1
|
Chang CJ, Lin MH, Chen LY, Tseng SH, Hwang AC, Huang CY, Yen KH, Chen LK, Peng LN. High muscle-to-fat ratio predicts slow muscle strength decline in middle-aged and older adults: Longitudinal aging study of Taipei. J Chin Med Assoc 2025; 88:367-374. [PMID: 40128155 DOI: 10.1097/jcma.0000000000001229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Handgrip strength is a vital marker of muscle function and predictor of health outcomes in older adults. This study investigated the relationship between the muscle-to-fat ratio and 3-year decrease in handgrip strength in community-dwelling adults aged ≥50 years. METHODS Data were obtained from the Longitudinal Aging Study of Taipei (LAST), a cohort study of adults aged 50 years and older. Measurements from two waves, 3 years apart, were analyzed. Demographics, laboratory data, and handgrip strength data were collected. Appendicular skeletal muscle mass was assessed using bioimpedance analysis, and the relative appendicular skeletal muscle mass index was calculated by dividing appendicular muscle mass by height squared. The muscle-to-fat ratio was derived by dividing appendicular muscle mass by total body fat. Handgrip strength decrease was divided into quartiles; slow decliners experienced the smallest decrease, whereas rapid decliners had the greatest decrease. Associations between the muscle-to-fat ratio and other risk factors were analyzed. RESULTS Over 3 years, the Charlson Comorbidity Index, medication use, waist-to-hip ratio, and fat percentage increased, whereas skeletal muscle mass, the muscle-to-fat ratio, and handgrip strength decreased. Rapid decliners were less likely to be male (21.6% vs 33.3%, p = 0.008) or alcohol drinkers (53.8% vs 66.2%, p = 0.01) and had lower skeletal muscle mass (6.3 ± 0.9 vs 6.6 ± 1.0, p = 0.006) and muscle-to-fat ratios (1.0 ± 0.4 vs 1.1 ± 0.5, p = 0.004) but greater fat percentages (30.4 ± 6.6 vs 29.0 ± 7.6, p = 0.045). A greater muscle-to-fat ratio (odds ratio [OR] = 3.751, p = 0.047), greater physical activity (OR = 1.694, p = 0.04), and lower glycated hemoglobin (HbA1c; OR = 0.61, p = 0.008) reduced the risk of rapid decline. CONCLUSION The muscle-to-fat ratio, together with physical activity and glycemic control, predicts a decrease in handgrip strength, highlighting its potential as a biomarker of intrinsic capacity and muscle-fat interplay. Further research is needed to explore the underlying biological mechanisms involved.
Collapse
Affiliation(s)
- Ching-Jen Chang
- Department of Family Medicine, Feng-Yuan Hospital, Ministry of Health and Welfare, Taiwan, Taipei, Taiwan, ROC
| | - Ming-Hsien Lin
- Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Liang-Yu Chen
- Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Sung-Hua Tseng
- Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - An-Chun Hwang
- Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chung-Yu Huang
- Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Ko-Han Yen
- Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Liang-Kung Chen
- Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Taipei Municipal Gan-Dau Hospital (Managed by Taipei Veterans General Hospital), Taipei, Taiwan, ROC
| | - Li-Ning Peng
- Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| |
Collapse
|
2
|
Moustafa Mahmoud M, Abdel Hameed NQ, Adel Al Dreny Abd Al Latef B, Samir Kamar S, Ahmed Rashed L, Abdelhameed Gouda SA. High-intensity exercise alongside insulin alleviates muscle atrophy in type 1 diabetes mellitus concomitant with modulation of mitophagy-related proteins in skeletal muscle. Arch Physiol Biochem 2025; 131:293-305. [PMID: 39382178 DOI: 10.1080/13813455.2024.2410791] [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: 04/26/2024] [Revised: 08/08/2024] [Accepted: 09/15/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Diabetes patients' quality of life can be severely impacted by diabetic muscle atrophy. AIM This study aimed to explore the impact of high-intensity exercise (HIE) alongside insulin treatment on muscle atrophy in a rat model of type 1 diabetes mellitus (T1DM). METHODOLOGY Fifty rats were allocated into five groups; Group 1, control sedentary (CS), T1DM was elicited in the rest of the groups by giving them Streptozotocin (STZ) (60 mg/kg), where group 2 (DS) remained sedentary, while groups 3,4,5 were treated with insulin after induction of diabetes. Group 4 (DI+MIE) and 5 (DI+ HIE) underwent moderate and high-intensity exercise, respectively. RESULTS HIE for 14 days combined with insulin treatment significantly restored muscle strength and mass with a significant modification in the mitophagy-related proteins and fibroblast growth factor 21 (FGF 21) compared to other treated groups. CONCLUSION This study concluded that there is a therapeutic role for HIE with insulin against T1DM-induced muscle atrophy.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Muscular Atrophy/therapy
- Muscular Atrophy/etiology
- Muscular Atrophy/metabolism
- Muscular Atrophy/pathology
- Insulin/therapeutic use
- Insulin/pharmacology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/drug effects
- Rats
- Mitophagy/drug effects
- Physical Conditioning, Animal
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/therapy
- Male
- Rats, Wistar
- Hypoglycemic Agents/therapeutic use
- Hypoglycemic Agents/pharmacology
- Streptozocin
- Muscle Strength/drug effects
Collapse
Affiliation(s)
| | | | | | - Samaa Samir Kamar
- Histology department, Faculty of Medicine- Cairo University, Cairo, Egypt
- Histology department, Armed Forces College of Medicine, Cairo, Egypt
| | - Laila Ahmed Rashed
- Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
3
|
Rai V, Deepu V, Agrawal DK. Targeting RAGE-signaling pathways in the repair of rotator-cuff injury. Mol Cell Biochem 2025; 480:2539-2554. [PMID: 39395136 PMCID: PMC11961478 DOI: 10.1007/s11010-024-05132-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024]
