1
|
Huang CJ, Choo KB. Circular RNAs and host genes act synergistically in regulating cellular processes and functions in skeletal myogenesis. Gene 2024; 940:149189. [PMID: 39724991 DOI: 10.1016/j.gene.2024.149189] [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: 08/03/2024] [Revised: 11/14/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
Circular RNAs (circRNAs) are post-transcriptional regulators generated from backsplicing of pre-mRNAs of host genes. A major circRNA regulatory mechanism involves microRNA (miRNA) sequestering, relieving miRNA-blocked mRNAs for translation and functions. To investigate possible circRNA-host gene relationship, skeletal myogenesis is chosen as a study model for its developmental importance and for readily available muscle tissues from farm animals for studies at different myogenic stages. This review aims to provide an integrated interpretations on methodologies, regulatory mechanisms and possible host gene-circRNA synergistic functional relationships in skeletal myogenesis, focusing on myoblast differentiation and proliferation, core drivers of muscle formation in myogenesis, while other myogenic processes that play supportive roles in the structure, maintenance and function of muscle tissues are also briefly discussed. On literature review,thirty-two circRNAs derived from thirty-one host genes involved in various myogenic stages are identified; twenty-two (68.6 %) of these circRNAs regulate myogenesis by sequestering miRNAs to engage PI3K/AKT and other signaling pathways while four (12.5 %) are translated into proteins for functions. In circRNA-host gene relationship,ten (32.3 %) host genes are shown to regulate myogenesis,nine (29.0 %) are specific to skeletal muscle functions,and twelve (38.8 %) are linked to skeletal muscle disorders.Our analysis of skeletal myogenesis suggests that circRNAs and host genes act synergistically to regulate cellular functions. Such circRNA-host gene functional synergism may also be found in other major cellular processes. CircRNAs may have evolved later than miRNAs to counteract the suppressive effects of miRNAs and to augment host gene functions to further fine-tune gene regulation.
Collapse
Affiliation(s)
- Chiu-Jung Huang
- Department of Animal Science & Graduate Institute of Biotechnology, College of Environmental Planning & Bioresources (former School of Agriculture), Chinese Culture University, Taipei, Taiwan.
| | - Kong Bung Choo
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
2
|
Huang Y, Wang W, Fan X, Liu X, Liu W, Wang Z, Li Y, Yang Y, Tang Z. The miR-6240 target gene Igf2bp3 promotes myoblast fusion by enhancing myomaker mRNA stability. Cell Mol Biol Lett 2024; 29:152. [PMID: 39639214 PMCID: PMC11622686 DOI: 10.1186/s11658-024-00650-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 10/11/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Myoblast fusion plays a crucial role in myogenesis. Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) functions as an RNA N6-methyladenosine reader and exerts important roles in various biological processes. While our prior study suggested Igf2bp3 contributes to myogenesis, its molecular regulatory mechanism is largely unclear. METHODS Real-time quantitative polymerase chain reaction (RT-qPCR) and western blot were used for gene expression analysis. siRNA and CRISPRi technologies were conducted to knockdown the expression of Igf2bp3. CRISPR/Cas9 technology was performed to knockout Igf2bp3. The Igf2bp3 overexpression vector was designed using the pcDNA3.1(+) vector. Immunofluorescence detection was employed for subcellular localization and cell differentiation analysis. Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) assays were conducted for cell proliferation and fusion detection. The dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were utilized for regulatory mechanism analysis of Igf2bp3. RESULTS The overexpression of Igf2bp3 enhances myoblast fusion while knockdown of Igf2bp3 blocks the formation of myotubes. miR-6240 promotes myoblast proliferation while preventing myoblast differentiation and fusion by targeting the 3' untranslated rgion (UTR) of Igf2bp3. Notably, the impacts of miR-6240 mimics on myoblast proliferation, differentiation, and fusion can be effectively counteracted by the overexpression of Igf2bp3. Moreover, our findings elucidate a direct interaction between Igf2bp3 and the myoblast fusion factor myomaker (Mymk). Igf2bp3 binds to Mymk to enhance its mRNA stability. This interaction results in increased expression of Mymk and heightened myoblast fusion. CONCLUSIONS Our study unveils Igf2bp3 as a novel post-transcriptional regulator of myoblast fusion through the miR-6240/Mymk axis, significantly contributing to our understanding of skeletal muscle development.
Collapse
Affiliation(s)
- Yuxin Huang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention; College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Wei Wang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Xinhao Fan
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Xiaoqin Liu
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Weiwei Liu
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention; College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Zishuai Wang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Yixing Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention; College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi, China
| | - Yalan Yang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China.
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China.
| | - Zhonglin Tang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Foshan, 528226, China.
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China.
| |
Collapse
|
3
|
Chinvattanachot G, Rivas D, Duque G. Mechanisms of muscle cells alterations and regeneration decline during aging. Ageing Res Rev 2024; 102:102589. [PMID: 39566742 DOI: 10.1016/j.arr.2024.102589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/27/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Skeletal muscles are essential for locomotion and body metabolism regulation. As muscles age, they lose strength, elasticity, and metabolic capability, leading to ineffective motion and metabolic derangement. Both cellular and extracellular alterations significantly influence muscle aging. Satellite cells (SCs), the primary muscle stem cells responsible for muscle regeneration, become exhausted, resulting in diminished population and functionality during aging. This decline in SC function impairs intercellular interactions as well as extracellular matrix production, further hindering muscle regeneration. Other muscle-resident cells, such as fibro-adipogenic progenitors (FAPs), pericytes, and immune cells, also deteriorate with age, reducing local growth factor activities and responsiveness to stress or injury. Systemic signaling, including hormonal changes, contributes to muscle cellular catabolism and disrupts muscle homeostasis. Collectively, these cellular and environmental components interact, disrupting muscle homeostasis and regeneration in advancing age. Understanding these complex interactions offers insights into potential regenerative strategies to mitigate age-related muscle degeneration.
Collapse
Affiliation(s)
- Guntarat Chinvattanachot
- Department of Orthopedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| | - Daniel Rivas
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Gustavo Duque
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
4
|
Li D, Qu Y, Wang B, Zhang H, Qin L. Spatio-temporal expression of Sox2 + progenitor cells regulates the regeneration of rat submandibular gland. Arch Oral Biol 2024; 168:106080. [PMID: 39217919 DOI: 10.1016/j.archoralbio.2024.106080] [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: 02/28/2024] [Revised: 04/12/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Sox2 plays crucial roles in tissues homeostasis and regeneration. However, there are lack of a comprehensive examination of Sox2 expression and its functional role in submandibular gland regeneration. Therefore, we aimed to elucidate the impact of Sox2 on submandibular gland regeneration. MATERIALS AND METHODS A Sprague-Dawley rat submandibular gland duct ligation/de-ligation regeneration model was conducted in this study. Sox2-shRNA vectors were retro-ductally administered into the submandibular gland to establish a stable Sox2 knockdown model. Conventional histopathological and molecular biological methods were used to investigate phenotypic changes. RESULTS The submandibular gland normalized completely 28 days after ligature removal (following 7 days of duct ligation). AQP5 expression gradually increased after ligation removal until returning to normal levels. In submandibular gland regeneration, Sox2 re-expressed and co-expressed with AQP5+ acinar cells, and Sox2 expression peaked on day 14, recovered to normal on day 28, reproducing the developmental pattern. Sox2 knockdown hindered gland regeneration and induced irreversible fibrosis. The AQP5 expression was significantly lower than the contemporaneous solely ligated group, while the blue collagen deposition and the Vimentin expression increased prominently. The expression of CD68, IL-1β, TNF-α and IL-17A increased significantly, and epithelial cells in the Sox2 knockdown group expressed higher levels of IL-17A. CONCLUSIONS These findings highlight Sox2 as a crucial regulator of the acinar cell lineage. Sox2+ progenitor cells are pivotal for acinar cell maintenance, which is indispensable for submandibular gland regeneration. Collectively, our findings may help develop targeted interventions for enhancing tissue repair and preventing irreversible fibrosis in salivary gland disorders.
Collapse
Affiliation(s)
- Dan Li
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, China
| | - Yi Qu
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, China
| | - Bin Wang
- Department of Head and Neck Oncology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Workers' New Village No.3, Taiyuan, 030013, China
| | - Haoyang Zhang
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, China
| | - Lizheng Qin
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, China.
| |
Collapse
|
5
|
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
|
6
|
Wei Y, Huang Y, Wen C, Wei K, Peng L, Wei X. Theabrownin/whey protein isolate complex coacervate strengthens C 2C 12 cell proliferation via modulation of energy metabolism and mitochondrial apoptosis. Int J Biol Macromol 2024; 283:137686. [PMID: 39561831 DOI: 10.1016/j.ijbiomac.2024.137686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/30/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
Theabrownin (TB)-whey protein isolate (WPI) complex coacervates (TW) were firstly prepared to investigate the regulatory effects on skeletal muscle. The binding of TB to WPI reached saturation with the strongest electrostatic interaction at the ratio of 10:1. The formation of TW was driven by electrostatic interactions with the aid of hydrogen bonding and hydrophobic interactions, and the digestion behavior of TW was investigated based on in vitro gastrointestinal and CaCO2 cell models. The regulatory effect of TW on muscle cells was investigated by C2C12 cell assay. Cell cycle analysis showed that TW promoted the transition of skeletal muscle cells from proliferative state to differentiated state. Immunofluorescence and gene expression revealed that TW positively regulated myogenic regulatory factors, contributing to myofiber formation. Moreover, TW activated the intracellular TCA cycling and oxidative phosphorylation, providing energy for skeletal muscle regeneration and repair. Mechanistically, TW inhibited the release of cytochrome C from mitochondria to cytoplasm through the Bcl-2/Cytochrome C/Cleaved-Caspase-3 pathway, exhibiting a protective effect on skeletal muscle cells. In the future, the molecular mechanism of TW enhancing skeletal muscle function should be validated through aging animal models and clinical trials and expand its therapeutic application for muscle health in functional food and dietary supplements.
Collapse
Affiliation(s)
- Yang Wei
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Yi Huang
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Caican Wen
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Kang Wei
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Lanlan Peng
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Xinlin Wei
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China.
| |
Collapse
|
7
|
Wang L, Meng Q, Su CH. From Food Supplements to Functional Foods: Emerging Perspectives on Post-Exercise Recovery Nutrition. Nutrients 2024; 16:4081. [PMID: 39683475 DOI: 10.3390/nu16234081] [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: 10/21/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Effective post-exercise recovery is vital for optimizing athletic performance, focusing on muscle repair, glycogen replenishment, rehydration, and inflammation management. This review explores the evolving trend from traditional supplements, such as protein, carbohydrates, creatine, and branched-chain amino acids (BCAAs), toward functional foods rich in bioactive compounds. Evidence highlights the benefits of functional foods like tart cherry juice (anthocyanins), turmeric-seasoned foods, and sources of omega-3 fatty acids, including fish, flaxseeds, chia seeds, and walnuts, for mitigating oxidative stress and inflammation. Additionally, probiotics and prebiotics support gut health and immune function, which are integral to effective recovery. Personalized nutrition, informed by genetic and metabolic profiling, is examined as a promising approach to tailor recovery strategies. A systematic search across PubMed, Web of Science, and Google Scholar (2000-2024) identified studies with high empirical rigor and relevance to recovery outcomes. Findings underscore the need for further research into nutrient interactions, dosage optimization, and long-term effects on athletic performance. Integrating functional foods with personalized nutrition presents a comprehensive framework for enhanced recovery, greater resilience to physical stress, and sustained performance in athletes.
Collapse
Affiliation(s)
- Lifeng Wang
- Public Sports Department, Xuhai College, China University of Mining and Technology, Xuzhou 221008, China
| | - Qing Meng
- School of Physical Education, Huaqiao University, Xiamen 361021, China
- Sport and Health Research Center, Huaqiao University, Xiamen 361021, China
| | - Chun-Hsien Su
- Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, Taipei 111396, Taiwan
| |
Collapse
|
8
|
Prylutskyy Y, Nozdrenko D, Motuziuk O, Prylutska S, Bogutska K, Abramchuk O, Morenko A, Franskevych D, Scharff P, Ritter U. C 60 Fullerene Reduces the Development of Post-Traumatic Dysfunction in Rat Soleus Muscle. Int J Mol Sci 2024; 25:12206. [PMID: 39596273 PMCID: PMC11594679 DOI: 10.3390/ijms252212206] [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: 10/24/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Traumatic skeletal muscle injury is a complex pathology caused by high-energy trauma to muscle tissue. Previously, a positive effect was established when C60 fullerene was administered against the background of muscle ischemia, mechanical muscle injury, and other muscle dysfunctions, which probably protected the muscle tissue from damage caused by oxidative stress. Using tensiometry and biochemical analysis, the biomechanical parameters of skeletal muscle contraction and biochemical indices of the blood of rats 15 days after traumatic injury of the soleus muscle caused by myocyte destruction by compression were studied. The intraperitoneal administration of C60 fullerene aqueous solution (C60FAS) in a daily dose of 1 mg/kg improved its contractile function by 28-40 ± 2% and the values of the investigated biochemical indices of the animals' blood by 15-34 ± 2% relative to the trauma group. The obtained results indicate the potential ability of C60 fullerenes, as powerful antioxidants, to reduce the development of post-traumatic dysfunction of the soleus muscle.
