
- Mao Kuriki
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| | - Amaury Korb
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| | - Glenda Comai
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| | - Shahragim Tajbakhsh
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| |
Collapse
|
44
|
Silva LMG, Gouveia VA, Campos GRS, Dale CS, da Palma RK, de Oliveira APL, Marcos RL, Duran CCG, Cogo JC, Silva Junior JA, Zamuner SR. Photobiomodulation mitigates Bothrops jararacussu venom-induced damage in myoblast cells by enhancing myogenic factors and reducing cytokine production. PLoS Negl Trop Dis 2024; 18:e0012227. [PMID: 38814992 PMCID: PMC11192417 DOI: 10.1371/journal.pntd.0012227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 06/21/2024] [Accepted: 05/20/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Photobiomodulation has exhibited promise in mitigating the local effects induced by Bothrops snakebite envenoming; however, the mechanisms underlying this protection are not yet fully understood. Herein, the effectiveness of photobiomodulation effects on regenerative response of C2C12 myoblast cells following exposure to Bothrops jararacussu venom (BjsuV), as well as the mechanisms involved was investigated. METHODOLOGY/PRINCIPAL FINDINGS C2C12 myoblast cells were exposed to BjsuV (12.5 μg/mL) and irradiated once for 10 seconds with laser light of 660 nm (14.08 mW; 0.04 cm2; 352 mW/cm2) or 780 nm (17.6 mW; 0.04 cm2; 440 mW/ cm2) to provide energy densities of 3.52 and 4.4 J/cm2, and total energies of 0.1408 and 0.176 J, respectively. Cell migration was assessed through a wound-healing assay. The expression of MAPK p38-α, NF-Кβ, Myf5, Pax-7, MyoD, and myogenin proteins were assessed by western blotting analysis. In addition, interleukin IL1-β, IL-6, TNF-alfa and IL-10 levels were measured in the supernatant by ELISA. The PBM applied to C2C12 cells exposed to BjsuV promoted cell migration, increase the expression of myogenic factors (Pax7, MyF5, MyoD and myogenin), reduced the levels of proinflammatory cytokines, IL1-β, IL-6, TNF-alfa, and increased the levels of anti-inflammatory cytokine IL-10. In addition, PBM downregulates the expression of NF-kB, and had no effect on p38 MAKP. CONCLUSION/SIGNIFICANCE These data demonstrated that protection of the muscle cell by PBM seems to be related to the increase of myogenic factors as well as the modulation of inflammatory mediators. PBM therapy may offer a new therapeutic strategy to address the local effects of snakebite envenoming by promoting muscle regeneration and reducing the inflammatory process.
Collapse
Affiliation(s)
| | - Viviane Almeida Gouveia
- Postgraduate Program in Medicine-Biophotonics, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | | | - Camila Squarzone Dale
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Renata Kelly da Palma
- Facultad De Ciencias De la Salud de Manresa, Universitat de Vic-Universitat Central De Catalunya (UVic-UCC), Barcelona, Spain
- Tissue Repair and Regeneration Laboratory (TR2Lab), Institute for Research and Innovation in Life and Health Sciences in Central Catalonia (Iris-CC). Vic, Spain
| | | | - Rodrigo Labat Marcos
- Postgraduate Program in Medicine-Biophotonics, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | - Cinthya Cosme Gutierrez Duran
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
- Postgraduate Program in Medicine-Biophotonics, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | - José Carlos Cogo
- Programa de Mestrado em Bioengenharia do Instituto de Ciências e Tecnologia da Universidade Brasil, São Paulo, Brazil
| | - José Antônio Silva Junior
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
- Postgraduate Program in Medicine-Biophotonics, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | - Stella Regina Zamuner
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
- Postgraduate Program in Medicine-Biophotonics, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| |
Collapse
|
45
|
Ni M, He J, Li T, Zhao G, Ji Z, Ren F, Leng J, Wu M, Huang R, Li P, Hou L. Establishment and Characterization of SV40 T-Antigen Immortalized Porcine Muscle Satellite Cell. Cells 2024; 13:703. [PMID: 38667318 PMCID: PMC11049531 DOI: 10.3390/cells13080703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/06/2024] [Accepted: 04/06/2024] [Indexed: 04/28/2024] Open
Abstract
Muscle satellite cells (MuSCs) are crucial for muscle development and regeneration. The primary pig MuSCs (pMuSCs) is an ideal in vitro cell model for studying the pig's muscle development and differentiation. However, the long-term in vitro culture of pMuSCs results in the gradual loss of their stemness, thereby limiting their application. To address this conundrum and maintain the normal function of pMuSCs during in vitro passaging, we generated an immortalized pMuSCs (SV40 T-pMuSCs) by stably expressing SV40 T-antigen (SV40 T) using a lentiviral-based vector system. The SV40 T-pMuSCs can be stably sub-cultured for over 40 generations in vitro. An evaluation of SV40 T-pMuSCs was conducted through immunofluorescence staining, quantitative real-time PCR, EdU assay, and SA-β-gal activity. Their proliferation capacity was similar to that of primary pMuSCs at passage 1, and while their differentiation potential was slightly decreased. SiRNA-mediated interference of SV40 T-antigen expression restored the differentiation capability of SV40 T-pMuSCs. Taken together, our results provide a valuable tool for studying pig skeletal muscle development and differentiation.
