1
|
Gentile GM, Blue RE, Goda GA, Guzman BB, Szymanski RA, Lee EY, Engels NM, Hinkle ER, Wiedner HJ, Bishop AN, Harrison JT, Zhang H, Wehrens XH, Dominguez D, Giudice J. Alternative splicing of the Snap23 microexon is regulated by MBNL, QKI, and RBFOX2 in a tissue-specific manner and is altered in striated muscle diseases. RNA Biol 2025; 22:1-20. [PMID: 40207498 PMCID: PMC12064062 DOI: 10.1080/15476286.2025.2491160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/05/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
The reprogramming of alternative splicing networks during development is a hallmark of tissue maturation and identity. Alternative splicing of microexons (small, genomic regions ≤ 51 nucleotides) functionally regulate protein-protein interactions in the brain and is altered in several neuronal diseases. However, little is known about the regulation and function of alternatively spliced microexons in striated muscle. Here, we investigated alternative splicing of a microexon in the synaptosome-associated protein 23 (Snap23) encoded gene. We found that inclusion of this microexon is developmentally regulated and tissue-specific, as it occurs exclusively in adult heart and skeletal muscle. The alternative region is highly conserved in mammalian species and encodes an in-frame sequence of 11 amino acids. Furthermore, we showed that alternative splicing of this microexon is mis-regulated in mouse models of heart and skeletal muscle diseases. We identified the RNA-binding proteins (RBPs) quaking (QKI) and RNA binding fox-1 homolog 2 (RBFOX2) as the primary splicing regulators of the Snap23 microexon. We found that QKI and RBFOX2 bind downstream of the Snap23 microexon to promote its inclusion, and this regulation can be escaped when the weak splice donor is mutated to the consensus 5' splice site. Finally, we uncovered the interplay between QKI and muscleblind-like splicing regulator (MBNL) as an additional, but minor layer of Snap23 microexon splicing control. Our results are one of the few reports detailing microexon alternative splicing regulation during mammalian striated muscle development.
Collapse
Affiliation(s)
- Gabrielle M. Gentile
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - R. Eric Blue
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Grant A. Goda
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bryan B. Guzman
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rachel A. Szymanski
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eunice Y. Lee
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nichlas M. Engels
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emma R. Hinkle
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hannah J. Wiedner
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Aubriana N. Bishop
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jonathan T. Harrison
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hua Zhang
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xander H.T. Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Daniel Dominguez
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- RNA Discovery Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jimena Giudice
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- RNA Discovery Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Wu P, Song N, Xiang Y, Tao Z, Mao B, Guo R, Wang X, Wu D, Zhang Z, Chen X, Ma D, Zhang T, Hao B, Ma J. FOXK2 in skeletal muscle development: a new pathogenic gene for congenital myopathy with ptosis. EMBO Mol Med 2025:10.1038/s44321-025-00247-x. [PMID: 40410591 DOI: 10.1038/s44321-025-00247-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/26/2025] [Accepted: 04/29/2025] [Indexed: 05/25/2025] Open
Abstract
Congenital ptosis, a genetic disorder involving levator palpebrae muscle dysfunction, is often associated with congenital myopathy. The genetic causes of this condition remain poorly understood. In this study, we identified FOXK2 mutations in five pedigrees with congenital myopathy and ptosis through whole exome sequencing and Sanger sequencing. Zebrafish with foxk2 deficiency exhibited underdeveloped skeletal muscles and reduced mobility, while mice with Foxk2 deletion in skeletal muscle stem cells (MuSCs) showed generalized skeletal muscle abnormalities. Further analysis revealed that FOXK2 deficiency impaired myogenic differentiation in C2C12 cells and disrupted mitochondrial homeostasis in both mouse MuSCs and C2C12 cells. Rescue experiments confirmed the loss-of-function effects of FOXK2 mutation. Coenzyme Q10 treatment improved mitochondrial function and alleviated skeletal muscle development defects in Foxk2-deficient mice. Preliminary omics analysis suggested FOXK2 directly regulates the expression of mitochondrial function-related genes by modulating chromatin accessibility at its binding sites. Our study identifies FOXK2 as a novel pathogenic gene for congenital myopathy with ptosis and highlights its essential role in skeletal muscle development and mitochondrial homeostasis, offering insights for potential diagnostics and therapies.
Collapse
Affiliation(s)
- Peixuan Wu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics, Fudan University, Shanghai, 200032, China
| | - Nan Song
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics, Fudan University, Shanghai, 200032, China
| | - Yang Xiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics, Fudan University, Shanghai, 200032, China
| | - Zhe Tao
- Dalian Women and Children's Medical Group Neurology Department, Dalian, 116012, China
| | - Bing Mao
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ruochen Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xin Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics, Fudan University, Shanghai, 200032, China
| | - Dan Wu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics, Fudan University, Shanghai, 200032, China
| | - Zhenzhen Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics, Fudan University, Shanghai, 200032, China
| | - Xin Chen
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics, Fudan University, Shanghai, 200032, China
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics, Fudan University, Shanghai, 200032, China.
| | - Tianyu Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics, Fudan University, Shanghai, 200032, China.
| | - Bingtao Hao
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- Henan Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450000, China.
| | - Jing Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Ostaszewska A, Michalska Z, Dzierzynska M, Fularczyk M, Bielak K, Morawska A, Kosmala M, Kulig I, Morytz J, Trusiak H, Zimowska M, Rodziewicz-Motowidlo S, Ciemerych MA, Archacka K, Brzoska E. Beneficial but diverse influence of custom-designed hydrogels modified with IL-4 and SDF-1 peptides on selected populations of cells essential for skeletal muscle regeneration. Int J Biol Macromol 2025:144282. [PMID: 40381767 DOI: 10.1016/j.ijbiomac.2025.144282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/28/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Under specific circumstances, such as extensive injuries, the development of degenerative diseases, or aging, the naïve potential of skeletal muscle to regenerate may be limited. For this reason, different tools and approaches are being tested which could result in the improvement of skeletal muscle reconstruction. Among them are hydrogels, investigated also by us, additionally functionalized with fragments of proteins known to support skeletal muscle regeneration, i.e., stromal-derived factor 1 or interleukin 4. In the current study, we evaluated the impact of such custom-designed hydrogels on different human cells important for efficient muscle regeneration, i.e., myoblasts, crucial for myofiber reconstruction, fibroblasts, ensuring ECM formation, and endothelial cells, securing new vessel development in regenerated muscles. Our results indicate that hydrogels functionalized with SDF-1 and IL-4 peptides induce beneficial but diverse effects in analyzed cell types, influencing either their proliferation, migration, or differentiation. Most importantly, hydrogels tested by us do not harm analyzed cell types, indicating that in vivo skeletal muscle regeneration might be improved by them.
Collapse
Affiliation(s)
- Anna Ostaszewska
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Zuzanna Michalska
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Maria Dzierzynska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Poland
| | - Martyna Fularczyk
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Poland
| | - Kacper Bielak
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Agnieszka Morawska
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Magdalena Kosmala
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Izabela Kulig
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Justyna Morytz
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Hanna Trusiak
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Małgorzata Zimowska
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | | | - Maria A Ciemerych
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Poland
| | - Karolina Archacka
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Faculty of Biology, Institute of Developmental Biology and Biomedical Sciences, University of Warsaw, Poland
| |
Collapse
|
4
|
Ranat K, Phan H, Ellythy S, Kenter M, Akkouch A. Advancements in Musculoskeletal Tissue Engineering: The Role of Melt Electrowriting in 3D-Printed Scaffold Fabrication. J Funct Biomater 2025; 16:163. [PMID: 40422828 DOI: 10.3390/jfb16050163] [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: 03/18/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/28/2025] Open
Abstract
Musculoskeletal tissue injuries of the bone, cartilage, ligaments, tendons, and skeletal muscles are among the most common injuries experienced in medicine and become increasingly problematic in cases of significant tissue damage, such as nonunion bone defects and volumetric muscle loss. Current gold standard treatment options for musculoskeletal injuries, although effective, have limited capability to fully restore native tissue structure and function. To overcome this challenge, three-dimensional (3D) printing techniques have emerged as promising therapeutic options for tissue regeneration. Melt electrowriting (MEW), a recently developed advanced 3D printing technique, has gained significant traction in the field of tissue regeneration because of its ability to fabricate complex customizable scaffolds via high-precision microfiber deposition. The tailorability at microscale levels offered by MEW allows for enhanced recapitulation of the tissue microenvironment. Here, we survey the recent contributions of MEW in advancing musculoskeletal tissue engineering. More specifically, we briefly discuss the principles and technical aspects of MEW, provide an overview of current printers on the market, review in-depth the latest biomedical applications in musculoskeletal tissue regeneration, and, lastly, examine the limitations of MEW and offer future perspectives.
Collapse
Affiliation(s)
- Kunal Ranat
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| | - Hong Phan
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| | - Suhaib Ellythy
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| | - Mitchell Kenter
- Department of Surgical Services, Division of Medical Engineering, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| | - Adil Akkouch
- Department of Surgical Services, Division of Medical Engineering, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
- Department of Surgical Services, Division of Orthopaedic Surgery, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| |
Collapse
|
5
|
Zhou H, Loo LSW, Ong FYT, Lou X, Wang J, Myint MK, Thong A, Seow DCS, Wibowo M, Ng S, Lv Y, Kwang LG, Bennie RZ, Pang KT, Dobson RCJ, Domigan LJ, Kanagasundaram Y, Yu H. Cost-effective production of meaty aroma from porcine cells for hybrid cultivated meat. Food Chem 2025; 473:142946. [PMID: 39864181 DOI: 10.1016/j.foodchem.2025.142946] [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: 10/14/2024] [Revised: 01/04/2025] [Accepted: 01/15/2025] [Indexed: 01/28/2025]
Abstract
Cultivated meats are typically hybrids of animal cells and plant proteins, but their high production costs limit their scalability. This study explores a cost-effective alternative by hypothesizing that controlling the Maillard and lipid thermal degradation reactions in pure cells can create a meaty aroma that could be extracted from minimal cell quantities. Using spontaneously immortalized porcine myoblasts and fibroblasts adapted to suspension culture with a 1 % serum concentration, we developed a method to isolate flavor precursors via freeze-thawing. Thermal reaction conditions were optimized to enhance aroma compound production. Chemical profiling demonstrates that myoblasts produce an aroma profile closer to pork meat than fibroblasts, although serum reduction decreased aroma yield. Sensory analysis supported these findings. Incorporating the optimized aroma extract - derived from just 1.2 % (w/w) cells - into plant proteins resulted in a hybrid cultivated meat with 78.5 % sensory similarity to pork meat, but with a significant 80 % reduction in production costs.
Collapse
Affiliation(s)
- Hanzhang Zhou
- Department of Physiology, The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, Singapore 117593, Singapore; Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore; Institute of Bioengineering and Bioimaging (IBB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Larry Sai Weng Loo
- Institute of Bioengineering and Bioimaging (IBB), Agency for Science, Technology and Research (A*STAR), Singapore; Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore 138669, Singapore; Food, Chemical and Biotechnology Cluster, Singapore Institute of Technology, 10 Dover Drive, Singapore 138683, Singapore; Department of Food Science & Technology, Faculty of Science, National University of Singapore, Singapore 117543, Singapore
| | - Francesca Yi Teng Ong
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Xuanming Lou
- Department of Physiology, The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, Singapore 117593, Singapore
| | - Jiahao Wang
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Matthew Khine Myint
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore 138668, Singapore
| | - Aaron Thong
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore 138669, Singapore
| | - Deborah Chwee San Seow
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore 138669, Singapore
| | - Mario Wibowo
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore 138669, Singapore
| | - Shengyong Ng
- Ants Innovate Pte. Ltd., Temasek Boulevard, Singapore 038987, Singapore
| | - Yunbo Lv
- Nanyang Environment And Water Research Institute (NEWRI), Nanyang Technological University, 1 Cleantech Loop, Singapore 637141, Singapore
| | - Leng Gek Kwang
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Rachel Z Bennie
- School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand; Biomolecular Interaction Centre, School of Biological Sciences, University of Canterbury, Christchurch 8140, New Zealand; The Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Kuin Tian Pang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117583, Singapore; Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore 138668, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore 138671, Singapore; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637371, Singapore
| | - Renwick C J Dobson
- School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand; Biomolecular Interaction Centre, School of Biological Sciences, University of Canterbury, Christchurch 8140, New Zealand; The Riddet Institute, Massey University, Palmerston North, New Zealand; Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Laura J Domigan
- Biomolecular Interaction Centre, School of Biological Sciences, University of Canterbury, Christchurch 8140, New Zealand; The Riddet Institute, Massey University, Palmerston North, New Zealand; Department of Chemical and Materials Engineering, University of Auckland, Auckland 1142, New Zealand
| | - Yoganathan Kanagasundaram
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore 138669, Singapore
| | - Hanry Yu
- Department of Physiology, The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, Singapore 117593, Singapore; Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore; Institute of Bioengineering and Bioimaging (IBB), Agency for Science, Technology and Research (A*STAR), Singapore; Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore; CAMP, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, Singapore 138602, Singapore.
| |
Collapse
|
6
|
Villaronga-Luque A, Savill RG, López-Anguita N, Bolondi A, Garai S, Gassaloglu SI, Rouatbi R, Schmeisser K, Poddar A, Bauer L, Alves T, Traikov S, Rodenfels J, Chavakis T, Bulut-Karslioglu A, Veenvliet JV. Integrated molecular-phenotypic profiling reveals metabolic control of morphological variation in a stem-cell-based embryo model. Cell Stem Cell 2025; 32:759-777.e13. [PMID: 40245869 DOI: 10.1016/j.stem.2025.03.012] [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: 12/20/2023] [Revised: 11/27/2024] [Accepted: 03/21/2025] [Indexed: 04/19/2025]
Abstract
Considerable phenotypic variation under identical culture conditions limits the potential of stem-cell-based embryo models (SEMs) in basic and applied research. The biological processes causing this seemingly stochastic variation remain unclear. Here, we investigated the roots of phenotypic variation by parallel recording of transcriptomic states and morphological history in individual structures modeling embryonic trunk formation. Machine learning and integration of time-resolved single-cell RNA sequencing with imaging-based phenotypic profiling identified early features predictive of phenotypic end states. Leveraging this predictive power revealed that early imbalance of oxidative phosphorylation and glycolysis results in aberrant morphology and a neural lineage bias, which we confirmed by metabolic measurements. Accordingly, metabolic interventions improved phenotypic end states. Collectively, our work establishes divergent metabolic states as drivers of phenotypic variation and offers a broadly applicable framework to chart and predict phenotypic variation in organoids and SEMs. The strategy can be used to identify and control underlying biological processes, ultimately increasing reproducibility.