Abstract
Rotator cuff injury (RCI) is a common musculoskeletal problem that can have a significant impact on the quality of life and functional abilities of those affected. Novel therapies, including proteomics-based, stem cells, platelet-rich plasma, and exosomes, are being developed to promote rotator-cuff healing. The receptor for advanced glycation end-products (RAGE) is a multifunctional receptor that is expressed on several cell types and is implicated in several physiologic and pathological processes, such as tissue repair, inflammation, and degeneration. Because of its capacity to bind with a variety of ligands and initiate signaling pathways that lead to inflammatory responses in RCI, RAGE plays a crucial role in inflammation. In this critical review article, we discussed the role of RAGE-mediated persistent inflammation in RCI followed by novel factors including PKCs, TIRAP, DIAPH1, and factors related to muscle injury with their therapeutic potential in RCI. These factors involve various aspects of muscle injury and signaling and the possibility of targeting these factors to improve the clinical outcomes in RCI still needs further investigation.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Vinitha Deepu
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
4
|
Wang YJ, Chen ZH, Shen YT, Wang KX, Han YM, Zhang C, Yang XM, Chen BQ. Stem cell therapy: A promising therapeutic approach for skeletal muscle atrophy. World J Stem Cells 2025; 17:98693. [DOI: 10.4252/wjsc.v17.i2.98693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/09/2024] [Accepted: 01/23/2025] [Indexed: 02/24/2025] Open
Abstract
Skeletal muscle atrophy results from disruptions in the growth and metabolism of striated muscle, leading to a reduction or loss of muscle fibers. This condition not only significantly impacts patients’ quality of life but also imposes substantial socioeconomic burdens. The complex molecular mechanisms driving skeletal muscle atrophy contribute to the absence of effective treatment options. Recent advances in stem cell therapy have positioned it as a promising approach for addressing this condition. This article reviews the molecular mechanisms of muscle atrophy and outlines current therapeutic strategies, focusing on mesenchymal stem cells, induced pluripotent stem cells, and their derivatives. Additionally, the challenges these stem cells face in clinical applications are discussed. A deeper understanding of the regenerative potential of various stem cells could pave the way for breakthroughs in the prevention and treatment of muscle atrophy.
Collapse
Affiliation(s)
- Ying-Jie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Ze-Hao Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Yun-Tian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Ke-Xin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Yi-Min Han
- Medical College, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Chen Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226000, Jiangsu Province, China
| | - Xiao-Ming Yang
- Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong 226000, Jiangsu Province, China
- Research and Development Center for E-Learning, Ministry of Education, Beijing 100816, China
| | - Bing-Qian Chen
- Department of Orthopaedics, Changshu Hospital Affiliated to Soochow University, Changshu 215500, Jiangsu Province, China
| |
Collapse
|
5
|
Zhang S, Guo S, Wang P, Song Y, Yang L, Sun Q, Huang Q, Zhang Y. Dapagliflozin attenuates skeletal muscle atrophy in diabetic nephropathy mice through suppressing Gasdermin D-mediated pyroptosis. Int Immunopharmacol 2025; 148:114088. [PMID: 39837016 DOI: 10.1016/j.intimp.2025.114088] [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: 08/17/2024] [Revised: 12/30/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Skeletal muscle atrophy is a clinical concern in diabetic nephropathy, and without effective therapeutic approaches. Massive evidence has demonstrated that dapagliflozin, a sodium-glucose co-transporter 2 inhibitor can relieve diabetic nephropathy by inhibiting glucose re-absorption or podocyte pyroptosis. Nevertheless, whether dapagliflozin could treat skeletal muscle atrophy or the potential protection mechanism in diabetic nephropathy mice is unclear. METHODS The variety of approaches were used to assess the particular histology-associated characteristics, mRNA, and protein expression. These included examing the morphology of renal and skeletal muscle tissues through H&E staining, detecting mRNA and protein expression through real-time PCR and Western blot analysis, and monitoring fasting blood glucose levels by using Blood Glucose Monitor Test Kits. RESULTS Dapagliflozin mitigated renal tissue injury with podocyte protein-nephrin and skeletal muscle atrophy effectively with mitochondrial-related proteins. Meanwhile, our research revealed that Casp3 was the target gene and dapagliflozin could decrease the expressions. Subsequently, we verified that dapagliflozin can effectively decrease canonical pyroptosis pathway proteins, which include Gasdermin D, NLRP3, Casp1, and ASC. Meanwhile, Palmitic acid (PA) induced Gasdermin E-N fragment (non-canonical pyroptosis protein) in C2C12 cells, and then released the inflammatory molecules such as IL-1β, IL-18, and NF-kappaB, which were suppressed by dapagliflozin treatment. Aside from that, dapagliflozin demonstrated a good binding affinity to the Casp3 and Gasdermin D protein, whereas it had a less binding affinity with NLRP3, Casp1, ASC, and Gasdermin E. At last, the Gasdermin D inhibitor can reverse the therapeutic effect of dapagliflozin. CONCLUSION Dapagliflozin alleviates skeletal muscle atrophy in diabetic nephropathy mice, which is through the Gasdermin D-mediated canonical pyroptosis pathway.