Collapse
Affiliation(s)
- Mengru Ni
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.N.); (J.H.); (T.L.); (G.Z.); (Z.J.); (F.R.); (J.L.); (M.W.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
| | - Jingqing He
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.N.); (J.H.); (T.L.); (G.Z.); (Z.J.); (F.R.); (J.L.); (M.W.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
| | - Tao Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.N.); (J.H.); (T.L.); (G.Z.); (Z.J.); (F.R.); (J.L.); (M.W.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
| | - Gan Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.N.); (J.H.); (T.L.); (G.Z.); (Z.J.); (F.R.); (J.L.); (M.W.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhengyu Ji
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.N.); (J.H.); (T.L.); (G.Z.); (Z.J.); (F.R.); (J.L.); (M.W.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Fada Ren
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.N.); (J.H.); (T.L.); (G.Z.); (Z.J.); (F.R.); (J.L.); (M.W.); (R.H.); (P.L.)
| | - Jianxin Leng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.N.); (J.H.); (T.L.); (G.Z.); (Z.J.); (F.R.); (J.L.); (M.W.); (R.H.); (P.L.)
| | - Mengyan Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.N.); (J.H.); (T.L.); (G.Z.); (Z.J.); (F.R.); (J.L.); (M.W.); (R.H.); (P.L.)
| | - Ruihua Huang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.N.); (J.H.); (T.L.); (G.Z.); (Z.J.); (F.R.); (J.L.); (M.W.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
- Huai’an Academy, Nanjing Agricultural University, Huai’an 223001, China
| | - Pinghua Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.N.); (J.H.); (T.L.); (G.Z.); (Z.J.); (F.R.); (J.L.); (M.W.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
- Huai’an Academy, Nanjing Agricultural University, Huai’an 223001, China
| | - Liming Hou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.N.); (J.H.); (T.L.); (G.Z.); (Z.J.); (F.R.); (J.L.); (M.W.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
- Huai’an Academy, Nanjing Agricultural University, Huai’an 223001, China
| |
Collapse
|
46
|
Liu F, Cao Y, Wang X, Zhang K, Li N, Su Y, Zhang Y, Meng Q. Islr regulates satellite cells asymmetric division through the SPARC/p-ERK1/2 signaling pathway. FASEB J 2024; 38:e23534. [PMID: 38597911 DOI: 10.1096/fj.202302614r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/03/2024] [Accepted: 02/20/2024] [Indexed: 04/11/2024]
Abstract
Satellite cells (SCs) are adult muscle stem cells responsible for muscle regeneration after acute and chronic muscle injuries. The balance between stem cell self-renewal and differentiation determines the kinetics and efficiency of skeletal muscle regeneration. This study assessed the function of Islr in SC asymmetric division. The deletion of Islr reduced muscle regeneration in adult mice by decreasing the SC pool. Islr is pivotal for SC proliferation, and its deletion promoted the asymmetric division of SCs. A mechanistic search revealed that Islr bound to and degraded secreted protein acidic and rich in cysteine (SPARC), which activated p-ERK1/2 signaling required for asymmetric division. These findings demonstrate that Islr is a key regulator of SC division through the SPARC/p-ERK1/2 signaling pathway. These data provide a basis for treating myopathy.