Collapse
Affiliation(s)
- Alba Villaronga-Luque
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Faculty of Biology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ryan G Savill
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Faculty of Biology, Technische Universität Dresden, 01307 Dresden, Germany
| | | | - Adriano Bolondi
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Sumit Garai
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01307 Dresden, Germany
| | - Seher Ipek Gassaloglu
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Roua Rouatbi
- MOSAIC Group, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Center for Systems Biology Dresden, 01307 Dresden, Germany; Faculty of Computer Science, Technische Universität Dresden, 01062 Dresden, Germany; Center for Scalable Data Analytics and Artificial Intelligence Dresden/Leipzig, 01062 Dresden, Germany
| | - Kathrin Schmeisser
- Energetics of Biological Systems Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Aayush Poddar
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Lisa Bauer
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Tiago Alves
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sofia Traikov
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jonathan Rodenfels
- Cluster of Excellence Physics of Life, Technische Universität Dresden, 01307 Dresden, Germany; Center for Systems Biology Dresden, 01307 Dresden, Germany; Energetics of Biological Systems Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | | | - Jesse V Veenvliet
- Stembryogenesis Laboratory, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01307 Dresden, Germany; Center for Systems Biology Dresden, 01307 Dresden, Germany.
| |
Collapse
|
7
|
Meng Y, Zeng W, Zhu X, Bao L, Pan Y, Li H, Zhang J, Liu L, Gao Z, Du Z, Chu W. The Role of Twist2 in Myoblast Proliferation, Fusion, and Its Impact on Muscle Structure During the Growth of Chinese Perch ( Siniperca chuatsi). Animals (Basel) 2025; 15:1177. [PMID: 40282011 PMCID: PMC12024327 DOI: 10.3390/ani15081177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/12/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Twist2 plays a pivotal regulatory role in the growth of skeletal muscle across various organisms. Nonetheless, the specific mechanism by which Twist2 governs skeletal muscle function in fish, particularly in the economically significant Chinese perch (Siniperca chuatsi), remains unclear. Within the muscle injury model in Chinese perch, we observed that Twist2 expression was upregulated during the repair phase of fast muscle tissue, exhibiting an expression pattern analogous to that of Pax7. Following the knockdown of Twist2 using Twist2-specific in vivo-siRNA in fast muscle tissues, the expression of myogenic regulatory factors (MRFs) and Myomaker was significantly reduced in the Twist2-siRNA-treated group compared with the control group, whereas no significant differences were observed for Pax3 and Pax7. Furthermore, the diameter of myofibers and the number of nuclei in single myofibers were reduced, and concurrently, the number of BrdU-positive cells (proliferating cells) was significantly reduced in the Twist2-siRNA-treated group. Taken together, this study demonstrates that Twist2 promotes myoblast proliferation and fusion, thereby regulating fast muscle growth in juvenile Chinese perch. These findings provide a clear direction for further exploration of molecular mechanisms underlying skeletal muscle growth in economic fish species.
Collapse
Affiliation(s)
- Yangyang Meng
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China; (Y.M.); (W.Z.); (L.B.); (Y.P.); (H.L.); (J.Z.)
| | - Wei Zeng
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China; (Y.M.); (W.Z.); (L.B.); (Y.P.); (H.L.); (J.Z.)
| | - Xin Zhu
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China; (Y.M.); (W.Z.); (L.B.); (Y.P.); (H.L.); (J.Z.)
| | - Lingsheng Bao
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China; (Y.M.); (W.Z.); (L.B.); (Y.P.); (H.L.); (J.Z.)
| | - Yaxiong Pan
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China; (Y.M.); (W.Z.); (L.B.); (Y.P.); (H.L.); (J.Z.)
| | - Honghui Li
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China; (Y.M.); (W.Z.); (L.B.); (Y.P.); (H.L.); (J.Z.)
| | - Jianshe Zhang
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China; (Y.M.); (W.Z.); (L.B.); (Y.P.); (H.L.); (J.Z.)
| | - Lusha Liu
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (L.L.); (Z.G.)
| | - Zexia Gao
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (L.L.); (Z.G.)
| | - Zhenyu Du
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai 200241, China;
| | - Wuying Chu
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China; (Y.M.); (W.Z.); (L.B.); (Y.P.); (H.L.); (J.Z.)
| |
Collapse
|
8
|
Cillo M, Buonomo V, Vainshtein A, Grumati P. Autophagy, ER-phagy and ER Dynamics During Cell Differentiation. J Mol Biol 2025:169151. [PMID: 40222412 DOI: 10.1016/j.jmb.2025.169151] [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: 11/30/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
The endoplasmic reticulum (ER) is a multifunctional organelle essential for protein and lipid synthesis, ion transport and inter-organelle communication. It comprises a highly dynamic network of membranes that continuously reshape to support a wide range of cellular processes. During cellular differentiation, extensive remodelling of both ER architecture and its proteome is required to accommodate alterations in cell morphology and function. Autophagy, and ER-phagy in particular, plays a pivotal role in reshaping the ER, enabling cells to meet their evolving needs and adapt to developmental cues. Despite the ER's critical role in cellular differentiation, the mechanisms responsible for regulating its dynamics are not fully understood. Emerging evidence suggests that transcriptional and post-translational regulation play a role in fine-tuning ER-phagy and the unfolded protein response (UPR). This review explores the molecular basis of autophagy and ER-phagy, highlighting their role in ER remodelling during cellular differentiation. A deeper understanding of these processes could open new avenues for targeted therapeutic approaches in conditions where ER remodelling is impaired.
Collapse
Affiliation(s)
- Michele Cillo
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Viviana Buonomo
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | | | - Paolo Grumati
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy.
| |
Collapse
|
9
|
Heisser RH, Bawa M, Shah J, Bu A, Raman R. Soft Biological Actuators for Meter-Scale Homeostatic Biohybrid Robots. Chem Rev 2025; 125:3976-4007. [PMID: 40138615 DOI: 10.1021/acs.chemrev.4c00785] [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: 03/29/2025]
Abstract
Skeletal muscle's elegant protein-based architecture powers motion throughout the animal kingdom, with its constituent actomyosin complexes driving intra- and extra-cellular motion. Classical motors and recently developed soft actuators cannot match the packing density and contractility of individual muscle fibers that scale to power the motion of ants and elephants alike. Accordingly, the interdisciplinary fields of robotics and tissue engineering have combined efforts to build living muscle actuators that can power a new class of robots to be more energy-efficient, dexterous, and safe than existing motor-powered and hydraulic paradigms. Doing so ethically and at scale─creating meter-scale tissue constructs from sustainable muscle progenitor cell lines─has inspired innovations in biomaterials and tissue culture methodology. We weave discussions of muscle cell biology, materials chemistry, tissue engineering, and biohybrid design to review the state of the art in soft actuator biofabrication. Looking forward, we outline a vision for meter-scale biohybrid robotic systems and tie discussions of recent progress to long-term research goals.
Collapse
Affiliation(s)
- Ronald H Heisser
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Maheera Bawa
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Jessica Shah
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 45 Carleton St., Cambridge, Massachusetts 02142, United States of America
| | - Angel Bu
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Ritu Raman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| |
Collapse
|
10
|
Valdebenito GE, Chacko AR, Chung CY, Sheshadri P, Chi H, O'Callaghan B, Madej MJ, Houlden H, Rouse H, Morales V, Bianchi K, Tedesco FS, Pitceathly RDS, Duchen MR. Metabolic remodeling in hiPSC-derived myofibers carrying the m.3243A>G mutation. Stem Cell Reports 2025; 20:102448. [PMID: 40086445 PMCID: PMC12069895 DOI: 10.1016/j.stemcr.2025.102448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/16/2025] Open
Abstract
Mutations in mitochondrial DNA cause severe multisystem disease frequently associated with muscle weakness. The m.3243A>G mutation is the major cause of mitochondrial encephalomyopathy lactic acidosis and stroke-like episodes (MELAS). Experimental models that recapitulate the disease phenotype in vitro for disease modeling or drug screening are very limited. We have therefore generated hiPSC-derived muscle fibers with variable heteroplasmic mtDNA mutation load without significantly affecting muscle differentiation potential. The cells exhibit physiological characteristics of muscle fibers and show a well-organized myofibrillar structure. In cells carrying the m.3243A>G mutation, the mitochondrial membrane potential and oxygen consumption were reduced in relation to the mutant load. We have shown through proteomic, phosphoproteomic, and metabolomic analyses that the m.3243A>G mutation variably affects the cell phenotype in relation to the mutant load. This variation is reflected by an increase in the NADH/NAD+ ratio, which in turn influences key nutrient-sensing pathways in the myofibers. This model enables a detailed study of the impact of the mutation on cellular bioenergetics and on muscle physiology with the potential to provide a platform for drug screening.
Collapse
Affiliation(s)
- Gabriel E Valdebenito
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK; Consortium for Mitochondrial Research, UCL, Gower Street, London WC1E 6BT, UK.
| | - Anitta R Chacko
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK; Consortium for Mitochondrial Research, UCL, Gower Street, London WC1E 6BT, UK
| | - Chih-Yao Chung
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK; Consortium for Mitochondrial Research, UCL, Gower Street, London WC1E 6BT, UK
| | - Preethi Sheshadri
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK; Consortium for Mitochondrial Research, UCL, Gower Street, London WC1E 6BT, UK
| | - Haoyu Chi
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK; Consortium for Mitochondrial Research, UCL, Gower Street, London WC1E 6BT, UK
| | - Benjamin O'Callaghan
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Monika J Madej
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK; Ryvu Therapeutics S.A., Krakow, Poland
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Hannah Rouse
- Bart's Cancer Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Valle Morales
- Bart's Cancer Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Katiuscia Bianchi
- Bart's Cancer Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK; Stem Cells and Neuromuscular Regeneration Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital for Children, London, UK
| | - Robert D S Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK; NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Michael R Duchen
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK; Consortium for Mitochondrial Research, UCL, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
11
|
Zhang Y, Wang T, Wang Z, Shi X, Jin J. Functions and Therapeutic Potentials of Long Noncoding RNA in Skeletal Muscle Atrophy and Dystrophy. J Cachexia Sarcopenia Muscle 2025; 16:e13747. [PMID: 40034097 PMCID: PMC11876862 DOI: 10.1002/jcsm.13747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 12/23/2024] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Skeletal muscle is the most abundant tissue in the human body and is responsible for movement, metabolism, energy production and longevity. Muscle atrophy is a frequent complication of several diseases and occurs when protein degradation exceeds protein synthesis. Genetics, ageing, nerve injury, weightlessness, cancer, chronic diseases, the accumulation of metabolic byproducts and other stimuli can lead to muscle atrophy. Muscular dystrophy is a neuromuscular disorder, part of which is caused by the deficiency of dystrophin protein and is mostly related to genetics. Muscle atrophy and muscular dystrophy are accompanied by dynamic changes in transcriptomic, translational and epigenetic regulation. Multiple signalling pathways, such as the transforming growth factor-β (TGF-β) signalling pathway, the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) pathway, inflammatory signalling pathways, neuromechanical signalling pathways, endoplasmic reticulum stress and glucocorticoids signalling pathways, regulate muscle atrophy. A large number of long noncoding RNAs (lncRNAs) have been found to be abnormally expressed in atrophic muscles and dystrophic muscles and regulate the balance of muscle protein synthesis and degradation or dystrophin protein expression. These lncRNAs may serve as potential targets for treating muscle atrophy and muscular dystrophy. In this review, we summarized the known lncRNAs related to muscular dystrophy and muscle atrophy induced by denervation, ageing, weightlessness, cachexia and abnormal myogenesis, along with their molecular mechanisms. Finally, we explored the potential of using these lncRNAs as therapeutic targets for muscle atrophy and muscular dystrophy, including the methods of discovery and clinical application prospects for functional lncRNAs.
Collapse
Affiliation(s)
- Yidi Zhang
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Teng Wang
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Ziang Wang
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Xin'e Shi
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Jianjun Jin
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| |
Collapse
|
12
|
Cheng K, Chen C, Zhou Q, Chen X, Xie P. Deficit of neuronal EAAT2 impairs hippocampus CA3 neuron's activity and may induce depressive like behaviors. J Adv Res 2025:S2090-1232(25)00176-6. [PMID: 40096942 DOI: 10.1016/j.jare.2025.03.016] [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: 05/25/2023] [Revised: 01/20/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
INTRODUCTION Major depressive disorder (MDD) is a severe neuropsychiatric disease that is accompanied by hippocampal dysfunction. Currently, the complex neuronal types and molecules involved in the various hippocampal subfields in patients with depression remain unclear. OBJECTIVES We focused on the role of hippocampal excitatory amino acid transporter 2 (EAAT2) in chronic stress. METHODS We studied two chronic stress models, the chronic unpredictable mild stress (CUMS) and the chronic social defeat stress (CSDS) models, and performed pharmacological inhibition, genetic manipulations to examine overexpression of neuron-specific solute carrier family 1 member 2 (SLC1A2), the gene encoding EAAT2, in the dorsal CA3 and conditional Slc1a2 knockout in CA3, whole-cell recording, and behavioral tests. RESULTS Our results indicated that decreased EAAT2 expression and specific inhibition were associated with depression-like behavior and enhanced CA3 pyramidal neuron activity. In addition, neuron-specific EAAT2 overexpression in the CA3 yielded antidepressant-like effects and inhibited CA3 pyramidal neuron hyperactivity, whereas conditional CA3 EAAT2 knockout showed opposite effects at both behavioral and functional levels. We also found that the single-nucleotide polymorphism, rs77619780, in the SLCA1A2 gene was associated with lower MDD risk. CONCLUSION Our findings revealed that EAAT2 deficit in the CA3 induces depression-like behavior, which offers novel insight into MDD pathophysiology.