Collapse
Affiliation(s)
- Shuo Zhang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Shuang Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Pengyu Wang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Yan Song
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Leiming Yang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Qiyu Sun
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Qi Huang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China.
| | - Youzhi Zhang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China; Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China.
| |
Collapse
|
6
|
Chang M, Liu R, Chen B, Xu J, Wang W, Ji Y, Gao Z, Liu B, Yao X, Sun H, Xu F, Shen Y. hBMSC-EVs alleviate weightlessness-induced skeletal muscle atrophy by suppressing oxidative stress and inflammation. Stem Cell Res Ther 2025; 16:46. [PMID: 39901193 PMCID: PMC11792267 DOI: 10.1186/s13287-025-04175-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Muscle disuse and offloading in microgravity are likely the primary factors mediating spaceflight-induced muscle atrophy, for which there is currently no effective treatment other than exercise. Extracellular vesicles derived from bone marrow mesenchymal stem cells (BMSC-EVs) possess anti-inflammatory and antioxidant properties, offering a potential strategy for combating weightless muscular atrophy. METHODS In this study, human BMSCs-EVs (hBMSC-EVs) were isolated using super-centrifugation and characterized. C2C12 myotube nutrition-deprivation and mice tail suspension models were established. Subsequently, the diameter of C2C12 myotubes, Soleus mass, cross-sectional area (CSA) of muscle fibers, and grip strength in mice were assessed to investigate the impact of hBMSC-EVs on muscle atrophy. Immunostaining, transmission electron microscopy observation, and western blot analysis were employed to assess the impact of hBMSC-EVs on muscle fiber types, ROS levels, inflammation, ubiquitin-proteasome system activity, and autophagy lysosome pathway activation in skeletal muscle atrophy. RESULTS The active hBMSC-EVs can be internalized by C2C12 myotubes and skeletal muscle. hBMSC-EVs can effectively reduce C2C12 myotube atrophy caused by nutritional deprivation, with a concentration of 10 × 108 particles/mL showing the best effect (P < 0.001). Additionally, hBMSC-EVs can down-regulate the protein levels associated with UPS and oxidative stress. Moreover, intravenous administration of hBMSC-EVs at a concentration of 1 × 1010 particles/mL can effectively reverse the reduction in soleus mass (P < 0.001), CSA (P < 0.01), and grip strength (P < 0.001) in mice caused by weightlessness. They demonstrate the ability to inhibit protein degradation mediated by UPS and autophagy lysosome pathway, along with the suppression of oxidative stress and inflammatory responses. Furthermore, hBMSC-EVs impede the transition of slow muscle fibers to fast muscle fibers via upregulation of Sirt1 and PGC-1α protein levels. CONCLUSIONS Our findings indicate that hBMSC-EVs are capable of inhibiting excessive activation of the UPS and autophagy lysosome pathway, suppressing oxidative stress and inflammatory response, reversing muscle fiber type transformation, effectively delaying hindlimb unloading-induced muscle atrophy and enhancing muscle function. Our study has further advanced the understanding of the molecular mechanism underlying muscle atrophy in weightlessness and has demonstrated the protective effect of hBMSC-EVs on muscle atrophy.
Collapse
Affiliation(s)
- Mengyuan Chang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Ruiqi Liu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Bingqian Chen
- Department of Orthopedics, First People's Hospital of Changshu City, Changshu Hospital Affiliated to Soochow University, Changshu, 215500, Jiangsu Province, People's Republic of China
| | - Jin Xu
- Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang, 222000, Jiangsu Province, People's Republic of China
| | - Wei 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, 226001, Jiangsu Province, People's Republic of China
| | - Yanan Ji
- 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, 226001, Jiangsu Province, People's Republic of China
| | - Zihui Gao
- 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, 226001, Jiangsu Province, People's Republic of China
| | - Boya 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, 226001, Jiangsu Province, People's Republic of 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, 226001, Jiangsu Province, People's Republic of 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, 226001, Jiangsu Province, People's Republic of China.
- Research and Development Center for E-Learning, Ministry of Education, Beijing, People's Republic of China.
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu Province, People's Republic of 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, 226001, Jiangsu Province, People's Republic of China.
| |
Collapse
|
7
|
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
|
8
|
Wang X, Tang X, Wang Y, Zhao S, Xu N, Wang H, Kuang M, Han S, Jiang Z, Zhang W. Plant-Derived Treatments for Different Types of Muscle Atrophy. Phytother Res 2025; 39:1107-1138. [PMID: 39743857 PMCID: PMC11832362 DOI: 10.1002/ptr.8420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/24/2024] [Accepted: 12/01/2024] [Indexed: 01/04/2025]
Abstract
With the development of medicine and chemistry, an increasing number of plant-derived medicines have been shown to exert beneficial therapeutic on the treatment of various physical and psychological diseases. In particular, by using physical chemistry methods, we are able to examine the chemical components of plants and the effects of these substances on the human body. Muscle atrophy (MA) is characterized by decreased muscle mass and function, is caused by multiple factors and severely affects the quality of life of patients. The multifactorial and complex pathogenesis of MA hinders drug research and disease treatment. However, phytotherapy has achieved significant results in the treatment of MA. We searched PubMed and the Web of Science for articles related to plant-derived substances and muscle atrophy. After applying exclusion and inclusion criteria, 166 and 79 articles met the inclusion criteria, respectively. A total of 173 articles were included in the study after excluding duplicates. The important role of phytoactives such as curcumin, resveratrol, and ginsenosides in the treatment of MA (e.g., maintaining a positive nitrogen balance in muscles and exerting anti-inflammatory and antioxidant effects) has been extensively studied. Unfortunately, MA dose not have to a single cause, and each cause has its own unique mechanism of injury. This review focuses on the therapeutic mechanisms of active plant components in MA and provides insights into the personalized treatment of MA.
