1
|
Chi J, Fan B, Li Y, Jiao Q, Li GY. Mitochondrial transplantation: a promising strategy for the treatment of retinal degenerative diseases. Neural Regen Res 2025; 20:3370-3387. [PMID: 39851134 PMCID: PMC11974652 DOI: 10.4103/nrr.nrr-d-24-00851] [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: 07/30/2024] [Revised: 09/11/2024] [Accepted: 10/24/2024] [Indexed: 01/26/2025] Open
Abstract
The retina, a crucial neural tissue, is responsible for transforming light signals into visual information, a process that necessitates a significant amount of energy. Mitochondria, the primary powerhouses of the cell, play an integral role in retinal physiology by fulfilling the high-energy requirements of photoreceptors and secondary neurons through oxidative phosphorylation. In a healthy state, mitochondria ensure proper visual function by facilitating efficient conversion and transduction of visual signals. However, in retinal degenerative diseases, mitochondrial dysfunction significantly contributes to disease progression, involving a decline in membrane potential, the occurrence of DNA mutations, increased oxidative stress, and imbalances in quality-control mechanisms. These abnormalities lead to an inadequate energy supply, the exacerbation of oxidative damage, and the activation of cell death pathways, ultimately resulting in neuronal injury and dysfunction in the retina. Mitochondrial transplantation has emerged as a promising strategy for addressing these challenges. This procedure aims to restore metabolic activity and function in compromised cells through the introduction of healthy mitochondria, thereby enhancing the cellular energy production capacity and offering new strategies for the treatment of retinal degenerative diseases. Although mitochondrial transplantation presents operational and safety challenges that require further investigation, it has demonstrated potential for reviving the vitality of retinal neurons. This review offers a comprehensive examination of the principles and techniques underlying mitochondrial transplantation and its prospects for application in retinal degenerative diseases, while also delving into the associated technical and safety challenges, thereby providing references and insights for future research and treatment.
Collapse
Affiliation(s)
- Jing Chi
- Department of Ophthalmology, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Bin Fan
- Department of Ophthalmology, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yulin Li
- Department of Ophthalmology, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qing Jiao
- Department of Ophthalmology, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Guang-Yu Li
- Department of Ophthalmology, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
2
|
Deschenes MR, Rackley M, Fernandez S, Heidebrecht M. Mature and Juvenile Neuromuscular Plasticity in Response to Unloading. Dev Neurobiol 2025; 85:e22966. [PMID: 40343402 PMCID: PMC12060605 DOI: 10.1002/dneu.22966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/05/2024] [Accepted: 04/04/2025] [Indexed: 05/11/2025]
Abstract
The neuromuscular junction (NMJ) is the synapse that enables the requisite electrical communication between the motor nervous system and the myofibers that respond to such electrical stimulation with movement and force development. Changes in an NMJ's normal activity pattern have been demonstrated to remodel both the synapse and the myofibers that comprise the NMJ. Significant amounts of research have been devoted to the study of aging on the neuromuscular system. Far less, however, has been focused on revealing the effects of reduced activity on the NMJ and myofibers comprising juvenile neuromuscular systems. In the present investigation, the consequences of decreased activity imposed by muscle unloading (UL) via hindlimb suspension for 2 weeks (a period known to induce muscle remodeling) were examined in both young adult, that is, mature (8 mo), and juvenile (3 mo) neuromuscular systems. In total, 4 treatment groups comprised of 10 animals (Juvenile-Control, Juvenile-Unloaded, Mature-Control, and Mature-Unloaded) were studied. Immunofluorescent procedures, coupled with confocal microscopy, were used to quantify remodeling of both the pre- and postsynaptic features of NMJs, as well as assessing the myofiber profiles of the soleus muscles housing the NMJs of interest. Results of ANOVA procedures revealed that there were significant (p < 0.05) main effects for both treatment, whereby UL consistently led to expanded size of the NMJ, and Age where expanded NMJ dimensions were consistently linked with mature compared to juvenile neuromuscular systems. Moreover, only sporadically was interaction between the main effects of Age and Treatment noted. Importantly, one variable that remained impressively resistant to the effects of both Age and Treatment was the critical parameter of pre- to postsynaptic coupling suggesting stability in effective communication at the NMJ throughout the lifespan and despite changes in activity patterns. The data presented here suggest that further inquiry must be performed regarding disuse-related plasticity of the neuromuscular system in adolescent individuals as those individuals regularly suffer injuries resulting in periods of muscle UL.
Collapse
Affiliation(s)
- Michael R. Deschenes
- Department of Kinesiology & Health SciencesCollege of William & MaryWilliamsburgVirginiaUSA
- Program in NeuroscienceCollege of William & MaryWilliamsburgVirginiaUSA
| | - Max Rackley
- Department of Kinesiology & Health SciencesCollege of William & MaryWilliamsburgVirginiaUSA
| | - Sophie Fernandez
- Department of Kinesiology & Health SciencesCollege of William & MaryWilliamsburgVirginiaUSA
| | - Megan Heidebrecht
- Department of Kinesiology & Health SciencesCollege of William & MaryWilliamsburgVirginiaUSA
| |
Collapse
|
3
|
Singh AK, Sudhan YG, Ramakrishna R, Durairajan SSK. Viral agents in neuromuscular pathology. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:397-434. [PMID: 40414639 DOI: 10.1016/bs.irn.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
In recent years, viral infections have been increasingly identified as major players in neuromuscular pathologies. This chapter presents an overview of the evidence and future directions for virus-induced neuromuscular disorders. Information is integrated on the global burden of these diseases related to epidemiology, clinical features, diagnosis, treatment, and preventive strategies was integrated. Responsible viruses include enteroviruses, flaviviruses, herpesviruses, and emerging pathogens such as SARS-CoV-2. It represents a broad spectrum of neuromuscular disorders, including Guillain-Barré syndrome, viral myositis, and critical illness neuropathy/myopathy. The book chapter discusses different diagnostic approaches, therapy strategies, and rehabilitation methods, in addition to early intervention and preventive measures. This has led to new insights into novel therapies, unmet research needs, and future perspectives on viral neuromuscular disorders. This chapter demonstrates that supporting both clinical care and patient management with clinical research entails a profound understanding of the difficult interactions between the viruses concerned and the neuromuscular system.
Collapse
Affiliation(s)
- Abhay Kumar Singh
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, India
| | - Yemgadda Goutham Sudhan
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, India
| | - Ramakrisha Ramakrishna
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, India
| | | |
Collapse
|
4
|
Xu D, Liu Q, Wang J, Yin E, Zhou B, Li X, Shi Y. Muscle-Derived Mitochondria as a Novel Therapy for Muscle Degeneration After Rotator Cuff Tears. J Bone Joint Surg Am 2025:00004623-990000000-01436. [PMID: 40279441 DOI: 10.2106/jbjs.24.01322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
BACKGROUND Rotator cuff tears (RCTs) commonly lead to muscle atrophy, fatty infiltration, and fibrosis, resulting in pain, weakness, and impaired shoulder mobility. These pathological changes are often irreversible and pose substantial treatment challenges. The aim of this study was to evaluate the therapeutic potential of muscle-derived mitochondria (Mito) in mitigating muscle degeneration and fibrosis following RCTs. METHODS Sprague Dawley rats were assigned to 3 groups: sham surgery, RCTs treated with Mito, or RCTs treated with phosphate-buffered saline solution (PBS). Following RCTs, in vivo Mito or PBS treatments were administered to the supraspinatus muscles (SSPs) of the rats immediately and then biweekly for 12 weeks. Data were collected on muscle morphology, fibrosis, fatty infiltration, oxidative stress, mitochondrial function, macrophage phenotypes, and serum inflammatory cytokines. In vitro experiments included mitochondria tracking in bone marrow-derived macrophages (BMDMs), characterization of macrophage polarization, and inflammatory cytokine profiling. RESULTS Isolated mitochondria preserved their morphology and function. Mito treatment improved muscle wet weight (p < 0.0001) and fiber cross-sectional area (p < 0.0001) while reducing fibrosis (p < 0.0001) and fatty infiltration (p < 0.0001). It upregulated mitochondrial markers cytochrome c oxidase (COX IV) and translocase of outer mitochondrial membrane 20 (TOMM20) (p < 0.0001) and enhanced antioxidative activity, as shown by increased superoxide dismutase (SOD) activity (p < 0.0001), elevated glutathione peroxidase (GSH-PX) levels (p = 0.038), and decreased malondialdehyde (MDA) levels (p = 0.0002). Mitochondrial density and morphology were restored in SSPs after Mito treatment. Additionally, Mito treatment induced an anti-inflammatory macrophage phenotype and reduced pro-inflammatory cytokines in vivo and in vitro. CONCLUSIONS Mito treatment mitigated muscle degeneration, improved mitochondrial function, and fostered an anti-inflammatory environment through macrophage modulation, demonstrating its potential as a cell-free therapeutic strategy for RCT-related muscle pathologies. CLINICAL RELEVANCE Although this is a preclinical study, its approach offers a novel avenue for improving RCT treatment outcomes. However, further validation in large animal models is needed to address the translational applicability of these findings, given the inherent regenerative capacity of rodent muscles.
Collapse
Affiliation(s)
- Ding Xu
- Department of Orthopedic Surgery, Ningbo No.6 Hospital, Ningbo, People's Republic of China
| | - Qing Liu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Jingzeng Wang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Enzhi Yin
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Biaohuan Zhou
- Department of Anorectal Surgery, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, and The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People's Republic of China
| | - Xiao Li
- Priority Medical Department, General Hospital of Central Theater Command, Wuhan, People's Republic of China
| | - Yulong Shi
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
5
|
Chen P, Jia F, Wang M, Yang S. Analysis of the mechanism of skeletal muscle atrophy from the pathway of decreased protein synthesis. Front Physiol 2025; 16:1533394. [PMID: 40352150 PMCID: PMC12061715 DOI: 10.3389/fphys.2025.1533394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/18/2025] [Indexed: 05/14/2025] Open
Abstract
Skeletal muscle atrophy is associated with denervation, cancer, diabetes, aging, immobilization, and inflammation, which can significantly impair mobility. It is primarily attributable to increased protein catabolism alongside reduced protein synthesis, although the precise mechanisms underlying this process are not yet fully known. Unlike in the pathway driving increased catabolism, fewer studies have explored the mechanism underpinning muscle atrophy under reduced protein synthesis. Therefore, this study aimed to focus on summarizing relevant studies on the reduction of protein synthesis leading to skeletal muscle atrophy, as driven by alterations in pathways such as the insulin-like growth factor-1-phosphatidylinositol 3-kinase-protein kinase B-rapamycin signaling pathway, glycogen synthase kinase-3, glucocorticoids, 5'-adenosine monophosphate-activated protein kinase, branched-chain amino acid sensors, myostatin, long-term proinflammatory factors, oxidative stress and mitochondrial dysfunction, calciumion concentration, activating transcription factor 4, and glycyl-tRNA synthetase alterations. Consolidating these data will provide a foundation and theoretical basis for further investigation into the mechanisms of muscle atrophy from the perspective of reduced protein synthesis pathways.