Collapse
Affiliation(s)
- Ke Cheng
- Chongqing Key Laboratory of Cerebrovascular Disease Research, Chongqing 402160, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160,China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Chong Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qinji Zhou
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, 404010, China
| | - Xiangyu Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
13
|
Wu Y, Liu X, Fan Y, Zuo H, Niu X, Zuo B, Xu Z. MiR-34b Regulates Muscle Growth and Development by Targeting SYISL. Cells 2025; 14:379. [PMID: 40072107 PMCID: PMC11898696 DOI: 10.3390/cells14050379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Non-coding genes, such as microRNA and lncRNA, which have been widely studied, play an important role in the regulatory network of skeletal muscle development. However, the functions and mechanisms of most non-coding RNAs in skeletal muscle regulatory networks are unclear. This study investigated the function and mechanism of miR-34b in muscle growth and development. MiR-34b overexpression and interference tests were performed in C2C12 myoblasts and animal models. It was demonstrated that miR-34b significantly promoted mouse muscle growth and development in vivo, while miR-34b inhibited myoblast proliferation and promoted myoblast differentiation in vitro. Bioinformatics prediction using TargetScan for miRNA target identification and Bibiserv2 for potential miRNA-gene interaction analysis revealed a miR-34b binding site in the SYlSL sequence. The molecular mechanism of miR-34b regulating muscle growth and development was studied by co-transfection experiment, luciferase reporter gene detection, RNA immunoprecipitation, and RNA pull-down. MiR-34b can directly bind to SYISL and AGO2 proteins and regulate the expression of SYISL target genes p21 and MyoG by targeting SYISL, thereby regulating muscle growth and development. This study highlights that, as a novel regulator of myogenesis, miR-34b regulates muscle growth and development by targeting SYISL.
Collapse
Affiliation(s)
- Yuting Wu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiao Liu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yonghui Fan
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Hao Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaoyu Niu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Bo Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430068, China
| | - Zaiyan Xu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Afairs, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (X.L.); (Y.F.); (H.Z.); (X.N.)
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
14
|
Su Y, Deng K, Liu Z, Zhang Z, Liu Z, Huang Z, Gao Y, Gao K, Fan Y, Zhang Y, Wang F. m6A modified pre-miR-503-5p contributes to myogenic differentiation through the activation of mTOR pathway. Int J Biol Macromol 2025; 294:139517. [PMID: 39756749 DOI: 10.1016/j.ijbiomac.2025.139517] [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: 10/03/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/07/2025]
Abstract
The post-transcriptional regulation of epigenetic modification is a hot topic in skeletal muscle development research. Both m6A modifications and miRNAs have been well-established as crucial regulators in skeletal muscle development. However, the interacting regulatory mechanisms between m6A modifications and miRNAs in skeletal muscle development remain unclear. In this study, miRNA sequencing analysis of goat primary myoblasts (GPMs) pre- and post-differentiation revealed that miR-503-5p was upregulated during myogenic differentiation, and its precursor was identified to contain m6A modification sites. Combined analysis of RIP, qRT-PCR and mRNA stability assay showed that Ythdf2 could recognize and bind the m6A site on pre-miR-503-5p, thereby facilitating the maturation of pre-miR-503-5p in an m6A-dependent manner. Moreover, the overexpression of miR-503-5p significantly inhibits the proliferation of GPMs, promotes myogenic differentiation, and enhances mitochondrial biogenesis while activating the mTOR pathway. However, the suppression of mTOR activity can effectively counteract the accelerated myogenic differentiation induced by miR-503-5p overexpression. Collectively, our results indicate that Ythdf2-dependent m6A modification facilitates the maturation of pre-miR-503-5p, thereby promoting skeletal muscle differentiation through the activation of the mTOR pathway. These insights lay a valuable foundation for further investigation into the complexities of skeletal muscle development and the potential implications of epigenetic regulation in this process.
Collapse
Affiliation(s)
- Yalong Su
- Sanya Research Institute of Nanjing Agricultural University, Nanjing Agricultural University, Sanya 572025, China; Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Kaiping Deng
- Sanya Research Institute of Nanjing Agricultural University, Nanjing Agricultural University, Sanya 572025, China; Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhipeng Liu
- Sanya Research Institute of Nanjing Agricultural University, Nanjing Agricultural University, Sanya 572025, China; Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhen Zhang
- Sanya Research Institute of Nanjing Agricultural University, Nanjing Agricultural University, Sanya 572025, China; Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhilin Liu
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zidi Huang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuhao Gao
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ke Gao
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yixuan Fan
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yanli Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Feng Wang
- Sanya Research Institute of Nanjing Agricultural University, Nanjing Agricultural University, Sanya 572025, China; Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
15
|
Shi DL, Grifone R, Zhang X, Li H. Rbm24-mediated post-transcriptional regulation of skeletal and cardiac muscle development, function and regeneration. J Muscle Res Cell Motil 2025; 46:53-65. [PMID: 39614020 DOI: 10.1007/s10974-024-09685-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/20/2024] [Indexed: 12/01/2024]
Abstract
RNA-binding proteins are critically involved in the post-transcriptional control of gene expression during embryonic development and in adult life, contributing to regulating cell differentiation and maintaining tissue homeostasis. Compared to the relatively well documented functions of transcription factors, the regulatory roles of RNA-binding proteins in muscle development and function remain largely elusive. However, deficiency of many RNA-binding proteins has been associated with muscular defects, neuromuscular disorders and heart diseases, such as myotonic dystrophy, amyotrophic lateral sclerosis, and cardiomyopathy. Rbm24 is highly conserved among vertebrates and is one of the best characterized RNA-binding proteins with crucial implication in the myogenic and cardiomyogenic programs. It presents the distinctive particularity of displaying highly restricted expression in both skeletal and cardiac muscles, with changes in subcellular localization during the process of differentiation. Functional analyses using different vertebrate models have clearly demonstrated its requirement for skeletal muscle differentiation and regeneration as well as for myocardium organization and cardiac function, by regulating the expression of both common and distinct target genes in these tissues. The challenge remains to decipher the dynamic feature of post-transcriptional circuits regulated by Rbm24 during skeletal myogenesis, cardiomyogenesis, and muscle repair. This review discusses current understanding of its function in striated muscles and its possible implication in human disease, with the aim of identifying research gaps for future investigation.
Collapse
Affiliation(s)
- De-Li Shi
- Laboratoire de Biologie du Développement, Sorbonne Université, CNRS UMR7622, INSERM U1156, LBD, Paris, F-75005, France.
| | - Raphaëlle Grifone
- Laboratoire de Biologie du Développement, Sorbonne Université, CNRS UMR7622, INSERM U1156, LBD, Paris, F-75005, France
| | - Xiangmin Zhang
- College of Marine Life Sciences, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Hongyan Li
- College of Marine Life Sciences, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| |
Collapse
|
16
|
Lopez AN, Olivarez MA, Stenhouse C, Moses RM, Newton MG, Sah N, Seo H, Cain J, Lefevre C, Ross A, Ryan P, Wiegert JG, Wu G, Johnson GA, Bazer FW. Effects of dietary supplementation of creatine on fetal development in gilts at d 60 and d 90 of gestation. J Anim Sci Biotechnol 2025; 16:31. [PMID: 40022146 PMCID: PMC11871691 DOI: 10.1186/s40104-025-01166-0] [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: 09/18/2024] [Accepted: 01/15/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND The creatine-creatine kinase-phosphocreatine (Cr-CK-PCr) system maintains intracellular ratios of ATP/ADP for support of cellular functions and has been characterized at the placental-uterine interface of rodents, primates, swine and sheep, and thus may support fetal development. This study determined effects of dietary supplementation of creatine (Cr) to gestating gilts on fetal development, the number and ratio of primary and secondary muscle fibers, and on protein expression in endometrium and fetal biceps-femoris muscle, respectively in fetal pigs on d 60 and d 90 of gestation. METHODS Reproductively mature gilts were synchronized to estrus using Matrix, observed for estrus (d 0), and artificially inseminated 12 h and 36 h later. Gilts were individually housed and fed 0.86 kg of 14% crude protein diet twice daily that meets nutritional requirements for pregnant gilts. Gilts were assigned to either basal diet control (CON) group, or Cr supplemented group (provided 30 g Cr monohydrate daily) from d 10 to either d 60 or d 90 of gestation. Gilts were euthanized and hysterectomized on either d 60 or d 90 of gestation. These protocols were completed in two replicates, as gilts were bred in spring and euthanized in summer or bred in fall and euthanized in winter (n = 20 gilts/replicate). Litter size, crown-rump length, sex, and fetal weight was recorded. Three female and male fetuses closest to mean litter weight were selected to assess effects of treatment on weight of fetal brain, kidney, liver, spleen, and biceps-femoris muscle. Data were analyzed to determine effects of treatment, days of gestation, replicate, and sex on litter size, fetal measurements, and incidence of intrauterine growth restriction. RESULTS Dietary Cr supplementation increased fetal brain weight to body weight ratios on d 90 of gestation (P < 0.05) and fetal kidney weight to body weight ratios on d 60 of gestation (P < 0.01), while days of gestation had significant effect on expression of mitochondrial CK isoform in gilt endometria (P < 0.05). CONCLUSIONS Results suggest that dietary supplementation of Cr in gestating gilts enhanced development of select fetal organs and contribute to understanding roles of the Cr-CK-PCr system in pregnancy.
Collapse
Affiliation(s)
- Arianna N Lopez
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Maddison A Olivarez
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Claire Stenhouse
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
- Department of Animal Science, Pennsylvania State University, University Park, PA, 16802, USA
| | - Robyn M Moses
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Makenzie G Newton
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Nirvay Sah
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
- Department of Pathology, University of California-San Diego, San Diego, CA, 92093, USA
| | - Heewon Seo
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Joseph Cain
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Carli Lefevre
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Alexandria Ross
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Patrick Ryan
- Department of Kinesiology, Texas A&M University, College Station, TX, 77843, USA
| | - Jeffrey G Wiegert
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Gregory A Johnson
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
17
|
Cui M, Tzioufa F, Bruton J, Westerblad H, Munic Kos V. The impact of bisphenol AF on skeletal muscle function and differentiation in vitro. Toxicol In Vitro 2025; 103:105975. [PMID: 39586365 DOI: 10.1016/j.tiv.2024.105975] [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: 03/25/2024] [Revised: 11/15/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Various environmental chemicals have been identified as contributors to metabolic diseases. Bisphenol AF (BPAF), a substitute for bisphenol A, has been associated with changes in glucose metabolism and incidence of type 2 diabetes mellitus in humans. However, its mode of action remains unclear. Considering that skeletal muscle is the primary tissue for glucose utilization and the development of insulin resistance, yet largely neglected in toxicological assessments, we investigated the impact of BPAF on skeletal muscle function and differentiation. We examined the effects of BPAF (0.01-10 μM) on glucose uptake, response to insulin, production of reactive oxygen species (ROS), intracellular calcium, and myocyte differentiation, during hyperglycemia, insulin stimulation, and muscle contraction. We used the rat myoblast cell line L6 differentiated into myotubes, and murine primary isolated muscle fibers. In myotubes and contracting adult fibers, BPAF increased mitochondrial ROS. Basal glucose uptake was increased in myotubes while cells' ability to respond to insulin was decreased. Additionally, in developing myotubes, differentiation markers were downregulated with BPAF, along with impaired formation of tube structures. These effects were primarily observed at 10 μM concentration, which is markedly higher than reported human exposure concentrations. The results provide an insight into potential hazards associated with BPAF in terms of metabolic disruption in skeletal muscle. The developed in vitro methods show promise for future usage in assessments of new chemicals and their mixtures.
Collapse
Affiliation(s)
- Minying Cui
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Foteini Tzioufa
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Joseph Bruton
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Westerblad
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
18
|
Seong EJ, Kim Y, Su ZY, Kang HT, Lee JH. Combined Treatment of Metformin and Resveratrol Promotes Myogenesis Through Increased Irisin Release in C2C12 Cells. Pharm Res 2025; 42:419-428. [PMID: 40011370 DOI: 10.1007/s11095-025-03834-2] [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: 12/02/2024] [Accepted: 02/04/2025] [Indexed: 02/28/2025]
Abstract
PURPOSE This study aimed to investigate the additive effects of a combination of metformin and resveratrol on irisin expression in C2C12 cells. METHODS The study involved treating C2C12 cells with metformin and resveratrol, either alone or in combination, and analyzing their effects on myogenesis and irisin release. The activation of signaling pathways, including AMPK/SIRT1/PGC1α, as well as the relative mRNA and protein expression levels of MyoD, myogenin, and Myh were also assessed. RESULTS Combination treatment with metformin and resveratrol significantly increased MyoD, myogenin, Myh, and FNDC5 expression compared with the group treated with metformin alone. The increase in irisin production was associated with phosphorylation of AMPK and upregulation of PGC-1α and SIRT1, indicating activation of the AMPK/SIRT1/PGC-1α pathway. The mRNA and protein expression levels of MyoD, myogenin, and Myh were also significantly higher in the combination treatment group compared to the metformin alone group. CONCLUSION The combination of metformin and resveratrol effectively increased irisin release through the AMPK/Sirt1/PGC-1α pathway, suggesting that this combination treatment could enhance myogenesis.