Collapse
Affiliation(s)
- Yuriy Prylutskyy
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine; (Y.P.); (D.N.); (O.M.); (K.B.); (D.F.)
| | - Dmytro Nozdrenko
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine; (Y.P.); (D.N.); (O.M.); (K.B.); (D.F.)
| | - Olexandr Motuziuk
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine; (Y.P.); (D.N.); (O.M.); (K.B.); (D.F.)
- Faculty of Biology and Forestry, Lesya Ukrainka Volyn National University, 43025 Lutsk, Ukraine; (O.A.); (A.M.)
| | - Svitlana Prylutska
- Faculty of Plant Protection, Biotechnology and Ecology, National University of Life and Environmental Science of Ukraine, 03041 Kyiv, Ukraine;
| | - Kateryna Bogutska
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine; (Y.P.); (D.N.); (O.M.); (K.B.); (D.F.)
| | - Olga Abramchuk
- Faculty of Biology and Forestry, Lesya Ukrainka Volyn National University, 43025 Lutsk, Ukraine; (O.A.); (A.M.)
| | - Alevtyna Morenko
- Faculty of Biology and Forestry, Lesya Ukrainka Volyn National University, 43025 Lutsk, Ukraine; (O.A.); (A.M.)
| | - Daria Franskevych
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine; (Y.P.); (D.N.); (O.M.); (K.B.); (D.F.)
| | - Peter Scharff
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, 98693 Ilmenau, Germany;
| | - Uwe Ritter
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, 98693 Ilmenau, Germany;
| |
Collapse
|
9
|
Mihaly E, Chellu N, Iyer SR, Su EY, Altamirano DE, Dias ST, Grayson WL. Neuromuscular Regeneration of Volumetric Muscle Loss Injury in Response to Agrin-Functionalized Tissue Engineered Muscle Grafts and Rehabilitative Exercise. Adv Healthc Mater 2024:e2403028. [PMID: 39523723 DOI: 10.1002/adhm.202403028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Neuromuscular deficits compound the loss of contractile tissue in volumetric muscle loss (VML). Two avenues for promoting recovery are neuromuscular junction (NMJ)-promoting substrates (e.g., agrin) and endurance exercise. Although mechanical stimulation enhances agrin-induced NMJ formation, the two modalities have yet to be evaluated combinatorially. It is hypothesized that the implantation of human myogenic progenitor-seeded tissue-engineered muscle grafts (hTEMGs) in combination with agrin treatment and/or exercise will enhance neuromuscular recovery after VML. The hTEMGs alone transplant into VML defects promote significant regeneration with minimal scarring. A sex-appropriate, low-intensity continuous running exercise paradigm increases acetylcholine receptor (AChR) cluster density in male mice twofold relative to hTEMG alone after 7 weeks of treadmill training (p < 0.05). To further promote neuromuscular recovery, agrin is incorporated into the scaffolds via covalent tethering. In vitro, agrin increases the proliferation of hMPs, and trends toward greater myogenic maturity and AChR clustering. Upon transplantation, both hTEMGs + agrin and hTEMGs + exercise induce near 100% recovery of muscle mass and increase twitch and tetanic force output (p > 0.05). However, agrin treatment in combination with exercise produces no additional benefit. These data highlight the unprecedented regenerative potential of using hTEMGs together with either agrin or exercise supplementation to treat VML injuries.
Collapse
Affiliation(s)
- Eszter Mihaly
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD, 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Neha Chellu
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD, 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Shama R Iyer
- School of Science, Mathematics & Engineering, Marymount University, Arlington, VA, 22207, USA
| | - Eileen Y Su
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD, 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Dallas E Altamirano
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD, 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Shaquielle T Dias
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD, 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Warren L Grayson
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD, 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Materials Science & Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Chemical & Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD, 21218, USA
| |
Collapse
|
10
|
Xue X, Chen Y, Yu Z, Feng Y, Zhang L, Han C, Yin X, Lu B, Shu H. Effects of Diet Supplemented With Hydrolyzable Tannin on the Growth Performance, Antioxidant Capacity, and Muscle Nutritional Quality of Juvenile Mastacembelus armatus. AQUACULTURE NUTRITION 2024; 2024:8266189. [PMID: 39555515 PMCID: PMC11554411 DOI: 10.1155/2024/8266189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024]
Abstract
In this study, four groups of diet were prepared, with eel commercial diet without hydrolyzable tannin (HT) as the control group (H0), and the other three groups were fed with diet containing 0.05% (H1), 0.1% (H2), and 0.2% (H3) doses of HT to juvenile Mastacembelus armatus with an initial body weight of (0.40 ± 0.005) g. Juvenile fish in all groups were fed continuously for 60 days. Growth indices, hepatopancreatic antioxidant enzymes, biochemical indices (including total superoxide dismutase [T-SOD], catalase [CAT], malondialdehyde [MDA], total antioxidant capacity [T-AOC], alanine aminotransferase [ALT], aspartate aminotransferase [AST], alkaline phosphatase [AKP], and triglyceride [TG]), the content of muscle amino acids and fatty acids, stomach and intestine enzyme activities (pepsin, amylase, lipase), and genes expressions were evaluated. The results showed that 0.1% HT significantly improved the growth performance, hepatopancreatic antioxidant capacity, as well as muscle quality and lipase activity of juvenile M. armatus. In summary, the optimal addition level of HT in the diet of juvenile M. armatus is 0.1%, which helps to improve aquaculture efficiency and improve the muscle quality of M. armatus. However, the long-term effects of feeding HT on M. armatus and its physiological reaction mechanism need to be further explored.
Collapse
Affiliation(s)
- Xiaowen Xue
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Yiman Chen
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Zhide Yu
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Yuwei Feng
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Linan Zhang
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Chong Han
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Xiaoli Yin
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Baoyue Lu
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Hu Shu
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| |
Collapse
|
11
|
Zhe Y, Wu Z, Yasenjian S, Zhong J, Jiang H, Zhang M, Chai Z, Xin J. Effect of NR1D1 on the proliferation and differentiation of yak skeletal muscle satellite cells. Front Vet Sci 2024; 11:1428117. [PMID: 39559540 PMCID: PMC11571325 DOI: 10.3389/fvets.2024.1428117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024] Open
Abstract
The severe conditions at high altitudes, where yaks inhabit, contribute to delayed muscular growth and compromised tenderness of their muscle tissue. Myosatellite cells are responsible for the growth and regeneration of skeletal muscle after birth and have the potential to proliferate and differentiate, its development is closely related to meat quality, and the nuclear receptor gene NR1D1 is involved in muscle formation and skeletal muscle regulation. Therefore, in order to understand the effect of NR1D1 on muscle satellite cells, we identified the mRNA expression levels of marker genes specifically expressed in muscle satellite cells at different stages to determine the type of cells isolated. Eventually, we successfully constructed a primary cell line of yak muscle satellite cells. Then we constructed NR1D1 overexpression vector and interference RNA, and introduced them into isolated yak skeletal muscle satellite cells. We performed qPCR, CCK8, and fluorescence-specific to detect the expression of genes or abundance of proteins as markers of cell proliferation and differentiation. Compared with those in the control group, the expression levels of proliferation marker genes KI-67, CYCLIND1, and CYCLINA were significantly inhibited after NR1D1 overexpression, which was also supported by the CCK-8 test, whereas differentiation marker genes MYOD, MYOG, and MYF5 were significantly inhibited. Fluorescence-specific staining showed that KI-67 protein abundance and the number of microfilaments both decreased, while the opposite trend was observed after NR1D1 interference. In conclusion, we confirmed that NR1D1 inhibited the proliferation and differentiation of yak skeletal muscle satellite cells, which provides a theoretical basis for further research on the effect of NR1D1 on improving meat quality traits and meat production performance of yaks.
Collapse
Affiliation(s)
- Yuqi Zhe
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Zhijuan Wu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Sibinuer Yasenjian
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Hui Jiang
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Institute of Animal Science and Veterinary Research, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Ming Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Zhixin Chai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- Sichuan Qinghai Tibet Plateau Herbivore Livestock Engineering Technology Center, Chengdu, China
| | - Jinwei Xin
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Institute of Animal Science and Veterinary Research, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| |
Collapse
|
12
|
Santocildes G, Viscor G, Pagès T, Torrella JR. Simulated altitude is medicine: intermittent exposure to hypobaric hypoxia and cold accelerates injured skeletal muscle recovery. J Physiol 2024; 602:5855-5878. [PMID: 38153352 DOI: 10.1113/jp285398] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023] Open
Abstract
Muscle injuries are the leading cause of sports casualties. Because of its high plasticity, skeletal muscle can respond to different stimuli to maintain and improve functionality. Intermittent hypobaric hypoxia (IHH) improves muscle oxygen delivery and utilization. Hypobaria coexists with cold in the biosphere, opening the possibility to consider the combined use of both environmental factors to achieve beneficial physiological adjustments. We studied the effects of IHH and cold exposure, separately and simultaneously, on muscle regeneration. Adult male rats were surgically injured in one gastrocnemius and randomly assigned to the following groups: (1) CTRL: passive recovery; (2) COLD: intermittently exposed to cold (4°C); (3) HYPO: submitted to IHH (4500 m); (4) COHY: exposed to intermittent simultaneous cold and hypoxia. Animals were subjected to these interventions for 4 h/day for 9 or 21 days. COLD and COHY rats showed faster muscle regeneration than CTRL, evidenced after 9 days at histological (dMHC-positive and centrally nucleated fibre reduction) and functional levels after 21 days. HYPO rats showed a full recovery from injury (at histological and functional levels) after 9 days, while COLD and COHY needed more time to induce a total functional recovery. IHH can be postulated as an anti-fibrotic treatment since it reduces collagen I deposition. The increase in the pSer473Akt/total Akt ratio observed after 9 days in COLD, HYPO and COHY, together with the increase in the pThr172AMPKα/total AMPKα ratio observed in the gastrocnemius of HYPO, provides clues to the molecular mechanisms involved in the improved muscle regeneration. KEY POINTS: Only intermittent hypobaric exposure accelerated muscle recovery as early as 9 days following injury at histological and functional levels. Injured muscles from animals treated with intermittent (4 h/day) cold, hypobaric hypoxia or a simultaneous combination of both stimuli regenerated histological structure and recovered muscle function 21 days after injury. The combination of cold and hypoxia showed a blunting effect as compared to hypoxia alone in the time course of the muscle recovery. The increased expression of the phosphorylated forms of Akt observed in all experimental groups could participate in the molecular cascade of events leading to a faster regeneration. The elevated levels of phosphorylated AMPKα in the HYPO group could play a key role in the modulation of the inflammatory response during the first steps of the muscle regeneration process.
Collapse
Affiliation(s)
- Garoa Santocildes
- Secció de Fisiologia, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Ginés Viscor
- Secció de Fisiologia, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Teresa Pagès
- Secció de Fisiologia, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Joan Ramon Torrella
- Secció de Fisiologia, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
13
|
Davenport ML, Fong A, Albury KN, Henley-Beasley CS, Barton ER, Maden M, Swanson MS. Spiny mice are primed but fail to regenerate volumetric skeletal muscle loss injuries. Skelet Muscle 2024; 14:26. [PMID: 39468576 PMCID: PMC11520498 DOI: 10.1186/s13395-024-00358-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND In recent years, the African spiny mouse Acomys cahirinus has been shown to regenerate a remarkable array of severe internal and external injuries in the absence of a fibrotic response, including the ability to regenerate full-thickness skin excisions, ear punches, severe kidney injuries, and complete transection of the spinal cord. While skeletal muscle is highly regenerative in adult mammals, Acomys displays superior muscle regeneration properties compared with standard laboratory mice following several injuries, including serial cardiotoxin injections of skeletal muscle and volumetric muscle loss (VML) of the panniculus carnosus muscle following full-thickness excision injuries. VML is an extreme muscle injury defined as the irrecoverable ablation of muscle mass, most commonly resulting from combat injuries or surgical debridement. Barriers to the treatment of VML injury include early and prolonged inflammatory responses that promote fibrotic repair and the loss of structural and mechanical cues that promote muscle regeneration. While the regeneration of the panniculus carnosus in Acomys is impressive, its direct relevance to the study of VML in patients is less clear as this muscle has largely been lost in humans, and, while striated, is not a true skeletal muscle. We therefore sought to test the ability of Acomys to regenerate a skeletal muscle more commonly used in VML injury models. METHODS We performed two different VML injuries of the Acomys tibialis anterior muscle and compared the regenerative response to a standard laboratory mouse strain, Mus C57BL6/J. RESULTS Neither Acomys nor Mus recovered lost muscle mass or myofiber number within three months following VML injury, and Acomys also failed to recover force production better than Mus. In contrast, Acomys continued to express eMHC within the injured area even three months following injury, whereas Mus ceased expressing eMHC less than one-month post-injury, suggesting that Acomys muscle was primed, but failed, to regenerate. CONCLUSIONS While the panniculus carnosus muscle in Acomys regenerates following VML injury in the context of full-thickness skin excision, this regenerative ability does not translate to regenerative repair of a skeletal muscle.
Collapse
Affiliation(s)
- Mackenzie L Davenport
- Department of Molecular Genetics and Microbiology, University of Florida, College of Medicine, Gainesville, FL, 32610, USA.
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA.
- UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA.
- Myology Institute, University of Florida, Gainesville, FL, USA.
| | - Amaya Fong
- Department of Molecular Genetics and Microbiology, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA
- UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Kaela N Albury
- Department of Molecular Genetics and Microbiology, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA
- UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA
| | - C Spencer Henley-Beasley
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, FL, USA
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, FL, USA
| | - Malcolm Maden
- UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA
- Department of Biology, University of Florida, Gainesville, FL, 32611, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, University of Florida, College of Medicine, Gainesville, FL, 32610, USA.