Collapse
Affiliation(s)
| | | | | | - Shengbo Yang
- Department of Anatomy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
6
|
Ma S, Lu Y, Sui S, Yang JS, Fu BB, Tan PX, Chai Y, Lv J, Kong L, Wu X, Gao YB, Yan T. Unraveling the triad of immunotherapy, tumor microenvironment, and skeletal muscle biomechanics in oncology. Front Immunol 2025; 16:1572821. [PMID: 40242775 PMCID: PMC12000078 DOI: 10.3389/fimmu.2025.1572821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 04/18/2025] Open
Abstract
The intricate interaction between skeletal muscle biomechanics, the tumor microenvironment, and immunotherapy constitutes a pivotal research focus oncology. This work provides a comprehensive review of methodologies for evaluating skeletal muscle biomechanics, including handheld dynamometry, advanced imaging techniques, electrical impedance myography, elastography, and single-fiber experiments to assess muscle quality and performance. Furthermore, it elucidates the mechanisms, applications, and limitations of various immunotherapy modalities, including immune checkpoint inhibitors, adoptive cell therapy, cancer vaccines, and combined chemoimmunotherapy, while examining their effects on skeletal muscle function and systemic immune responses. Key findings indicate that although immunotherapy is effective in augmenting antitumor immunity, it frequently induces muscle-related adverse effects such as weakness, fatigue, or damage, primarily mediated by cytokine release and immune activation. This work underscores the significance of immune niches within the tumor microenvironment in influencing treatment outcomes and proposes strategies to optimize therapy through personalized regimens and combinatorial approaches. This review highlights the need for further research on the formation of immune niches and interactions muscle-tumor. Our work is crucial for advancing the efficacy of immunotherapy, reducing adverse effects, and ultimately improving survival rates and quality of life of patients with cancer.
Collapse
Affiliation(s)
- Shuang Ma
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Ying Lu
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Shang Sui
- St. John’s Kilmarnock School, Breslau, ON, Canada
| | - Jia-shuo Yang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing-bing Fu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei-xin Tan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Chai
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Jiaqi Lv
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Lingyu Kong
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Xiaolin Wu
- School of Mathematics and Statistics, Liaoning University, Shenyang, China
| | - Yi-bo Gao
- Department of Oral and Maxillofacial Surgery, Taikang Bybo Dental, Beijing, China
| | - Tao Yan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Liu H, Wang K, Shang T, Cai Z, Lu C, Shen M, Yu S, Yao X, Shen Y, Chen X, Xu F, Sun H. Astragaloside IV Improves Muscle Atrophy by Modulating the Activity of UPS and ALP via Suppressing Oxidative Stress and Inflammation in Denervated Mice. Mol Neurobiol 2025; 62:4689-4704. [PMID: 39480556 DOI: 10.1007/s12035-024-04590-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Peripheral nerve injury is common clinically and can lead to neuronal degeneration and atrophy and fibrosis of the target muscle. The molecular mechanisms of muscle atrophy induced by denervation are complex and not fully understood. Inflammation and oxidative stress play an important triggering role in denervated muscle atrophy. Astragaloside IV (ASIV), a monomeric compound purified from astragalus membranaceus, has antioxidant and anti-inflammatory properties. The aim of this study was to investigate the effect of ASIV on denervated muscle atrophy and its molecular mechanism, so as to provide a new potential therapeutic target for the prevention and treatment of denervated muscle atrophy. In this study, an ICR mouse model of muscle atrophy was generated through sciatic nerve dissection. We found that ASIV significantly inhibited the reduction of tibialis anterior muscle mass and muscle fiber cross-sectional area in denervated mice, reducing ROS and oxidative stress-related protein levels. Furthermore, ASIV inhibits the increase in inflammation-associated proteins and infiltration of inflammatory cells, protecting the denervated microvessels in skeletal muscle. We also found that ASIV reduced the expression levels of MAFbx, MuRF1 and FoxO3a, while decreasing the expression levels of autophagy-related proteins, it inhibited the activation of ubiquitin-proteasome and autophagy-lysosome hydrolysis systems and the slow-to-fast myofiber shift. Our results show that ASIV inhibits oxidative stress and inflammatory responses in skeletal muscle due to denervation, inhibits mitophagy and proteolysis, improves microvascular circulation and reverses the transition of muscle fiber types; Therefore, the process of skeletal muscle atrophy caused by denervation can be effectively delayed.
Collapse
Affiliation(s)
- Hua Liu
- 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, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, Nantong, Jiangsu Province, 226600, P. R. China
| | - Kexin Wang
- 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, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Tongxin Shang
- 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, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Zhigang Cai
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, Nantong, Jiangsu Province, 226600, P. R. China
| | - Chunfeng Lu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, Jiangsu Province, 226006, P. R. China
| | - Mi 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, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Shu Yu
- 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, 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, 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, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Xiaofang Chen
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, Nantong, Jiangsu Province, 226600, P. R. China.
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, Jiangsu Province, 226006, 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, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China.
| |
Collapse
|
8
|
Eun SY, Lee CH, Cheon YH, Chung CH, Lee MS, Kim JY. Dual Action of Pueraria montana var. lobata Extract on Myogenesis and Muscle Atrophy. Nutrients 2025; 17:1217. [PMID: 40218975 PMCID: PMC11990788 DOI: 10.3390/nu17071217] [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: 03/14/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Muscle atrophy, defined by diminished muscle mass and function, is a notable concern associated with aging, disease, and glucocorticoid treatment. Pueraria montana var. lobata extract (PMLE) demonstrates multiple bioactive properties, such as antioxidant, anti-inflammatory, and metabolic regulatory activities; however, its role in muscle atrophy has not been extensively investigated to date. This study examined how PMLE influences both muscle cell differentiation and dexamethasone (DEX)-induced muscle degeneration by focusing on the underlying molecular mechanisms. Methods: This study examined the effects of PMLE on myogenic differentiation and DEX-induced muscle atrophy. C2C12 myoblasts were treated with PMLE (10-100 ng/mL) and assessed for changes in the expression of myogenesis-related genes and activation of Akt/mTOR and AMPK/SIRT1/PGC-1α signaling cascades. In vivo, a DEX-induced muscle atrophy model was used to assess muscle mass, fiber morphology, and molecular changes. Results: PMLE PMLE promoted muscle cell development by increasing the expression of MyHC, MyoD, and myogenin while activating protein synthesis and mitochondrial biogenesis pathways. PMLE counteracted DEX-induced myotube atrophy, restoring myotube diameter and promoting cellular fusion in vitro. In vivo, PMLE mitigated muscle degradation in fast-twitch muscle groups and reversed DEX-induced suppression of key anabolic and mitochondrial pathways. Conclusions: These findings suggest that PMLE promotes myogenic differentiation and protects against muscle atrophy by regulating critical molecular pathways, indicating its promise as a treatment candidate for conditions involving muscle wasting. Further studies are required to assess its clinical application and long-term safety efficacy.
Collapse
Affiliation(s)
- So Young Eun
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea; (S.Y.E.); (C.H.L.); (Y.-H.C.); (C.H.C.)
- Department of Pharmacology, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea
| | - Chang Hoon Lee
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea; (S.Y.E.); (C.H.L.); (Y.-H.C.); (C.H.C.)
- Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, 460 Iksandae-ro, Iksan 54538, Republic of Korea
| | - Yoon-Hee Cheon
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea; (S.Y.E.); (C.H.L.); (Y.-H.C.); (C.H.C.)
| | - Chong Hyuk Chung
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea; (S.Y.E.); (C.H.L.); (Y.-H.C.); (C.H.C.)
- Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, 460 Iksandae-ro, Iksan 54538, Republic of Korea
| | - Myeung Su Lee
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea; (S.Y.E.); (C.H.L.); (Y.-H.C.); (C.H.C.)
- Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, 460 Iksandae-ro, Iksan 54538, Republic of Korea
| | - Ju-Young Kim
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea; (S.Y.E.); (C.H.L.); (Y.-H.C.); (C.H.C.)
| |
Collapse
|
9
|
Zhou X, Li S, Wang L, Wang J, Zhang P, Chen X. The emerging role of exercise preconditioning in preventing skeletal muscle atrophy. Front Physiol 2025; 16:1559594. [PMID: 40206380 PMCID: PMC11979144 DOI: 10.3389/fphys.2025.1559594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Skeletal muscle atrophy, characterized by the loss of muscle mass and function, can result from disuse, aging, disease, drug. Exercise preconditioning-a form of exercise training performed before these harmful threats-induces notable remodeling and extensive biochemical adaptations in skeletal muscle, creating a protective phenotype in muscle fibers, and thus serving as an effective intervention for preventing skeletal muscle atrophy. Here, we review the current understanding relating to how exercise preconditioning protects skeletal muscle from damage caused by inactivity, sarcopenia, disease, or pharmacological intervention, with an emphasis on the cellular mechanisms involved. Key mechanisms highlighted as making a significant contribution to the protective effects of exercise on skeletal muscle fibers include mitochondria; the expression of cytoprotective proteins such as HSP72, SOD2, SESN2, PGC-1α and AMPK; and the regulation of oxidative stress. These findings underscore the potential of exercise preconditioning as a non-pharmacological intervention for preserving muscle mass and function, as well as preventing muscular atrophy, ultimately improving the quality of life for at-risk populations.
Collapse
Affiliation(s)
- Xu Zhou
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Shiming Li
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Lu Wang
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Jun Wang
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Peng Zhang
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoping Chen
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
10
|
Gao X, Chen Y, Wang J, Xu J, Wan H, Li X, Shi Y. Mitochondria-Rich Extracellular Vesicles From Bone Marrow Stem Cells Mitigate Muscle Degeneration in Rotator Cuff Tears in a Rat Model Through Macrophage M2 Phenotype Conversion. Arthroscopy 2025:S0749-8063(25)00229-4. [PMID: 40147598 DOI: 10.1016/j.arthro.2025.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Abstract
PURPOSE To investigate the protective effects of extracellular vesicles derived from bone marrow stem cells (BMSC-EVs) on muscle degeneration in a rat model of rotator cuff tendon and suprascapular nerve (SSN) transection (termed the RCT-SSN model), focusing on mitochondrial transfer. METHODS The EVs were identified and characterized. The RCT-SSN model was established by transecting the supraspinatus, infraspinatus tendons, and suprascapular nerve. Ninety-six rats were divided into 4 groups (n = 24 each): sham surgery, RCT-SSN treated with BMSC-EVs, RCT-SSN treated with EVs from rhodamine 6G-pretreated BMSCs (Rho-EVs), or phosphate-buffered saline. Intramuscular injections were administered every 2 weeks. After 12 weeks, supraspinatus muscles were analyzed for atrophy, fibrosis, oxidative stress, macrophage phenotypes, serum cytokines, and mitochondrial characteristics. In vitro experiments included EVs tracking in macrophages, macrophage phenotype characterization, and inflammatory cytokine profiling. RESULTS BMSC-EVs and Rho-EVs displayed similar morphology, but only BMSC-EVs contained functional mitochondria. BMSC-EVs significantly reduced muscle weight loss (0.047 ± 0.010% vs 0.145 ± 0.013%, P < .001), increased muscle fiber cross-sectional area (2037 ± 231.9 μm2 vs 527.9 ± 92.01 μm2, P < .001), and decreased fibrosis (12.09 ± 3.31% vs 25.69 ± 4.84%, P < .001) compared with phosphate-buffered saline. BMSC-EVs enhanced superoxide dismutase activity (93.3 ± 11.8 U/mg protein vs 53.4 ± 8.0 U/mg protein, P < .001), improved mitochondrial function, density and structure, and induced an anti-inflammatory macrophage shift, suppressing proinflammatory cytokines in vitro and in vivo. Rho-EVs showed no such effects. CONCLUSIONS This study showed that transecting the supraspinatus, infraspinatus tendons, and suprascapular nerve in a rat model induced muscle degeneration and fibrosis. BMSC-EVs, but not Rho-EVs, mitigated these effects by promoting an anti-inflammatory macrophage phenotype and protecting mitochondrial function through mitochondrial transfer. CLINICAL RELEVANCE Mitochondrial transfer via BMSC-EVs may offer a therapeutic strategy to prevent muscle degeneration in patients with rotator cuff tear.