Collapse
Affiliation(s)
- Eun Ji Seong
- Department of Food Science and Biotechnology, College of Bio-Nano Technology, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea
| | - Yejin Kim
- Department of Food Science and Biotechnology, College of Bio-Nano Technology, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea
| | - Zheng-Yuan Su
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan City, 320314, Taiwan
| | - Hee-Taik Kang
- Department of Family Medicine, Severance Hospital, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea.
| | - Jong Hun Lee
- Department of Food Science and Biotechnology, College of Bio-Nano Technology, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea.
| |
Collapse
|
19
|
Viola V, Samanta T, Nava M, Celli A, Armamento‐Villareal R, Nguyen N, Colleluori G, Barnouin Y, Napoli N, Qualls C, Kaipparettu B, Villareal D. Testosterone Modulation of Muscle Transcriptomic Profile During Lifestyle Therapy in Older Men with Obesity and Hypogonadism. J Cachexia Sarcopenia Muscle 2025; 16:e13697. [PMID: 40035362 PMCID: PMC11924803 DOI: 10.1002/jcsm.13697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/23/2024] [Accepted: 11/29/2024] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Testosterone replacement therapy (TRT) added to lifestyle therapy can mitigate weight-loss-induced reduction of muscle mass and bone mineral density (BMD) in older men with obesity and hypogonadism. OBJECTIVE To investigate the molecular mechanisms underlying the attenuation of muscle and BMD loss in response to TRT during intensive lifestyle intervention in this high-risk older population. METHODS Among 83 older (≥ 65 years) men with obesity (BMI ≥ 30 kg/m2) and hypogonadism (early AM testosterone persistently < 300 ng/dL) associated with frailty (Modified Physical Performance Test score ≤ 31) randomized into 26-week lifestyle therapy plus testosterone (LT+TRT) or placebo (LT+Pbo) in the LITROS trial, 38 underwent serial muscle biopsies for the muscle transcriptomics substudy. RESULTS Despite similar ~10% weight loss, lean body mass and thigh muscle volume decreased less in LT+TRT than LT+Pbo (-2% vs. -4%, respectively; p = 0.04). Hip BMD was preserved in LT+TRT compared with LT+Pbo (0.4% vs. -1.3%; p = 0.03). Muscle strength increased similarly in LT+TRT and LT+Pbo (23% vs. 24%; p = 0.95). Total testosterone increased more in LT+TRT than LT+Pbo (133% vs. 32%; p = 0.005). Based on Next Generation Sequencing, of the 39 160 and 39 115 genes detected in LT+TRT and LT+Pbo, respectively, 195 were differentially expressed in LT+TRT and 158 in LT+Pbo. Gene Ontology enrichment analyses revealed that in LT+TRT, just four muscle-related pathways (muscle organ development, muscle organ morphogenesis, regulation of skeletal muscle contraction, muscle atrophy) were downregulated and one pathway (muscle system process) was upregulated. In contrast, in LT+Pbo, nine muscle-related pathways (muscle system process, muscle tissue development, muscle organ development, skeletal muscle tissue development, skeletal muscle organ development, skeletal muscle cell differentiation, muscle organ morphogenesis, response to stimuli involved in regulation of muscle adaptation, muscle atrophy) and one pathway related to bone (bone mineralization involved in bone maturation) were downregulated. Muscle system process was upregulated in LT+TRT but downregulated in LT+Pbo. RT-PCR analyses showed that LT+TRT resulted in a higher expression of MYOD1 (p = 0.02) and WNT4 (p = 0.02), key genes involved in muscle and bone metabolism, respectively, compared with LT+Pbo. We also observed significantly higher mRNA expression of MYBPH (p = 0.006), SCN3B (p = 0.02) and DSC2 (p = 0.01), genes involved in the muscle system process, in response to LT+TRT compared with LT+Pbo. CONCLUSION The addition of TRT to lifestyle therapy mitigates the weight-loss-induced reduction of muscle mass and BMD via countering the weight-loss-induced downregulation of genes involved in muscle and bone anabolism.
Collapse
Affiliation(s)
- Viola Viola
- Center for Translational Research on Inflammatory DiseasesMichael E DeBakey VA Medical CenterHoustonTexasUSA
- Division of Endocrinology, Diabetes and MetabolismBaylor College of MedicineHoustonTexasUSA
- Operative Research Unit of Osteo‐Metabolic and Thyroid DiseasesFondazione Policlinico Universitario Campus Bio‐MedicoRomeItaly
| | - Tagari Samanta
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTexasUSA
| | - Maria Liza Duremdes Nava
- Center for Translational Research on Inflammatory DiseasesMichael E DeBakey VA Medical CenterHoustonTexasUSA
- Division of Endocrinology, Diabetes and MetabolismBaylor College of MedicineHoustonTexasUSA
| | - Alessandra Celli
- Center for Translational Research on Inflammatory DiseasesMichael E DeBakey VA Medical CenterHoustonTexasUSA
- Division of Endocrinology, Diabetes and MetabolismBaylor College of MedicineHoustonTexasUSA
| | - Reina Armamento‐Villareal
- Center for Translational Research on Inflammatory DiseasesMichael E DeBakey VA Medical CenterHoustonTexasUSA
- Division of Endocrinology, Diabetes and MetabolismBaylor College of MedicineHoustonTexasUSA
| | - Ngoc Ho Lam Nguyen
- Center for Translational Research on Inflammatory DiseasesMichael E DeBakey VA Medical CenterHoustonTexasUSA
- Division of Endocrinology, Diabetes and MetabolismBaylor College of MedicineHoustonTexasUSA
| | - Georgia Colleluori
- Center for Translational Research on Inflammatory DiseasesMichael E DeBakey VA Medical CenterHoustonTexasUSA
- Division of Endocrinology, Diabetes and MetabolismBaylor College of MedicineHoustonTexasUSA
| | - Yoann Barnouin
- Center for Translational Research on Inflammatory DiseasesMichael E DeBakey VA Medical CenterHoustonTexasUSA
- Division of Endocrinology, Diabetes and MetabolismBaylor College of MedicineHoustonTexasUSA
| | - Nicola Napoli
- Operative Research Unit of Osteo‐Metabolic and Thyroid DiseasesFondazione Policlinico Universitario Campus Bio‐MedicoRomeItaly
- Division of Bone and Mineral DiseasesWashington University in St LouisSt. LouisMissouriUSA
| | - Clifford Qualls
- Department of Mathematics and StatisticsUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | | | - Dennis T. Villareal
- Center for Translational Research on Inflammatory DiseasesMichael E DeBakey VA Medical CenterHoustonTexasUSA
- Division of Endocrinology, Diabetes and MetabolismBaylor College of MedicineHoustonTexasUSA
| |
Collapse
|
20
|
Zhao J, Fang Y, Qu J, He J, Yi J, Chen R, Yang Q, Zhang K, Wu W, Sun D, Fang B. Utilizing zebrafish models to elucidate mechanisms and develop therapies for skeletal muscle atrophy. Life Sci 2025; 362:123357. [PMID: 39756508 DOI: 10.1016/j.lfs.2024.123357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/04/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025]
Abstract
Skeletal muscle atrophy, resulting from an imbalance in muscle protein synthesis and degradation, compromises muscle quality and function, imposing significant burdens on movement and metabolic stability. Animal models are crucial for understanding the mechanisms of skeletal muscle atrophy and developing clinical prevention and treatment strategies. Zebrafish, as small aquatic vertebrates, exhibit high genetic homology with humans and offer advantages such as rapid reproduction, development, and transparent embryos. Their physiological and anatomical similarities to mammals, including a substantial proportion of skeletal muscle and observable swimming behavior reflecting body dysfunction, make zebrafish an ideal model for studying skeletal muscle-related diseases. This review outlines the development of zebrafish skeletal muscle and highlights key pathways regulating muscle proteins, emphasizing their anatomical and genetic consistency with humans. Various zebrafish models of skeletal muscle atrophy created through physical, chemical, and gene-editing methods are systematically summarized. Current challenges and proposed improvement strategies are also discussed to enhance the reliability and applicability of zebrafish models, providing a comprehensive reference for advancing research on skeletal muscle atrophy.
Collapse
Affiliation(s)
- Jing Zhao
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Junying Qu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiaxuan He
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jia Yi
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, 999077, Hong Kong
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China; Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Bin Fang
- Department of Orthopedic Surgery, Yiwu Central Hospital, the Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu 322000, China.
| |
Collapse
|
21
|
Olenic M, Deelkens C, Heyman E, De Vlieghere E, Zheng X, van Hengel J, De Schauwer C, Devriendt B, De Smet S, Thorrez L. Review: Livestock cell types with myogenic differentiation potential: Considerations for the development of cultured meat. Animal 2025; 19 Suppl 1:101242. [PMID: 39097434 DOI: 10.1016/j.animal.2024.101242] [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: 12/08/2023] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 08/05/2024] Open
Abstract
With the current environmental impact of large-scale animal production and societal concerns about the welfare of farm animals, researchers are questioning whether we can cultivate animal cells for the purpose of food production. This review focuses on a pivotal aspect of the cellular agriculture domain: cells. We summarised information on the various cell types from farm animals currently used for the development of cultured meat, including mesenchymal stromal cells, myoblasts, and pluripotent stem cells. The review delves into the advantages and limitations of each cell type and considers factors like the selection of the appropriate cell source, as well as cell culture conditions that influence cell performance. As current research in cultured meat seeks to create muscle fibers to mimic the texture and nutritional profile of meat, we focused on the myogenic differentiation capacity of the cells. The most commonly used cell type for this purpose are myoblasts or satellite cells, but given their limited proliferation capacity, efforts are underway to formulate myogenic differentiation protocols for mesenchymal stromal cells and pluripotent stem cells. The multipotent character of the latter cell types might enable the creation of other tissues found in meat, such as adipose and connective tissues. This review can help guiding the selection of a cell type or culture conditions in the context of cultured meat development.
Collapse
Affiliation(s)
- M Olenic
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium; Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - C Deelkens
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium; Medical Cell Biology, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - E Heyman
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - E De Vlieghere
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium; Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Belgium
| | - X Zheng
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium
| | - J van Hengel
- Medical Cell Biology, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - C De Schauwer
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - B Devriendt
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - S De Smet
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - L Thorrez
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium.
| |
Collapse
|
22
|
Buonomo V, Lohachova K, Reggio A, Cano-Franco S, Cillo M, Santorelli L, Venditti R, Polishchuk E, Peluso I, Brunello L, Cirillo C, Petrosino S, Silva M, De Cegli R, Di Bartolomeo S, Gargioli C, Swuec P, Cortese M, Stolz A, Bhaskara RM, Grumati P. Two FAM134B isoforms differentially regulate ER dynamics during myogenesis. EMBO J 2025; 44:1039-1073. [PMID: 39762646 PMCID: PMC11832904 DOI: 10.1038/s44318-024-00356-2] [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: 05/09/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 02/19/2025] Open
Abstract
Endoplasmic reticulum (ER) plasticity and ER-phagy are intertwined processes essential for maintaining ER dynamics. We investigated the interplay between two isoforms of the ER-phagy receptor FAM134B in regulating ER remodeling in differentiating myoblasts. During myogenesis, the canonical FAM134B1 is degraded, while its isoform FAM134B2 is transcriptionally upregulated. The switch, favoring FAM134B2, is an important regulator of ER morphology during myogenesis. FAM134B2 partial reticulon homology domain, with its rigid conformational characteristics, enables efficient ER reshaping. FAM134B2 action increases in the active phase of differentiation leading to ER restructuring via ER-phagy, which then reverts to physiological levels when myotubes are mature and the ER is reorganized. Knocking out both FAM134B isoforms in myotubes results in an aberrant proteome landscape and the formation of dilated ER structures, both of which are rescued by FAM134B2 re-expression. Our results underscore how the fine-tuning of FAM134B isoforms and ER-phagy orchestrate the ER dynamics during myogenesis providing insights into the molecular mechanisms governing ER homeostasis in muscle cells.
Collapse
Affiliation(s)
- Viviana Buonomo
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy
- Department of Clinical Medicine and Surgery, Federico II University, 80131, Naples, Italy
| | - Kateryna Lohachova
- Institute of Biochemistry II, School of Medicine, Goethe University, 60590, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, 60438, Frankfurt am Main, Germany
| | - Alessio Reggio
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy
- Saint Camillus International University of Health Sciences, 00131, Rome, Italy
| | - Sara Cano-Franco
- Institute of Biochemistry II, School of Medicine, Goethe University, 60590, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, 60438, Frankfurt am Main, Germany
| | - Michele Cillo
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy
- Department of Clinical Medicine and Surgery, Federico II University, 80131, Naples, Italy
| | - Lucia Santorelli
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy
| | - Rossella Venditti
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, 80131, Naples, Italy
| | - Elena Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy
| | - Ivana Peluso
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy
| | - Lorene Brunello
- Institute of Biochemistry II, School of Medicine, Goethe University, 60590, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, 60438, Frankfurt am Main, Germany
| | - Carmine Cirillo
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy
| | - Sara Petrosino
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of Sacred Heart, 00136, Rome, Italy
| | - Malan Silva
- Cryo-Electron Microscopy Unit, National Facility for Structural Biology, Human Technopole, 20157, Milan, Italy
| | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy
| | | | - Cesare Gargioli
- Department of Biology, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Paolo Swuec
- Cryo-Electron Microscopy Unit, National Facility for Structural Biology, Human Technopole, 20157, Milan, Italy
| | - Mirko Cortese
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy
- DISTABiF, University Luigi Vanvitelli, 81100, Caserta, Italy
| | - Alexandra Stolz
- Institute of Biochemistry II, School of Medicine, Goethe University, 60590, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, 60438, Frankfurt am Main, Germany
| | - Ramachandra M Bhaskara
- Institute of Biochemistry II, School of Medicine, Goethe University, 60590, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, 60438, Frankfurt am Main, Germany
| | - Paolo Grumati
- Telethon Institute of Genetics and Medicine (TIGEM), 80078, Pozzuoli, Italy.
- Department of Clinical Medicine and Surgery, Federico II University, 80131, Naples, Italy.
| |
Collapse
|
23
|
Simon A, Djeddi S, Bournon P, Reiss D, Thompson J, Laporte J. Transcriptomic characterization of postnatal muscle maturation. Dis Model Mech 2025; 18:DMM052098. [PMID: 39945189 PMCID: PMC11911633 DOI: 10.1242/dmm.052098] [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: 09/07/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025] Open
Abstract
Gene differential expression and alternative splicing are mechanisms that give rise to a plethora of tissue-specific transcripts. Although these mechanisms have been studied in various tissues, their role during muscle maturation is not well understood. Because this stage of development is impaired in multiple muscular diseases, we used RNA sequencing to analyze transcriptome remodeling in skeletal muscle from late embryonic stage [embryonic day (E)18.5] to adult mice (7 weeks). Major transcriptomic changes were detected, especially in the first 2 weeks after birth, with a total of 8571 differentially expressed genes and 3096 alternatively spliced genes. Comparison of the two mechanisms showed that they regulate different biological processes essential for the structure and function of skeletal muscle. Investigation of genes mutated in muscle disorders revealed previously unknown transcripts. In particular, we validated a novel exon in Lrp4, a gene mutated in congenital myasthenia, in mice and humans. Overall, the characterization of the transcriptome in disease-relevant tissues revealed key pathways in the regulation of tissue maturation and function. Importantly, the exhaustive description of alternative splicing and resulting transcripts can improve genetic diagnosis of muscular diseases.