Collapse
Affiliation(s)
- Fan Liu
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Yuxin Cao
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Xiong Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Kuo Zhang
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Na Li
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Yang Su
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Yali Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Qingyong Meng
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| |
Collapse
|
47
|
Lin F, Sun L, Zhang Y, Gao W, Chen Z, Liu Y, Tian K, Han X, Liu R, Li Y, Shen L. Mitochondrial stress response and myogenic differentiation. Front Cell Dev Biol 2024; 12:1381417. [PMID: 38681520 PMCID: PMC11055459 DOI: 10.3389/fcell.2024.1381417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 03/29/2024] [Indexed: 05/01/2024] Open
Abstract
Regeneration and repair are prerequisites for maintaining effective function of skeletal muscle under high energy demands, and myogenic differentiation is one of the key steps in the regeneration and repair process. A striking feature of the process of myogenic differentiation is the alteration of mitochondria in number and function. Mitochondrial dysfunction can activate a number of transcriptional, translational and post-translational programmes and pathways to maintain cellular homeostasis under different types and degrees of stress, either through its own signaling or through constant signaling interactions with the nucleus and cytoplasm, a process known as the mitochondrial stress responses (MSRs). It is now believed that mitochondrial dysfunction is closely associated with a variety of muscle diseases caused by reduced levels of myogenic differentiation, suggesting the possibility that MSRs are involved in messaging during myogenic differentiation. Also, MSRs may be involved in myogenesis by promoting bioenergetic remodeling and assisting myoblast survival during myogenic differentiation. In this review, we will take MSRs as an entry point to explore its concrete regulatory mechanisms during myogenic differentiation, with a perspective to provide a theoretical basis for the treatment and repair of related muscle diseases.
Collapse
Affiliation(s)
- Fu Lin
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Liankun Sun
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yu Zhang
- Experimental Teaching Center of Basic Medicine, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Weinan Gao
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zihan Chen
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
- Clinical Medical College of Jilin University, The First Hospital of Jilin University, Changchun, China
| | - Yanan Liu
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Kai Tian
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
- China Japan Union Hospital of Jilin University, Changchun, China
| | - Xuyu Han
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
- China Japan Union Hospital of Jilin University, Changchun, China
| | - Ruize Liu
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
- China Japan Union Hospital of Jilin University, Changchun, China
| | - Yang Li
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Luyan Shen
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
48
|
Wang K, Yang J, An Y, Wang J, Tan S, Xu H, Dong Y. MST1/2 regulates fibro/adipogenic progenitor fate decisions in skeletal muscle regeneration. Stem Cell Reports 2024; 19:501-514. [PMID: 38552635 PMCID: PMC11096422 DOI: 10.1016/j.stemcr.2024.02.010] [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: 07/03/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 04/12/2024] Open
Abstract
Defective skeletal muscle regeneration is often accompanied by fibrosis. Fibroblast/adipose progenitors (FAPs) are important in these processes, however, the regulation of FAP fate decisions is unclear. Here, using inducible conditional knockout mice, we show that blocking mammalian Ste20-like kinases 1/2 (MST1/2) of FAPs prevented apoptosis and reduced interleukin-6 secretion in vivo and in vitro, which impaired myoblast proliferation and differentiation, as well as impaired muscle regeneration. Deletion of Mst1/2 increased co-localization of Yes-associated protein (YAP) with Smad2/3 in nuclei and promoted differentiation of FAPs toward myofibroblasts, resulting in excessive collagen deposition and skeletal muscle fibrosis. Meanwhile, inhibition of MST1/2 increased YAP/Transcriptional co-activator with PDZ-binding motif activation, which promoted activation of the WNT/β-catenin pathway and impaired the differentiation of FAPs toward adipocytes. These results reveal a new mechanism for MST1/2 action in disrupted skeletal muscle regeneration and fibrosis via regulation of FAP apoptosis and differentiation. MST1/2 is a potential therapeutic target for the treatment of some myopathies.
Collapse
Affiliation(s)
- Kezhi Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jingjing Yang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yina An
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jing Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Shuyu Tan
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Hui Xu
- Department of Physical Education, China Agricultural University, Beijing 100193, China.