Collapse
Affiliation(s)
- Xing Gao
- Animal Experimental Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuanyuan Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China
| | - Jingzeng Wang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Xu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hu Wan
- Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiao Li
- Priority Medical Department, General Hospital of Central Theater command, Wuhan, China
| | - Yulong Shi
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
11
|
Dong M, Maturana AD. Effects of aging on calcium channels in skeletal muscle. Front Mol Biosci 2025; 12:1558456. [PMID: 40177518 PMCID: PMC11961898 DOI: 10.3389/fmolb.2025.1558456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
In skeletal muscle, calcium is not only essential to stimulate and sustain their contractions but also for muscle embryogenesis, regeneration, energy production in mitochondria, and fusion. Different ion channels contribute to achieving the various functions of calcium in skeletal muscles. Muscle contraction is initiated by releasing calcium from the sarcoplasmic reticulum through the ryanodine receptor channels gated mechanically by four dihydropyridine receptors of T-tubules. The calcium influx through store-operated calcium channels sustains the contraction and stimulates muscle regeneration. Mitochondrial calcium uniporter allows the calcium entry into mitochondria to stimulate oxidative phosphorylation. Aging alters the expression and activity of these different calcium channels, resulting in a reduction of skeletal muscle force generation and regeneration capacity. Regular physical training and bioactive molecules from nutrients can prevent the effects of aging on calcium channels. This review focuses on the current knowledge of the effects of aging on skeletal muscles' calcium channels.
Collapse
Affiliation(s)
| | - Andrés Daniel Maturana
- Department of Applied Biosciences, Graduate School of Bioagricultural Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
12
|
Cho HB, Kim H, Lee S, Cho CW, Park J, Youn S, So G, Kang S, Kim HJ, Park K. Near Infrared-Mediated Intracellular NADH Delivery Strengthens Mitochondrial Function and Stability in Muscle Dysfunction Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415303. [PMID: 39887582 PMCID: PMC11948086 DOI: 10.1002/advs.202415303] [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: 11/20/2024] [Revised: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Mitochondrial transfer emerges as a promising therapy for the restoration of mitochondrial function in damaged cells, mainly due to its limited immunogenicity. However, isolated mitochondria rapidly lose function because they produce little energy outside cells. Therefore, this study investigates whether near infrared (NIR)-mediated nicotinamide adenine dinucleotide (NADH) pre-treatment enhances mitochondrial function and stability in mitochondria-donor cells prior to transplantation. Clinical applications of NADH, an essential electron donor in the oxidative phosphorylation process, are restricted due to the limited cellular uptake of NADH. To address this, a photo-mediated method optimizes direct NADH delivery into cells and increases NADH absorption. L6 cells treated with NADH and irradiated with NIR enhanced NADH uptake, significantly improving mitochondrial energy production and function. Importantly, the improved functional characteristics of the mitochondria are maintained even after isolation from cells. Primed mitochondria, i.e., those enhanced by NIR-mediated NADH uptake (P-MT), are encapsulated in fusogenic liposomes and delivered into muscle cells with mitochondrial dysfunction. Compared to conventional mitochondria, P-MT mitochondria promote greater mitochondrial recovery and muscle regeneration. These findings suggest that NIR-mediated NADH delivery is an effective strategy for improving mitochondrial function, and has the potential to lead to novel treatments for mitochondrial disorders and muscle degeneration.
Collapse
Affiliation(s)
- Hui Bang Cho
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Hye‐Ryoung Kim
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Sujeong Lee
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Chae Won Cho
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Ji‐In Park
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Seulki Youn
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Gyuwon So
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Sumin Kang
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Hye Jin Kim
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Keun‐Hong Park
- Department of Nano‐regenerative Medical EngineeringCollege of Life ScienceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| |
Collapse
|
13
|
Wang X, Gao X, Deng C, Xu D, Chen Y, Huang J, Li X, Shi Y. Platelet-derived mitochondria attenuate muscle atrophy following rotator cuff tears in a rat model. J Shoulder Elbow Surg 2025:S1058-2746(25)00172-7. [PMID: 39986534 DOI: 10.1016/j.jse.2025.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Rotator cuff tears (RCTs) often result in muscle atrophy, compromising surgical outcomes and recovery. Mitochondrial dysfunction is implicated in this process, suggesting potential for mitochondria-based therapies. This study aimed to investigate the effects of platelet-derived mitochondria (Plt-Mito) administration into the supraspinatus muscle (SSP) following RCTs. METHODS Seventy-two male Sprague-Dawley rats were allocated into 3 distinct groups: (1) a sham surgery group, (2) a group with RCTs treated with Plt-Mito, and (3) a group with RCTs treated with phosphate-buffered saline. Treatments were administered every 2 weeks. After 12 weeks, the SSPs were analyzed for wet muscle weight ratio, muscle fiber cross-sectional area, fibrosis, antioxidant activity, mitochondrial markers, capillary density, and mitochondrial structure. RESULTS Plt-Mito successfully incorporated into SSP, maintaining functional integrity. Compared to the phosphate-buffered saline group, Plt-Mito treatment significantly preserved wet muscle weight, increased mean muscle fiber cross-sectional area, promoted muscle regeneration, reduced fibrosis, enhanced antioxidant activity (increased superoxide dismutase activity and decreased malondialdehyde activity), improved muscle vascularity (increased platelet endothelial cell adhesion molecule-1 and α-smooth muscle actin), increased expression of mitochondrial markers (C oxidase subunit IV and uncoupling protein 1) and maintained mitochondrial density and structure. CONCLUSIONS Our findings demonstrated Plt-Mito administration effectively halted muscle atrophy and fibrosis, while attenuating mitochondrial damage and dysfunction following RCTs.
Collapse
Affiliation(s)
- Xin Wang
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xing Gao
- Animal Experimental Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Xu
- Department of Orthopedic Surgery, Ningbo NO.6 Hospital, Ningbo, China
| | - Yuanyuan Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China
| | - Jiaqi Huang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China
| | - Xiao Li
- Priority Medical Department, General Hospital of Central Theater Command, Wuhan, China.
| | - Yulong Shi
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
14
|
Kim D, Shin Y, Baek YW, Kang H, Lim J, Bae ON. The effect of biocide chloromethylisothiazolinone/methylisothiazolinone (CMIT/MIT) mixture on C2C12 muscle cell damage attributed to mitochondrial reactive oxygen species overproduction and autophagy activation. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:137-151. [PMID: 39446036 DOI: 10.1080/15287394.2024.2420083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
The mixture of 5-chloro-2-methyl-4-isothiazolin-3-one and 2-methyl-4-isothiazolin-3-one (CMIT/MIT) is a biocide widely used as a preservative in various commercial products. This biocide has also been used as an active ingredient in humidifier disinfectants in South Korea, resulting in serious health effects among users. Recent evidence suggests that the underlying mechanism of CMIT/MIT-initiated toxicity might be associated with defects in mitochondrial functions. The aim of this study was to utilize the C2C12 skeletal muscle model to investigate the effects of CMIT/MIT on mitochondrial function and relevant molecular pathways associated with skeletal muscle dysfunction. Data demonstrated that exposure to CMIT/MIT during myogenic differentiation induced significant mitochondrial excess production of reactive oxygen species (ROS) and a decrease in intracellular ATP levels. Notably, CMIT/MIT significantly inhibited mitochondrial oxidative phosphorylation (Oxphos) and reduced mitochondrial mass at a lower concentration than the biocide amount, which diminished the viability of myotubes. CMIT/MIT induced activation of autophagy flux and decreased protein expression levels of myosin heavy chain (MHC). Taken together, CMIT/MIT exposure produced damage in C2C12 myotubes by impairing mitochondrial bioenergetics and activating autophagy. Our findings contribute to an increased understanding of the underlying mechanisms associated with CMIT/MIT-induced adverse skeletal muscle health effects.
Collapse
Affiliation(s)
- Donghyun Kim
- College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea
| | - Yusun Shin
- College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea
| | - Yong-Wook Baek
- Humidifier Disinfectant Health Center, Environmental Health Research, National Institute of Environmental Research, Incheon, Republic of Korea
| | - HanGoo Kang
- Humidifier Disinfectant Health Center, Environmental Health Research, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Jungyun Lim
- Humidifier Disinfectant Health Center, Environmental Health Research, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Ok-Nam Bae
- College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea
| |
Collapse
|
15
|
Huot JR, Jamnick NA, Pin F, Livingston PD, Callaway CS, Bonetto A. GL261 glioblastoma induces delayed body weight gain and stunted skeletal muscle growth in young mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.635159. [PMID: 39990490 PMCID: PMC11844426 DOI: 10.1101/2025.02.10.635159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Introduction The survival rate for children and adolescents has increased to over 85%. However, there is limited understanding of the impact of pediatric cancers on muscle development and physiology. Given that brain tumors alone account for 26% of all pediatric cancers, this study aimed to investigate the skeletal muscle consequences of tumor growth in young mice. Methods C2C12 myotubes were co-cultured with GL261 murine glioblastoma cells to assess myotube size. GL261 cells were then injected subcutaneously into 4-week-old male C57BL/6J mice. Animals were euthanized 28 days post-GL261 implantation. Muscle function was tested in vivo and ex vivo . Muscle protein synthesis was measured via the SUnSET method, and gene/protein expression levels were assessed via Western blotting and qPCR. Results In vitro , the C2C12 cultures exposed to GL261 exhibited myotube atrophy, consistent with a disrupted anabolic/catabolic balance. In vivo , carcass, heart, and fat mass were significantly reduced in the tumor-bearing mice. Skeletal muscle growth was impeded in the GL261 hosts, along with smaller muscle CSA. Both in vivo muscle torque and the ex vivo EDL muscle force were unchanged. At molecular level, the tumor hosts displayed reduced muscle protein synthesis and increased muscle protein ubiquitination, in disagreement with decreased muscle ubiquitin ligase mRNA expression. Conclusions Overall, we showed that GL261 tumors impact the growth of pediatric mice by stunting skeletal muscle development, decreasing muscle mass, reducing muscle fiber size, diminishing muscle protein synthesis, and altering protein catabolism signaling.
Collapse
|
16
|
Allen RJ, Kronemberger A, Shi Q, Pope M, Cuadra-Muñoz E, Son W, Song LS, Anderson EJ, Pereira RO, Lira VA. Altered relaxation and Mitochondria-Endoplasmic Reticulum Contacts Precede Major (Mal)adaptations in Aging Skeletal Muscle and are Prevented by Exercise. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.633043. [PMID: 39975407 PMCID: PMC11838400 DOI: 10.1101/2025.01.14.633043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Sarcopenia, or age-related muscle dysfunction, contributes to morbidity and mortality. Besides decreases in muscle force, sarcopenia is associated with atrophy and fast-to-slow fiber type switching, which is typically secondary to denervation in humans and rodents. However, very little is known about cellular changes preceding these important (mal)adaptations. To this matter, mitochondria and the sarcoplasmic reticulum are critical for tension generation in myofibers. They physically interact at the boundaries of sarcomeres forming subcellular hubs called mitochondria-endo/sarcoplasmic reticulum contacts (MERCs). Yet, whether changes at MERCs ultrastructure and proteome occur early in aging is unknown. Here, studying young adult and older mice we reveal that aging slows muscle relaxation leading to longer excitation-contraction-relaxation (ECR) cycles before maximal force decreases and fast-to-slow fiber switching takes place. We reveal that muscle MERC ultrastructure and mitochondria-associated ER membrane (MAM) protein composition are also affected early in aging and are closely associated with rate of muscle relaxation. Additionally, we demonstrate that regular exercise preserves muscle relaxation rate and MERC ultrastructure in early aging. Finally, we profile a set of muscle MAM proteins involved in energy metabolism, protein quality control, Ca2+ homeostasis, cytoskeleton integrity and redox balance that are inversely regulated early in aging and by exercise. These may represent new targets to preserve muscle function in aging individuals.