Collapse
Affiliation(s)
- Xingpeng Wang
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Xiaofu Tang
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Yunhui Wang
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Shengyin Zhao
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Ning Xu
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Haoyu Wang
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Mingjie Kuang
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Shijie Han
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Zhensong Jiang
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Wen Zhang
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| |
Collapse
|
9
|
Gaida D, Park YW, Kang YJ, Kim SG. Therapeutic potential of 4-hexylresorcinol in reducing sarcopenia in diabetic masseter muscle. Maxillofac Plast Reconstr Surg 2025; 47:2. [PMID: 39832033 PMCID: PMC11747031 DOI: 10.1186/s40902-025-00457-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 12/31/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND This study aimed to evaluate the effects of 4-hexylresorcinol (4HR), a synthetic compound with antioxidant and stress-modulating properties, on diabetic sarcopenia in the masseter muscle. METHODS A controlled, parallel-arm study was conducted using 38 Sprague-Dawley rats divided into diabetic and non-diabetic groups. Diabetes was induced with streptozotocin (STZ), and the groups were further subdivided to receive weekly subcutaneous injections of either 4HR or saline. Muscle volume was assessed using micro-computed tomography (μCT), and glycogen storage and protein expression were analyzed using periodic acid-Schiff (PAS) staining and immunohistochemistry. RESULTS μCT analysis revealed that diabetic rats exhibited significantly reduced masseter muscle volume compared to non-diabetic rats. However, 4HR treatment partially mitigated muscle volume loss in diabetic animals. Histological analysis showed higher PAS staining intensity in the diabetic group treated with 4HR compared to the untreated diabetic group, suggesting improved glycogen storage. Immunohistochemistry demonstrated that 4HR treatment significantly increased Glut4 and phosphorylated AMPKα (p-AMPKα) expression in diabetic muscle, indicating enhanced glucose uptake and metabolic activity. CONCLUSIONS 4HR effectively alleviates diabetes-induced sarcopenia by preserving muscle volume, enhancing glycogen storage, and upregulating Glut4 and p-AMPKα expression. These findings suggest that 4HR holds potential as a therapeutic agent for combating muscle wasting in diabetes.
Collapse
Affiliation(s)
- Dhouha Gaida
- Gangneung-Wonju National University KR, Gangneung-si, Gangwon-do, Republic of Korea
| | - Young-Wook Park
- Gangneung-Wonju National University KR, Gangneung-si, Gangwon-do, Republic of Korea
| | - Yei-Jin Kang
- Gangneung-Wonju National University KR, Gangneung-si, Gangwon-do, Republic of Korea
| | - Seong-Gon Kim
- Gangneung-Wonju National University KR, Gangneung-si, Gangwon-do, Republic of Korea.
| |
Collapse
|
10
|
Akhmetshina A, Schooltink L, Amor M, Kuentzel KB, Rainer S, Nandy A, Habisch H, Madl T, Rendina-Ruedy E, Leithner K, Vujić N, Kratky D. Loss or inhibition of lysosomal acid lipase in vitro leads to cholesteryl ester accumulation without affecting muscle formation or mitochondrial function. BBA ADVANCES 2024; 7:100135. [PMID: 39839442 PMCID: PMC11745973 DOI: 10.1016/j.bbadva.2024.100135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/23/2025] Open
Abstract
Skeletal muscle (SM) is essential for movement, stability, and overall body function, and it readily adapts to changes in energy demand. Myogenesis is energy-intensive and involves complex molecular and cellular events. We recently demonstrated that the absence of lysosomal acid lipase (LAL) in vivo significantly impacts the SM phenotype, primarily by disrupting energy homeostasis and reducing ATP production. As systemic LAL deficiency affects multiple organs, we hypothesized that the altered SM phenotype resulted from systemic rather than SM-specific loss of LAL activity. To distinguish between systemic and cell-intrinsic effects, we used primary myoblasts isolated from Lal-deficient (-/-) mice as well as C2C12 cells treated with the pharmacological inhibitor of LAL, Lalistat-2. We found a significant accumulation of cholesteryl esters in both models studied, highlighting the central role of LAL in lipid catabolism in the SM. However, lipid accumulation was absent under lipoprotein-deficient culture conditions. Neither genetic loss nor pharmacological inhibition of LAL affected myofiber formation or mitochondrial function in vitro, in contrast to what we observed in SM isolated from Lal-/- mice. Tracing [13C6]-labeled glucose in both cell culture models revealed only minor changes in tricarboxylic acid cycle metabolites. These results suggest that although LAL plays an essential role in lipid metabolism, its impact on the processes involved in muscle differentiation and cellular energy production is minor. We conclude that the cell-intrinsic effects of Lal-/- SM are unlikely to drive the SM phenotype observed in vivo.