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA.
- UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA.
- Myology Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
14
|
Tripathi G, Dourson A, Wayland J, Khanna S, Hoffmann M, Govindarajan T, Morales FM, Queme L, Millay D, Jankowski MP. Synaptic-like coupling of macrophages to myofibers regulates muscle repair. RESEARCH SQUARE 2024:rs.3.rs-5290399. [PMID: 39574892 PMCID: PMC11581056 DOI: 10.21203/rs.3.rs-5290399/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Peripheral injury responses essential for muscle repair and nociception require complex interactions of target tissues, immune cells and primary sensory neurons. Nociceptors and myofibers both react robustly to signals generated from circulating immune cells, which promote repair, growth, and regeneration of muscle while simultaneously modulating peripheral sensitization. Here, we found that macrophages form a synaptic-like contact with myofibers to hasten repair after acute incision injury and to facilitate regeneration after major muscle damage. Transient chemogenetic activation of macrophages enhanced calcium dependent membrane repair, induced muscle calcium waves in vivo , elicited low level electrical activity in the muscles and enhanced myonuclear accretion. Under severe injury, macrophage activation could also modulate pain-like behaviors. This study identifies a novel mechanism by which synaptic-like functions of macrophages impacts muscle repair after tissue damage.
Collapse
|
15
|
Xue S, Benvie AM, Blum JE, Kolba NJ, Cosgrove BD, Thalacker-Mercer A, Berry DC. Suppressing PDGFRβ Signaling Enhances Myocyte Fusion to Promote Skeletal Muscle Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618247. [PMID: 39464006 PMCID: PMC11507758 DOI: 10.1101/2024.10.15.618247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Muscle cell fusion is critical for forming and maintaining multinucleated myotubes during skeletal muscle development and regeneration. However, the molecular mechanisms directing cell-cell fusion are not fully understood. Here, we identify platelet-derived growth factor receptor beta (PDGFRβ) signaling as a key modulator of myocyte fusion in adult muscle cells. Our findings demonstrate that genetic deletion of Pdgfrβ enhances muscle regeneration and increases myofiber size, whereas PDGFRβ activation impairs muscle repair. Inhibition of PDGFRβ activity promotes myonuclear accretion in both mouse and human myotubes, whereas PDGFRβ activation stalls myotube development by preventing cell spreading to limit fusion potential. Transcriptomics analysis show that PDGFRβ signaling cooperates with TGFβ signaling to direct myocyte size and fusion. Mechanistically, PDGFRβ signaling requires STAT1 activation, and blocking STAT1 phosphorylation enhances myofiber repair and size during regeneration. Collectively, PDGFRβ signaling acts as a regenerative checkpoint and represents a potential clinical target to rapidly boost skeletal muscle repair.
Collapse
Affiliation(s)
- Siwen Xue
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | - Abigail M Benvie
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | - Jamie E Blum
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Current address: Department of Chemical Engineering; Stanford University; Stanford, CA
| | - Nikolai J Kolba
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | | | - Anna Thalacker-Mercer
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Department of Cell, Development and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel C Berry
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Corresponding author
| |
Collapse
|
16
|
Lu F, Zhang S, Dong S, Wang M, Pang K, Zhao Y, Huang J, Kang J, Liu N, Zhang X, Zhao D, Lu F, Zhang W. Exogenous hydrogen sulfide enhances myogenic differentiation of C2C12 myoblasts under high palmitate stress. Heliyon 2024; 10:e38661. [PMID: 39416846 PMCID: PMC11481675 DOI: 10.1016/j.heliyon.2024.e38661] [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: 06/26/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Skeletal muscle atrophy was one of main complications of type 2 diabetes mellitus. Hydrogen sulfide (H2S) is involved in various physiological functions, such as anti-hypertension and anti-oxidant. Skeletal muscle atrophy caused by type 2 diabetes could lead to the regeneration of muscle fibers. Wnt signaling pathway plays a crucial important role in this process. H2S maybe regulate the Wnt signaling pathway to alleviate skeletal muscle atrophy, however, this role has not been clarified. The aim of this study is to investigate the potential regulatory role of H2S in the Wnt signaling pathway. C2C12 myoblasts treated with 500 μmol palmitate as an in vitro model. Western blot was used to detect the levels of CSE, PKM1, β-catenin, MuRF1, MYOG, MYF6 and MYOD1. In addition, MuRF1 was mutated at Cys44 and MuRF1 S-sulfhydration was detected by biotin switch assay. The interaction between PKM1 and MuRF1 was assessed via Co-immunoprecipitation. Differentiation of C2C12 myoblasts was evaluated using LAMININ staining. These data showed the levels of CSE, β-catenin, PKM1, MYOG, MYF6 and MYOD1 were decreased in pal group, compared with control and pal + NaHS groups. MuRF1 Cys44 mutants increased the protein levels of β-catenin, MYOG, MYF6 and MYOD1 in pal group. Our results suggest that H2S regulates the S-sulfhydration levels of MuRF1 at Cys44, influencing the ubiquitination levels of PKM1 and ultimately promoting myoblast differentiation.
Collapse
Affiliation(s)
- Fangping Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- Department of Pathophysiology, Mudanjiang Medical University, Mudanjiang, China
| | - Shiwu Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shiyun Dong
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Mengyi Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Kemiao Pang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yajun Zhao
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Jiayi Huang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Jiaxin Kang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Ning Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Xueya Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Dechao Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Tanner GI, Schiltz L, Narra N, Figueiredo ML, Qazi TH. Granular Hydrogels Improve Myogenic Invasion and Repair after Volumetric Muscle Loss. Adv Healthc Mater 2024; 13:e2303576. [PMID: 38329892 DOI: 10.1002/adhm.202303576] [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: 10/17/2023] [Revised: 12/26/2023] [Indexed: 02/10/2024]
Abstract
Skeletal muscle injuries including volumetric muscle loss (VML) lead to excessive tissue scarring and permanent functional disability. Despite its high prevalence, there is currently no effective treatment for VML. Bioengineering interventions such as biomaterials that fill the VML defect to support cell and tissue growth are a promising therapeutic strategy. However, traditional biomaterials developed for this purpose lack the pore features needed to support cell infiltration. The present study investigates for the first time, the impact of granular hydrogels on muscle repair - hypothesizing that their flowability will permit conformable filling of the defect site and their inherent porosity will support the invasion of native myogenic cells, leading to effective muscle repair. Small and large microparticle fragments are prepared from photocurable hyaluronic acid polymer via extrusion fragmentation and facile size sorting. In assembled granular hydrogels, particle size and degree of packing significantly influence pore features, rheological behavior, and injectability. Using a mouse model of VML, it is demonstrated that, in contrast to bulk hydrogels, granular hydrogels support early-stage (satellite cell invasion) and late-stage (myofiber regeneration) muscle repair processes. Together, these results highlight the promising potential of injectable and porous granular hydrogels in supporting endogenous repair after severe muscle injury.
Collapse
Affiliation(s)
- Gabrielle I Tanner
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Leia Schiltz
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Niharika Narra
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Marxa L Figueiredo
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Taimoor H Qazi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
18
|
Zheng H, Yang Z, Zhou L, Zhang B, Cheng R, Zhang Q. Bioactive Nb 2C MXene-Functionalized Hydrogel with Microenvironment Remodeling and Enhanced Neurogenesis to Promote Skeletal Muscle Regeneration and Functional Restoration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310483. [PMID: 39254284 DOI: 10.1002/smll.202310483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/18/2024] [Indexed: 09/11/2024]
Abstract
The complete structure-functional repair of volumetric muscle loss (VML) remains a giant challenge and biomedical hydrogels to remodel microenvironment and enhance neurogenesis have appeared to be a promising direction. However, the current hydrogels for VML repair hardly achieve these two goals simultaneously due to their insufficient functionality and the challenge in high-cost of bioactive factors. In this study, a facile strategy using Nb2C MXene-functionalized hydrogel (OPTN) as a bioactive scaffold is proposed to promote VML repair with skeletal muscle regeneration and functional restoration. In vitro experiments show that OPTN scaffold can effectively scavenge reactive oxygen species (ROS), guide macrophages polarization toward M2 phenotype, and resist bacterial infection, providing a favorable microenvironment for myoblasts proliferation as well as the endothelial cells proliferation, migration, and tube formation. More importantly, OPTN scaffold with electroactive feature remarkably boosts myoblasts differentiation and mesenchymal stem cells neural differentiation. Animal experiments further confirm that OPTN scaffold can achieve a prominent structure-functional VML repair by attenuating ROS levels, alleviating inflammation, reducing fibrosis, and facilitating angiogenesis, newborn myotube formation, and neurogenesis. Collectively, this study provides a highly promising and effective strategy for the structure-functional VML repair through designing bioactive multifunctional hydrogel with microenvironment remodeling and enhanced neurogenesis.
Collapse
Affiliation(s)
- Hua Zheng
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710129, P. R. China
- Xi'an Key Laboratory of Functional Organic Porous Materials, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Zuoting Yang
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710129, P. R. China
- Xi'an Key Laboratory of Functional Organic Porous Materials, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Li Zhou
- Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Baoliang Zhang
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710129, P. R. China
- Xi'an Key Laboratory of Functional Organic Porous Materials, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Ruidong Cheng
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710129, P. R. China
- Xi'an Key Laboratory of Functional Organic Porous Materials, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Qiuyu Zhang
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710129, P. R. China
- Xi'an Key Laboratory of Functional Organic Porous Materials, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| |
Collapse
|
19
|
Ma S, Liu J, Zhao Y, Wang Y, Zhao R. In ovo betaine injection improves breast muscle growth in newly hatched goslings through FXR/IGF-2 pathway. Poult Sci 2024; 103:104075. [PMID: 39094501 PMCID: PMC11345595 DOI: 10.1016/j.psj.2024.104075] [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: 05/13/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024] Open
Abstract
Betaine has been shown to enhance growth performance and increase breast muscle yield in ducks and broilers through various mechanisms, including the modification of DNA methylation. However, the impact of in ovo betaine injection on muscle growth in newly hatched goslings remains unclear. In this study, fifty eggs were injected with saline or betaine at 7.5 mg/egg prior to incubation, and the subsequent effects on breast muscle growth in the newly hatched goslings were investigated. Betaine significantly increased (P < 0.05) the hatch weight, breast muscle weight, and breast muscle index, accompanied by an augmentation in muscle bundle cross-sectional area. Concurrently, betaine significantly upregulated (P < 0.05) the expression levels of myogenic regulatory factors, including myogenin (MyoG) and paired box 7 (Pax7) both mRNA and protein, while downregulating (P < 0.05) the mRNA and protein levels of myostatin (MSTN). Histological analysis revealed a higher abundance of proliferating cell nuclear antigen (PCNA) and Pax7 immune-positive cells in the breast muscle of the betaine group, consistent with elevated PCNA and Pax7 mRNA and protein levels. Additionally, significantly increased (P < 0.05) contents of insulin-like growth factor 1 (IGF-1) and insulin-like growth factor 2 (IGF-2) were observed in the breast muscle of the betaine group, so was mRNA expression of IGF-1, IGF-2, and insulin-like growth factor 1 receptor (IGF-1R). Betaine also significantly in8creased (P < 0.05) global DNA methylation of the breast muscle, accompanied by enhanced mRNA and protein levels of methionine cycle and DNA methylation-related enzymes, Interestingly, the promoter regions of IGF-1, IGF-2, and IGF-1R genes were significantly hypomethylated (P < 0.05). Moreover, in ovo betaine injection significantly upregulated (P < 0.05) the protein level of farnesoid X receptor (FXR) in breast muscle and FXR binding to the promoter of IGF-2 gene. These findings suggest that in ovo betaine injection promotes breast muscle growth during embryonic development in goslings through the FXR-mediated IGF-2 pathway, ultimately improving hatch weight and breast muscle weight.
Collapse
Affiliation(s)
- Shuai Ma
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Jie Liu
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Yulan Zhao
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Yan Wang
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China; National Key Laboratory of Meat Quality Control and Cultured Meat Development, Nanjing Agricultural University, Nanjing 210095, P. R. China.
| |
Collapse
|
20
|
Huang N, Zou K, Zhong Y, Luo Y, Wang M, Xiao L. Hotspots and trends in satellite cell research in muscle regeneration: A bibliometric visualization and analysis from 2010 to 2023. Heliyon 2024; 10:e37529. [PMID: 39309858 PMCID: PMC11415684 DOI: 10.1016/j.heliyon.2024.e37529] [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: 12/10/2023] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Background The incidence of muscle atrophy or sports injuries is increasing with time and population aging, thereby attracting considerable attention to muscle generation research. Muscle satellite cells, which play an important role in this process, lack comprehensive literature regarding their use for muscle regeneration. Hence, this study aimed to analyze the hotspots and trends in satellite cell research from 2010 to 2023, providing a reference for muscle regeneration research. Methods Studies on satellite cells' role in muscle regeneration from 2010 to 2023 were retrieved from the Web of Science Core Collection. Using CiteSpace and VOSviewer, we analyzed annual publications, authors and co-citing authors, countries and institutions, journals and co-citing journals, co-citing references, and keywords. Results From 2010 to 2023, 1468 papers were retrieved, indicating an overall increasing trend in the number of annual publications related to satellite cells in muscle regeneration. The United States had the highest number of publications, while the Institut National de la Santé et de la Recherche Médicale was the institution with the most publications. Among journals, " PloS One" had the highest number of published papers, and "Cell" emerged as the most co-cited journal. A total of 7425 authors were involved, with Michael A. Rudnicki being the author with the highest number of publications and the most co-cited author. The most cited reference was "Satellite cells and the muscle stem cell niche." Among keywords, "satellite cells" was the most common, with "heterogeneity" having the highest centrality. Frontier themes included "Duchenne muscular dystrophy," "skeletal muscle," "in-vivo," "muscle regeneration," "mice," "muscle atrophy," "muscle fibers," "inflammation," " mesenchymal stem cells," and "satellite cell." Conclusion This study presents the current status and trends in satellite cell research on muscle regeneration from 2010 to 2023 using bibliometric analyses, providing valuable insights into numerous future research directions.