Collapse
Affiliation(s)
- Alix Simon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM UMRS 1258, Université de Strasbourg, 67404 Illkirch, France
| | - Sarah Djeddi
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM UMRS 1258, Université de Strasbourg, 67404 Illkirch, France
| | - Pauline Bournon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM UMRS 1258, Université de Strasbourg, 67404 Illkirch, France
| | - David Reiss
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM UMRS 1258, Université de Strasbourg, 67404 Illkirch, France
| | - Julie Thompson
- Complex Systems and Translational Bioinformatics (CSTB), ICube laboratory – CNRS, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM UMRS 1258, Université de Strasbourg, 67404 Illkirch, France
| |
Collapse
|
24
|
Lee YE, Jeong ES, Kim YM, Gong SP. Investigation of scaffold manufacturing conditions for 3-dimensional culture of myogenic cell line derived from black sea bream ( Acanthopagrus schlegelii). Cytotechnology 2025; 77:18. [PMID: 39676766 PMCID: PMC11635079 DOI: 10.1007/s10616-024-00676-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024] Open
Abstract
Culturing fish myogenic cells in vitro holds significant potential to revolutionize aquaculture practices and support sustainable food production. However, advancement in in vitro culture technologies for skeletal muscle-derived myogenic cells have predominantly focused on mammals, with limited studies on fish. Scaffold-based three-dimensional (3D) culture systems for fish myogenic cells remain underexplored, highlighting a critical research gap compared to mammalian systems. This study evaluated the effects of scaffold composition and manufacturing methods on cellular growth in the 3D culture of black sea bream (Acanthopagrus schlegelii) myogenic cells. Scaffolds were manufactured using three natural polymers: black sea bream-derived extracellular matrix (ECM), sodium alginate, and gelatin. Two scaffold types were tested: "cell-laden scaffolds" prepared by mixing cells into the pre-scaffold solution followed by gelation, and "cell-seeding scaffolds" produced by freezing, gelation, and lyophilization before cell inoculation. Scaffold characteristics, including pore size, porosity, swelling ratio, and degradation rate, were assessed. Cell-seeding scaffolds exhibited relatively larger pore size, higher porosity, and higher degradation rate, while cell-laden scaffolds had higher swelling ratios. When black sea bream myogenic cells were cultured in these scaffolds, cell-seeding scaffolds supported cellular growth, particularly when composed of 3% sodium alginate and 4% gelatin with any concentration of ECM. In contrast, cell-laden scaffolds did not support cellular growth regardless of their composition. These findings provide fundamental insights for optimizing scaffold properties to develop more optimized conditions for 3D culture of fish muscle lineage cells.
Collapse
Affiliation(s)
- Ye-Eun Lee
- Department of Fisheries Biology, Pukyong National University, Busan, 48513 Korea
| | - Eun Soo Jeong
- Department of Fisheries Biology, Pukyong National University, Busan, 48513 Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan, 48513 Korea
| | - Seung Pyo Gong
- Department of Fisheries Biology, Pukyong National University, Busan, 48513 Korea
- Major in Aquaculture and Applied Life Science, Division of Fisheries Life Science, Pukyong National University, 45 Yongso-ro, Nam-Gu, Busan, 48513 Korea
| |
Collapse
|
25
|
Zuo H, Jiang W, Gao J, Ma Z, Li C, Peng Y, Jin J, Zhan X, Lv W, Liu X, Hu J, Zhang M, Jia Y, Xu Z, Tang J, Zheng R, Zuo B. SYISL Knockout Promotes Embryonic Muscle Development of Offspring by Modulating Maternal Gut Microbiota and Fetal Myogenic Cell Dynamics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410953. [PMID: 39680624 PMCID: PMC11809340 DOI: 10.1002/advs.202410953] [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: 09/09/2024] [Revised: 12/02/2024] [Indexed: 12/18/2024]
Abstract
Embryonic muscle fiber formation determines post-birth muscle fiber totals. The previous research shows SYISL knockout significantly increases muscle fiber numbers and mass in mice, but the mechanism remains unclear. This study confirms that the SYISL gene, maternal gut microbiota, and their interaction significantly affect the number of muscle fibers in mouse embryos through distinct mechanisms, as SYISL knockout alters maternal gut microbiota composition and boosts butyrate levels in embryonic serum. Both fecal microbiota transplantation and butyrate feeding significantly increase muscle fiber numbers in offspring, with butyrate inhibiting histone deacetylases and increasing histone acetylation in embryonic muscle. Combined analysis of RNA-seq between wild-type and SYISL knockout mice with ChIP-seq for H3K9ac and H3K27ac reveals that SYISL and maternal microbiota interaction regulates myogenesis via the butyrate-HDAC-H3K9ac/H3K27ac pathway. Furthermore, scRNA-seq analysis shows that SYISL knockout alone significantly increases the number and proportion of myogenic cells and their dynamics, independently of regulating histone acetylation levels. Cell communication analysis suggests that this may be due to the downregulation of signaling pathways such as MSTN and TGFβ. Overall, multiple pathways are highlighted through which SYISL influences embryonic muscle development, offering valuable insights for treating muscle diseases and improving livestock production.
Collapse
Affiliation(s)
- Hao Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryWuhan430070China
| | - Wei Jiang
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Jianwei Gao
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Zhibo Ma
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Chen Li
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Yaxin Peng
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Jianjun Jin
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Xizhen Zhan
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Wei Lv
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Xiao Liu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Jingjing Hu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Mengdi Zhang
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Yiming Jia
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Zaiyan Xu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Department of Basic Veterinary MedicineCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhan430070China
| | - Junming Tang
- Hubei Key Laboratory of Embryonic Stem Cell ResearchSchool of Basic Medicine ScienceHubei University of MedicineShiyan442000China
| | - Rong Zheng
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
| | - Bo Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural AffairsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of EducationHuazhong Agricultural UniversityWuhan430070China
- Hubei Hongshan LaboratoryWuhan430070China
| |
Collapse
|
26
|
Wang K, Xu M, Han X, Liu H, Han J, Sun W, Zhou H. Transcriptome analysis of muscle atrophy in Leizhou black goats: identification of key genes and insights into limb-girdle muscular dystrophy. BMC Genomics 2025; 26:80. [PMID: 39871147 PMCID: PMC11773938 DOI: 10.1186/s12864-025-11282-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: 08/07/2024] [Accepted: 01/23/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND The Leizhou Black Goat (LZBG), a prominent breed in tropical China's meat goat industry, frequently exhibits inherent muscle atrophy and malnutrition-related traits. Particularly, muscles critical for support, such as the legs, often display severe symptoms. This study aimed to investigate the differential genes and signaling pathways influencing muscle development and atrophy across various muscle locations in LZBG from a muscular atrophy-affected family. RESULTS Differential expression analysis revealed 536 mRNAs with significant differences across three muscle groups. Marked variations in mRNA expression patterns were observed between leg and back muscles versus abdominal muscles, reflecting characteristics similar to those found in limb-girdle muscular dystrophy. The analysis identified several key differentially expressed genes implicated in muscle development and atrophy, including PITX1, COLQ, ZIC1, SBK2, and TBX1, showed Significant difference expression levels and expression patterns with normal individuals. Functional annotation and protein interaction network analysis indicated enrichment of these genes in muscle-related pathways. Protein interaction network analysis identified five key clusters related to muscle function and development. CONCLUSION The mRNA expression patterns of the leg and back muscles in LZBG from a muscular atrophy-affected family differed significantly from those of the abdominal muscle, displaying typical characteristics of limb-girdle muscular dystrophy. Genes such as PITX1, TBX1, SBK2, TCAP, and COLQ were identified as key regulators of muscle development and contributors to muscle atrophy. These findings enhance our understanding of the mechanisms underlying muscular atrophy in LZBGs. The identification of key genes and pathways provides valuable insights for developing future breeding strategies aimed at improving meat production efficiency.
Collapse
Affiliation(s)
- Ke Wang
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, 524013, China.
- Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Sanya, 572025, China.
| | - Mengning Xu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaotao Han
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, 524013, China
| | - Hu Liu
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, 524013, China
| | - Jiancheng Han
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, 524013, China
| | - Wei Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou, 225009, China
| | - Hanlin Zhou
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, 524013, China.
- Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Sanya, 572025, China.
| |
Collapse
|
27
|
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
|
28
|
Tourel A, Reynaud-Dulaurier R, Brocard J, Fauré J, Marty I, Petiot A. RyR1 Is Involved in the Control of Myogenesis. Cells 2025; 14:158. [PMID: 39936950 PMCID: PMC11817019 DOI: 10.3390/cells14030158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
The RyR1 calcium release channel is a key player in skeletal muscle excitation-contraction coupling. Mutations in the RYR1 gene are associated with congenital myopathies. Recently, a role of RyR1 in myotubes differentiation has been proposed and attributed to its calcium channel function, which nonetheless remains to be clearly demonstrated. In order to clarify RyR1 role in myogenesis, we have developed an in vitro model, the so-called RyR1-Rec myotubes, which are mouse primary myotubes with an inducible decrease in RyR1 protein amount and in RyR1-mediated calcium release. Using this model, we showed that the RyR1 protein decrease was responsible for an increase in both differentiation and fusion, from the RNA level to the morphological level, without affecting the myogenic factors MyoD and MyoG. Although an increase in mTOR pathway was observed in RyR1-Rec myotubes, it did not seem to be responsible for the role of RyR1 in myogenesis. Additionally, even if modulation of intracellular calcium level affected RyR1-Rec myotubes differentiation, we have shown that the role of RyR1 in myogenesis was independent of its calcium channel function. Therefore, our findings indicate that, besides its pivotal role as a calcium channel responsible for muscle contraction, RyR1 fulfills a calcium-independent inhibitor function of myogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Isabelle Marty
- University Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institute Neurosciences, 38000 Grenoble, France (R.R.-D.); (J.B.); (J.F.)
| | - Anne Petiot
- University Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institute Neurosciences, 38000 Grenoble, France (R.R.-D.); (J.B.); (J.F.)
| |
Collapse
|
29
|
Nunes OBDS, Buranello TW, Farias FDA, Rosero J, Recchia K, Bressan FF. Can cell-cultured meat from stem cells pave the way for sustainable alternative protein? Curr Res Food Sci 2025; 10:100979. [PMID: 40040753 PMCID: PMC11878651 DOI: 10.1016/j.crfs.2025.100979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 03/06/2025] Open
Abstract
As the global population grows, the demand for food and animal-derived products rises significantly, posing a notable challenge to the progress of society in general. Alternative protein production may adequately address such a challenge, and cell-based meat production emerges as a promising solution. This review investigates methodologies for in vitro myogenesis and adipogenesis from stem cells (adult, embryonic, or induced pluripotent stem cells - iPSCs) across different animal species, as well as the remaining challenges for scalability, the possibility of genetic modification, along with safety concerns regarding the commercialization of cell-cultured meat. Regarding such complexities, interdisciplinary approaches will be vital for assessing the potential of cell-cultured meat as a sustainable protein source, mimicking the sensory and nutritional attributes of conventional livestock meat whilst meeting the demands of a growing global population while mitigating environmental impacts.
Collapse
Affiliation(s)
- Octavio Bignardi da Silva Nunes
- Department of Food Engineering, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
| | - Tiago Willian Buranello
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Postgraduate Program in Anatomy of Domestic and Wils Species, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, SP, Brazil
| | - Fabiana de Andrade Farias
- Department of Food Engineering, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
| | - Jenyffer Rosero
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Postgraduate Program in Anatomy of Domestic and Wils Species, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, SP, Brazil
| | - Kaiana Recchia
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Postgraduate Program in Anatomy of Domestic and Wils Species, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, SP, Brazil
| | - Fabiana Fernandes Bressan
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Postgraduate Program in Anatomy of Domestic and Wils Species, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, SP, Brazil
| |
Collapse
|
30
|
Gustafson AL, Durbin AD, Artinger KB, Ford HL. Myogenesis gone awry: the role of developmental pathways in rhabdomyosarcoma. Front Cell Dev Biol 2025; 12:1521523. [PMID: 39902277 PMCID: PMC11788348 DOI: 10.3389/fcell.2024.1521523] [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: 11/01/2024] [Accepted: 12/23/2024] [Indexed: 02/05/2025] Open
Abstract
Rhabdomyosarcoma is a soft-tissue sarcoma that occurs most frequently in pediatric patients and has poor survival rates in patients with recurrent or metastatic disease. There are two major sub-types of RMS: fusion-positive (FP-RMS) and fusion-negative (FN-RMS); with FP-RMS typically containing chromosomal translocations between the PAX3/7-FOXO1 loci. Regardless of subtype, RMS resembles embryonic skeletal muscle as it expresses the myogenic regulatory factors (MRFs), MYOD1 and MYOG. During normal myogenesis, these developmental transcription factors (TFs) orchestrate the formation of terminally differentiated, striated, and multinucleated skeletal muscle. However, in RMS these TFs become dysregulated such that they enable the sustained properties of malignancy. In FP-RMS, the PAX3/7-FOXO1 chromosomal translocation results in restructured chromatin, altering the binding of many MRFs and driving an oncogenic state. In FN-RMS, re-expression of MRFs, as well as other myogenic TFs, blocks terminal differentiation and holds cells in a proliferative, stem-cell-like state. In this review, we delve into the myogenic transcriptional networks that are dysregulated in and contribute to RMS progression. Advances in understanding the mechanisms through which myogenesis becomes stalled in RMS will lead to new tumor-specific therapies that target these aberrantly expressed developmental transcriptional pathways.