Collapse
Affiliation(s)
- Alena Akhmetshina
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laszlo Schooltink
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Melina Amor
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Katharina B. Kuentzel
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Silvia Rainer
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Ananya Nandy
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hansjoerg Habisch
- Otto Loewi Research Center, Division of Medicinal Chemistry, Medical University of Graz, Graz, Austria
| | - Tobias Madl
- Otto Loewi Research Center, Division of Medicinal Chemistry, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Elizabeth Rendina-Ruedy
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Katharina Leithner
- BioTechMed-Graz, Graz, Austria
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Nemanja Vujić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
11
|
Kobayashi Y, Huang J, Barnett BK, Falcon CY, Falcon PA, Hirschberg CS, Fine DH, Ye Y, Shimizu E. Delayed Tooth Development and the Impaired Differentiation of Stem/Progenitor Cells in Incisors from Type 2 Diabetes Mice. Int J Mol Sci 2024; 25:13619. [PMID: 39769381 PMCID: PMC11728242 DOI: 10.3390/ijms252413619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Patients with diabetes mellitus (DM) have an increased risk of tooth decay caused by alterations in their tooth development and their oral environment, as well as a tendency to present with pulp infection due to compromised immune response. The present study analyzed the characteristic alterations in tooth development under DM conditions using incisors from db/db type 2 diabetic mouse model (T2DM mice). In micro-CT analyses, T2DM mice showed delayed dentin and enamel formation. Through transcriptomic analyses, pre-ameloblast- and pre-odontoblast-specific genes were found to be significantly decreased in the incisors of T2DM mice, whereas major ameloblast- and mature odontoblast-specific genes were not changed. Stem cell markers were decreased in T2DM mice compared to those from the control mice, suggesting that the stemness of dental pulp cells (DPCs) is attenuated in T2DM mice. In vitro analyses demonstrated that DPCs from T2DM mice have lower colony-forming units (CFU), slower propagation, and diminished differentiation characteristics compared to the control. These data suggest that T2DM conditions could impair the differentiation property of multiple progenitor/stem cells in the tooth, resulting in delayed tooth development in T2DM mice.
Collapse
Affiliation(s)
- Yoshifumi Kobayashi
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ 07103, USA; (Y.K.); (J.H.); (D.H.F.)
| | - Jia Huang
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ 07103, USA; (Y.K.); (J.H.); (D.H.F.)
| | - Brandon K. Barnett
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ 07103, USA (C.Y.F.); (P.A.F.); (C.S.H.)
| | - Carla Y. Falcon
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ 07103, USA (C.Y.F.); (P.A.F.); (C.S.H.)
| | - Paul A. Falcon
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ 07103, USA (C.Y.F.); (P.A.F.); (C.S.H.)
| | - Craig S. Hirschberg
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ 07103, USA (C.Y.F.); (P.A.F.); (C.S.H.)
| | - Daniel H. Fine
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ 07103, USA; (Y.K.); (J.H.); (D.H.F.)
| | - Yi Ye
- Bluestone Center for Clinical Research, New York University College of Dentistry, New York, NY 10010, USA;
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY 10010, USA
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Emi Shimizu
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ 07103, USA; (Y.K.); (J.H.); (D.H.F.)
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ 07103, USA (C.Y.F.); (P.A.F.); (C.S.H.)
| |
Collapse
|
12
|
Bahadoran Z, Mirmiran P, Ghasemi A. Type 2 diabetes-related sarcopenia: role of nitric oxide. Nutr Metab (Lond) 2024; 21:107. [PMID: 39695784 DOI: 10.1186/s12986-024-00883-z] [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: 09/07/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Sarcopenia, characterized by progressive and generalized loss of skeletal muscle (SkM) mass, strength, and physical performance, is a prevalent complication in type 2 diabetes (T2D). Nitric oxide (NO), a multifunctional gasotransmitter involved in whole-body glucose and insulin homeostasis, plays key roles in normal SkM physiology and function. Here, we highlight the role of NO in SkM mass maintenance and its potential contribution to the development of T2D-related sarcopenia. Physiologic NO level, primarily produced by sarcolemmal neuronal nitric oxide synthase (nNOSμ isoform), is involved in protein synthesis in muscle fibers and maintenance of SkM mass. The observed effect of nNOSμ on SkM mass is muscle-type specific and sex-dependent. Impaired NO homeostasis [due to a diminished nNOSμ-NO availability and excessive NO production through inducible NOS (iNOS) in response to atrophic stimuli, e.g., inflammatory cytokines] in SkM occurred during the development and progression of T2D, may cause sarcopenia. Theoretically, restoration of NO through nNOS overexpression, supplying NOS substrates (e.g., L-arginine and L-citrulline), phosphodiesterase (PDE) inhibition, and supplementation with NO donors (e.g., inorganic nitrate) may be potential therapeutic approaches to preserve SkM mass and prevents sarcopenia in T2D.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Micronutrient Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Sahid-Erabi St, Yemen St, Chamran Exp, P.O. Box 19395-4763, Tehran, Iran.
| |
Collapse
|
13
|
Zhao T, Mao W, Hu M, Yu Q, Peng X, Ji J, Qiu J, Wu J. Advances in sarcopenia and urologic disorders. Front Nutr 2024; 11:1475977. [PMID: 39568720 PMCID: PMC11578050 DOI: 10.3389/fnut.2024.1475977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
Sarcopenia is a loss of muscle strength, muscle mass, and function that can increase a patient's risk of injury, illness, and can even severely impair quality of life and increase a patient's risk of death. A growing body of research suggests that sarcopenia and urinary tract disorders are closely related. In this review, we aimed to emphasize the definition of skeletal sarcopenia, summarize the methods used to diagnose skeletal sarcopenia, discuss the advances in the study of sarcopenia in benign diseases of the urinary system, discuss the advances in the study of sarcopenia in malignant diseases of the urinary system. Sarcopenia and urologic diseases interact with each other; urologic diseases cause sarcopenia, and sarcopenia aggravates the condition of the original disease, thus falling into a vicious circle. This review provides a comprehensive understanding of sarcopenia in urologic diseases, which is very important for the management and prognosis of urologic diseases.