Collapse
Affiliation(s)
- Ryan J. Allen
- Department of Health and Human Physiology, Fraternal Order of Eagles Diabetes Research Center, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, USA
| | - Ana Kronemberger
- Department of Health and Human Physiology, Fraternal Order of Eagles Diabetes Research Center, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, USA
| | - Qian Shi
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Marshall Pope
- Proteomics Facility, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Elizabeth Cuadra-Muñoz
- Department of Health and Human Physiology, Fraternal Order of Eagles Diabetes Research Center, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, USA
| | - Wangkuk Son
- Department of Health and Human Physiology, Fraternal Order of Eagles Diabetes Research Center, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, USA
| | - Long-Sheng Song
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Ethan J. Anderson
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| | - Renata O. Pereira
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Vitor A. Lira
- Department of Health and Human Physiology, Fraternal Order of Eagles Diabetes Research Center, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
17
|
Xu H, Brown JL, Bhaskaran S, Van Remmen H. Reactive oxygen species in the pathogenesis of sarcopenia. Free Radic Biol Med 2025; 227:446-458. [PMID: 39613046 PMCID: PMC11816180 DOI: 10.1016/j.freeradbiomed.2024.11.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
One of the most critical factors impacting healthspan in the elderly is the loss of muscle mass and function, clinically referred to as sarcopenia. Muscle atrophy and weakness lead to loss of mobility, increased risk of injury, metabolic changes and loss of independence. Thus, defining the underlying mechanisms of sarcopenia is imperative to enable the development of effective interventions to preserve muscle function and quality in the elderly and improve healthspan. Over the past few decades, understanding the roles of mitochondrial dysfunction and oxidative stress has been a major focus of studies seeking to reveal critical molecular pathways impacted during aging. In this review, we will highlight how oxidative stress might contribute to sarcopenia by discussing the impact of oxidative stress on the loss of innervation and alteration in the neuromuscular junction (NMJ), on muscle mitochondrial function and atrophy pathways, and finally on muscle contractile function.
Collapse
Affiliation(s)
- Hongyang Xu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Jacob L Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States
| | - Shylesh Bhaskaran
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States.
| |
Collapse
|
18
|
Flaherty S, Song L, Albuquerque B, Rinaldi A, Piper M, Shanthappa D, Chen X, Stansfield J, Asano S, Pashos E, Ross T, Jagarlapudi S, Sheikh A, Zhang B, Wu Z. GDF15 Neutralization Ameliorates Muscle Atrophy and Exercise Intolerance in a Mouse Model of Mitochondrial Myopathy. J Cachexia Sarcopenia Muscle 2025; 16:e13715. [PMID: 39976232 PMCID: PMC11840706 DOI: 10.1002/jcsm.13715] [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: 12/21/2023] [Revised: 11/27/2024] [Accepted: 12/25/2024] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Primary mitochondrial myopathies (PMMs) are disorders caused by mutations in genes encoding mitochondrial proteins and proteins involved in mitochondrial function. PMMs are characterized by loss of muscle mass and strength as well as impaired exercise capacity. Growth/Differentiation Factor 15 (GDF15) was reported to be highly elevated in PMMs and cancer cachexia. Previous studies have shown that GDF15 neutralization is effective in improving skeletal muscle mass and function in cancer cachexia. It remains to be determined if the inhibition of GDF15 could be beneficial for PMMs. The purpose of the present study is to assess whether treatment with a GDF15 neutralizing antibody can alleviate muscle atrophy and physical performance impairment in a mouse model of PMM. METHODS The effects of GDF15 neutralization on PMM were assessed using PolgD257A/D257A (POLG) mice. These mice express a proofreading-deficient version of the mitochondrial DNA polymerase gamma, leading to an increased rate of mutations in mitochondrial DNA (mtDNA). These animals display increased circulating GDF15 levels, reduced muscle mass and function, exercise intolerance, and premature aging. Starting at 9 months of age, the mice were treated with an anti-GDF15 antibody (mAB2) once per week for 12 weeks. Body weight, food intake, body composition, and muscle mass were assessed. Muscle function and exercise capacity were evaluated using in vivo concentric max force stimulation assays, forced treadmill running and voluntary home-cage wheel running. Mechanistic investigations were performed via muscle histology, bulk transcriptomic analysis, RT-qPCR and western blotting. RESULTS Anti-GDF15 antibody treatment ameliorated the metabolic phenotypes of the POLG animals, improving body weight (+13% ± 8%, p < 0.0001), lean mass (+13% ± 15%, p < 0.001) and muscle mass (+35% ± 24%, p < 0.001). Additionally, the treatment improved skeletal muscle max force production (+35% ± 43%, p < 0.001) and exercise performance, including treadmill (+40% ± 29%, p < 0.05) and voluntary wheel running (+320% ± 19%, p < 0.05). Mechanistically, the beneficial effects of GDF15 neutralization are linked to the reversal of the transcriptional dysregulation in genes involved in autophagy and proteasome signalling. The treatment also appears to dampen glucocorticoid signalling by suppressing circulating corticosterone levels in the POLG animals. CONCLUSIONS Our findings highlight the potential of GDF15 neutralization with a monoclonal antibody as a therapeutic avenue to enhance physical performance and mitigate adverse clinical outcomes in patients with PMM.
Collapse
Affiliation(s)
- Stephen E. Flaherty
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Obesity and ComplicationsEli LillyBostonMassachusettsUSA
| | - LouJin Song
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Bina Albuquerque
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Anthony Rinaldi
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Program Mamager, Preclinical Sciences, ToxicologyVertex PharmaceuticalsBostonMassachusettsUSA
| | - Mary Piper
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | | | - Xian Chen
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - John Stansfield
- Biostatistics, Early Clinical DevelopmentPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Shoh Asano
- Inflammation and Immunology Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Evanthia Pashos
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Trenton Thomas Ross
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Srinath Jagarlapudi
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Abdul Sheikh
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Bei Zhang
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Zhidan Wu
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| |
Collapse
|
19
|
Qie B, Tuo J, Chen F, Ding H, Lyu L. Gene therapy for genetic diseases: challenges and future directions. MedComm (Beijing) 2025; 6:e70091. [PMID: 39949979 PMCID: PMC11822459 DOI: 10.1002/mco2.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Genetic diseases constitute the majority of rare human diseases, resulting from abnormalities in an individual's genetic composition. Traditional treatments offer limited relief for these challenging conditions. In contrast, the rapid advancement of gene therapy presents significant advantages by directly addressing the underlying causes of genetic diseases, thereby providing the potential for precision treatment and the possibility of curing these disorders. This review aims to delineate the mechanisms and outcomes of current gene therapy approaches in clinical applications across various genetic diseases affecting different body systems. Additionally, genetic muscular disorders will be examined as a case study to investigate innovative strategies of novel therapeutic approaches, including gene replacement, gene suppression, gene supplementation, and gene editing, along with their associated advantages and limitations at both clinical and preclinical levels. Finally, this review emphasizes the existing challenges of gene therapy, such as vector packaging limitations, immunotoxicity, therapy specificity, and the subcellular localization and immunogenicity of therapeutic cargos, while discussing potential optimization directions for future research. Achieving delivery specificity, as well as long-term effectiveness and safety, will be crucial for the future development of gene therapies targeting genetic diseases.
Collapse
Affiliation(s)
- Beibei Qie
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Jianghua Tuo
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Feilong Chen
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Haili Ding
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Lei Lyu
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| |
Collapse
|
20
|
Li X, Yao X, Zhao W, Wei B, Zhang R, Yan G, Ma M, Wang Z, Liu X, Liu Y, Wang G, Li H, Kong Q, Wang J, Mu L. Muscle fiber types switched during the development of experimental autoimmune myasthenia gravis via the PI3K/Akt signaling pathway. Mol Immunol 2025; 178:41-51. [PMID: 39832429 DOI: 10.1016/j.molimm.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/25/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
As one of the largest organs of our human body, skeletal muscle has good research prospects in myasthenia gravis (MG), the symptoms of which include systemic skeletal muscle weakness. Skeletal muscle is composed of two types of muscle fibers. Different fiber subtypes can be converted into each other; however, the underlying mechanism is not yet clear. In this paper, we firstly established an experimental autoimmune myasthenia gravis (EAMG) rat model and found that the skeletal muscle fibers of the EAMG group were atrophied, with a change in the proportion of fiber subtypes, which switched from type IIa to type I in the EAMG group at the peak stage, as verified by histological and molecular analyses. Second-generation sequencing results predicted that the PI3K-Akt signaling pathway might be involved in the switch, and the mRNA expression levels of the PI3K-Akt pathway-related genesNr4a1, IL2rb, Col1A1 and Ddit4 were significantly different. In conclusion, this study indicates that the switch of muscle fiber subtypes in MG via the PI3K-Akt signaling pathway may be a potential target for the treatment of MG-related skeletal muscle atrophy in the future.
Collapse
MESH Headings
- Animals
- Signal Transduction
- Proto-Oncogene Proteins c-akt/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Myasthenia Gravis, Autoimmune, Experimental/pathology
- Myasthenia Gravis, Autoimmune, Experimental/metabolism
- Myasthenia Gravis, Autoimmune, Experimental/immunology
- Rats
- Muscle Fibers, Skeletal/pathology
- Muscle Fibers, Skeletal/metabolism
- Female
- Muscle, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Rats, Inbred Lew
- Muscular Atrophy/pathology
Collapse
Affiliation(s)
- Xinrong Li
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Xiuhua Yao
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Wei Zhao
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Bo Wei
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Ran Zhang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Geng Yan
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Mingyu Ma
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Zhenhai Wang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Xijun Liu
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Yumei Liu
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Guangyou Wang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Qingfei Kong
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China
| | - Jinghua Wang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China; Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Lili Mu
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, School of Basic Medical Science, Harbin Medical University, Heilongjiang, China.
| |
Collapse
|
21
|
Song C, Zheng W, Liu G, Xu Y, Deng Z, Xiu Y, Zhang R, Yang L, Zhang Y, Yu G, Su Y, Luo J, He B, Xu J, Dai H. Sarcopenic obesity is attenuated by E-syt1 inhibition via improving skeletal muscle mitochondrial function. Redox Biol 2025; 79:103467. [PMID: 39675068 PMCID: PMC11699297 DOI: 10.1016/j.redox.2024.103467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024] Open
Abstract
In aging and metabolic disease, sarcopenic obesity (SO) correlates with intramuscular adipose tissue (IMAT). Using bioinformatics analysis, we found a potential target protein Extended Synaptotagmin 1 (E-syt1) in SO. To investigate the regulatory role of E-syt1 in muscle metabolism, we performed in vivo and in vitro experiments through E-syt1 loss- and gain-of-function on muscle physiology. When E-syt1 is overexpressed in vitro, myoblast proliferation, differentiation, mitochondrial respiration, biogenesis, and mitochondrial dynamics are impaired, which were alleviated by the silence of E-syt1. Furthermore, overexpression of E-syt1 inhibited mitophagic flux. Mechanistically, E-syt1 overexpression leads to mitochondrial calcium overload and mitochondrial ROS burst, inhibits the fusion of mitophagosomes with lysosomes, and impedes the acidification of lysosomes. Animal experiments demonstrated the inhibition of E-syt1 increased the capacity of endurance exercise, muscle mass, mitochondrial function, and oxidative capacity of the muscle fibers in OVX mice. These findings establish E-syt1 as a novel contributor to the pathogenesis of skeletal muscle metabolic disorders in SO. Consequently, targeting E-syt1-induced dysfunction may serve as a viable strategy for attenuating SO.