Collapse
Affiliation(s)
- Annika L. Gustafson
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Adam D. Durbin
- Division of Molecular Oncology, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Kristin B. Artinger
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN, United States
| | - Heide L. Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
31
|
Fu Y, Hao X, Shang P, Nie J, Chamba Y, Zhang B, Zhang H. MUSTN1 Interaction With SMPX Regulates Muscle Development and Regeneration. Cell Prolif 2025:e13809. [PMID: 39828423 DOI: 10.1111/cpr.13809] [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: 09/11/2024] [Revised: 12/28/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
Pigs are important agricultural animals whose growth rate and meat production performance are related to muscle development. Musculoskeletal embryonic nuclear protein 1 (MUSTN1) participates in various biological processes, including myogenesis and growth in animals, but the physiological functions and mechanisms of porcine MUSTN1 on muscle development are unclear; thus, we aimed to elucidate them. We found that MUSTN1 was highly expressed in the muscles of fast-growing pigs. Functionally, MUSTN1 promoted myoblast proliferation and differentiation. MUSTN1 knockout mice exhibited reduced muscle mass and fibre cross-sectional area, decreased exercise endurance, and delayed muscle regeneration. Small muscle protein X-linked (SMPX) was identified as an interacting protein of MUSTN1, and its promotion of myogenic differentiation depended on MUSTN1. Furthermore, MUSTN1 stabilised SMPX and maintained myofiber morphology. This study suggests that MUSTN1 is a critical regulator in the control of muscle development and regeneration and is a potential target for animal genetic improvement and the treatment of human muscle disease.
Collapse
Affiliation(s)
- Yu Fu
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing, China
| | - Xin Hao
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing, China
| | - Peng Shang
- Department of Animal Husbandry, Xizang Agricultural and Animal Husbandry University, Linzhi, China
| | - Jingru Nie
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing, China
| | - Yangzom Chamba
- Department of Animal Husbandry, Xizang Agricultural and Animal Husbandry University, Linzhi, China
| | - Bo Zhang
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing, China
| | - Hao Zhang
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
32
|
Nguyen MT, Ly QK, Ngo THP, Lee W. Calponin 3 Regulates Myoblast Proliferation and Differentiation Through Actin Cytoskeleton Remodeling and YAP1-Mediated Signaling in Myoblasts. Cells 2025; 14:142. [PMID: 39851570 PMCID: PMC11764405 DOI: 10.3390/cells14020142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
An actin-binding protein, known as Calponin 3 (CNN3), modulates the remodeling of the actin cytoskeleton, a fundamental process for the maintenance of skeletal muscle homeostasis. Although the roles of CNN3 in actin remodeling have been established, its biological significance in myoblast differentiation remains largely unknown. This study investigated the functional significance of CNN3 in myogenic differentiation, along with its effects on actin remodeling and mechanosensitive signaling in C2C12 myoblasts. CNN3 knockdown led to a marked increase in filamentous actin, which promoted the nuclear localization of Yes-associated protein 1 (YAP1), a mechanosensitive transcriptional coactivator required for response to the mechanical cues that drive cell proliferation. Subsequently, CNN3 depletion enhanced myoblast proliferation by upregulating the expression of the YAP1 target genes related to cell cycle progression, such as cyclin B1, cyclin D1, and PCNA. According to a flow cytometry analysis, CNN3-deficient cells displayed higher S and G2/M phase fractions, which concurred with elevated proliferation rates. Furthermore, CNN3 knockdown impaired myogenic differentiation, as evidenced by reduced levels of MyoD, MyoG, and MyHC, key markers of myogenic commitment and maturation, and immunocytochemistry showed that myotube formation was diminished in CNN3-suppressed cells, which was supported by lower differentiation and fusion indices. These findings reveal that CNN3 is essential for myogenic differentiation, playing a key role in regulating actin remodeling and cellular localization of YAP1 to orchestrate the proliferation and differentiation in myogenic progenitor cells. This study highlights CNN3 as a critical regulator of skeletal myogenesis and suggests its therapeutic potential as a target for muscle atrophy and related disorders.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (T.H.P.N.)
| | - Quoc Kiet Ly
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (T.H.P.N.)
| | - Thanh Huu Phan Ngo
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (T.H.P.N.)
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (T.H.P.N.)
- Section of Molecular and Cellular Medicine, Medical Institute of Dongguk University, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
| |
Collapse
|
33
|
Wenlun W, Chaohang Y, Yan H, Wenbin L, Nanqing Z, Qianmin H, Shengcai W, Qing Y, Shirui Y, Feng Z, Lingyun Z. Developing a ceRNA-based lncRNA-miRNA-mRNA regulatory network to uncover roles in skeletal muscle development. FRONTIERS IN BIOINFORMATICS 2025; 4:1494717. [PMID: 39882307 PMCID: PMC11774864 DOI: 10.3389/fbinf.2024.1494717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
The precise role of lncRNAs in skeletal muscle development and atrophy remain elusive. We conducted a bioinformatic analysis of 26 GEO datasets from mouse studies, encompassing embryonic development, postnatal growth, regeneration, cell proliferation, and differentiation, using R and relevant packages (limma et al.). LncRNA-miRNA relationships were predicted using miRcode and lncBaseV2, with miRNA-mRNA pairs identified via miRcode, miRDB, and Targetscan7. Based on the ceRNA theory, we constructed and visualized the lncRNA-miRNA-mRNA regulatory network using ggalluvial among other R packages. GO, Reactome, KEGG, and GSEA explored interactions in muscle development and regeneration. We identified five candidate lncRNAs (Xist, Gas5, Pvt1, Airn, and Meg3) as potential mediators in these processes and microgravity-induced muscle wasting. Additionally, we created a detailed lncRNA-miRNA-mRNA regulatory network, including interactions such as lncRNA Xist/miR-126/IRS1, lncRNA Xist/miR-486-5p/GAB2, lncRNA Pvt1/miR-148/RAB34, and lncRNA Gas5/miR-455-5p/SOCS3. Significant signaling pathway changes (PI3K/Akt, MAPK, NF-κB, cell cycle, AMPK, Hippo, and cAMP) were observed during muscle development, regeneration, and atrophy. Despite bioinformatics challenges, our research underscores the significant roles of lncRNAs in muscle protein synthesis, degradation, cell proliferation, differentiation, function, and metabolism under both normal and microgravity conditions. This study offers new insights into the molecular mechanisms governing skeletal muscle development and regeneration.
Collapse
Affiliation(s)
- Wang Wenlun
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Yu Chaohang
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Huang Yan
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Li Wenbin
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Zhou Nanqing
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Hu Qianmin
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Wu Shengcai
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Yuan Qing
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Yu Shirui
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Zhang Feng
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Zhu Lingyun
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| |
Collapse
|
34
|
Stevens BT, Hatley ME. Developmental Heterogeneity of Rhabdomyosarcoma. Cold Spring Harb Perspect Med 2025; 15:a041583. [PMID: 38772705 PMCID: PMC11694754 DOI: 10.1101/cshperspect.a041583] [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] [Indexed: 05/23/2024]
Abstract
Rhabdomyosarcoma (RMS) is a pediatric embryonal solid tumor and the most common pediatric soft tissue sarcoma. The histology and transcriptome of RMS resemble skeletal muscle progenitor cells that have failed to terminally differentiate. Thus, RMS is typically thought to arise from corrupted skeletal muscle progenitor cells during development. However, RMS can occur in body regions devoid of skeletal muscle, suggesting the potential for nonmyogenic cells of origin. Here, we discuss the interplay between RMS driver mutations and cell(s) of origin with an emphasis on driving location specificity. Additionally, we discuss the mechanisms governing RMS transformation events and tumor heterogeneity through the lens of transcriptional networks and epigenetic control. Finally, we reimagine Waddington's developmental landscape to include a plane of transformation connecting distinct lineage landscapes to more accurately reflect the phenomena observed in pediatric cancers.
Collapse
Affiliation(s)
- Bradley T Stevens
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
- St. Jude Graduate School of Biomedical Sciences, Memphis, Tennessee 38105, USA
| | - Mark E Hatley
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| |
Collapse
|
35
|
Fu X, Zhang F, Dong X, Pu L, Feng Y, Xu Y, Gao F, Liang T, Kang J, Sun H, Hong T, Liu Y, Zhou H, Jiang J, Yin D, Hu X, Wang DZ, Ding J, Chen J. Adapting cytoskeleton-mitochondria patterning with myocyte differentiation by promyogenic PRR33. Cell Death Differ 2025; 32:177-193. [PMID: 39147882 PMCID: PMC11742405 DOI: 10.1038/s41418-024-01363-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: 08/07/2023] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Coordinated cytoskeleton-mitochondria organization during myogenesis is crucial for muscle development and function. Our understanding of the underlying regulatory mechanisms remains inadequate. Here, we identified a novel muscle-enriched protein, PRR33, which is upregulated during myogenesis and acts as a promyogenic factor. Depletion of Prr33 in C2C12 represses myoblast differentiation. Genetic deletion of Prr33 in mice reduces myofiber size and decreases muscle strength. The Prr33 mutant mice also exhibit impaired myogenesis and defects in muscle regeneration in response to injury. Interactome and transcriptome analyses reveal that PRR33 regulates cytoskeleton and mitochondrial function. Remarkably, PRR33 interacts with DESMIN, a key regulator of cytoskeleton-mitochondria organization in muscle cells. Abrogation of PRR33 in myocytes substantially abolishes the interaction of DESMIN filaments with mitochondria, leading to abnormal intracellular accumulation of DESMIN and mitochondrial disorganization/dysfunction in myofibers. Together, our findings demonstrate that PRR33 and DESMIN constitute an important regulatory module coordinating mitochondrial organization with muscle differentiation.
Collapse
Affiliation(s)
- Xuyang Fu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Feng Zhang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Xiaoxuan Dong
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Linbin Pu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Yan Feng
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yang Xu
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Feng Gao
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Tian Liang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Jianmeng Kang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Hongke Sun
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Tingting Hong
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yunxia Liu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Hongmei Zhou
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jun Jiang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Deling Yin
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Xinyang Hu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Da-Zhi Wang
- University of South Florida Health Heart Institute, Center for Regenerative Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33602, USA
| | - Jian Ding
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Jinghai Chen
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China.
| |
Collapse
|
36
|
Fumoto Y, Takada S, Onodera Y, Hatakeyama S, Oikawa T. Development of a Myogenin minimal promoter-based system for visualizing the degree of myogenic differentiation. Biochem Biophys Res Commun 2024; 741:151091. [PMID: 39622159 DOI: 10.1016/j.bbrc.2024.151091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/11/2024]
Abstract
Myogenic differentiation plays a fundamental role in myogenesis during development and in muscle regeneration. Sequential expression of myogenic regulatory factors (MRFs) including myogenin in the progenitor cells triggers the expression of effector proteins such as myosin heavy chain (MHC), leading to the terminal muscle differentiation. Although we have a snapshot-like understanding of molecules at each stage of the differentiation, how these molecules are interrelated in the continuum of myogenic differentiation remains poorly understood. In this study, we analyzed the dynamics of the minimal Myogenin promoter activity in live myoblasts. With the development of a new co-expression analysis method, we were able to reveal in detail the relationship between this Myogenin promoter activity and the expression of endogenous myogenin or MHC, as differentiation markers. Consequently, we found that our visualization system of myogenic differentiation is suitable for monitoring the transition from myoblasts to myotubes, in which the Myogenin promoter activity quantitatively represents the degree of myogenic differentiation. Thus, this system allows simultaneous observation of the degree of myoblast differentiation in relation to other molecules, which would contribute to deepening our understanding of myogenic differentiation as a continuous process.
Collapse
Affiliation(s)
- Yoshizuki Fumoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Japan.
| | - Shingo Takada
- Department of Sports Education, Faculty of Lifelong Sport, Hokusho University, Japan
| | - Yasuhito Onodera
- Department of Molecular and Cellular Dynamics Research, Graduate School of Biomedical Science and Engineering, Hokkaido University, Japan
| | - Shigetsugu Hatakeyama
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan
| | - Tsukasa Oikawa
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Japan.
| |
Collapse
|
37
|
Liao Y, Peng Z, Fu S, Hua Y, Luo W, Liu R, Chen Y, Gu W, Zhao P, Zhao J, Wang Y, Wang H. Elevated EBF2 in mouse but not pig drives the progressive brown fat lineage specification via chromatin activation. J Adv Res 2024:S2090-1232(24)00624-6. [PMID: 39736442 DOI: 10.1016/j.jare.2024.12.046] [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: 06/28/2024] [Revised: 12/08/2024] [Accepted: 12/28/2024] [Indexed: 01/01/2025] Open
Abstract
Brown adipose tissue (BAT) is responsible for non-shivering thermogenesis, but it is absent in some mammals, including pigs. During development, BAT progenitors are derived from paired box 7 (Pax7)-expressing somitic mesodermal stem cells, which also give rise to skeletal muscle. However, the intrinsic mechanisms underlying the fate decisions between brown fat and muscle progenitors remain elusive across species. In this study, we analyzed the dynamics of chromatin landscape during the segregation and specification of brown fat and muscle lineages from Pax7+ multipotent mesodermal stem cells, aiming to uncover epigenetic factors that drive de novo BAT formation. Notably, myogenic progenitors were specified at embryonic day (E) 12.5, exhibiting high levels of H3K4me3 and low H3K27me3 at muscle-related genes. In contrast, the specification of the BAT lineage occurred much later, with coordinated step-wise depositions of histone modifications at BAT-associated genes from E10.5 to E14.5. We identified the transcription factor early B-cell factor 2 (EBF2) as a key driver of the progressive specification of brown fat lineage and the simultaneous deviation away from the muscle lineage. Mechanistically, EBF2 interacts with transcriptional co-activators CREB binding protein/ E1A-binding protein p300 (CBP/P300) to induce H3K27ac deposition and chromatin activation at BAT-associated genes to promote brown adipogenesis. Both mouse and pig EBF2 could potently stimulate adipogenesis in unspecified multipotent mesodermal stem cells. However, in pigs, EBF2 expression was depleted during the critical lineage specification time window, thus preventing the embryonic formation and development of porcine BAT. Hence, the elevation of EBF2 in mice, but not in pigs, promote chromatin activation to drive the progressive specification of brown fat lineage.