Collapse
Affiliation(s)
- Tonglei Zhao
- Southeast University School of Medicine, Nanjing, China
| | - Weipu Mao
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
- Department of Urology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University School of Medicine, Nanjing, China
| | - Mingjin Hu
- Department of Urology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University School of Medicine, Nanjing, China
| | - Qingyang Yu
- Southeast University School of Medicine, Nanjing, China
| | - Xinyang Peng
- Southeast University School of Medicine, Nanjing, China
| | - Jie Ji
- Southeast University School of Medicine, Nanjing, China
| | - Jianguo Qiu
- Department of Urology, Lianshui People's Hospital of Kangda College Affiliated to Nanjing Medical University, Jiang Su, China
| | - Jianping Wu
- Department of Urology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University School of Medicine, Nanjing, China
| |
Collapse
|
14
|
Wang C, Liu X, Hu X, Wu T, Duan R. Therapeutic targeting of GDF11 in muscle atrophy: Insights and strategies. Int J Biol Macromol 2024; 279:135321. [PMID: 39236952 DOI: 10.1016/j.ijbiomac.2024.135321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/29/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
The exploration of novel therapeutic avenues for skeletal muscle atrophy is imperative due to its significant health impact. Recent studies have spotlighted growth differentiation factor 11 (GDF11), a TGFβ superfamily member, for its rejuvenating role in reversing age-related tissue dysfunction. This review synthesizes current findings on GDF11, elucidating its distinct biological functions and the ongoing debates regarding its efficacy in muscle homeostasis. By addressing discrepancies in current research outcomes and its ambiguous role due to its homological identity to myostatin, a negative regulator of muscle mass, this review aims to clarify the role of GDF11 in muscle homeostasis and its potential as a therapeutic target for muscle atrophy. Through a thorough examination of GDF11's mechanisms and effects, this review provides insights that could pave the way for innovative treatments for muscle atrophy, emphasizing the need and strategies to boost endogenous GDF11 levels for therapeutic potential.
Collapse
Affiliation(s)
- Chuanzhi Wang
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Xiaocao Liu
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Xilong Hu
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Tao Wu
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Rui Duan
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China.
| |
Collapse
|
15
|
Christensen S, Robinson K, Thomas S, Williams DR. Dietary intake by patients taking GLP-1 and dual GIP/GLP-1 receptor agonists: A narrative review and discussion of research needs. OBESITY PILLARS 2024; 11:100121. [PMID: 39175746 PMCID: PMC11340591 DOI: 10.1016/j.obpill.2024.100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024]
Abstract
Background Obesity and type 2 diabetes mellitus (T2DM) are increasingly common in the United States and worldwide. Because both conditions are associated with serious health consequences, weight reduction is recommended by professional medical and nutrition societies to improve outcomes. Due to the striking efficacy of glucagon-like peptide receptor agonists (GLP-1RAs) and dual mechanism glucose-dependent insulinotropic polypeptide/glucagon-like peptide receptor agonists (GIP/GLP-1RAs) for weight reduction and glycemic control, there is increased utilization for patients with obesity and/or T2DM. Yet, the impact of these medications on dietary intake is less understood. Methods This narrative literature review summarizes clinical studies quantifying and characterizing dietary intake in people with obesity and/or T2DM using GLP-1 or GIP/GLP-1 RAs. Results Though data from these studies reveal that total caloric intake was reduced by 16-39 %, few studies evaluated the actual composition of the diet. Conclusions Further research is needed to understand the unique nutritional needs of adults on GLP-1 or dual GIP/GLP-1RAs and to support the development of nutritional guidelines for these individuals.
Collapse
Affiliation(s)
- Sandra Christensen
- Integrative Medical Weight Management, 2611 NE 125th St, Suite 100B, Seattle, WA, USA
| | - Katie Robinson
- Abbott Laboratories, 2900 Easton Square Place, ES1, Columbus, OH, USA
| | - Sara Thomas
- Abbott Laboratories, 2900 Easton Square Place, ES1, Columbus, OH, USA
| | | |
Collapse
|
16
|
Chan DC, Lin YC, Tzeng HP, Yang RS, Chiang MT, Liu SH. Exendin-4, a glucagon-like peptide-1 receptor agonist, alleviates muscular dysfunction and wasting in a streptozotocin-induced diabetic mouse model compared to metformin. Tissue Cell 2024; 89:102479. [PMID: 39018713 DOI: 10.1016/j.tice.2024.102479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/13/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
Diabetic muscular atrophy is becoming a fast-growing problem worldwide, including sarcopenia, which is associated with substantial mortality and morbidity risk. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have been marketed and suggested to exert protective effects on not only glycemic control but also diabetic complications in diabetic patients. In this study, we investigated the therapeutic use of GLP-1RAs exendin-4, compared to antidiabetic drug metformin, for the intervention of muscular dysfunction during diabetic conditions using a streptozotocin (STZ)-induced diabetic mouse model. The results showed that both exendin-4 and metformin could effectively alleviate hyperglycemia in diabetic mice, and also counteract diabetes-induced muscle weight loss, weaker grip, and changes in muscle fiber cross-sectional area distribution. Unexpectedly, exendin-4, but not metformin, enhanced the increased kidney weight and histological change in diabetic mice. Taken together, these findings suggest that both exendin-4 and metformin could effectively improve the diabetic hyperglycemia and muscular dysfunction; but exendin-4 may aggravate the nephropathy in STZ-induced diabetic mice.