Collapse
Affiliation(s)
- Nan Huang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Intelligent Rehabilitation Technology Innovation Center, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Kang Zou
- Department of Critical Care Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Yanbiao Zhong
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Yun Luo
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Maoyuan Wang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Li Xiao
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| |
Collapse
|
21
|
Zhao Z, Xiao M, Xu X, Song M, Dai D, Zhan S, Cao J, Guo J, Zhong T, Wang L, Li L, Zhang H. ADAR1 Promotes Myogenic Proliferation and Differentiation of Goat Skeletal Muscle Satellite Cells. Cells 2024; 13:1607. [PMID: 39404371 PMCID: PMC11475720 DOI: 10.3390/cells13191607] [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: 08/20/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
As one of the most important economic traits for domestic animal husbandry, skeletal muscle is regulated by an intricate molecular network. Adenosine deaminase acting on RNA (ADAR1) involves various physiological processes and diseases, such as innate immunity and the development of lung adenocarcinoma, breast cancer, gastric cancer, etc. However, its role in skeletal muscle growth requires further clarification. Here, we explored the functions of ADAR1 in the myogenic process of goat skeletal muscle satellite cells (MuSCs). The ADAR1 transcripts were noticeably enriched in goat visceral tissues compared to skeletal muscle. Additionally, its levels in slow oxidative muscles like the psoas major and minor muscles were higher than in the fast oxidative glycolytic and fast glycolytic muscles. Among the two common isoforms from ADAR1, p110 is more abundant than p150. Moreover, overexpressing ADAR1 enhanced the proliferation and myogenic differentiation of MuSCs. The mRNA-seq performed on MuSCs' knockdown of ADAR1 obtained 146 differentially expressed genes (DEGs), 87 upregulated and 59 downregulated. These DEGs were concentrated in muscle development and process pathways, such as the MAPK and cAMP signaling pathways. Furthermore, many DEGs as the key nodes defined by protein-protein interaction networks (PPI), including STAT3, MYH3/8, TGFβ2, and ACTN4, were closely related to the myogenic process. Finally, RNA immunoprecipitation combined with qPCR (RIP-qPCR) showed that ADAR1 binds to PAX7 and MyoD mRNA. This study indicates that ADAR1 promotes the myogenic development of goat MuSCs, which provides a useful scientific reference for further exploring the ADAR1-related regulatory networks underlying mammal skeletal muscle growth.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Li Li
- Farm Animal Genetic Resources Exploration Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Z.Z.); (M.X.); (X.X.); (M.S.); (D.D.); (S.Z.); (J.C.); (J.G.); (T.Z.); (L.W.)
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Z.Z.); (M.X.); (X.X.); (M.S.); (D.D.); (S.Z.); (J.C.); (J.G.); (T.Z.); (L.W.)
| |
Collapse
|
22
|
Liu Z, Deng K, Su Y, Zhang Z, Shi C, Wang J, Fan Y, Zhang G, Wang F. IGF2BP1-mediated the stability and protein translation of FGFR1 mRNA regulates myogenesis through the ERK signaling pathway. Int J Biol Macromol 2024; 280:135989. [PMID: 39326619 DOI: 10.1016/j.ijbiomac.2024.135989] [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: 03/03/2024] [Revised: 08/21/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
N6-methyladenosine (m6A) is the most prevalent post-transcriptional modification of RNAs and plays a key regulatory role in various biological processes. As a member of the insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs) family, IGF2BP1 has recently demonstrated its ability to specifically bind m6A-modified sites within mRNAs and effectively regulate their mRNA stability. However, the precise roles of IGF2BP1 in mammalian skeletal muscle development, along with its downstream mRNA targets during myogenesis, have yet to be fully elucidated. Here, we observed that IGF2BP1 expression significantly decreased during myogenic differentiation. Knockdown of IGF2BP1 significantly inhibited myoblast proliferation while promoted myogenic differentiation. In contrast, IGF2BP1 overexpression robustly stimulated myoblast proliferation but suppressed their differentiation. Combined analysis of high-throughput sequencing and RNA stability assays revealed that IGF2BP1 can enhance fibroblast growth factor receptor 1 (FGFR1) mRNA stability and promote its translation in an m6A-dependent manner, thereby regulating its expression level and the Extracellular Signal-Regulated Kinase (ERK) pathway. Additionally, knockdown of FGFR1 rescued the phenotypic changes (namely increased cell proliferation and suppressed differentiation) induced by IGF2BP1 overexpression via attenuating ERK signaling. Taken together, our findings suggest that IGF2BP1 maintains the stability and translation of FGFR1 mRNA in an m6A-dependent manner, thereby inhibiting skeletal myogenesis through activation of the ERK signaling pathway. This study further enriches the understanding of the molecular mechanisms by which RNA methylation regulates myogenesis, providing valuable insights into the role of IGF2BP1-mediated post-transcriptional regulation in muscle development.
Collapse
Affiliation(s)
- Zhipeng Liu
- Sanya Research Institute of Nanjing Agricultural University & Hainan Seed Industry laborator, Nanjing Agricultural University, Sanya 572025, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Kaiping Deng
- Sanya Research Institute of Nanjing Agricultural University & Hainan Seed Industry laborator, Nanjing Agricultural University, Sanya 572025, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Yalong Su
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhen Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Congyu Shi
- Sanya Research Institute of Nanjing Agricultural University & Hainan Seed Industry laborator, Nanjing Agricultural University, Sanya 572025, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Jingang Wang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Yixuan Fan
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Guoming Zhang
- Sanya Research Institute of Nanjing Agricultural University & Hainan Seed Industry laborator, Nanjing Agricultural University, Sanya 572025, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China; College of veterinary medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Feng Wang
- Sanya Research Institute of Nanjing Agricultural University & Hainan Seed Industry laborator, Nanjing Agricultural University, Sanya 572025, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
23
|
Zhang H, Chang M, Chen D, Yang J, Zhang Y, Sun J, Yao X, Sun H, Gu X, Li M, Shen Y, Dai B. Congenital myopathies: pathophysiological mechanisms and promising therapies. J Transl Med 2024; 22:815. [PMID: 39223631 PMCID: PMC11370226 DOI: 10.1186/s12967-024-05626-5] [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: 01/03/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Congenital myopathies (CMs) are a kind of non-progressive or slow-progressive muscle diseases caused by genetic mutations, which are currently defined and categorized mainly according to their clinicopathological features. CMs exhibit pleiotropy and genetic heterogeneity. Currently, supportive treatment and pharmacological remission are the mainstay of treatment, with no cure available. Some adeno-associated viruses show promising prospects in the treatment of MTM1 and BIN1-associated myopathies; however, such gene-level therapeutic interventions target only specific mutation types and are not generalizable. Thus, it is particularly crucial to identify the specific causative genes. Here, we outline the pathogenic mechanisms based on the classification of causative genes: excitation-contraction coupling and triadic assembly (RYR1, MTM1, DNM2, BIN1), actin-myosin interaction and production of myofibril forces (NEB, ACTA1, TNNT1, TPM2, TPM3), as well as other biological processes. Furthermore, we provide a comprehensive overview of recent therapeutic advancements and potential treatment modalities of CMs. Despite ongoing research endeavors, targeted strategies and collaboration are imperative to address diagnostic uncertainties and explore potential treatments.
Collapse
Affiliation(s)
- Han Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Mengyuan Chang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Daiyue Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Jiawen Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Yijie Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Jiacheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Xinlei Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Meiyuan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China.
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China.
| | - Bin Dai
- Department of Orthopedics, Binhai County People's Hospital, Binhai, Jiangsu Province, 224500, P. R. China.
| |
Collapse
|
24
|
Wensveen FM, Šestan M, Polić B. The immunology of sickness metabolism. Cell Mol Immunol 2024; 21:1051-1065. [PMID: 39107476 PMCID: PMC11364700 DOI: 10.1038/s41423-024-01192-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/29/2024] [Indexed: 09/01/2024] Open
Abstract
Everyone knows that an infection can make you feel sick. Although we perceive infection-induced changes in metabolism as a pathology, they are a part of a carefully regulated process that depends on tissue-specific interactions between the immune system and organs involved in the regulation of systemic homeostasis. Immune-mediated changes in homeostatic parameters lead to altered production and uptake of nutrients in circulation, which modifies the metabolic rate of key organs. This is what we experience as being sick. The purpose of sickness metabolism is to generate a metabolic environment in which the body is optimally able to fight infection while denying vital nutrients for the replication of pathogens. Sickness metabolism depends on tissue-specific immune cells, which mediate responses tailored to the nature and magnitude of the threat. As an infection increases in severity, so do the number and type of immune cells involved and the level to which organs are affected, which dictates the degree to which we feel sick. Interestingly, many alterations associated with metabolic disease appear to overlap with immune-mediated changes observed following infection. Targeting processes involving tissue-specific interactions between activated immune cells and metabolic organs therefore holds great potential for treating both people with severe infection and those with metabolic disease. In this review, we will discuss how the immune system communicates in situ with organs involved in the regulation of homeostasis and how this communication is impacted by infection.
Collapse
Affiliation(s)
| | - Marko Šestan
- University of Rijeka Faculty of Medicine, Rijeka, Croatia
| | - Bojan Polić
- University of Rijeka Faculty of Medicine, Rijeka, Croatia
| |
Collapse
|
25
|
Gahlawat S, Oruc D, Paul N, Ragheb M, Patel S, Fasasi O, Sharma P, Shreiber DI, Freeman JW. Tissue Engineered 3D Constructs for Volumetric Muscle Loss. Ann Biomed Eng 2024; 52:2325-2347. [PMID: 39085677 PMCID: PMC11329418 DOI: 10.1007/s10439-024-03541-w] [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: 03/19/2024] [Accepted: 05/08/2024] [Indexed: 08/02/2024]
Abstract
Severe injuries to skeletal muscles, including cases of volumetric muscle loss (VML), are linked to substantial tissue damage, resulting in functional impairment and lasting disability. While skeletal muscle can regenerate following minor damage, extensive tissue loss in VML disrupts the natural regenerative capacity of the affected muscle tissue. Existing clinical approaches for VML, such as soft-tissue reconstruction and advanced bracing methods, need to be revised to restore tissue function and are associated with limitations in tissue availability and donor-site complications. Advancements in tissue engineering (TE), particularly in scaffold design and the delivery of cells and growth factors, show promising potential for regenerating damaged skeletal muscle tissue and restoring function. This article provides a brief overview of the pathophysiology of VML and critiques the shortcomings of current treatments. The subsequent section focuses on the criteria for designing TE scaffolds, offering insights into various natural and synthetic biomaterials and cell types for effectively regenerating skeletal muscle. We also review multiple TE strategies involving both acellular and cellular scaffolds to encourage the development and maturation of muscle tissue and facilitate integration, vascularization, and innervation. Finally, the article explores technical challenges hindering successful translation into clinical applications.
Collapse
Affiliation(s)
- Sonal Gahlawat
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Doga Oruc
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Nikhil Paul
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Mark Ragheb
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Swati Patel
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Oyinkansola Fasasi
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Peeyush Sharma
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - David I Shreiber
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA
| | - Joseph W Freeman
- Department of Biomedical Engineering, Rutgers University-New Brunswick, Piscataway, NJ, USA.
| |
Collapse
|
26
|
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 2024:10.1038/s41418-024-01363-w. [PMID: 39147882 DOI: 10.1038/s41418-024-01363-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
27
|
Sicherer ST, Haque N, Parikh Y, Grasman J. Current methodologies for inducing aligned myofibers in tissue constructs for skeletal muscle tissue regeneration. Adv Wound Care (New Rochelle) 2024. [PMID: 39126403 DOI: 10.1089/wound.2024.0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024] Open
Abstract
SIGNIFICANCE Volumetric muscle loss (VML) results in the loss of large amounts of tissue that inhibits muscle regeneration. Existing therapies, such as autologous muscle transfer and physical therapy, are incapable of returning full function and force production to injured muscle. RECENT ADVANCES Skeletal muscle tissue constructs may provide an alternative to existing therapies currently used to treat VML. Unlike autologous muscle transplants, muscle constructs can be cultured in vitro and are not reliant on intact muscle tissue. Skeletal muscle constructs can be generated from small muscle biopsies and could be used to generate skeletal muscle tissue constructs to replace injured tissues. CRITICAL ISSUES To serve as effective therapies, muscle constructs must be capable of generating contractile forces that can assist the function of host skeletal muscle. The contractile force of native muscle arises in part as a consequence of the highly aligned, bundled architecture of myofibers. Attempts to induce similar alignment include: applications of tension/strain across hydrogels, inducing aligned architectures within scaffolds, casting tissues in straited molds, and 3D printing. While all these methods have demonstrated efficacy towards inducing myofiber alignment, the extent of myofiber alignment, tissue formation, and force production varies. This manuscript critically reviews the advantages and limitations of these methods, and specifically discusses their ability to impart mechanical and architectural cues to induce alignment within constructs. FUTURE DIRECTIONS As tissue synthesizing techniques continue to improve, muscle constructs must include more cell types than simply myoblasts, such as the addition of neuronal and endothelial cells. Higher level tissue organization is critical to the success of these constructs. Many of these technologies have yet to be implanted into host tissue to understand engraftment and how they can contribute to traumatic injury, and as such continued collaboration between surgeons and tissue engineers is necessary to ultimately result in clinical translation.