| | - Yanjun Dong
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
49
|
Song H, Tian X, He L, Liu D, Li J, Mei Z, Zhou T, Liu C, He J, Jia X, Yang Z, Yan C, Han Y. CREG1 deficiency impaired myoblast differentiation and skeletal muscle regeneration. J Cachexia Sarcopenia Muscle 2024; 15:587-602. [PMID: 38272853 PMCID: PMC10995283 DOI: 10.1002/jcsm.13427] [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: 05/08/2023] [Revised: 11/20/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND CREG1 (cellular repressor of E1A-stimulated genes 1) is a protein involved in cellular differentiation and homeostasis regulation. However, its role in skeletal muscle satellite cells differentiation and muscle regeneration is poorly understood. This study aimed to investigate the role of CREG1 in myogenesis and muscle regeneration. METHODS RNA sequencing data (GSE8479) was analysed from the Gene Expression Omnibus database (GEO, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi). We generated Creg1 knockdown and skeletal muscle satellite cells specific Creg1 overexpression mice mediated by adeno-associated virus serotype 9 (AAV9), skeletal muscle mature myofibre Creg1 knockout mice (myoblast/Creg1MKO), and control mice Creg1flox/flox (Creg1fl/fl) as in vivo models. The mice were injected into tibialis anterior (TA) muscle with 100 μL of 10 μM cardiotoxin to establish a muscle regeneration model. Creg1fl/fl and Creg1MKO mice were treated with AAV-sh-C-Cbl (2 × 1010 genomic copies/mouse) to silence C-Cbl in the TA muscle. 293T and C2C12 cells were transfected with plasmids using lipofectamine RNAi MAX in vitro. Mass spectrometry analyses and RNA sequencing transcriptomic assay were performed. RESULTS We analysed the transcriptional profiles of the skeletal muscle biopsies from healthy older (N = 25) and younger (N = 26) adult men and women in GSE8479 database, and the results showed that Creg1 was associated with human sarcopenia. We found that Creg1 knockdown mice regenerated less newly formed fibres in response to cardiotoxin injection (~30% reduction, P < 0.01); however, muscle satellite cells specific Creg1 overexpression mice regenerated more newly formed fibres (~20% increase, P < 0.05). AMPKa1 is known as a key mediator in the muscle regeneration process. Our results revealed that CREG1 deficiency inhibited AMPKa1 signalling through C-CBL E3-ubiquitin ligase-mediated AMPKa1 degradation (P < 0.01). C-CBL-mediated AMPKa1 ubiquitination was attributed to the K48-linked polyubiquitination of AMPKa1 at K396 and that the modification played an important role in the regulation of AMPKa1 protein stability. We also found that Creg1MKO mice regenerated less newly formed fibres compared with Creg1fl/fl mice (~30% reduction, P < 0.01). RNA-seq analysis showed that CREG1 deletion in impaired muscles led to the upregulation of inflammation and DKK3 expression. The TA muscles of Creg1MKO mice were injected with AAV-vector or AAV-shC-Cbl, silencing C-CBL (P < 0.01) in the skeletal muscles of Creg1MKO mice significantly improved muscle regeneration induced by CTX injury (P < 0.01). CONCLUSIONS Our findings suggest that CREG1 may be a potential therapeutic target for skeletal muscle regeneration.
Collapse
Affiliation(s)
- Haixu Song
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Xiaoxiang Tian
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Lianqi He
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Dan Liu
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Jiayin Li
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Zhu Mei
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Ting Zhou
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Chunying Liu
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Jiaqi He
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Xiaodong Jia
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Zheming Yang
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Chenghui Yan
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| | - Yaling Han
- Department of Cardiology, Cardiovascular Research Institute, State Key Laboratory of Frigid Zone Cardiovascular DiseaseGeneral Hospital of Northern Theater CommandShenyangChina
| |
Collapse
|
50
|
Cao X, Xue L, Yu X, Yan Y, Lu J, Luo X, Wang H, Wang J. Myogenic exosome miR-140-5p modulates skeletal muscle regeneration and injury repair by regulating muscle satellite cells. Aging (Albany NY) 2024; 16:4609-4630. [PMID: 38428405 PMCID: PMC10968704 DOI: 10.18632/aging.205617] [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: 10/16/2023] [Accepted: 01/23/2024] [Indexed: 03/03/2024]
Abstract
Muscle satellite cells (SCs) play a crucial role in the regeneration and repair of skeletal muscle injuries. Previous studies have shown that myogenic exosomes can enhance satellite cell proliferation, while the expression of miR-140-5p is significantly reduced during the repair process of mouse skeletal muscle injuries induced by BaCl2. This study aims to investigate the potential of myogenic exosomes carrying miR-140-5p inhibitors to activate SCs and influence the regeneration of injured muscles. Myogenic progenitor cell exosomes (MPC-Exo) and contained miR-140-5p mimics/inhibitors myogenic exosomes (MPC-Exo140+ and MPC-Exo140-) were employed to treat SCs and use the model. The results demonstrate that miR-140-5p regulates SC proliferation by targeting Pax7. Upon the addition of MPC-Exo and MPC-Exo140-, Pax7 expression in SCs significantly increased, leading to the transition of the cell cycle from G1 to S phase and an enhancement in cell proliferation. Furthermore, the therapeutic effect of MPC-Exo140- was validated in animal model, where the expression of muscle growth-related genes substantially increased in the gastrocnemius muscle. Our research demonstrates that MPC-Exo140- can effectively activate dormant muscle satellite cells, initiating their proliferation and differentiation processes, ultimately leading to the formation of new skeletal muscle cells and promoting skeletal muscle repair and remodeling.
Collapse
Affiliation(s)
- Xiaorui Cao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Linli Xue
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Xiuju Yu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Yi Yan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Jiayin Lu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Xiaomao Luo
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Juan Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| |
Collapse
|