Collapse
Affiliation(s)
- Chao Song
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China; School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, 350001, China
| | - Wu Zheng
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Guoming Liu
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Yiyang Xu
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Zhibo Deng
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Yu Xiu
- Department of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Rongsheng Zhang
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Linhai Yang
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Yifei Zhang
- Department of Pediatrics, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Guoyu Yu
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Yibin Su
- Department of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Jun Luo
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China
| | - Bingwei He
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China; School of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, 350001, China.
| | - Jie Xu
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China.
| | - Hanhao Dai
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, 350001, China.
| |
Collapse
|
22
|
Wu ZJ, Li YC, Zheng Y, Zhou MQ, Li H, Wu SX, Zhao XY, Yang YH, Du L. Differential effects of EPA and DHA on aging-related sarcopenia in mice and possible mechanisms involved. Food Funct 2025; 16:601-616. [PMID: 39704327 DOI: 10.1039/d4fo04341c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Sarcopenia frequently occurs with aging and leads to major adverse impacts in elderly individuals. The protective effects of omega-3 polyunsaturated fatty acids against aging-related sarcopenia have been demonstrated; however, the effect and underlying mechanism of EPA or DHA alone remain inconclusive. Hence, the present study was aimed to clarify the differential effects and possible mechanisms of EPA and DHA on aging-related sarcopenia. In this study, two-month-old and eighteen-month-old male C57BL/6J mice were fed with an AIN-93M diet and an AIN-93M diet containing 1% EPA or 1% DHA for 24 weeks, respectively. The results revealed that EPA and DHA supplementation effectively alleviated the decline in grip strength, skeletal muscle mass, and myofiber cross-sectional areas in aged mice, with EPA exhibiting a better effect against aging-related sarcopenia than DHA. The ROS scavenging role of EPA in aged skeletal muscle was also superior to that of DHA. Additionally, EPA showed a stronger role in improving protein turnover and myogenesis in aged skeletal muscle, as evidenced by suppressing the activation of FoxO3a and NF-κB, blunting the expression levels of muscle atrophy markers MAFbx and MuRF1, activating the PI3K/Akt/mTOR signaling pathway, and elevating MyoD expression. Moreover, EPA also revealed a better effect on inhibiting mitochondria- and endoplasmic reticulum stress-mediated apoptosis in aged skeletal muscle. Furthermore, EPA manifested a more pronounced effect on improving mitochondrial damage of aged skeletal muscle than DHA, and the reason might be due to its superior capability of regulating mitochondrial quality control, as clearly shown by enhancing mitochondrial biogenesis through the AMPK/PGC-1α-dependent pathway, restraining the loss of mitochondrial fusion and fission proteins including Opa1, Mfn2, and Fis1, and promoting mitophagy via the PINK1/Parkin-dependent pathway.
Collapse
Affiliation(s)
- Zi-Jian Wu
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, No. 105 Jiefang Road, Jinan, Shandong, 250013, China.
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Ying-Chao Li
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Yan Zheng
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, No. 105 Jiefang Road, Jinan, Shandong, 250013, China
| | - Meng-Qing Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong, 250012, China
- Suzhou Centers for Diseases Prevention and Control, No. 498 Qingyunbei Road, Suzhou, Anhui, 234000, China
| | - Hui Li
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Shi-Xiang Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Xin-Yue Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Yu-Hong Yang
- School of Food Science and Engineering, Qilu University of Technology (Shandong Academy of Sciences), No. 3501 Daxue Road, Jinan, Shandong, 250353, China.
- Shandong Haizhibao Ocean Science and Technology Co., Ltd., No. 259 Pinghai East Road, Rongcheng City, Shandong, 264300, China
| | - Lei Du
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, No. 105 Jiefang Road, Jinan, Shandong, 250013, China.
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong, 250012, China
| |
Collapse
|
23
|
Yao H, Xie Q, Yang Y, Zhou C, Zeng Z, Zhang W. Melatonin attenuates sepsis-induced muscle atrophy by regulating the PI3K/Akt signaling pathway. Int Immunopharmacol 2025; 144:113619. [PMID: 39602954 DOI: 10.1016/j.intimp.2024.113619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/27/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND In intensive care units, sepsis-related muscle atrophy is a severe complication of numerous diseases, yet the underlying mechanism and potential therapeutic options remain elusive. Recent research has identified melatonin as a promising candidate for attenuating organ dysfunction triggered by sepsis. METHODS We used in vitro and in vivo models to simulate sepsis, C2C12 myotubes were treated with LPS, and the mice underwent cecal ligation and puncture (CLP) surgery. Following a pretreatment regimen involving melatonin and the AKT inhibitor MK-2206 2HCl, we analyzed changes in p-Akt and MuRF1 protein levels, fiber cross-sectional areas, and myotube diameters. The analyses included RNA sequencing, Western blotting, qRT-PCR, and immunofluorescence staining. RESULTS Activation of the PI3K/Akt pathway in skeletal muscle occurred 24 h post-CLP surgery in mice. This was accompanied by upregulated MuRF1 expression and reduced muscle fiber cross-sectional area, which culminated in muscle atrophy. However, these detrimental effects were attenuated when the mice were pretreated with melatonin via intraperitoneal injection for seven consecutive days. Similarly, LPS treatment of C2C12 myotubes activated the PI3K/Akt pathway, elevated MuRF1 expression, and markedly reduced myotube diameter after 48 h, leading to muscle atrophy. Pretreatment of C2C12 myotubes with melatonin 24 h in advance mitigated these adverse effects. However, cotreatment of C2C12 myotubes with melatonin and MK-2206 2HCl attenuated the beneficial effects of melatonin. CONCLUSION Melatonin can attenuate sepsis-induced muscle atrophy by regulating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Huiming Yao
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Qian Xie
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yuting Yang
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Chaoqi Zhou
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Zhenguo Zeng
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
24
|
Lilley T, Camera DM, Kwa FAA. Repairing muscle with broccoli-derived sulforaphane: A preclinical evaluation for the treatment of mitochondrial myopathies. Drug Discov Today 2025; 30:104283. [PMID: 39736463 DOI: 10.1016/j.drudis.2024.104283] [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: 10/18/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
Skeletal muscle health relies on the production of adenosine triphosphate (ATP) in the mitochondria. ATP production is accompanied by oxidative phosphorylation, which generates reactive oxygen species (ROS). When there is an imbalance in ROS levels, oxidative stress and subsequent mitochondrial dysfunction, mitochondrial myopathies including sarcopenia, chronic progressive external ophthalmoplegia, and proximal myopathy can result. Such incurable myopathies are characterised by aberrant metabolism, limited ATP production, and muscle atrophy. Broccoli-derived sulforaphane has emerged as a novel treatment for mitochondrial myopathies because of its antioxidant and anti-inflammatory properties. This review discusses preclinical studies that reveal sulforaphane's potential therapeutic benefits and limitations in treating mitochondrial myopathies.
Collapse
Affiliation(s)
- Thomas Lilley
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia
| | - Donny M Camera
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia
| | - Faith A A Kwa
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia.
| |
Collapse
|
25
|
Zhang G, Hu F, Huang T, Ma X, Cheng Y, Liu X, Jiang W, Dong B, Fu C. The recent development, application, and future prospects of muscle atrophy animal models. MEDCOMM – FUTURE MEDICINE 2024; 3. [DOI: 10.1002/mef2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/01/2024] [Indexed: 01/06/2025]
Abstract
AbstractMuscle atrophy, characterized by the loss of muscle mass and function, is a hallmark of sarcopenia and cachexia, frequently associated with aging, malignant tumors, chronic heart failure, and malnutrition. Moreover, it poses significant challenges to human health, leading to increased frailty, reduced quality of life, and heightened mortality risks. Despite extensive research on sarcopenia and cachexia, consensus in their assessment remains elusive, with inconsistent conclusions regarding their molecular mechanisms. Muscle atrophy models are crucial tools for advancing research in this field. Currently, animal models of muscle atrophy used for clinical and basic scientific studies are induced through various methods, including aging, genetic editing, nutritional modification, exercise, chronic wasting diseases, and drug administration. Muscle atrophy models also include in vitro and small organism models. Despite their value, each of these models has certain limitations. This review focuses on the limitations and diverse applications of muscle atrophy models to understand sarcopenia and cachexia, and encourage their rational use in future research, therefore deepening the understanding of underlying pathophysiological mechanisms, and ultimately advancing the exploration of therapeutic strategies for sarcopenia and cachexia.
Collapse
Affiliation(s)
- Gongchang Zhang
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Fengjuan Hu
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Tingting Huang
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Xiaoqing Ma
- Longkou People Hospital Longkou Shandong Province China
| | - Ying Cheng
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Xiaolei Liu
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Wenzhou Jiang
- Longkou People Hospital Longkou Shandong Province China
| | - Birong Dong
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
| | - Chenying Fu
- Geriatric Health Care and Medical Research Center West China Hospital, Sichuan University Chengdu Sichuan Province China
- National Clinical Research Center for Geriatrics West China Hospital, Sichuan University Chengdu Sichuan Province China
- Department of Laboratory of Aging and Geriatric Medicine National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University Chengdu Sichuan Province China
| |
Collapse
|
26
|
Kubat GB, Ulger O, Atalay O, Fatsa T, Turkel I, Ozerklig B, Celik E, Ozenc E, Simsek G, Tuncer M. The effects of exercise and mitochondrial transplantation alone or in combination against Doxorubicin-induced skeletal muscle atrophy. J Muscle Res Cell Motil 2024; 45:233-251. [PMID: 38822935 DOI: 10.1007/s10974-024-09676-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 05/22/2024] [Indexed: 06/03/2024]
Abstract
Doxorubicin (DOX) is a chemotherapy drug used to treat various types of cancer, but it is associated with significant side effects such as skeletal muscle atrophy. Exercise has been found to prevent skeletal muscle atrophy through the modulation of mitochondrial pathways. Mitochondrial transplantation (MT) may mitigate toxicity, neurological disorders, kidney and liver injury, and skeletal muscle atrophy. The objective of this study was to evaluate the effects of MT, exercise, and MT with exercise on DOX-induced skeletal muscle atrophy. Male Sprague Dawley rats were randomly assigned to the following groups: control, DOX, MT with DOX, exercise with DOX, and exercise with MT and DOX. A 10-day treadmill running exercise and MT (6.5 µg/100 µL) to tibialis anterior (TA) muscle were administered prior to a single injection of DOX (20 mg/kg). Our data showed that exercise and MT with exercise led to an increase in cross-sectional area of the TA muscle. Exercise, MT and MT with exercise reduced inflammation and maintained mitochondrial enzyme activity. Additionally, exercise and MT have been shown to regulate mitochondrial fusion/fission. Our findings revealed that exercise and MT with exercise prevented oxidative damage. Furthermore, MT and MT with exercise decreased apoptosis and MT with exercise triggered mitochondrial biogenesis. These findings demonstrate the importance of exercise in the prevention of skeletal muscle atrophy and emphasize the significant benefits of MT with exercise. To the best of our knowledge, this is the first study to demonstrate the therapeutic effects of MT with exercise in DOX-induced skeletal muscle atrophy.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, Ankara, Turkey.
- Gulhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey.
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, Ankara, Turkey
- Gulhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Ozbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Tugba Fatsa
- Gulhane Health Sciences Institute, University of Health Sciences, Ankara, Turkey
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Turkey
| | - Ertugrul Celik
- Department of Pathology, Gulhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Emrah Ozenc
- Department of Pathology, Gulhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Gulcin Simsek
- Department of Pathology, Gulhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
27
|
Meacci E, Chirco A, Garcia-Gil M. Potential Vitamin E Signaling Mediators in Skeletal Muscle. Antioxidants (Basel) 2024; 13:1383. [PMID: 39594525 PMCID: PMC11591548 DOI: 10.3390/antiox13111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Vitamin E (Vit E) deficiency studies underline the relevance of this vitamin in skeletal muscle (SkM) homeostasis. The knowledge of the effectors and modulators of Vit E action in SkM cells is limited, especially in aging and chronic diseases characterized by a decline in musculoskeletal health. Vit E comprises eight fat-soluble compounds grouped into tocopherols and tocotrienols, which share the basic chemical structure but show different biological properties and potentials to prevent diseases. Vit E has antioxidant and non-antioxidant activities and both favorable and adverse effects depending on the specific conditions and tissues. In this review, we focus on the actual knowledge of Vit E forms in SkM functions and new potential signaling effectors (i.e., bioactive sphingolipids and myokines). The possible advantages of Vit E supplementation in counteracting SkM dysfunctions in sarcopenia and under microgravity will also be discussed.
Collapse
Affiliation(s)
- Elisabetta Meacci
- Department of Experimental and clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Firenze, Italy
- Interuniversity Institute of Myology, University of Florence, 50134 Firenze, Italy
| | - Antony Chirco
- Department of Experimental and clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Firenze, Italy
| | - Mercedes Garcia-Gil
- Department of Biology, Unit of Physiology, University of Pisa, Via S. Zeno 31, 56127 Pisa, Italy;
| |
Collapse
|
28
|
Jackson MJ. Reactive oxygen species in age-related musculoskeletal decline: implications for nutritional intervention. Proc Nutr Soc 2024:1-9. [PMID: 39512110 DOI: 10.1017/s0029665124004877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Musculoskeletal disorders and age-related musculoskeletal decline are major contributors to the burden of ill health seen in older subjects. Despite this increased burden, these chronic disorders of old age receive a relatively small proportion of national research funds. Much has been learned about fundamental processes involved in ageing from basic science research and this is leading to identification of key pathways that mediate ageing which may help the search for interventions to reduce age-related musculoskeletal decline. This short review will focus on the role of reactive oxygen species in age-related skeletal muscle decline and on the implications of this work for potential nutritional interventions in sarcopenia. The key physiological role of reactive oxygen species is now known to be in mediating redox signalling in muscle and other tissues and ageing leads to disruption of such pathways. In muscle, this is reflected in an age-related attenuation of specific adaptations and responses to contractile activity that impacts the ability of skeletal muscle from ageing individuals to respond to exercise. These pathways provides potential targets for identification of logical interventions that may help maintain muscle mass and function during ageing.
Collapse
Affiliation(s)
- Malcolm J Jackson
- MRC-Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing, Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
29
|
Andrade-Guerrero J, Martínez-Orozco H, Villegas-Rojas MM, Santiago-Balmaseda A, Delgado-Minjares KM, Pérez-Segura I, Baéz-Cortés MT, Del Toro-Colin MA, Guerra-Crespo M, Arias-Carrión O, Diaz-Cintra S, Soto-Rojas LO. Alzheimer's Disease: Understanding Motor Impairments. Brain Sci 2024; 14:1054. [PMID: 39595817 PMCID: PMC11592238 DOI: 10.3390/brainsci14111054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD), the most prevalent neurodegenerative disorder and the leading cause of dementia worldwide, profoundly impacts health and quality of life. While cognitive impairments-such as memory loss, attention deficits, and disorientation-predominate in AD, motor symptoms, though common, remain underexplored. These motor symptoms, including gait disturbances, reduced cardiorespiratory fitness, muscle weakness, sarcopenia, and impaired balance, are often associated with advanced stages of AD and contribute to increased mortality. Emerging evidence, however, suggests that motor symptoms may be present in earlier stages and can serve as predictive markers for AD in older adults. Despite a limited understanding of the underlying mechanisms driving these motor symptoms, several key pathways have been identified, offering avenues for further investigation. This review provides an in-depth analysis of motor symptoms in AD, discussing its progression, potential mechanisms, and therapeutic strategies. Addressing motor symptoms alongside cognitive decline may enhance patient functionality, improve quality of life, and support more comprehensive disease management strategies.
Collapse
Affiliation(s)
- Jesús Andrade-Guerrero
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.A.-G.); (M.M.V.-R.); (A.S.-B.); (K.M.D.-M.); (I.P.-S.); (M.T.B.-C.); (M.A.D.T.-C.)
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico;
| | - Humberto Martínez-Orozco
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico;
| | - Marcos M. Villegas-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.A.-G.); (M.M.V.-R.); (A.S.-B.); (K.M.D.-M.); (I.P.-S.); (M.T.B.-C.); (M.A.D.T.-C.)
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Alberto Santiago-Balmaseda
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.A.-G.); (M.M.V.-R.); (A.S.-B.); (K.M.D.-M.); (I.P.-S.); (M.T.B.-C.); (M.A.D.T.-C.)
| | - Karen M. Delgado-Minjares
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.A.-G.); (M.M.V.-R.); (A.S.-B.); (K.M.D.-M.); (I.P.-S.); (M.T.B.-C.); (M.A.D.T.-C.)
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Isaac Pérez-Segura
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.A.-G.); (M.M.V.-R.); (A.S.-B.); (K.M.D.-M.); (I.P.-S.); (M.T.B.-C.); (M.A.D.T.-C.)
| | - Mauricio T. Baéz-Cortés
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.A.-G.); (M.M.V.-R.); (A.S.-B.); (K.M.D.-M.); (I.P.-S.); (M.T.B.-C.); (M.A.D.T.-C.)
| | - Miguel A. Del Toro-Colin
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.A.-G.); (M.M.V.-R.); (A.S.-B.); (K.M.D.-M.); (I.P.-S.); (M.T.B.-C.); (M.A.D.T.-C.)
| | - Magdalena Guerra-Crespo
- Laboratorio de Medicina Regenerativa, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Oscar Arias-Carrión
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González, Ciudad de México 14080, Mexico;
| | - Sofía Diaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico;
| | - Luis O. Soto-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.A.-G.); (M.M.V.-R.); (A.S.-B.); (K.M.D.-M.); (I.P.-S.); (M.T.B.-C.); (M.A.D.T.-C.)
| |
Collapse
|
30
|
Sevegnani M, Lama A, Girardi F, Hess MW, Castelo MP, Pichler I, Biressi S, Piccoli G. Parkin R274W mutation affects muscle and mitochondrial physiology. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167302. [PMID: 38878834 DOI: 10.1016/j.bbadis.2024.167302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024]
Abstract
Recessive mutations in the Parkin gene (PRKN) are the most common cause of young-onset inherited parkinsonism. Parkin is a multifunctional E3 ubiquitin ligase that plays a variety of roles in the cell including the degradation of proteins and the maintenance of mitochondrial homeostasis, integrity, and biogenesis. In 2001, the R275W mutation in the PRKN gene was identified in two unrelated families with a multigenerational history of postural tremor, dystonia and parkinsonism. Drosophila models of Parkin R275W showed selective and progressive degeneration of dopaminergic neuronal clusters, mitochondrial abnormalities, and prominent climbing defects. In the Prkn mouse orthologue, the amino acid R274 corresponds to human R275. Here we described an age-related motor impairment and a muscle phenotype in R274W +/+ mice. In vitro, Parkin R274W mutation correlates with abnormal myoblast differentiation, mitochondrial defects, and alteration in mitochondrial mRNA and protein levels. Our data suggest that the Parkin R274W mutation may impact mitochondrial physiology and eventually myoblast proliferation and differentiation.
Collapse
Affiliation(s)
| | - Adriano Lama
- Department CIBIO, University of Trento, Trento, Italy
| | | | - Michael W Hess
- Innsbruck Medical University, Institute of Histology and Embryology, Medical University of Innsbruck, Austria
| | - Maria Paulina Castelo
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Irene Pichler
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | | | | |
Collapse
|
31
|
Du G, Zhang P, Guo J, Zhou X, Kan G, Jia J, Chen X, Liang J, Zhan Y. Exploring Radiomics Features Based on H&E Images as Potential Biomarkers for Evaluating Muscle Atrophy: A Preliminary Study. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:2324-2341. [PMID: 38653909 PMCID: PMC11522215 DOI: 10.1007/s10278-024-01122-w] [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: 12/20/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
Radiomics features have been widely used as novel biomarkers in the diagnosis of various diseases, but whether radiomics features derived from hematoxylin and eosin (H&E) images can evaluate muscle atrophy has not been studied. Therefore, this study aims to establish a new biomarker based on H&E images using radiomics methods to quantitatively analyze H&E images, which is crucial for improving the accuracy of muscle atrophy assessment. Firstly, a weightless muscle atrophy model was established by laying macaques in bed, and H&E images of the shank muscle fibers of the control and bed rest (BR) macaques were collected. Muscle fibers were accurately segmented by designing a semi-supervised segmentation framework based on contrastive learning. Then, 77 radiomics features were extracted from the segmented muscle fibers, and a stable subset of features was selected through the LASSO method. Finally, the correlation between radiomics features and muscle atrophy was analyzed using a support vector machine (SVM) classifier. The semi-supervised segmentation results show that the proposed method had an average Spearman's and intra-class correlation coefficient (ICC) of 88% and 86% compared to manually extracted features, respectively. Radiomics analysis showed that the AUC of the muscle atrophy evaluation model based on H&E images was 96.87%. For individual features, GLSZM_SZE outperformed other features in terms of AUC (91.5%) and ACC (84.4%). In summary, the feature extraction based on the semi-supervised segmentation method is feasible and reliable for subsequent radiomics research. Texture features have greater advantages in evaluating muscle atrophy compared to other features. This study provides important biomarkers for accurate diagnosis of muscle atrophy.