Collapse
Affiliation(s)
- Sydnee T Sicherer
- New Jersey Institute of Technology, Biomedical Engineering, Newark, New Jersey, United States;
| | - Noor Haque
- New Jersey Institute of Technology, Biomedical Engineering, Newark, New Jersey, United States;
| | - Yash Parikh
- New Jersey Institute of Technology, Biomedical Engineering, Newark, New Jersey, United States;
| | - Jonathan Grasman
- New Jersey Institute of Technology, Biomedical Engineering, 323 Dr Martin Luther King Jr Blvd, Newark, New Jersey, United States, 07102;
| |
Collapse
|
28
|
Emmert ME, Emmert AS, Goh Q, Cornwall R. Sexual dimorphisms in skeletal muscle: current concepts and research horizons. J Appl Physiol (1985) 2024; 137:274-299. [PMID: 38779763 PMCID: PMC11343095 DOI: 10.1152/japplphysiol.00529.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 05/25/2024] Open
Abstract
The complex compositional and functional nature of skeletal muscle makes this organ an essential topic of study for biomedical researchers and clinicians. An additional layer of complexity is added with the consideration of sex as a biological variable. Recent research advances have revealed sexual dimorphisms in developmental biology, muscle homeostasis, adaptive responses, and disorders relating to skeletal muscle. Many of the observed sex differences have hormonal and molecular mechanistic underpinnings, whereas others have yet to be elucidated. Future research is needed to investigate the mechanisms dictating sex-based differences in the various aspects of skeletal muscle. As such, it is necessary that skeletal muscle biologists ensure that both female and male subjects are represented in biomedical and clinical studies to facilitate the successful testing and development of therapeutics for all patients.
Collapse
Affiliation(s)
- Marianne E Emmert
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Andrew S Emmert
- Department of Orthopaedic Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Qingnian Goh
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Orthopaedic Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Roger Cornwall
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Orthopaedic Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| |
Collapse
|
29
|
Alvarez AM, Trufen CEM, Buri MV, de Sousa MBN, Arruda-Alves FI, Lichtenstein F, Castro de Oliveira U, Junqueira-de-Azevedo IDLM, Teixeira C, Moreira V. Tumor Necrosis Factor-Alpha Modulates Expression of Genes Involved in Cytokines and Chemokine Pathways in Proliferative Myoblast Cells. Cells 2024; 13:1161. [PMID: 38995013 PMCID: PMC11240656 DOI: 10.3390/cells13131161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/20/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024] Open
Abstract
Skeletal muscle regeneration after injury is a complex process involving inflammatory signaling and myoblast activation. Pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF-α) are key mediators, but their effects on gene expression in proliferating myoblasts are unclear. We performed the RNA sequencing of TNF-α treated C2C12 myoblasts to elucidate the signaling pathways and gene networks regulated by TNF-α during myoblast proliferation. The TNF-α (10 ng/mL) treatment of C2C12 cells led to 958 differentially expressed genes compared to the controls. Pathway analysis revealed significant regulation of TNF-α signaling, along with the chemokine and IL-17 pathways. Key upregulated genes included cytokines (e.g., IL-6), chemokines (e.g., CCL7), and matrix metalloproteinases (MMPs). TNF-α increased myogenic factor 5 (Myf5) but decreased MyoD protein levels and stimulated the release of MMP-9, MMP-10, and MMP-13. TNF-α also upregulates versican and myostatin mRNA. Overall, our study demonstrates the TNF-α modulation of distinct gene expression patterns and signaling pathways that likely contribute to enhanced myoblast proliferation while suppressing premature differentiation after muscle injury. Elucidating the mechanisms involved in skeletal muscle regeneration can aid in the development of regeneration-enhancing therapeutics.
Collapse
Affiliation(s)
- Angela María Alvarez
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
- Reproduction Group, Pharmacy Department, School of Pharmaceutical and Food Sciences, University of Antioquia—UdeA, Medellín 050010, Colombia
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo 04044-020, SP, Brazil;
| | - Carlos Eduardo Madureira Trufen
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, v.i, 252 50 Vestec, Czech Republic
| | - Marcus Vinicius Buri
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
| | - Marcela Bego Nering de Sousa
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo 04044-020, SP, Brazil;
| | - Francisco Ivanio Arruda-Alves
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
| | - Flavio Lichtenstein
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
| | - Ursula Castro de Oliveira
- Laboratório de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (U.C.d.O.); (I.d.L.M.J.-d.-A.)
| | | | - Catarina Teixeira
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
- Laboratório de Farmacologia, Butantan Institute, Sao Paulo 05503-900, SP, Brazil
| | - Vanessa Moreira
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, Sao Paulo 05503-900, SP, Brazil; (A.M.A.); (C.E.M.T.); (M.V.B.); (F.I.A.-A.); (F.L.)
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo 04044-020, SP, Brazil;
| |
Collapse
|
30
|
Chiang WY, Yu HW, Wu MC, Huang YM, Chen YQ, Lin JW, Liu YW, You LR, Chiou A, Kuo JC. Matrix mechanics regulates muscle regeneration by modulating kinesin-1 activity. Biomaterials 2024; 308:122551. [PMID: 38593710 DOI: 10.1016/j.biomaterials.2024.122551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
Sarcopenia, a prevalent muscle disease characterized by muscle mass and strength reduction, is associated with impaired skeletal muscle regeneration. However, the influence of the biomechanical properties of sarcopenic skeletal muscle on the efficiency of the myogenic program remains unclear. Herein, we established a mouse model of sarcopenia and observed a reduction in stiffness within the sarcopenic skeletal muscle in vivo. To investigate whether the biomechanical properties of skeletal muscle directly impact the myogenic program, we established an in vitro system to explore the intrinsic mechanism involving matrix stiffness control of myogenic differentiation. Our findings identify the microtubule motor protein, kinesin-1, as a mechano-transduction hub that senses and responds to matrix stiffness, crucial for myogenic differentiation and muscle regeneration. Specifically, kinesin-1 activity is positively regulated by stiff matrices, facilitating its role in transporting mitochondria and enhancing translocation of the glucose transporter GLUT4 to the cell surface for glucose uptake. Conversely, the softer matrices significantly suppress kinesin-1 activity, leading to the accumulation of mitochondria around nuclei and hindering glucose uptake by inhibiting GLUT4 membrane translocation, consequently impairing myogenic differentiation. The insights gained from the in-vitro system highlight the mechano-transduction significance of kinesin-1 motor proteins in myogenic differentiation. Furthermore, our study confirms that enhancing kinesin-1 activity in the sarcopenic mouse model restores satellite cell expansion, myogenic differentiation, and muscle regeneration. Taken together, our findings provide a potential target for improving muscle regeneration in sarcopenia.
Collapse
Affiliation(s)
- Wan-Yu Chiang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Helen Wenshin Yu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Ming-Chung Wu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Yi-Man Huang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Yin-Quan Chen
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Jong-Wei Lin
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Yen-Wenn Liu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Li-Ru You
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Arthur Chiou
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan; Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan.
| |
Collapse
|
31
|
Ozturk T, Mignot J, Gattazzo F, Gervais M, Relaix F, Rouard H, Didier N. Dual inhibition of P38 MAPK and JNK pathways preserves stemness markers and alleviates premature activation of muscle stem cells during isolation. Stem Cell Res Ther 2024; 15:179. [PMID: 38902774 PMCID: PMC11191274 DOI: 10.1186/s13287-024-03795-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/08/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Adult skeletal muscle contains resident muscle stem cells (MuSC) with high myogenic and engraftment potentials, making them suitable for cell therapy and regenerative medicine approaches. However, purification process of MuSC remains a major hurdle to their use in the clinic. Indeed, muscle tissue enzymatic dissociation triggers a massive activation of stress signaling pathways, among which P38 and JNK MAPK, associated with a premature loss of MuSC quiescence. While the role of these pathways in the myogenic progression of MuSC is well established, the extent to which their dissociation-induced activation affects the functionality of these cells remains unexplored. METHODS We assessed the effect of P38 and JNK MAPK induction on stemness marker expression and MuSC activation state during isolation by pharmacological approaches. MuSC functionality was evaluated by in vitro assays and in vivo transplantation experiments. We performed a comparative analysis of the transcriptome of human MuSC purified with pharmacological inhibitors of P38 and JNK MAPK (SB202190 and SP600125, respectively) versus available RNAseq resources. RESULTS We monitored PAX7 protein levels in murine MuSC during muscle dissociation and demonstrated a two-step decline partly dependent on P38 and JNK MAPK activities. We showed that simultaneous inhibition of these pathways throughout the MuSC isolation process preserves the expression of stemness markers and limits their premature activation, leading to improved survival and amplification in vitro as well as increased engraftment in vivo. Through a comparative RNAseq analysis of freshly isolated human MuSC, we provide evidence that our findings in murine MuSC could be relevant to human MuSC. Based on these findings, we implemented a purification strategy, significantly improving the recovery yields of human MuSC. CONCLUSION Our study highlights the pharmacological limitation of P38 and JNK MAPK activities as a suitable strategy to qualitatively and quantitatively ameliorate human MuSC purification process, which could be of great interest for cell-based therapies.
Collapse
Affiliation(s)
- Teoman Ozturk
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
| | - Julien Mignot
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
| | | | - Marianne Gervais
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
| | - Frédéric Relaix
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
- EnvA, IMRB, 94700, Maisons-Alfort, France
- AP-HP, Hopital Mondor, Service d'histologie, 94010, Creteil, France
| | - Hélène Rouard
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
- AP-HP, Hopital Mondor, Service d'histologie, 94010, Creteil, France
| | - Nathalie Didier
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France.
| |
Collapse
|
32
|
Lee SB, Woo TW, Baek DC, Son CG. A standardized herbal combination of Astragalus membranaceus and Paeonia japonica promotes skeletal muscle hypertrophy in a treadmill exercise mouse model. Front Nutr 2024; 11:1362550. [PMID: 38966418 PMCID: PMC11223055 DOI: 10.3389/fnut.2024.1362550] [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: 12/28/2023] [Accepted: 05/29/2024] [Indexed: 07/06/2024] Open
Abstract
Background Maintaining a normal range of muscle mass and function is crucial not only for sustaining a healthy life but also for preventing various disorders. Numerous nutritional or natural resources are being explored for their potential muscle hypertrophic properties. Aim We aimed to evaluate the muscle hypertrophic effects of APX, a 1:1 mixture of Astragalus membranaceus and Paeonia japonica. In addition to the myotube differentiation cell assay, we utilized a weighted exercise-based animal model and evaluated changes in muscle hypertrophy using dual-energy X-ray absorptiometry (DXA) and histological analysis. Results The 8-week treadmill exercise led to notable decreases in body weight and fat mass but an increase in muscle mass compared to the control group. Administration of APX significantly accelerated muscle mass gain (p < 0.05) without altering body weight or fat mass compared to the exercise-only group. This muscle hypertrophic effect of APX was consistent with the histologic size of muscle fibers in the gastrocnemius (p > 0.05) and rectus femoris (p < 0.05), as well as the regulation of myogenic transcription factors (MyoD and myogenin), respectively. Furthermore, APX demonstrated a similar action to insulin-like growth factor 1, influencing the proliferation of C2C12 myoblast cells (p < 0.01) and their differentiation into myotubes (p < 0.05) compared to the control group. Conclusion The present study provides experimental evidence that APX has muscle hypertrophic effects, and its underlying mechanisms would involve the modulation of MyoD and myogenin.
Collapse
Affiliation(s)
| | | | | | - Chang-Gue Son
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon, Republic of Korea
| |
Collapse
|
33
|
Nozdrenko D, Motuziuk O, Prylutska S, Matviienko T, Bogutska K, Franskevych D, Nurishchenko N, Abramchuk O, Prylutskyy Y. С 60 fullerene protective effect against the rat muscle soleus trauma. Heliyon 2024; 10:e32677. [PMID: 38961948 PMCID: PMC11219981 DOI: 10.1016/j.heliyon.2024.e32677] [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: 01/21/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024] Open
Abstract
Muscle trauma is one of the most common body injuries. Severe consequences of muscle trauma are ischemic injuries of the extremities. It is known that the intensification of free radical processes takes place in almost most acute diseases and conditions, including muscle trauma. C60 fullerene (C60) with powerful antioxidant properties can be considered a potential nanoagent for developing an effective therapy for skeletal muscle trauma. Here the water-soluble C60 was prepared and its structural organization has been studied by the atomic force microscopy and dynamic light scattering techniques. The selective biomechanical parameters of muscle soleus contraction and biochemical indicators of blood in rats were evaluated after intramuscular injection of C60 1 h before the muscle trauma initiation. Analysis of the force muscle response after C60 injection (1 mg kg-1 dose) showed its protective effect against ischemia and mechanical injury at the level of 30 ± 2 % and 17 ± 1 %, accordingly, relative to the pathology group. Analysis of biomechanical parameters that are responsible for correcting precise positioning confirmed the effectiveness of C60 at a level of more than 50 ± 3 % relative to the pathology group. Moreover, a decrease in the biochemical indicators of blood by about 33 ± 2 % and 10 ± 1 % in ischemia and mechanical injury, correspondingly, relative to the pathology group occurs. The results obtained demonstrate the ability of C60 to correct the functional activity of damaged skeletal muscle.