Collapse
Affiliation(s)
- Yinlong Liao
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China; Yazhouwan National Laboratory, Sanya, China
| | - Zhelun Peng
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Shanshan Fu
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China
| | - Yao Hua
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China
| | - Wenzhe Luo
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China
| | - Ruige Liu
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yingjin Chen
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wei Gu
- Shandong Provincial Key Laboratory of Animal Microecologics and Efficient Breeding of Livestock and Poultry, Shandong Baolai-Leelai Bio-Tech Co., Ltd, Taian, China
| | - Pengxiang Zhao
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China
| | - Jianguo Zhao
- Institute of Zoology, Chinese Academy of Science, Beijing, China
| | - Yanfang Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Heng Wang
- College of Animal Science, Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian, China; College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
38
|
Ly QK, Nguyen MT, Ngo THP, Lee W. Essential Role of Cortactin in Myogenic Differentiation: Regulating Actin Dynamics and Myocardin-Related Transcription Factor A-Serum Response Factor (MRTFA-SRF) Signaling. Int J Mol Sci 2024; 25:13564. [PMID: 39769327 PMCID: PMC11677934 DOI: 10.3390/ijms252413564] [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: 10/30/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Cortactin (CTTN) is an actin-binding protein regulating actin polymerization and stabilization, which are vital processes for maintaining skeletal muscle homeostasis. Despite the established function of CTTN in actin cytoskeletal dynamics, its role in the myogenic differentiation of progenitor cells remains largely unexplored. In this study, we investigated the role of CTTN in the myogenic differentiation of C2C12 myoblasts by analyzing its effects on actin cytoskeletal remodeling, myocardin-related transcription factor A (MRTFA) nuclear translocation, serum response factor (SRF) activation, expression of myogenic transcription factors, and myotube formation. CTTN expression declined during myogenic differentiation, paralleling the reduction in MyoD, suggesting a potential role in the early stages of myogenesis. We also found that CTTN knockdown in C2C12 myoblasts reduced filamentous actin, enhanced globular actin levels, and inhibited the nuclear translocation of MRTFA, resulting in suppressed SRF activity. This led to the subsequent downregulation of myogenic regulatory factors, such as MyoD and MyoG. Furthermore, CTTN knockdown reduced the nuclear localization of YAP1, a mechanosensitive transcription factor, further supporting its regulatory roles in cell cycle and proliferation. Consequently, CTTN depletion impeded proliferation, differentiation, and myotube formation in C2C12 myoblasts, highlighting its dual role in the coordination of cell cycle regulation and myogenic differentiation of progenitor cells during myogenesis. This study identifies CTTN as an essential regulator of myogenic differentiation via affecting the actin remodeling-MRTFA-SRF signaling axis and cell proliferation.
Collapse
Affiliation(s)
- Quoc Kiet Ly
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (Q.K.L.); (M.T.N.); (T.H.P.N.)
| | - Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (Q.K.L.); (M.T.N.); (T.H.P.N.)
| | - Thanh Huu Phan Ngo
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (Q.K.L.); (M.T.N.); (T.H.P.N.)
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (Q.K.L.); (M.T.N.); (T.H.P.N.)
- Section of Molecular and Cellular Medicine, Medical Institute of Dongguk University, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
| |
Collapse
|
39
|
Hüttemeister J, Rudolph F, Radke MH, Fink C, Friedrich D, Preibisch S, Falcke M, Wagner E, Lehnart SE, Gotthardt M. Visualizing sarcomere and cellular dynamics in skeletal muscle to improve cell therapies. eLife 2024; 13:e95597. [PMID: 39688479 DOI: 10.7554/elife.95597] [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: 12/27/2023] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The giant striated muscle protein titin integrates into the developing sarcomere to form a stable myofilament system that is extended as myocytes fuse. The logistics underlying myofilament assembly and disassembly have started to emerge with the possibility to follow labeled sarcomere components. Here, we generated the mCherry knock-in at titin's Z-disk to study skeletal muscle development and remodeling. We find titin's integration into the sarcomere tightly regulated and its unexpected mobility facilitating a homogeneous distribution of titin after cell fusion - an integral part of syncytium formation and maturation of skeletal muscle. In adult mCherry-titin mice, treatment of muscle injury by implantation of titin-eGFP myoblasts reveals how myocytes integrate, fuse, and contribute to the continuous myofilament system across cell boundaries. Unlike in immature primary cells, titin proteins are retained at the proximal nucleus and do not diffuse across the whole syncytium with implications for future cell-based therapies of skeletal muscle disease.
Collapse
Affiliation(s)
- Judith Hüttemeister
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| | - Franziska Rudolph
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Michael H Radke
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Claudia Fink
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Dhana Friedrich
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Stephan Preibisch
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Martin Falcke
- Computational Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Eva Wagner
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
| | - Stephan E Lehnart
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Gotthardt
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
40
|
Han JH, Jang SW, Kim YR, Na GR, Park JH, Choi HW. Comparative Analysis of Different Extracellular Matrices for the Maintenance of Bovine Satellite Cells. Animals (Basel) 2024; 14:3496. [PMID: 39682461 DOI: 10.3390/ani14233496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/16/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Cultured meat produced using satellite cells has emerged to address issues such as overpopulation, the ethical conundrums associated with the breeding environment, and the methane gas emissions associated with factory farming. To date, however, the challenges of maintaining satellite cells in vitro and reducing the costs of the culture media are still substantial. Gelatin, collagen, and fibronectin are commonly used extracellular matrices (ECMs) that facilitate signal integration with the cells and promote cell adhesion. In this study, we compared the proliferation, cell cycle, immunocytochemistry, and expression levels of Pax7, Pax3, Myf5, MyoD1, and MyoG genes in bovine satellite cells (BSCs) cultured on gelatin-, collagen- and fibronectin-coated dishes as part of short- and long-term cultures. We observed that BSCs cultured on gelatin-coated dishes showed higher levels of Pax7 expression than BSCs cultured on collagen- and fibronectin-coated dishes in both short- and long-term cultures, indicating that BSCs cultured on gelatin effectively maintained the satellite cell population in both the short- and long-term cultures. Our study highlights that gelatin is an effective ECM for the maintenance of BSCs and the production of cultured meat.
Collapse
Affiliation(s)
- Jae Ho Han
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Si Won Jang
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Ye Rim Kim
- Department of Animal Science, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Ga Rim Na
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Ji Hoon Park
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Hyun Woo Choi
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Department of Animal Science, Jeonbuk National University, Jeonju 54896, Republic of Korea
| |
Collapse
|
41
|
Diao Z, Jia S, Itoyama E, Yoshioka H, Murakami M, Funaba M. A possibility of uncoupling protein 1 induction with the enhancement of myogenesis related to ruminal fermentation. Sci Rep 2024; 14:29857. [PMID: 39622913 PMCID: PMC11612152 DOI: 10.1038/s41598-024-81272-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
The expression of uncoupling protein 1 (UCP1), which regulates energy expenditure, is limited to brown/beige adipocytes in most mammals; however, it is also detected in the skeletal muscles of cattle. We previously observed a positive relationship between Ucp1 and fast-twitch myosin heavy chain (Myh) expression in bovine skeletal muscles. In the present study, we explored the regulatory expression of Ucp1 in bovine myogenic cells using cell culture. Vitamin C and high-dose capsaicin, which induce the formation of fast-twitch myotubes in murine myogenic cells, did not stimulate myogenesis in bovine myosatellite cells. Treatment with 4-phenylbutyric acid (PBA), a histone deacetylase inhibitor that enhances histone acetylation, upregulates the expression of all myogenic regulatory factors (MRFs), except Myog, in bovine myogenic cells. Consistent with this, PBA increased the expression levels of acetylated lysine 27 of histone 3 (H3K27), the fast-twitch component MYH1/2, and Ucp1 in bovine myogenic cells. SB203580, an inhibitor of p38 MAP kinase, blocked PBA-induced myogenesis and Ucp1 upregulation. PBA is a butyric acid-related molecule, and cattle produce large amounts of volatile fatty acids (VFAs), including acetic acid, propionic acid, and butyric acid, through ruminal fermentation. Propionic acid treatment stimulated H3K27 acetylation, myogenesis, and Ucp1 induction. Thus, the upregulation of muscular Ucp1 may be related to myogenic stimulation through the modulation of histone acetylation status in cattle; we propose that the cattle-specific expression of muscular UCP1 results from VFA production through ruminal fermentation.
Collapse
Affiliation(s)
- Zhicheng Diao
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwakecho, Kyoto, 606-8502, Japan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Shunhua Jia
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwakecho, Kyoto, 606-8502, Japan
| | - Erina Itoyama
- Kyoto University Livestock Farm, Kyotanba, 622-0203, Japan
| | | | - Masaru Murakami
- Laboratory of Molecular Biology, Azabu University School of Veterinary Medicine, Sagamihara, 252-5201, Japan
| | - Masayuki Funaba
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwakecho, Kyoto, 606-8502, Japan.
- Kyoto University Livestock Farm, Kyotanba, 622-0203, Japan.
| |
Collapse
|
42
|
Ahmad SS, Lim JH, Choi I, Lee EJ. Biocomputational screening of natural compounds targeting 15-hydroxyprostaglandin dehydrogenase to improve skeletal muscle during aging. Mol Divers 2024; 28:4425-4439. [PMID: 38904907 DOI: 10.1007/s11030-024-10825-9] [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: 11/28/2023] [Accepted: 02/18/2024] [Indexed: 06/22/2024]
Abstract
Skeletal muscle (SM) contains a diverse population of muscle stem (or satellite) cells, which are essential for the maintenance of muscle tissue and positively regulated by prostaglandin E2 (PGE2). However, in aged SM, PGE2 levels are reduced due to increased prostaglandin catabolism by 15-hydroxyprostaglandin dehydrogenase (15-PGDH), a negative regulator of SM tissue repair and regeneration. Screening of a library of 80,617 natural compounds in the ZINC database against 15-PGDH was conducted from PyRx. Further, drug-likeness rules, including those of Lipinski, Ghose, Veber, Egan, and Muegge were performed. The selected complex was forwarded for MD simulations up to 100ns. Based on free energy of binding obtained from docking revealed that ZINC14557836 and ZINC14638400 more potently inhibiting to 15-PGDH than SW033291 (the control and high-affinity inhibitor of 15-PGDH). The free energies of binding obtained from PyRx for 15-PGDH-ZINC14557836, 15-PGDH-ZINC14638400, and 15-PGDH-SW033291 complexes were - 10.30, -9.80, and - 8.0 kcal/mol, respectively. Root mean square deviations (RMSDs), root mean square fluctuations (RMSFs), radii of gyration (Rg), solvent-accessible surface areas (SASAs), and H-bond parameters obtained by 100 ns MD simulations predicted ZINC14557836 and ZINC14638400 more stably complexed with 15-PGDH than SW033291. The several parameters, including physicochemical properties and drug-likenesses, were within acceptable limits, and ZINC14557836 and ZINC14638400 also satisfied other drug-likeness rules, including those of Lipinski, Ghose, Veber, Egan, and Muegge. These findings suggest that ZINC14557836 and ZINC14638400 provide starting points for the development of medications that increase SM regeneration and muscle stem (or satellite) cell numbers by inhibiting 15-PGDH.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Jeong Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea.
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea.
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea.
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea.
| |
Collapse
|
43
|
Hu X, Liu Y, Tang B, Hu J, He H, Liu H, Li L, Hu S, Wang J. Comparative transcriptomic analysis revealed potential mechanisms regulating the hypertrophy of goose pectoral muscles. Poult Sci 2024; 103:104498. [PMID: 39504833 PMCID: PMC11577216 DOI: 10.1016/j.psj.2024.104498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024] Open
Abstract
Pectoral muscle development is an important economic trait. According to the different essence, muscle development can be divided into 2 processes: embryonic muscle fiber generation and postnatal muscle fiber hypertrophy, and postnatal muscle fiber hypertrophy has a greater impact on muscle development than the number of muscle fibers formed during the embryonic phase in poultry. However, the underlying mechanisms regulating the hypertrophy of goose pectoral muscles have not been elucidated. Therefore, the purpose of the present study was to conduct transcriptome sequencing in pectoral muscles of both Landes (LD) and Sichuan White (SW) geese at 6, 10, and 30 weeks of age to reveal the molecular mechanisms regulating pectoral muscle hypertrophy through intra-breed and inter-breed bioinformatics analyses. Phenotypically, the pectoral muscle weight/index of LD and SW geese increased from 6 to 30 weeks of age, and except for the pectoral muscle index at 10 weeks of age (P = 0.962), at the same age, the pectoral muscle weight/index of LD geese were significantly higher than that of SW geese (P < 0.05). In transcriptional regulation, intra-breed bioinformatics analysis identified 3331 genes whose expression levels were opposite to the trend of pectoral muscle hypertrophy both in LD and SW geese, and the 3331 genes were mainly enriched into abundant KEGG pathways related to lipid metabolism, proliferation/apoptosis, and immune response. Moreover, 23 genes (including SLC2A10, TNFRSF1A, PRKAA1, SLC27A4, ITGB2, THY1, RHOA, MYL10, ACTB, PRKCB, PIK3R2, RAC2, DMD, LATS2, YAP1, WWTR1, SMAD7, CTGF, FGF1, AXIN2, GLI2, ID2, and CCND2) who were enriched in 6 crosstalk pathways named viral myocarditis, insulin resistance, sphingolipid signaling pathway, hippo signaling pathway, chemokine signaling pathway, and leukocyte transendothelial migration were identified as the key candidate genes regulating the hypertrophy of goose pectoral muscles. In inter-breed bioinformatics analysis, abundant different expression genes (DEGs) related to lipid metabolism, immune response, and proliferation/apoptosis were identified between LD and SW geese too, and compared with SW geese, the expression level of MYL10 in LD geese was lower, while the expression levels of GLI2/CTGF/SMAD7 in LD geese were higher. These results suggested that the hypertrophy of goose pectoral muscles might be achieved through more lipid deposition and less leukocyte infiltration to promote the proliferation of cells within the muscles, and the low expression of MYL10 and high expressions of GLI2/CTGF/SMAD7 might the keys to induce the pectoral muscle hypertrophy of LD geese from 6 to 30 weeks of age over that of SW geese. All data the present study obtained will provide new insights into the molecular mechanisms regulating the hypertrophy of goose pectoral muscles.