Collapse
Affiliation(s)
- Getao Du
- School of Life Science and Technology, & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Peng Zhang
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Jianzhong Guo
- Institute of Applied Acoustics, School of Physics and Information Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Xu Zhou
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Guanghan Kan
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Jiajie Jia
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Xiaoping Chen
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China.
| | - Jimin Liang
- School of Electronic Engineering, Xidian University, Xi'an, Shaanxi, 710071, China.
| | - Yonghua Zhan
- School of Life Science and Technology, & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xidian University, Xi'an, Shaanxi, 710126, China.
| |
Collapse
|
32
|
Li Q, Liu Q, Lin Z, Lin W, Huang F, Zhu P. Hypomethylation in promoters of PGC-1α involved in exercise-driven skeletal muscular alterations in old age. Open Life Sci 2024; 19:20220959. [PMID: 39290496 PMCID: PMC11406220 DOI: 10.1515/biol-2022-0959] [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: 05/28/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Exercise training can significantly improve skeletal muscle mitochondrial function and has been proven to be highly relevant to alterations in skeletal muscle DNA methylation. However, it remains unclear whether late-in-life exercise has an effect on promoter methylation of PGC-1α, a key regulator of mitochondrial biogenesis. Here we employed two distinct exercise modalities, constant medium intensity exercise training (CMIT) and high-intensity interval exercise training (HIIT), to investigate their impacts on PGC-1α expression and methylation regulation in skeletal muscle of aged mice. The results revealed a notable decrease in PGC-1α expression in skeletal muscle of aged mice, accompanied by elevated methylation levels of the PGC-1α promoter, and increased DNA methyltransferase (DNMT) protein expressions. However, both forms of exercise training significantly corrected PGC-1α epigenetic changes, increased PGC-1α expression, and ameliorated skeletal muscle reduction. Furthermore, exercise training led to elevated expression of proteins related to mitochondrial biogenesis and energy metabolism in skeletal muscle, improving mitochondrial structure and function. In conclusion, late-in-life exercise improved skeletal muscle function, morphology, and mitochondria biogenesis, which may be associated with hypomethylation in promoters of PGC-1α and increased content of skeletal muscle PGC-1α. Notably, there was no clear difference between HIIT and CMIT in PGC-1α expression and skeletal muscle function.
Collapse
Affiliation(s)
- Qiaowei Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, P. R. China
- Fujian Provincial Institute of Clinical Geriatrics, Fujian Provincial Hospital, Fuzhou, 350001, P. R. China
- Fujian Key Laboratory of Geriatrics, Fuzhou, 350001, P. R. China
- Fujian Provincial Center for Geriatrics, Fuzhou, 350001, P. R. China
| | - Qin Liu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, P. R. China
- Fujian Provincial Center for Geriatrics, Fuzhou, 350001, P. R. China
| | - Zhong Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, P. R. China
- Fujian Key Laboratory of Geriatrics, Fuzhou, 350001, P. R. China
- Fujian Provincial Center for Geriatrics, Fuzhou, 350001, P. R. China
| | - Wenwen Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, P. R. China
- Fujian Key Laboratory of Geriatrics, Fuzhou, 350001, P. R. China
| | - Feng Huang
- Shengli Clinical Medical College of Fujian Medical University, 134 East Street, Fuzhou, 350001, P. R. China
- Fujian Provincial Institute of Clinical Geriatrics, Fujian Provincial Hospital134 East Street, Fuzhou, 350001, P. R. China
- Fujian Key Laboratory of Geriatrics, 134 East Street, Fuzhou, 350001, P. R. China
- Fujian Provincial Center for Geriatrics, 134 East Street, Fuzhou, 350001, P. R. China
| | - Pengli Zhu
- Shengli Clinical Medical College of Fujian Medical University, 134 East Street, Fuzhou, 350001, P. R. China
- Fujian Provincial Institute of Clinical Geriatrics, Fujian Provincial Hospital134 East Street, Fuzhou, 350001, P. R. China
- Fujian Key Laboratory of Geriatrics, 134 East Street, Fuzhou, 350001, P. R. China
- Fujian Provincial Center for Geriatrics, 134 East Street, Fuzhou, 350001, P. R. China
| |
Collapse
|
33
|
Kubat GB, Picone P. Skeletal muscle dysfunction in amyotrophic lateral sclerosis: a mitochondrial perspective and therapeutic approaches. Neurol Sci 2024; 45:4121-4131. [PMID: 38676818 PMCID: PMC11306305 DOI: 10.1007/s10072-024-07508-6] [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: 02/07/2024] [Accepted: 03/29/2024] [Indexed: 04/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and fatal neuromuscular disease that results in the loss of motor neurons and severe skeletal muscle atrophy. The etiology of ALS is linked to skeletal muscle, which can activate a retrograde signaling cascade that destroys motor neurons. This is why satellite cells and mitochondria play a crucial role in the health and performance of skeletal muscles. This review presents current knowledge on the involvement of mitochondrial dysfunction, skeletal muscle atrophy, muscle satellite cells, and neuromuscular junction (NMJ) in ALS. It also discusses current therapeutic strategies, including exercise, drugs, stem cells, gene therapy, and the prospective use of mitochondrial transplantation as a viable therapeutic strategy.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, Ankara, Turkey
| | - Pasquale Picone
- Istituto Per La Ricerca E L'Innovazione Biomedica, Consiglio Nazionale Delle Ricerche, Via U. La Malfa 153, 0146, Palermo, Italy.
| |
Collapse
|
34
|
Ulger O, Eş I, Proctor CM, Algin O. Stroke studies in large animals: Prospects of mitochondrial transplantation and enhancing efficiency using hydrogels and nanoparticle-assisted delivery. Ageing Res Rev 2024; 100:102469. [PMID: 39191353 DOI: 10.1016/j.arr.2024.102469] [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: 06/12/2024] [Revised: 08/08/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
One of the most frequent reasons for mortality and disability today is acute ischemic stroke, which occurs by an abrupt disruption of cerebral circulation. The intricate damage mechanism involves several factors, such as inflammatory response, disturbance of ion balance, loss of energy production, excessive reactive oxygen species and glutamate release, and finally, neuronal death. Stroke research is now carried out using several experimental models and potential therapeutics. Furthermore, studies are being conducted to address the shortcomings of clinical care. A great deal of research is being done on novel pharmacological drugs, mitochondria targeting compounds, and different approaches including brain cooling and new technologies. Still, there are many unanswered questions about disease modeling and treatment strategies. Before these new approaches may be used in therapeutic settings, they must first be tested on large animals, as most of them have been done on rodents. However, there are several limitations to large animal stroke models used for research. In this review, the damage mechanisms in acute ischemic stroke and experimental acute ischemic stroke models are addressed. The current treatment approaches and promising experimental methods such as mitochondrial transplantation, hydrogel-based interventions, and strategies like mitochondria encapsulation and chemical modification, are also examined in this work.
Collapse
Affiliation(s)
- Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, Ankara 06010, Turkiye; Gulhane Training and Research Hospital, University of Health Sciences, Ankara 06010, Turkiye.
| | - Ismail Eş
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Oxford OX3 7DQ, UK
| | - Christopher M Proctor
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Oxford OX3 7DQ, UK
| | - Oktay Algin
- Interventional MR Clinical R&D Institute, Ankara University, Ankara 06100, Turkiye; Department of Radiology, Medical Faculty, Ankara University, Ankara 06100, Turkiye; National MR Research Center (UMRAM), Bilkent University, Ankara 06800, Turkiye
| |
Collapse
|
35
|
Li Z, Liang S, Ke L, Wang M, Gao K, Li D, Xu Z, Li N, Zhang P, Cheng W. Cell life-or-death events in osteoporosis: All roads lead to mitochondrial dynamics. Pharmacol Res 2024; 208:107383. [PMID: 39214266 DOI: 10.1016/j.phrs.2024.107383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria exhibit heterogeneous shapes and networks within and among cell types and tissues, also in normal or osteoporotic bone tissues with complex cell types. This dynamic characteristic is determined by the high plasticity provided by mitochondrial dynamics and is stemmed from responding to the survival and functional requirements of various bone cells in a specific microenvironments. In contrast, mitochondrial dysfunction, induced by dysregulation of mitochondrial dynamics, may act as a trigger of cell death signals, including common apoptosis and other forms of programmed cell death (PCD). These PCD processes consisting of tightly structured cascade gene expression events, can further influence the bone remodeling by facilitating the death of various bone cells. Mitochondrial dynamics, therefore, drive the bone cells to stand at the crossroads of life and death by integrating external signals and altering metabolism, shape, and signal-response properties of mitochondria. This implies that targeting mitochondrial dynamics displays significant potential in treatment of osteoporosis. Considerable effort has been made in osteoporosis to emphasize the parallel roles of mitochondria in regulating energy metabolism, calcium signal transduction, oxidative stress, inflammation, and cell death. However, the emerging field of mitochondrial dynamics-related PCD is not well understood. Herein, to bridge the gap, we outline the latest knowledge on mitochondrial dynamics regulating bone cell life or death during normal bone remodeling and osteoporosis.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050011, China
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000, China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300, China.
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
36
|
Ahmad K, Manongi NJ, Rajapandian R, Moti Wala S, Al Edani EM, Samuel EA, Arcia Franchini AP. Effectiveness of Coenzyme Q10 Supplementation in Statin-Induced Myopathy: A Systematic Review. Cureus 2024; 16:e68316. [PMID: 39350827 PMCID: PMC11441719 DOI: 10.7759/cureus.68316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/31/2024] [Indexed: 10/04/2024] Open
Abstract
Statins are among the most widely prescribed drugs for treating dyslipidemia and reducing the incidence of heart disease and stroke. However, they come with a wide range of side effects, from myopathy to necrotizing rhabdomyolysis, as well as diabetes, hepatotoxicity, and sleep problems. The most common side effect of statins is statin-induced myopathy, often leading to discontinuation of statin therapy and noncompliance in many patients. This study aims to assess the effectiveness of coenzyme Q10 (CoQ10) supplementation as a treatment for patients with statin-induced myopathy. This systematic review was conducted by following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 statement. Relevant studies were identified through searches of Medline, PMC, PubMed, Science Direct, and Google Scholar. Only randomized control trials and meta-analyses of oral CoQ10 supplementation versus placebo in adults with statin-associated myalgia were included. The risk of bias was assessed using the Cochrane Risk of Bias tool (The Cochrane Collaboration, London, England, UK) and the measurement tool for the "assessment of multiple systematic reviews" (AMSTAR tool). Out of 5,000 records identified, only five were selected for this review: one meta-analysis and four randomized controlled trials. All of these studies were conducted between 2010 and 2023, involving a total of 800 patients. All randomized controlled trials showed improvement in statin-associated myopathy with CoQ10 supplementation, along with or without a reduced dosage of statins, without any notable side effects of CoQ10. Therefore, it can be deduced that CoQ10 supplementation significantly ameliorates statin-induced musculoskeletal symptoms.
Collapse
Affiliation(s)
- Khoula Ahmad
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Naelijwa J Manongi
- Family Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Ramkumar Rajapandian
- Trauma and Orthopedics, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Sajida Moti Wala
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Esraa M Al Edani
- Dermatology, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Essa A Samuel
- Physical Medicine and Rehabilitation, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Ana P Arcia Franchini
- Research, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
37
|
Zeki NM, Mustafa YF. Natural linear coumarin-heterocyclic conjugates: A review of their roles in phytotherapy. Fitoterapia 2024; 175:105929. [PMID: 38548026 DOI: 10.1016/j.fitote.2024.105929] [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: 12/15/2023] [Revised: 03/17/2024] [Accepted: 03/25/2024] [Indexed: 05/26/2024]
Abstract
Heterocycle conjugates provide a fresh investigative scope to find novel molecules with enhanced phytotherapeutic characteristics. Coumarin-based products are widely used in the synthesis of several compounds with biological and medicinal properties since they are naturally occurring heterocycles with a broad dispersion. The investigation of coumarin-based phytochemicals with annulated heterocyclic rings is a promising approach to discovering novel conjugates with significant phytotherapeutic attributes. Due to the applicable coumarin extraction processes, a range of linear coumarin-heterocyclic conjugates were isolated from different natural resources and exhibited remarkable therapeutic efficacy. This review highlights the phytotherapeutic potential and origins of various natural linear coumarin-heterocyclic conjugates. We searched several databases, including Science Direct, Web of Science, Springer, Google Scholar, and PubMed. After sieving, we ultimately identified and included 118 pertinent studies published between 2000 and the middle of 2023. This will inspire medicinal chemists with extremely insightful ideas for designing and synthesizing therapeutically active lead compounds in the future that are built on the pharmacophores of coumarin-heterocyclic conjugates and have significant therapeutic attributes.