Collapse
Affiliation(s)
- Dmytro Nozdrenko
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601, Ukraine
| | - Olexandr Motuziuk
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601, Ukraine
- Faculty of Biology and Forestry, Lesya Ukrainka Volyn National University, Lutsk, 43025, Ukraine
| | - Svitlana Prylutska
- Faculty of Plant Protection, Biotechnology and Ecology, National University of Life and Environmental Science of Ukraine, Kyiv, 03041, Ukraine
| | - Tetiana Matviienko
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601, Ukraine
| | - Kateryna Bogutska
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601, Ukraine
| | - Daria Franskevych
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601, Ukraine
| | - Nataliya Nurishchenko
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601, Ukraine
| | - Olga Abramchuk
- Faculty of Biology and Forestry, Lesya Ukrainka Volyn National University, Lutsk, 43025, Ukraine
| | - Yuriy Prylutskyy
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 01601, Ukraine
| |
Collapse
|
34
|
Cockrell C, Vodovotz Y, Zamora R, An G. The Wound Environment Agent-based Model (WEABM): a digital twin platform for characterization and complex therapeutic discovery for volumetric muscle loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.595972. [PMID: 38895374 PMCID: PMC11185759 DOI: 10.1101/2024.06.04.595972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Volumetric Muscle Loss (VML) injuries are characterized by significant loss of muscle mass, usually due to trauma or surgical resection, often with a residual open wound in clinical settings and subsequent loss of limb function due to the replacement of the lost muscle mass with non-functional scar. Being able to regrow functional muscle in VML injuries is a complex control problem that needs to override robust, evolutionarily conserved healing processes aimed at rapidly closing the defect in lieu of restoration of function. We propose that discovering and implementing this complex control can be accomplished by the development of a Medical Digital Twin of VML. Digital Twins (DTs) are the subject of a recent report from the National Academies of Science, Engineering and Medicine (NASEM), which provides guidance as to the definition, capabilities and research challenges associated with the development and implementation of DTs. Specifically, DTs are defined as dynamic computational models that can be personalized to an individual real world "twin" and are connected to that twin via an ongoing data link. DTs can be used to provide control on the real-world twin that is, by the ongoing data connection, adaptive. We have developed an anatomic scale cell-level agent-based model of VML termed the Wound Environment Agent Based Model (WEABM) that can serve as the computational specification for a DT of VML. Simulations of the WEABM provided fundamental insights into the biology of VML, and we used the WEABM in our previously developed pipeline for simulation-based Deep Reinforcement Learning (DRL) to train an artificial intelligence (AI) to implement a robust generalizable control policy aimed at increasing the healing of VML with functional muscle. The insights into VML obtained include: 1) a competition between fibrosis and myogenesis due to spatial constraints on available edges of intact myofibrils to initiate the myoblast differentiation process, 2) the need to biologically "close" the wound from atmospheric/environmental exposure, which represents an ongoing inflammatory stimulus that promotes fibrosis and 3) that selective, multimodal and adaptive local mediator-level control can shift the trajectory of healing away from a highly evolutionarily beneficial imperative to close the wound via fibrosis. Control discovery with the WEABM identified the following design principles: 1) multimodal adaptive tissue-level mediator control to mitigate pro-inflammation as well as the pro-fibrotic aspects of compensatory anti-inflammation, 2) tissue-level mediator manipulation to promote myogenesis, 3) the use of an engineered extracellular matrix (ECM) to functionally close the wound and 4) the administration of an anti-fibrotic agent focused on the collagen-producing function of fibroblasts and myofibroblasts. The WEABM-trained DRL AI integrates these control modalities and provides design specifications for a potential device that can implement the required wound sensing and intervention delivery capabilities needed. The proposed cyber-physical system integrates the control AI with a physical sense-and-actuate device that meets the tenets of DTs put forth in the NASEM report and can serve as an example schema for the future development of Medical DTs.
Collapse
Affiliation(s)
- Chase Cockrell
- Department of Surgery, University of Vermont Larner College of Medicine
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh
- McGowan Institute of Regenerative Medicine, University of Pittsburgh
| | | | - Gary An
- Department of Surgery, University of Vermont Larner College of Medicine
| |
Collapse
|
35
|
Collins BC, Shapiro JB, Scheib MM, Musci RV, Verma M, Kardon G. Three-dimensional imaging studies in mice identify cellular dynamics of skeletal muscle regeneration. Dev Cell 2024; 59:1457-1474.e5. [PMID: 38569550 PMCID: PMC11153043 DOI: 10.1016/j.devcel.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 12/06/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
The function of many organs, including skeletal muscle, depends on their three-dimensional structure. Muscle regeneration therefore requires not only reestablishment of myofibers but also restoration of tissue architecture. Resident muscle stem cells (SCs) are essential for regeneration, but how SCs regenerate muscle architecture is largely unknown. We address this problem using genetic labeling of mouse SCs and whole-mount imaging to reconstruct, in three dimensions, muscle regeneration. Unexpectedly, we found that myofibers form via two distinct phases of fusion and the residual basement membrane of necrotic myofibers is critical for promoting fusion and orienting regenerated myofibers. Furthermore, the centralized myonuclei characteristic of regenerated myofibers are associated with myofibrillogenesis and endure months post injury. Finally, we elucidate two cellular mechanisms for the formation of branched myofibers, a pathology characteristic of diseased muscle. We provide a synthesis of the cellular events of regeneration and show that these differ from those used during development.
Collapse
Affiliation(s)
- Brittany C Collins
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Jacob B Shapiro
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Mya M Scheib
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Robert V Musci
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Mayank Verma
- Department of Pediatrics, Division of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
36
|
Wang H, Liu M, Tang H, Zhang Z, Wen H, He F. Identification and functional analysis of circpdlim5a generated from pdlim5a gene splicing in the skeletal muscle of Japanese flounder (Paralichthys olivaceus). Gen Comp Endocrinol 2024; 352:114500. [PMID: 38508470 DOI: 10.1016/j.ygcen.2024.114500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/11/2024] [Accepted: 03/17/2024] [Indexed: 03/22/2024]
Abstract
Circular RNAs (circRNAs) are non-coding RNAs with endogenous regulatory functions, including regulating skeletal muscle development. However, its role in the development of skeletal muscle in Japanese flounder (Paralichthys olivaceus) is not clear. Therefore we screened a candidate circpdlim5a, which is derived from the gene pdlim5a, from the skeletal muscle transcriptome of Japanese flounder. We characterized circpdlim5a, which was more stable compared to the linear RNA pdlim5a. Distributional characterization of circpdlim5a showed that circpdlim5a was predominantly distributed in the nucleus and was highly expressed in the skeletal muscle of adult Japanese flounder (24 months). When we further studied the circpdlim5a function, we found that it inhibited the expression of proliferation and differentiation genes according to the over-expression experiment of circpdlim5a in myoblasts. We concluded that circpdlim5a may inhibit the proliferation and differentiation of myoblasts and thereby inhibit skeletal muscle development in Japanese flounder. This experiment provides information for the study of circRNAs by identifying circpdlim5a and exploring its function, and offers clues for molecular breeding from an epigenetic perspective.
Collapse
Affiliation(s)
- Hao Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China
| | - Min Liu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China
| | - Hengtai Tang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China
| | - Zhirui Zhang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China
| | - Haishen Wen
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China
| | - Feng He
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China.
| |
Collapse
|
37
|
Luo Y, Zheng S, Xiao W, Zhang H, Li Y. Pannexins in the musculoskeletal system: new targets for development and disease progression. Bone Res 2024; 12:26. [PMID: 38705887 PMCID: PMC11070431 DOI: 10.1038/s41413-024-00334-8] [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: 12/08/2023] [Revised: 03/04/2024] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
During cell differentiation, growth, and development, cells can respond to extracellular stimuli through communication channels. Pannexin (Panx) family and connexin (Cx) family are two important types of channel-forming proteins. Panx family contains three members (Panx1-3) and is expressed widely in bone, cartilage and muscle. Although there is no sequence homology between Panx family and Cx family, they exhibit similar configurations and functions. Similar to Cxs, the key roles of Panxs in the maintenance of physiological functions of the musculoskeletal system and disease progression were gradually revealed later. Here, we seek to elucidate the structure of Panxs and their roles in regulating processes such as osteogenesis, chondrogenesis, and muscle growth. We also focus on the comparison between Cx and Panx. As a new key target, Panxs expression imbalance and dysfunction in muscle and the therapeutic potentials of Panxs in joint diseases are also discussed.
Collapse
Affiliation(s)
- Yan Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, 410008, China
| | - Shengyuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, 410008, China
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hang Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
38
|
Ahmad SS, Ahmad K, Lim JH, Shaikh S, Lee EJ, Choi I. Therapeutic applications of biological macromolecules and scaffolds for skeletal muscle regeneration: A review. Int J Biol Macromol 2024; 267:131411. [PMID: 38588841 DOI: 10.1016/j.ijbiomac.2024.131411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 04/10/2024]
Abstract
Skeletal muscle (SM) mass and strength maintenance are important requirements for human well-being. SM regeneration to repair minor injuries depends upon the myogenic activities of muscle satellite (stem) cells. However, losses of regenerative properties following volumetric muscle loss or severe trauma or due to congenital muscular abnormalities are not self-restorable, and thus, these conditions have major healthcare implications and pose clinical challenges. In this context, tissue engineering based on different types of biomaterials and scaffolds provides an encouraging means of structural and functional SM reconstruction. In particular, biomimetic (able to transmit biological signals) and several porous scaffolds are rapidly evolving. Several biological macromolecules/biomaterials (collagen, gelatin, alginate, chitosan, and fibrin etc.) are being widely used for SM regeneration. However, available alternatives for SM regeneration must be redesigned to make them more user-friendly and economically feasible with longer shelf lives. This review aimed to explore the biological aspects of SM regeneration and the roles played by several biological macromolecules and scaffolds in SM regeneration in cases of volumetric muscle loss.
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
| | - Khurshid 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
| | - Sibhghatulla Shaikh
- 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
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea.
| |
Collapse
|
39
|
Wang S, Tian B, Hu Y, Li T, Cui X, Zhang L, Luo X. Research progress on the biological regulatory mechanisms of selenium on skeletal muscle in broilers. Poult Sci 2024; 103:103646. [PMID: 38520938 PMCID: PMC10978542 DOI: 10.1016/j.psj.2024.103646] [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: 12/16/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 03/25/2024] Open
Abstract
As one of the indispensable trace elements for both humans and animals, selenium widely participates in multiple physiological processes and facilitates strong anti-inflammatory, antioxidant, and immune enhancing abilities. The biological functions of selenium are primarily driven by its presence in selenoproteins as a form of selenocysteine. Broilers are highly sensitive to selenium intake. Recent reports have demonstrated that selenium deficiency can adversely affect the quality of skeletal muscles and the economic value of broilers; the regulatory roles of several key selenoproteins (e.g., GPX1, GPX4, TXNRD1, TXNRD3, SelK, SelT, and SelW) have been identified. Starting from the selenium metabolism and its biological utilization in the skeletal muscle, the effect of the selenium antioxidant function on broiler meat quality is discussed in detail. The progress of research into the prevention of skeletal muscle injury by selenium and selenoproteins is also summarized. The findings emphasize the necessity of in vivo and in vitro research, and certain mechanism problems are identified, which aids their further examination. This mini-review will be helpful to provide a theoretical basis for the further study of regulatory mechanisms of selenium nutrition in edible poultry.
Collapse
Affiliation(s)
- Shengchen Wang
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Bing Tian
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Yun Hu
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Tingting Li
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Xiaoyan Cui
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Liyang Zhang
- Mineral Nutrition Research Division, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xugang Luo
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China.
| |
Collapse
|
40
|
Kiperman T, Ma K. Circadian Clock in Muscle Disease Etiology and Therapeutic Potential for Duchenne Muscular Dystrophy. Int J Mol Sci 2024; 25:4767. [PMID: 38731986 PMCID: PMC11083552 DOI: 10.3390/ijms25094767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Circadian clock and clock-controlled output pathways exert temporal control in diverse aspects of skeletal muscle physiology, including the maintenance of muscle mass, structure, function, and metabolism. They have emerged as significant players in understanding muscle disease etiology and potential therapeutic avenues, particularly in Duchenne muscular dystrophy (DMD). This review examines the intricate interplay between circadian rhythms and muscle physiology, highlighting how disruptions of circadian regulation may contribute to muscle pathophysiology and the specific mechanisms linking circadian clock dysregulation with DMD. Moreover, we discuss recent advancements in chronobiological research that have shed light on the circadian control of muscle function and its relevance to DMD. Understanding clock output pathways involved in muscle mass and function offers novel insights into the pathogenesis of DMD and unveils promising avenues for therapeutic interventions. We further explore potential chronotherapeutic strategies targeting the circadian clock to ameliorate muscle degeneration which may inform drug development efforts for muscular dystrophy.