Collapse
Affiliation(s)
- Xinyue Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Yali Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Bincheng Tang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Jiwei Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Hua He
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Hehe Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Liang Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Shenqiang Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Jiwen Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.
| |
Collapse
|
44
|
Wang YT, Zheng SY, Jiang SD, Luo Y, Wu YX, Naranmandakh S, Li YS, Liu SG, Xiao WF. Irisin in degenerative musculoskeletal diseases: Functions in system and potential in therapy. Pharmacol Res 2024; 210:107480. [PMID: 39490914 DOI: 10.1016/j.phrs.2024.107480] [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: 08/16/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Degenerative musculoskeletal diseases are a class of diseases related to the gradual structural and functional deterioration of muscles, joints, and bones, including osteoarthritis (OA), osteoporosis (OP), sarcopenia (SP), and intervertebral disc degeneration (IDD). As the proportion of aging people around the world increases, degenerative musculoskeletal diseases not only have a multifaceted impact on patients, but also impose a huge burden on the medical industry in various countries. Therefore, it is crucial to find key regulatory factors and potential therapeutic targets. Recent studies have shown that irisin plays an important role in degenerative musculoskeletal diseases, suggesting that it may become a key molecule in the prevention and treatment of degenerative diseases of the musculoskeletal system. Therefore, this review provides a comprehensive description of the release and basic functions of irisin, and summarizes the role of irisin in OA, OP, SP, and IDD from a cellular and tissue perspective, providing comprehensive basis for clinical application. In addition, we summarized the many roles of irisin as a key information molecule in bone-muscle-adipose crosstalk and a regulatory molecule involved in inflammation, senescence, and cell death, and proposed the interesting possibility of irisin in degenerative musculoskeletal diseases.
Collapse
Affiliation(s)
- Yu-Tong Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Sheng-Yuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shi-de Jiang
- The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Yan Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Yu-Xiang Wu
- School of Kinesiology, Jianghan University, Wuhan, Hubei, China
| | - Shinen Naranmandakh
- Department of chemistry, School of Arts and Sciences, National University of Mongolia, Ulaanbaatar 14201, Mongolia
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Shu-Guang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Wen-Feng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
45
|
Geng W, Guo Y, Chen B, Cheng X, Li S, Challioui MK, Tian W, Li H, Zhang Y, Li Z, Jiang R, Tian Y, Kang X, Liu X. IGFBP7 promotes the proliferation and differentiation of primary myoblasts and intramuscular preadipocytes in chicken. Poult Sci 2024; 103:104258. [PMID: 39293261 PMCID: PMC11426050 DOI: 10.1016/j.psj.2024.104258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/21/2024] [Accepted: 08/20/2024] [Indexed: 09/20/2024] Open
Abstract
Though it is well known that insulin-like growth factor (IGF) binding protein 7 (IGFBP7) plays an important role in myogenesis and adipogenesis in mammals, its impact on the proliferation, differentiation, and lipid deposition in chicken primary myoblasts (CPM) and intramuscular preadipocytes remains unexplored. In the present study, we firstly examined the correlation between SNPs within the genomic sequence of the IGFBP7 gene and carcass and blood chemical traits in a F2 resource population by genetic association analysis, and found that a significant correlation between the SNP (4_49499525) located in the intron region of IGFBP7 and serum high-density lipoproteins (HDL). We then examined the expression patterns of IGFBP7 across different stages of proliferation and differentiation in CPMs and intramuscular preadipocytes via qPCR, and explored the biological functions of IGFBP7 through gain- and loss-of-function experiments and a range of techniques including qPCR, CCK-8, EdU, flow cytometry, Western blot, immunofluorescence, and Oil Red O staining to detect the proliferation, differentiation, and lipid deposition in CPMs and intramuscular preadipocytes. We ascertained that the expression levels of the IGFBP7 gene increased as cell differentiation progresses in CPMs and intramuscular preadipocytes, and that IGFBP7 promotes the proliferation and differentiation of these cells, as well as facilitates intracellular lipid deposition. Furthermore, we investigated the regulatory mechanism of IGFBP7 expression by using co-transfection strategy and dual-luciferase reporter assay, and discovered that the myogenic transcription factors (MRF), myoblast determination factor (MyoD) and myogenin (MyoG), along with the adipocyte-specific transcription factor (TF) CCAAT/enhancer-binding protein α (C/EBPα), can bind to the core transcription activation region of the IGFBP7 promoter located 500 bp upstream from the transcription start site, thereby promoting IGFBP7 transcription and expression. Taken together, our study underscores the role of IGFBP7 as a positive regulator for myogenesis and adipogenesis, while also elucidating the functional and transcriptional regulatory mechanisms of IGFBP7 in chicken skeletal muscle development and intramuscular adipogenesis.
Collapse
Affiliation(s)
- Wanzhuo Geng
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yulong Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Botong Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Xi Cheng
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Shuohan Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Mohammed Kamal Challioui
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Animal Production and Biotechnology Department, Institut Agronomique et Vétérinaire Hassan II, Rabat P.O. Box 6202, Rabat, Morocco
| | - Weihua Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China; International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou 450046, China
| | - Hong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China; International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou 450046, China
| | - Yanhua Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China; International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou 450046, China
| | - Zhuanjian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China; International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou 450046, China
| | - Ruirui Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China; International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou 450046, China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China; International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou 450046, China
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China; International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou 450046, China
| | - Xiaojun Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China; International Joint Research Laboratory for Poultry Breeding of Henan, Zhengzhou 450046, China.
| |
Collapse
|
46
|
Wang X, Li E, Li C, Zhang C, Liang Z, Xu R, Liu Y, Chen M, Li Y, Wu HD, Yuan R, Xiong Y, Chen Y, Liu X, Mo D. Fibin is a crucial mitochondrial regulatory gene in skeletal muscle development. Int J Biol Macromol 2024; 283:137568. [PMID: 39547619 DOI: 10.1016/j.ijbiomac.2024.137568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/28/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Fin bud initiation factor homolog (Fibin) is a secreted protein that is relatively conserved among species. It is closely related to fin bud development and can regulate a variety of cellular processes. In our previous high-throughput chromosome conformation capture (Hi-C) study of pig embryonic muscle development, it was found that Fibin has high expression and activity during the development of pig primary muscle fibers. Therefore, we speculated Fibin participated in myogenesis severely. Specific deletion of Fibin in mouse skeletal muscle resulted in abnormal primary muscle fiber development during the embryonic period and a substantial decrease in skeletal muscle mass in adulthood. In vitro, knocking out Fibin in C2C12 cells promoted cell proliferation; however, after myogenic induction, cells lacking Fibin had almost no ability to differentiate into myotubes. During myogenic differentiation, loss of Fibin disrupts the normal function of mitochondria and impairs oxidative phosphorylation, resulting in decrease of NADH and FADH in the electron transport chain. Transmission electron microscopy also showed that mitochondrial morphology of Fibin-deficient C2C12 was impaired. In conclusion, our research has unveiled a novel mechanism of myogenesis regulation in mitochondrial function and potential target Fibin, and improved understanding of a broad range of mitochondrial muscle diseases.
Collapse
Affiliation(s)
- Xiaoyu Wang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Enru Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Chenggan Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Chong Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Ziyun Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Rong Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Yihao Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Meilin Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Yongpeng Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Hoika David Wu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Renqiang Yuan
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Yanyun Xiong
- College of Animal Science and Technology, Guangxi Agricultural Engineering Vocational Technical College, Chongzuo 532199, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China.
| |
Collapse
|
47
|
Chandrababu A, Puthumana J. CRISPR-edited, cell-based future-proof meat and seafood to enhance global food security and nutrition. Cytotechnology 2024; 76:619-652. [PMID: 39435422 PMCID: PMC11490478 DOI: 10.1007/s10616-024-00645-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 07/15/2024] [Indexed: 10/23/2024] Open
Abstract
Food security is a major concern due to the growing population and climate change. A method for increasing food production is the use of modern biotechnology, such as cell culture, marker-assisted selection, and genetic engineering. Cellular agriculture has enabled the production of cell-cultivated meat in bioreactors that mimic the properties of conventional meat. Furthermore, 3D food printing technology has improved food production by adding new nutritional and organoleptic properties. Marker-assisted selection and genetic engineering could play an important role in producing animals and crops with desirable traits. Therefore, integrating cellular agriculture with genetic engineering technology could be a potential strategy for the production of cell-based meat and seafood with high health benefits in the future. This review highlights the production of cell-cultivated meat derived from a variety of species, including livestock, birds, fish, and marine crustaceans. It also investigates the application of genetic engineering methods, such as CRISPR/Cas (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein), in the context of cellular agriculture. Moreover, it examines aspects such as food safety, regulatory considerations, and consumer acceptance of genetically engineered cell-cultivated meat and seafood.
Collapse
Affiliation(s)
- Aswathy Chandrababu
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Cochin, Kerala 16 India
| | - Jayesh Puthumana
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Cochin, Kerala 16 India
| |
Collapse
|
48
|
Xie Z, Liu C, Sun C, Liu Y, Peng J, Meng L, Deng J, Wang Z, Yang C, Yuan Y, Xie Z. Single-Nucleus RNA Sequencing Unravels Early Mechanisms of Human Becker Muscular Dystrophy. Ann Neurol 2024; 96:1070-1085. [PMID: 39192489 DOI: 10.1002/ana.27070] [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: 03/12/2024] [Revised: 08/04/2024] [Accepted: 08/11/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVE The transcriptional heterogeneity at a single-nucleus level in human Becker muscular dystrophy (BMD) dystrophic muscle has not been explored. Here, we aimed to understand the transcriptional heterogeneity associated with myonuclei, as well as other mononucleated cell types that underly BMD pathogenesis by performing single-nucleus RNA sequencing. METHODS We profiled single-nucleus transcriptional profiles of skeletal muscle samples from 7 BMD patients and 3 normal controls. RESULTS A total of 17,216 nuclei (12,879 from BMD patients and 4,337 from controls) were classified into 13 known cell types, including 9 myogenic lineages and 4 non-myogenic lineages, and 1 unclassified nuclear type according to their cell identities. Among them, type IIx myonuclei were the first to degenerate in response to dystrophin reduction. Differential expression analysis revealed that the fibro-adipogenic progenitors (FAPs) population had the largest transcriptional changes among all cell types. Sub-clustering analysis identified a significantly compositional increase in the activated FAPs (aFAPs) subpopulation in BMD muscles. Pseudotime analysis, regulon inference, and deconvolution analysis of bulk RNA-sequencing data derived from 29 BMD patients revealed that the aFAPs subpopulation, a distinctive and previously unrecognized mononuclear subtype, was profibrogenic and expanded in BMD patients. Muscle quantitative real-time polymerase chain reaction and immunofluorescence analysis confirmed that the mRNA and protein levels of the aFAPs markers including LUM, DCN, and COL1A1 in BMD patients were significantly higher than those in controls, respectively. INTERPRETATION Our results provide insights into the transcriptional diversity of human BMD muscle at a single-nucleus resolution and new potential targets for anti-fibrosis therapies in BMD. ANN NEUROL 2024;96:1070-1085.
Collapse
Affiliation(s)
- Zhihao Xie
- Department of Neurology, Peking University First Hospital, Beijing, China
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chang Liu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Chengyue Sun
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Yilin Liu
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Jieru Peng
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lingchao Meng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Chunxia Yang
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhiying Xie
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
49
|
Zhang D, Yin L, Lin Z, Yu C, Li J, Ren P, Yang C, Qiu M, Liu Y. miR-136-5p/FZD4 axis is critical for Wnt signaling-mediated myogenesis and skeletal muscle regeneration. J Cell Physiol 2024; 239:e31046. [PMID: 37218742 DOI: 10.1002/jcp.31046] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/24/2023]
Abstract
Skeletal muscle can undergo a regenerative process in response to injury or disease to maintain muscle quality and function. Myogenesis depends on the proliferation and differentiation of myoblasts, and miRNAs can maintain the balance between them by precisely regulating many key factors in the myogenic network. Here, we found that miR-136-5p was significantly upregulated during the proliferation and differentiation of C2C12 cells. We demonstrate that miR-136-5p acts as a myogenic negative regulator during the development of mouse C2C12 myoblasts. In terms of mechanism, miR-136-5p inhibits the formation of β-catenin/LEF/TCF DNA-binding factor transcriptional regulatory complex by targeting FZD4, a gating protein in the Wnt signaling pathway, thereby enhancing downstream myogenic factors and finally promoting myoblast proliferation and differentiation. In addition, in BaCl2-induced muscle injury mouse model, miR-136-5p knockdown accelerated the regeneration of skeletal muscle after injury, and further led to the improvement of gastrocnemius muscle mass and muscle fiber diameter, while being suppressed by shFZD4 lentivirus infection. In summary, these results demonstrate the essential role of miR-136-5p/FZD4 axis in skeletal muscle regeneration. Given the conservation of miR-136-5p among species, miR-136-5p may be a new target for treating human skeletal muscle injury and improving the production of animal meat products.
Collapse
Affiliation(s)
- Donghao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lingqian Yin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhongzhen Lin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Chunlin Yu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Jingjing Li
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Peng Ren
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Chaowu Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Mohan Qiu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Yiping Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
50
|
Qaisar R. The emerging roles of necroptosis in skeletal muscle health and disease. Pflugers Arch 2024; 476:1645-1651. [PMID: 39037477 DOI: 10.1007/s00424-024-02994-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Necroptosis is a regulated form of cell death with implications in various physiological and pathological processes in multiple tissues. However, the relevant findings from post-mitotic tissues, such as skeletal muscle, are scarce. This review summarizes the potential contributions of necroptosis to skeletal muscle health and diseases. It first discusses the physiological roles of necroptosis in muscle regeneration and development. It then summarizes the contributions of necroptosis to the pathogenesis of multiple muscle diseases, including muscular dystrophies, inflammatory myopathies, cachexia, and neuromuscular disorders. Lastly, it unravels the gaps in our understanding and therapeutic challenges of inhibiting necroptosis as a potential intervention for muscle diseases. Specifically, the findings from the transgenic animal models and the use of pharmacological inhibitors of necroptosis are discussed with relevance to improving the structure and/or function of skeletal muscle in various diseases. Recent developments from experimental animal models and clinical data are presented to discuss the roles of necroptosis in skeletal muscle health and diseases.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates.
- Space Medicine Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates.
- Cardiovascular Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|