Collapse
Affiliation(s)
- Nameer Mazin Zeki
- Department of Pharmacology, College of Medicine, Ninevah University, Mosul, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq.
| |
Collapse
|
38
|
Bellanti F, Lo Buglio A, Pannone G, Pedicillo MC, De Stefano IS, Pignataro A, Capurso C, Vendemiale G. An Amino Acid Mixture to Counteract Skeletal Muscle Atrophy: Impact on Mitochondrial Bioenergetics. Int J Mol Sci 2024; 25:6056. [PMID: 38892242 PMCID: PMC11173258 DOI: 10.3390/ijms25116056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Skeletal muscle atrophy (SMA) is caused by a rise in muscle breakdown and a decline in protein synthesis, with a consequent loss of mass and function. This study characterized the effect of an amino acid mixture (AA) in models of SMA, focusing on mitochondria. C57/Bl6 mice underwent immobilization of one hindlimb (I) or cardiotoxin-induced muscle injury (C) and were compared with controls (CTRL). Mice were then administered AA in drinking water for 10 days and compared to a placebo group. With respect to CTRL, I and C reduced running time and distance, along with grip strength; however, the reduction was prevented by AA. Tibialis anterior (TA) muscles were used for histology and mitochondria isolation. I and C resulted in TA atrophy, characterized by a reduction in both wet weight and TA/body weight ratio and smaller myofibers than those of CTRL. Interestingly, these alterations were lightly observed in mice treated with AA. The mitochondrial yield from the TA of I and C mice was lower than that of CTRL but not in AA-treated mice. AA also preserved mitochondrial bioenergetics in TA muscle from I and C mice. To conclude, this study demonstrates that AA prevents loss of muscle mass and function in SMA by protecting mitochondria.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (F.B.); (A.L.B.); (C.C.)
| | - Aurelio Lo Buglio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (F.B.); (A.L.B.); (C.C.)
| | - Giuseppe Pannone
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (G.P.); (M.C.P.); (I.S.D.S.); (A.P.)
| | - Maria Carmela Pedicillo
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (G.P.); (M.C.P.); (I.S.D.S.); (A.P.)
| | - Ilenia Sara De Stefano
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (G.P.); (M.C.P.); (I.S.D.S.); (A.P.)
| | - Angela Pignataro
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (G.P.); (M.C.P.); (I.S.D.S.); (A.P.)
| | - Cristiano Capurso
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (F.B.); (A.L.B.); (C.C.)
| | - Gianluigi Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (F.B.); (A.L.B.); (C.C.)
| |
Collapse
|
39
|
Song Y, Ren S, Chen X, Li X, Chen L, Zhao S, Zhang Y, Shen X, Chen Y. Inhibition of MFN1 restores tamoxifen-induced apoptosis in resistant cells by disrupting aberrant mitochondrial fusion dynamics. Cancer Lett 2024; 590:216847. [PMID: 38583647 DOI: 10.1016/j.canlet.2024.216847] [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: 12/10/2023] [Revised: 03/18/2024] [Accepted: 03/31/2024] [Indexed: 04/09/2024]
Abstract
Tamoxifen (TAM) resistance presents a major clinical obstacle in the management of estrogen-sensitive breast cancer, highlighting the need to understand the underlying mechanisms and potential therapeutic approaches. We showed that dysregulated mitochondrial dynamics were involved in TAM resistance by protecting against mitochondrial apoptosis. The dysregulated mitochondrial dynamics were associated with increased mitochondrial fusion and decreased fission, thus preventing the release of mitochondrial cytochrome c to the cytoplasm following TAM treatment. Dynamin-related GTPase protein mitofusin 1 (MFN1), which promotes fusion, was upregulated in TAM-resistant cells, and high MFN1 expression indicated a poor prognosis in TAM-treated patients. Mitochondrial translocation of MFN1 and interaction between MFN1 and mitofusin 2 (MFN2) were enhanced to promote mitochondrial outer membrane fusion. The interaction of MFN1 and cristae-shaping protein optic atrophy 1 (OPA1) and OPA1 oligomerization were reduced due to augmented OPA1 proteolytic cleavage, and their apoptosis-promoting function was reduced due to cristae remodeling. Furthermore, the interaction of MFN1 and BAK were increased, which restrained BAK activation following TAM treatment. Knockdown or pharmacological inhibition of MFN1 blocked mitochondrial fusion, restored BAK oligomerization and cytochrome c release, and amplified activation of caspase-3/9, thus sensitizing resistant cells to apoptosis and facilitating the therapeutic effects of TAM both in vivo and in vitro. Conversely, overexpression of MFN1 alleviated TAM-induced mitochondrial apoptosis and promoted TAM resistance in sensitive cells. These results revealed that dysregulated mitochondrial dynamics contributes to the development of TAM resistance, suggesting that targeting MFN1-mediated mitochondrial fusion is a promising strategy to circumvent TAM resistance.
Collapse
Affiliation(s)
- Yuxuan Song
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China
| | - Shuang Ren
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China
| | - Xingmei Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China
| | - Xuhong Li
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China
| | - Lin Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China
| | - Shijie Zhao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China
| | - Yue Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China.
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China.
| | - Yan Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China; Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou 561113, China.
| |
Collapse
|
40
|
Zeki NM, Mustafa YF. 6,7-Coumarin-heterocyclic hybrids: A comprehensive review of their natural sources, synthetic approaches, and bioactivity. J Mol Struct 2024; 1303:137601. [DOI: 10.1016/j.molstruc.2024.137601] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
41
|
Lei Y, Gan M, Qiu Y, Chen Q, Wang X, Liao T, Zhao M, Chen L, Zhang S, Zhao Y, Niu L, Wang Y, Zhu L, Shen L. The role of mitochondrial dynamics and mitophagy in skeletal muscle atrophy: from molecular mechanisms to therapeutic insights. Cell Mol Biol Lett 2024; 29:59. [PMID: 38654156 PMCID: PMC11036639 DOI: 10.1186/s11658-024-00572-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: 01/09/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
Skeletal muscle is the largest metabolic organ of the human body. Maintaining the best quality control and functional integrity of mitochondria is essential for the health of skeletal muscle. However, mitochondrial dysfunction characterized by mitochondrial dynamic imbalance and mitophagy disruption can lead to varying degrees of muscle atrophy, but the underlying mechanism of action is still unclear. Although mitochondrial dynamics and mitophagy are two different mitochondrial quality control mechanisms, a large amount of evidence has indicated that they are interrelated and mutually regulated. The former maintains the balance of the mitochondrial network, eliminates damaged or aged mitochondria, and enables cells to survive normally. The latter degrades damaged or aged mitochondria through the lysosomal pathway, ensuring cellular functional health and metabolic homeostasis. Skeletal muscle atrophy is considered an urgent global health issue. Understanding and gaining knowledge about muscle atrophy caused by mitochondrial dysfunction, particularly focusing on mitochondrial dynamics and mitochondrial autophagy, can greatly contribute to the prevention and treatment of muscle atrophy. In this review, we critically summarize the recent research progress on mitochondrial dynamics and mitophagy in skeletal muscle atrophy, and expound on the intrinsic molecular mechanism of skeletal muscle atrophy caused by mitochondrial dynamics and mitophagy. Importantly, we emphasize the potential of targeting mitochondrial dynamics and mitophagy as therapeutic strategies for the prevention and treatment of muscle atrophy, including pharmacological treatment and exercise therapy, and summarize effective methods for the treatment of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Yuhang Lei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Mailin Gan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yanhao Qiu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qiuyang Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xingyu Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Tianci Liao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Mengying Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lei Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shunhua Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Ye Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lili Niu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Li Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Linyuan Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
42
|
Lei T, Rui Y, Xiaoshuang Z, Jinglan Z, Jihong Z. Mitochondria transcription and cancer. Cell Death Discov 2024; 10:168. [PMID: 38589371 PMCID: PMC11001877 DOI: 10.1038/s41420-024-01926-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Mitochondria are major organelles involved in several processes related to energy supply, metabolism, and cell proliferation. The mitochondria function is transcriptionally regulated by mitochondria DNA (mtDNA), which encodes the key proteins in the electron transport chain that is indispensable for oxidative phosphorylation (OXPHOS). Mitochondrial transcriptional abnormalities are closely related to a variety of human diseases, such as cardiovascular diseases, and diabetes. The mitochondria transcription is regulated by the mtDNA, mitochondrial RNA polymerase (POLRMT), two transcription factors (TFAM and TF2BM), one transcription elongation (TEFM), and one known transcription termination factor (mTERFs). Dysregulation of these factors directly leads to altered expression of mtDNA in tumor cells, resulting in cellular metabolic reprogramming and mitochondrial dysfunction. This dysregulation plays a role in modulating tumor progression. Therefore, understanding the role of mitochondrial transcription in cancer can have implications for cancer diagnosis, prognosis, and treatment. Targeting mitochondrial transcription or related pathways may provide potential therapeutic strategies for cancer treatment. Additionally, assessing mitochondrial transcriptional profiles or biomarkers in cancer cells or patient samples may offer diagnostic or prognostic information.
Collapse
Affiliation(s)
- Tang Lei
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Yu Rui
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Zhou Xiaoshuang
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Zhang Jinglan
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Zhang Jihong
- Medical School, Kunming University of Science and Technology, Kunming, China.
- Yunnan Province Clinical Research Center for Hematologic Disease, Kunming, China.
| |
Collapse
|
43
|
Pesta D. Mitochondrial density in skeletal and cardiac muscle. Mitochondrion 2024; 75:101838. [PMID: 38158151 DOI: 10.1016/j.mito.2023.101838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/27/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Kubat et al. provide a review on the role Mitochondrial density in skeletal and cardiac muscle of mitochondrial dysfunction in muscle atrophy. They stress mitochondria's pivotal function, citing a 52 % density in skeletal muscle. However, the reference to Park et al.'s work misinterprets their findings. Park et al. report citrate synthase (CS) activity, indicating mitochondrial density as 222 ± 13 μmol.min-1.mg-1 for cardiac muscle and 115 ± 2 μmol.min-1.mg-1 for skeletal muscle. Thus, the authors should clarify that skeletal muscle density is approximately 52 % of cardiac muscle, not an absolute 52 %. Mitochondrial volume density assessment, predominantly through TEM, establishes cardiomyocytes at 25-30 % and untrained skeletal muscle at 2-6 %, increasing to 11 % in trained athletes. However, this remains modest compared to myofibrils' 75 %-85 % of muscle fiber volume. Although the utility of CS activity is evident, TEM and other novel approaches such as three-dimensional focused ion beam scanning electron microscopy are likely superior for assessing mitochondrial volume density and morphology.
Collapse
Affiliation(s)
- D Pesta
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Centre for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|