Collapse
Affiliation(s)
| | - Ke Ma
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA;
| |
Collapse
|
41
|
Dar A, Li A, Petrigliano FA. Lineage tracing reveals a novel PDGFRβ + satellite cell subset that contributes to myo-regeneration of chronically injured rotator cuff muscle. Sci Rep 2024; 14:9668. [PMID: 38671006 PMCID: PMC11053018 DOI: 10.1038/s41598-024-58926-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Massive rotator cuff (RC) tendon tears are associated with progressive fibro-adipogenesis and muscle atrophy that altogether cause shoulder muscle wasting. Platelet derived growth factor β (PDGFRβ) lineage cells, that co-express PDGFRα have previously been shown to directly contribute to scar formation and fat accumulation in a mouse model of irreversible tendon and nerve transection (TTDN). Conversely, PDGFRβ+ lineage cells have also been shown to be myogenic in cultures and in other models of skeletal muscle injury. We therefore hypothesized that PDGFRβ demarcates two distinct RC residing subpopulations, fibro-adipogenic and myogenic, and aimed to elucidate the identity of the PDGFRβ myogenic precursors and evaluate their contribution, if any, to RC myo-regeneration. Lineage tracing revealed increasing contribution of PDGFRβ+ myo-progenitors to the formation of GFP+ myofibers, which were the most abundant myofiber type in regenerated muscle at 2 weeks post-TTDN. Muscle regeneration preceded muscle atrophy and both advanced from the lateral site of tendon transection to the farthest medial region. GFP+/PDGFRβ+Sca-1-lin-CXCR4+Integrin-β1+ marked a novel subset of satellite cells with confirmed myogenic properties. Further studies are warranted to identify the existence of PDGFRβ+ satellite cells in human and other mouse muscles and to define their myo-regenerative potential following acute and chronic muscle injury.
Collapse
Affiliation(s)
- Ayelet Dar
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Angela Li
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Frank A Petrigliano
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
42
|
Carnes ME, Gonyea CR, Coburn JM, Pins GD. A biomimetic approach to modulating the sustained release of fibroblast growth factor 2 from fibrin microthread scaffolds. EXPLORATION OF BIOMAT-X 2024; 1:58-83. [PMID: 39070763 PMCID: PMC11274095 DOI: 10.37349/ebmx.2024.00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2024]
Abstract
Aim The pleiotropic effect of fibroblast growth factor 2 (FGF2) on promoting myogenesis, angiogenesis, and innervation makes it an ideal growth factor for treating volumetric muscle loss (VML) injuries. While an initial delivery of FGF2 has demonstrated enhanced regenerative potential, the sustained delivery of FGF2 from scaffolds with robust structural properties as well as biophysical and biochemical signaling cues has yet to be explored for treating VML. The goal of this study is to develop an instructive fibrin microthread scaffold with intrinsic topographic alignment cues as well as regenerative signaling cues and a physiologically relevant, sustained release of FGF2 to direct myogenesis and ultimately enhance functional muscle regeneration. Methods Heparin was passively adsorbed or carbodiimide-conjugated to microthreads, creating a biomimetic binding strategy, mimicking FGF2 sequestration in the extracellular matrix (ECM). It was also evaluated whether FGF2 incorporated into fibrin microthreads would yield sustained release. It was hypothesized that heparin-conjugated and co-incorporated (co-inc) fibrin microthreads would facilitate sustained release of FGF2 from the scaffold and enhance in vitro myoblast proliferation and outgrowth. Results Toluidine blue staining and Fourier transform infrared spectroscopy confirmed that carbodiimide-conjugated heparin bound to fibrin microthreads in a dose-dependent manner. Release kinetics revealed that heparin-conjugated fibrin microthreads exhibited sustained release of FGF2 over a period of one week. An in vitro assay demonstrated that FGF2 released from microthreads remained bioactive, stimulating myoblast proliferation over four days. Finally, a cellular outgrowth assay suggests that FGF2 promotes increased outgrowth onto microthreads. Conclusions It was anticipated that the combined effects of fibrin microthread structural properties, topographic alignment cues, and FGF2 release profiles will facilitate the fabrication of a biomimetic scaffold that enhances the regeneration of functional muscle tissue for the treatment of VML injuries.
Collapse
Affiliation(s)
- Meagan E. Carnes
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Cailin R. Gonyea
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Jeannine M. Coburn
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - George D. Pins
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| |
Collapse
|
43
|
Gil-Melgosa L, Llombart-Blanco R, Extramiana L, Lacave I, Abizanda G, Miranda E, Agirre X, Prósper F, Pineda-Lucena A, Pons-Villanueva J, Pérez-Ruiz A. HDACi vorinostat protects muscle from degeneration after acute rotator cuff injury in mice. Bone Joint Res 2024; 13:169-183. [PMID: 38618868 PMCID: PMC11017234 DOI: 10.1302/2046-3758.134.bjr-2023-0292.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/16/2024] Open
Abstract
Aims Rotator cuff (RC) injuries are characterized by tendon rupture, muscle atrophy, retraction, and fatty infiltration, which increase injury severity and jeopardize adequate tendon repair. Epigenetic drugs, such as histone deacetylase inhibitors (HDACis), possess the capacity to redefine the molecular signature of cells, and they may have the potential to inhibit the transformation of the fibro-adipogenic progenitors (FAPs) within the skeletal muscle into adipocyte-like cells, concurrently enhancing the myogenic potential of the satellite cells. Methods HDACis were added to FAPs and satellite cell cultures isolated from mice. The HDACi vorinostat was additionally administered into a RC injury animal model. Histological analysis was carried out on the isolated supra- and infraspinatus muscles to assess vorinostat anti-muscle degeneration potential. Results Vorinostat, a HDACi compound, blocked the adipogenic transformation of muscle-associated FAPs in culture, promoting myogenic progression of the satellite cells. Furthermore, it protected muscle from degeneration after acute RC in mice in the earlier muscle degenerative stage after tenotomy. Conclusion The HDACi vorinostat may be a candidate to prevent early muscular degeneration after RC injury.
Collapse
Affiliation(s)
- Lara Gil-Melgosa
- Orthopedic Surgery Department of Clínica Universidad de Navarra (CUN) and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Rafael Llombart-Blanco
- Orthopedic Surgery Department of Clínica Universidad de Navarra (CUN) and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Leire Extramiana
- Technological Innovation Division, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV) and IdiSNA, Pamplona, Spain
| | | | - Gloria Abizanda
- Technological Innovation Division, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV) and IdiSNA, Pamplona, Spain
| | | | - Xabier Agirre
- Hemato-Oncology Program, FIMA-UNAV and IdiSNA, Pamplona, Spain
| | - Felipe Prósper
- Hemato-Oncology Program, FIMA-UNAV and IdiSNA, Pamplona, Spain
- Haematology Department, Clinica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | | | - Juan Pons-Villanueva
- Orthopedic Surgery Department of Clínica Universidad de Navarra (CUN) and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Ana Pérez-Ruiz
- Technological Innovation Division, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV) and IdiSNA, Pamplona, Spain
| |
Collapse
|
44
|
Li D, Yue Y, Feng X, Lv W, Fan Y, Sha P, Zhao T, Lin Y, Xiong X, Li J, Xiong Y. MicroRNA-542-3p targets Pten to inhibit the myoblasts proliferation but suppresses myogenic differentiation independent of targeted Pten. BMC Genomics 2024; 25:325. [PMID: 38561670 PMCID: PMC10983626 DOI: 10.1186/s12864-024-10260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Non-coding RNA is a key epigenetic regulation factor during skeletal muscle development and postnatal growth, and miR-542-3p was reported to be conserved and highly expressed in the skeletal muscle among different species. However, its exact functions in the proliferation of muscle stem cells and myogenesis remain to be determined. METHODS Transfection of proliferative and differentiated C2C12 cells used miR-542-3p mimic and inhibitor. RT-qPCR, EdU staining, immunofluorescence staining, cell counting kit 8 (CCK-8), and Western blot were used to evaluate the proliferation and myogenic differentiation caused by miR-542-3p. The dual luciferase reporter analysis and rescued experiment of the target gene were used to reveal the molecular mechanism. RESULTS The data shows overexpression of miR-542-3p downregulation of mRNA and protein levels of proliferation marker genes, reduction of EdU+ cells, and cellular vitality. Additionally, knocking it down promoted the aforementioned phenotypes. For differentiation, the miR-542-3p gain-of-function reduced both mRNA and protein levels of myogenic genes, including MYOG, MYOD1, et al. Furthermore, immunofluorescence staining immunized by MYHC antibody showed that the myotube number, fluorescence intensity, differentiation index, and myotube fusion index all decreased in the miR-542-3p mimic group, compared with the control group. Conversely, these phenotypes exhibited an increased trend in the miR-542-3p inhibitor group. Mechanistically, phosphatase and tensin homolog (Pten) was identified as the bona fide target gene of miR-542-3p by dual luciferase reporter gene assay, si-Pten combined with miR-542-3p inhibitor treatments totally rescued the promotion of proliferation by loss-function of miR-542-3p. CONCLUSIONS This study indicates that miR-542-3p inhibits the proliferation and differentiation of myoblast and Pten is a dependent target gene of miR-542-3p in myoblast proliferation, but not in differentiation.
Collapse
Grants
- No. 31902154 the National Natural Sciences Foundation of China
- No. 31902154 the National Natural Sciences Foundation of China
- No. 31902154 the National Natural Sciences Foundation of China
- No. 31902154 the National Natural Sciences Foundation of China
- No. 31902154 the National Natural Sciences Foundation of China
- No. 31902154 the National Natural Sciences Foundation of China
- No. 31902154 the National Natural Sciences Foundation of China
- No. 31902154 the National Natural Sciences Foundation of China
- No. 31902154 the National Natural Sciences Foundation of China
- No. 31902154 the National Natural Sciences Foundation of China
- No. 31902154 the National Natural Sciences Foundation of China
- No. 23NSFSC1804 the Natural Science Foundation of Sichuan Province
- No. 23NSFSC1804 the Natural Science Foundation of Sichuan Province
- No. 23NSFSC1804 the Natural Science Foundation of Sichuan Province
- No. 23NSFSC1804 the Natural Science Foundation of Sichuan Province
- No. 23NSFSC1804 the Natural Science Foundation of Sichuan Province
- No. 23NSFSC1804 the Natural Science Foundation of Sichuan Province
- No. 23NSFSC1804 the Natural Science Foundation of Sichuan Province
- No. 23NSFSC1804 the Natural Science Foundation of Sichuan Province
- No. 23NSFSC1804 the Natural Science Foundation of Sichuan Province
- No. 23NSFSC1804 the Natural Science Foundation of Sichuan Province
- No. 23NSFSC1804 the Natural Science Foundation of Sichuan Province
- No. 23ZDYF3118 the Key Research and Development Program of Sichuan Province
- No. 23ZDYF3118 the Key Research and Development Program of Sichuan Province
- No. 23ZDYF3118 the Key Research and Development Program of Sichuan Province
- No. 23ZDYF3118 the Key Research and Development Program of Sichuan Province
- No. 23ZDYF3118 the Key Research and Development Program of Sichuan Province
- No. 23ZDYF3118 the Key Research and Development Program of Sichuan Province
- No. 23ZDYF3118 the Key Research and Development Program of Sichuan Province
- No. 23ZDYF3118 the Key Research and Development Program of Sichuan Province
- No. 23ZDYF3118 the Key Research and Development Program of Sichuan Province
- No. 23ZDYF3118 the Key Research and Development Program of Sichuan Province
- No. 23ZDYF3118 the Key Research and Development Program of Sichuan Province
- No. XM2023004 the Southwest Minzu University Double World-Class Project
- No. XM2023004 the Southwest Minzu University Double World-Class Project
- No. XM2023004 the Southwest Minzu University Double World-Class Project
- No. XM2023004 the Southwest Minzu University Double World-Class Project
- No. XM2023004 the Southwest Minzu University Double World-Class Project
- No. XM2023004 the Southwest Minzu University Double World-Class Project
- No. XM2023004 the Southwest Minzu University Double World-Class Project
- No. XM2023004 the Southwest Minzu University Double World-Class Project
- No. XM2023004 the Southwest Minzu University Double World-Class Project
- No. XM2023004 the Southwest Minzu University Double World-Class Project
- No. XM2023004 the Southwest Minzu University Double World-Class Project
- No. 2023NYXXS130 the Fundamental Research Funds for the Central Universities, Southwest Minzu University
- No. 2023NYXXS130 the Fundamental Research Funds for the Central Universities, Southwest Minzu University
- No. 2023NYXXS130 the Fundamental Research Funds for the Central Universities, Southwest Minzu University
- No. 2023NYXXS130 the Fundamental Research Funds for the Central Universities, Southwest Minzu University
- No. 2023NYXXS130 the Fundamental Research Funds for the Central Universities, Southwest Minzu University
- No. 2023NYXXS130 the Fundamental Research Funds for the Central Universities, Southwest Minzu University
- No. 2023NYXXS130 the Fundamental Research Funds for the Central Universities, Southwest Minzu University
- No. 2023NYXXS130 the Fundamental Research Funds for the Central Universities, Southwest Minzu University
- No. 2023NYXXS130 the Fundamental Research Funds for the Central Universities, Southwest Minzu University
- No. 2023NYXXS130 the Fundamental Research Funds for the Central Universities, Southwest Minzu University
- No. 2023NYXXS130 the Fundamental Research Funds for the Central Universities, Southwest Minzu University
Collapse
Affiliation(s)
- Dandan Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education/Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Yongqi Yue
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, 712100, China
| | - Xinxin Feng
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education/Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
- Chongxin County Animal Husbandry and Veterinary Center, Pingliang, 744200, China
| | - Weibing Lv
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education/Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Yilin Fan
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education/Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Peiran Sha
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education/Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Te Zhao
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education/Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Yaqiu Lin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education/Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Xianrong Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education/Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Jian Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education/Sichuan Province, Southwest Minzu University, Chengdu, 610041, China
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China
| | - Yan Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education/Sichuan Province, Southwest Minzu University, Chengdu, 610041, China.