Collapse
Affiliation(s)
- Ding-Cheng Chan
- Department of Geriatrics and Gerontology, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Yuan-Cheng Lin
- Institute of Toxicology, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Huei-Ping Tzeng
- Institute of Toxicology, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Rong-Sen Yang
- Department of Orthopedics, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Meng-Tsan Chiang
- Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan, Republic of China.
| | - Shing-Hwa Liu
- Institute of Toxicology, National Taiwan University, Taipei, Taiwan, Republic of China; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, Republic of China; Department of Pediatrics, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan, Republic of China.
| |
Collapse
|
17
|
Gong L, Tong T, Zhang E. Editorial: Exercise intervention on metabolic syndrome: focus on pyroptosis. Front Endocrinol (Lausanne) 2024; 15:1461283. [PMID: 39135620 PMCID: PMC11317435 DOI: 10.3389/fendo.2024.1461283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Affiliation(s)
- Lijing Gong
- Key Laboratory of Exercise and Physical Fitness of Ministry of Education, Beijing Sport University, Beijing, China
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Tao Tong
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Enming Zhang
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| |
Collapse
|
18
|
Qiao R, Guo J, Zhang C, Wang S, Fang J, Geng R, Kang SG, Huang K, Tong T. Diabetes-induced muscle wasting: molecular mechanisms and promising therapeutic targets. Crit Rev Food Sci Nutr 2024:1-17. [PMID: 39049742 DOI: 10.1080/10408398.2024.2382348] [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: 07/27/2024]
Abstract
Diabetes has become a serious public health crisis, presenting significant challenges to individuals worldwide. As the largest organ in the human body, skeletal muscle is a significant target of this chronic disease, yet muscle wasting as a complication of diabetes is still not fully understood and effective treatment methods have yet to be developed. Here, we discuss the targets involved in inducing muscle wasting under diabetic conditions, both validated targets and emerging targets. Diabetes-induced skeletal muscle wasting is known to involve changes in various signaling molecules and pathways, such as protein degradation pathways, protein synthesis pathways, mitochondrial function, and oxidative stress inflammation. Recent studies have shown that some of these present potential as promising therapeutic targets, including the neuregulin 1/epidermal growth factor receptor family, advanced glycation end-products, irisin, ferroptosis, growth differentiation factor 15 and more. This study's investigation and discussion of such pathways and their potential applications provides a theoretical basis for the development of clinical treatments for diabetes-induced muscle wasting and a foundation for continued focus on this disease.
Collapse
Affiliation(s)
- Ruixue Qiao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Jingya Guo
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Chengmei Zhang
- Guizhou Academy of Testing and Analysis, Guiyang, The People's Republic of China
| | - Sirui Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Jingjing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Ruixuan Geng
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Seong-Gook Kang
- Department of Food Engineering and Solar Salt Research Center, Mokpo National University, Muangun, Republic of Korea
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, The People's Republic of China
- Beijing Laboratory for Food Quality and Safety, Beijing, The People's Republic of China
| | - Tao Tong
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, The People's Republic of China
- Beijing Laboratory for Food Quality and Safety, Beijing, The People's Republic of China
| |
Collapse
|
19
|
Jeong YJ, Kim JH, Jung YJ, Kwak MS, Sung MH, Imm JY. KL-Biome (Postbiotic Formulation of Lactiplantibacillus plantarum KM2) Improves Dexamethasone-Induced Muscle Atrophy in Mice. Int J Mol Sci 2024; 25:7499. [PMID: 39000606 PMCID: PMC11242066 DOI: 10.3390/ijms25137499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Sarcopenia refers to an age-related decrease in muscle mass and strength. The gut-muscle axis has been proposed as a promising target to alleviate muscle atrophy. The effect of KL-Biome-a postbiotic preparation comprising heat-killed Lactiplantibacillus plantarum KM-2, its metabolites, and an excipient (soybean powder)-on muscle atrophy was evaluated using dexamethasone (DEX)-induced atrophic C2C12 myoblasts and C57BL/6J mice. KL-Biome significantly downregulated the expression of genes (Atrogin-1 and MuRF1) associated with skeletal muscle degradation but increased the anabolic phosphorylation of FoxO3a, Akt, and mTOR in C2C12 cells. Oral administration of KL-Biome (900 mg/kg) for 8 weeks significantly improved muscle mass, muscle function, and serum lactate dehydrogenase levels in DEX-treated mice. KL-Biome administration increased gut microbiome diversity and reversed DEX-mediated gut microbiota alterations. Furthermore, it significantly increased the relative abundances of the genera Subdologranulum, Alistipes, and Faecalibacterium prausnitzii, which are substantially involved in short-chain fatty acid production. These findings suggest that KL-Biome exerts beneficial effects on muscle atrophy by regulating gut microbiota.