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China.
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, 610041, China.
| |
Collapse
|
45
|
Ducommun S, Jannig PR, Cervenka I, Murgia M, Mittenbühler MJ, Chernogubova E, Dias JM, Jude B, Correia JC, Van Vranken JG, Ocana-Santero G, Porsmyr-Palmertz M, McCann Haworth S, Martínez-Redondo V, Liu Z, Carlström M, Mann M, Lanner JT, Teixeira AI, Maegdefessel L, Spiegelman BM, Ruas JL. Mustn1 is a smooth muscle cell-secreted microprotein that modulates skeletal muscle extracellular matrix composition. Mol Metab 2024; 82:101912. [PMID: 38458566 PMCID: PMC10950823 DOI: 10.1016/j.molmet.2024.101912] [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: 12/19/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024] Open
Abstract
OBJECTIVE Skeletal muscle plasticity and remodeling are critical for adapting tissue function to use, disuse, and regeneration. The aim of this study was to identify genes and molecular pathways that regulate the transition from atrophy to compensatory hypertrophy or recovery from injury. Here, we have used a mouse model of hindlimb unloading and reloading, which causes skeletal muscle atrophy, and compensatory regeneration and hypertrophy, respectively. METHODS We analyzed mouse skeletal muscle at the transition from hindlimb unloading to reloading for changes in transcriptome and extracellular fluid proteome. We then used qRT-PCR, immunohistochemistry, and bulk and single-cell RNA sequencing data to determine Mustn1 gene and protein expression, including changes in gene expression in mouse and human skeletal muscle with different challenges such as exercise and muscle injury. We generated Mustn1-deficient genetic mouse models and characterized them in vivo and ex vivo with regard to muscle function and whole-body metabolism. We isolated smooth muscle cells and functionally characterized them, and performed transcriptomics and proteomics analysis of skeletal muscle and aorta of Mustn1-deficient mice. RESULTS We show that Mustn1 (Musculoskeletal embryonic nuclear protein 1, also known as Mustang) is highly expressed in skeletal muscle during the early stages of hindlimb reloading. Mustn1 expression is transiently elevated in mouse and human skeletal muscle in response to intense exercise, resistance exercise, or injury. We find that Mustn1 expression is highest in smooth muscle-rich tissues, followed by skeletal muscle fibers. Muscle from heterozygous Mustn1-deficient mice exhibit differences in gene expression related to extracellular matrix and cell adhesion, compared to wild-type littermates. Mustn1-deficient mice have normal muscle and aorta function and whole-body glucose metabolism. We show that Mustn1 is secreted from smooth muscle cells, and that it is present in arterioles of the muscle microvasculature and in muscle extracellular fluid, particularly during the hindlimb reloading phase. Proteomics analysis of muscle from Mustn1-deficient mice confirms differences in extracellular matrix composition, and female mice display higher collagen content after chemically induced muscle injury compared to wild-type littermates. CONCLUSIONS We show that, in addition to its previously reported intracellular localization, Mustn1 is a microprotein secreted from smooth muscle cells into the muscle extracellular space. We explore its role in muscle ECM deposition and remodeling in homeostasis and upon muscle injury. The role of Mustn1 in fibrosis and immune infiltration upon muscle injury and dystrophies remains to be investigated, as does its potential for therapeutic interventions.
Collapse
Affiliation(s)
- Serge Ducommun
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Paulo R Jannig
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Igor Cervenka
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Marta Murgia
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B, 35131 Padua, Italy; Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Melanie J Mittenbühler
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ekaterina Chernogubova
- Department of Medicine, Cardiovascular Unit, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - José M Dias
- Department of Cell and Molecular Biology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden; Nanomedicine and Spatial Biology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Baptiste Jude
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jorge C Correia
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Gabriel Ocana-Santero
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Margareta Porsmyr-Palmertz
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sarah McCann Haworth
- Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Vicente Martínez-Redondo
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Zhengye Liu
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Matthias Mann
- Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Johanna T Lanner
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ana I Teixeira
- Nanomedicine and Spatial Biology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Lars Maegdefessel
- Department of Medicine, Cardiovascular Unit, Karolinska Institutet, 171 77 Stockholm, Sweden; Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; German Center for Cardiovascular Research DZHK, Partner Site Munich Heart Alliance, 10785 Berlin, Germany
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Jorge L Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Pharmacology and Stanley and Judith Frankel Institute for Heart & Brain Health, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
46
|
Teng H, Zheng J, Liang Y, Zhao J, Yan Y, Li S, Li S, Tong H. Podocan promoting skeletal muscle post-injury regeneration by inhibiting TGF-β signaling pathway. FASEB J 2024; 38:e23502. [PMID: 38430223 DOI: 10.1096/fj.202302158rr] [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: 10/24/2023] [Revised: 01/20/2024] [Accepted: 02/06/2024] [Indexed: 03/03/2024]
Abstract
Podocan, the fifth member of Small Leucine-Rich Proteoglycan (SLRP) family of extracellular matrix components, is poorly known in muscle development. Previous studies have shown that Podocan promotes C2C12 differentiation in mice. In this study, we elucidated the effect of Podocan on skeletal muscle post-injury regeneration and its underlying mechanism. Injection of Podocan protein promoted the process of mice skeletal muscle post-injury regeneration. This effect seemed to be from the acceleration of muscle satellite cell differentiation in vivo. Meanwhile, Podocan promoted myogenic differentiation in vitro by binding with TGF-β1 to inhibit the activity of the TGF-β signaling pathway. These results indicated that Podocan had the potential roles to enhance skeletal muscle post-injury regeneration. Its mechanism is likely the regulation of the expression of p-Smad2 and p-Smad4 related to the TGF-β signaling pathway by interacting with TGF-β1.
Collapse
Affiliation(s)
- Huaixin Teng
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Jingxian Zheng
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Yanyan Liang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Jingwen Zhao
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Yunqin Yan
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Shufeng Li
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Shuang Li
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Huili Tong
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| |
Collapse
|
47
|
Cheng M, Nie Y, Song M, Chen F, Yu Y. Forkhead box O proteins: steering the course of stem cell fate. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:7. [PMID: 38466341 DOI: 10.1186/s13619-024-00190-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/26/2024] [Indexed: 03/13/2024]
Abstract
Stem cells are pivotal players in the intricate dance of embryonic development, tissue maintenance, and regeneration. Their behavior is delicately balanced between maintaining their pluripotency and differentiating as needed. Disruptions in this balance can lead to a spectrum of diseases, underscoring the importance of unraveling the complex molecular mechanisms that govern stem cell fate. Forkhead box O (FOXO) proteins, a family of transcription factors, are at the heart of this intricate regulation, influencing a myriad of cellular processes such as survival, metabolism, and DNA repair. Their multifaceted role in steering the destiny of stem cells is evident, as they wield influence over self-renewal, quiescence, and lineage-specific differentiation in both embryonic and adult stem cells. This review delves into the structural and regulatory intricacies of FOXO transcription factors, shedding light on their pivotal roles in shaping the fate of stem cells. By providing insights into the specific functions of FOXO in determining stem cell fate, this review aims to pave the way for targeted interventions that could modulate stem cell behavior and potentially revolutionize the treatment and prevention of diseases.
Collapse
Affiliation(s)
- Mengdi Cheng
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Yujie Nie
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Min Song
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Fulin Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Yuan Yu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China.
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
48
|
Zheng G, Li C, Chen X, Deng Z, Xie T, Huo Z, Wei X, Huang Y, Zeng X, Luo Y, Bai J. HDAC9 inhibition reduces skeletal muscle atrophy and enhances regeneration in mice with cigarette smoke-induced COPD. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167023. [PMID: 38218381 DOI: 10.1016/j.bbadis.2024.167023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Cigarette smoke (CS) is the major risk factor for chronic obstructive pulmonary disease (COPD), and sarcopenia is one of the significant comorbidities of COPD. However, the pathogenesis of CS-related deficient skeletal muscle regeneration has yet to be clarified. The impact of CS on myoblast differentiation was examined, and then we determined which HDAC influenced the myogenic process and muscle atrophy in vitro and in vivo. Finally, we further investigated the potential mechanisms via RNA sequencing. Long-term CS exposure activated skeletal muscle primary satellite cells (SCs) while inhibiting differentiation, and defective myogenesis was also observed in C2C12 cells treated with CS extract (CSE). The level of HDAC9 changed in vitro and in vivo in CS exposure models as well as COPD patients, as detected by bioinformatics analysis. Our data showed that CSE impaired myogenic capacity and myotube formation in C2C12 cells via HDAC9. Moreover, inhibition of HDAC9 in mice exposed to CS prevented skeletal muscle dysfunction and promoted SC differentiation. The results of RNA-Seq analysis and verification indicated that HDAC9 knockout improved muscle differentiation in CS-exposed mice, probably by acting on the AKT/mTOR pathway and inhibiting the P53/P21 pathway. More importantly, the serum of HDAC9 KO mice exposed to CS alleviated the differentiation impairment of C2C12 cells caused by serum intervention in CS-exposed mice, and this effect was inhibited by LY294002 (an AKT/mTOR pathway inhibitor). These results suggest that HDAC9 plays an essential role in the defective regeneration induced by chronic exposure to CS.
Collapse
Affiliation(s)
- Guixian Zheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Chao Li
- Department of Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, Hunan 410219, China
| | - Xiaoli Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhaohui Deng
- Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Zhuzhou, Hunan 412000, China
| | - Ting Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zengyu Huo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xinyan Wei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yanbing Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xia Zeng
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, China
| | - Yu Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, China
| | - Jing Bai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
49
|
Wan R, Liu S, Feng X, Luo W, Zhang H, Wu Y, Chen S, Shang X. The Revolution of exosomes: From biological functions to therapeutic applications in skeletal muscle diseases. J Orthop Translat 2024; 45:132-139. [PMID: 38544740 PMCID: PMC10966453 DOI: 10.1016/j.jot.2024.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 11/11/2024] Open
Abstract
Skeletal muscle diseases, a broad category encompassing a myriad of afflictions such as acute muscle injury and muscular dystrophies, pose a significant health burden globally. These conditions often lead to muscle weakness, compromised mobility, and a diminished quality of life. In light of this, innovative and effective therapeutic strategies are fervently sought after. Exosomes, naturally extracellular vesicles with a diameter of 30-150 nm, pervade biological fluids. These microscopic entities harbor a host of biological molecules, including proteins, nucleic acids, and lipids, bearing a significant resemblance to their parent cells. The roles they play in the biological theater are manifold, influencing crucial physiological and pathological processes within the organism. In the context of skeletal muscle diseases, their potential extends beyond these roles, as they present a promising therapeutic target and a vehicle for targeted drug delivery. This potentially paves the way for significant clinical applications. This review aims to elucidate the mechanisms underpinning exosome action, their myriad biological functions, and the strides made in exosome research and application. A comprehensive exploration of the part played by exosomes in skeletal muscle repair and regeneration is undertaken. In addition, we delve into the use of exosomes in the therapeutic landscape of skeletal muscle diseases, providing a valuable reference for a deeper understanding of exosome applications in this realm. The concluding section encapsulates the prospective avenues for exosome research and the promising future they hold, underscoring the tremendous potential these diminutive vesicles possess in the field of skeletal muscle diseases. The Translational Potential of this Article. The comprehensive exploration of exosome's diverse biological functions and translational potential in the context of skeletal muscle diseases presented in this review underscores their promising future as a therapeutic target with significant clinical applications, thus paving the way for innovative and effective therapeutic strategies in this realm.
Collapse
Affiliation(s)
- Renwen Wan
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shan Liu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xinting Feng
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wei Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Hanli Zhang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yang Wu
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiliang Shang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|
50
|
Yadava RS, Mandal M, Mahadevan MS. Studying the Effect of MBNL1 and MBNL2 Loss in Skeletal Muscle Regeneration. Int J Mol Sci 2024; 25:2687. [PMID: 38473933 PMCID: PMC10931579 DOI: 10.3390/ijms25052687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Loss of function of members of the muscleblind-like (MBNL) family of RNA binding proteins has been shown to play a key role in the spliceopathy of RNA toxicity in myotonic dystrophy type 1 (DM1), the most common muscular dystrophy affecting adults and children. MBNL1 and MBNL2 are the most abundantly expressed members in skeletal muscle. A key aspect of DM1 is poor muscle regeneration and repair, leading to dystrophy. We used a BaCl2-induced damage model of muscle injury to study regeneration and effects on skeletal muscle satellite cells (MuSCs) in Mbnl1∆E3/∆E3 and Mbnl2∆E2/∆E2 knockout mice. Similar experiments have previously shown deleterious effects on these parameters in mouse models of RNA toxicity. Muscle regeneration in Mbnl1 and Mbnl2 knockout mice progressed normally with no obvious deleterious effects on MuSC numbers or increased expression of markers of fibrosis. Skeletal muscles in Mbnl1∆E3/∆E3/ Mbnl2∆E2/+ mice showed increased histopathology but no deleterious reductions in MuSC numbers and only a slight increase in collagen deposition. These results suggest that factors beyond the loss of MBNL1/MBNL2 and the associated spliceopathy are likely to play a key role in the defects in skeletal muscle regeneration and deleterious effects on MuSCs that are seen in mouse models of RNA toxicity due to expanded CUG repeats.
Collapse
Affiliation(s)
| | | | - Mani S. Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA; (R.S.Y.)
| |
Collapse
|