Collapse
Affiliation(s)
- Yu-Jin Jeong
- Department of Foods and Nutrition, Kookmin University, Seoul 02707, Republic of Korea;
| | - Jong-Hoon Kim
- KookminBio Corporation, Seoul 02826, Republic of Korea; (J.-H.K.); (Y.-J.J.); (M.-S.K.); (M.-H.S.)
| | - Ye-Jin Jung
- KookminBio Corporation, Seoul 02826, Republic of Korea; (J.-H.K.); (Y.-J.J.); (M.-S.K.); (M.-H.S.)
| | - Mi-Sun Kwak
- KookminBio Corporation, Seoul 02826, Republic of Korea; (J.-H.K.); (Y.-J.J.); (M.-S.K.); (M.-H.S.)
| | - Moon-Hee Sung
- KookminBio Corporation, Seoul 02826, Republic of Korea; (J.-H.K.); (Y.-J.J.); (M.-S.K.); (M.-H.S.)
| | - Jee-Young Imm
- Department of Foods and Nutrition, Kookmin University, Seoul 02707, Republic of Korea;
| |
Collapse
|
20
|
Hetherington-Rauth M, Johnson E, Migliavacca E, Langsetmo L, Hepple RT, Ryan TE, Ferrucci L, Breuillé D, Corthesy J, Lane NE, Feige JN, Napoli N, Tramontana F, Orwoll ES, Cawthon PM. The Mediating Role of Kynurenine Pathway Metabolites on the Relationship Between Inflammation and Muscle Mass in Oldest-Old Men. J Gerontol A Biol Sci Med Sci 2024; 79:glae131. [PMID: 38761087 PMCID: PMC11163921 DOI: 10.1093/gerona/glae131] [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: 12/13/2023] [Indexed: 05/20/2024] Open
Abstract
Tryptophan (TRP) metabolites along the kynurenine (KYN) pathway (KP) have been found to influence muscle. Proinflammatory cytokines are known to stimulate the degradation of TRP down the KP. Given that both inflammation and KP metabolites have been connected with loss of muscle, we assessed the potential mediating role of KP metabolites on inflammation and muscle mass in older men. Five hundred and five men (85.0 ± 4.2 years) from the Osteoporotic Fractures in Men cohort study with measured D3-creatine dilution (D3Cr) muscle mass, KP metabolites, and inflammation markers (C-reactive protein [CRP], alpha-1-acid glycoprotein [AGP] and a subsample [n = 305] with interleukin [IL-6, IL-1β, IL-17A] and tumor necrosis factor-α [TNF-α]) were included in the analysis. KP metabolites and inflammatory markers were measured using liquid chromatography-tandem mass spectrometry and immunoassays, respectively. 23%-92% of the inverse relationship between inflammatory markers and D3Cr muscle mass was mediated by KP metabolites (indirect effect p < .05). 3-hydroxyanthranilic acid (3-HAA), quinolinic acid (QA), TRP, xanthurenic acid (XA), KYN/TRP, 3-hydroxykynurenine (3-HK)/3-HAA, QA/3-HAA, and nicotinamide (NAM)/QA mediated the AGP relationship. 3-HAA, QA, KYN/TRP, 3-HK/XA, HKr ratio, 3-HK/3-HAA, QA/3-HAA, and NAM/QA mediated the CRP. KYN/TRP, 3-HK/XA, and NAM/QA explained the relationship for IL-6 and 3-HK/XA and QA/3-HAA for TNF-α. No mediation effect was observed for the other cytokines (indirect effect p > .05). KP metabolites, particularly higher ratios of KYN/TRP, 3-HK/XA, 3-HK/3-HAA, QA/3-HAA, and a lower ratio of NAM/QA, mediated the relationship between inflammation and low muscle mass. Our preliminary cross-sectional data suggest that interventions to alter D3Cr muscle mass may focus on KP metabolites rather than inflammation per se.
Collapse
Affiliation(s)
| | - Eileen Johnson
- California Pacific Medical Center, Research Institute, San Francisco, California, USA
| | | | - Lisa Langsetmo
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Russell T Hepple
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Gaithersburg, Maryland, USA
| | - Denis Breuillé
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Lausanne, Switzerland
| | - John Corthesy
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Lausanne, Switzerland
| | - Nancy E Lane
- Division of Rheumatology, Department of Medicine, University of California, Davis, Davis, California, USA
| | - Jérôme N Feige
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nicola Napoli
- Department of Medicine and Surgery, Research Unit of Endocrinology and Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
- Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Flavia Tramontana
- Department of Medicine and Surgery, Research Unit of Endocrinology and Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
- Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Eric S Orwoll
- Department of Medicine, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Peggy M Cawthon
- California Pacific Medical Center, Research Institute, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
21
|
Umek N, Pušnik L, Ugwoke CK, Šink Ž, Horvat S, Janáček J, Cvetko E. Skeletal muscle myosin heavy chain expression and 3D capillary network changes in streptozotocin-induced diabetic female mice. BIOMOLECULES & BIOMEDICINE 2024; 24:582-592. [PMID: 37902457 PMCID: PMC11088899 DOI: 10.17305/bb.2023.9843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 10/31/2023]
Abstract
It is not well-understood how type 1 diabetes (T1DM) affects skeletal muscle histological phenotype, particularly capillarisation. This study aimed to analyze skeletal muscle myosin heavy chain (MyHC) fibre type changes and 3D capillary network characteristics in experimental T1DM mice. Female C57BL/6J-OlaHsd mice were categorized into streptozotocin (STZ)-induced diabetic (n = 12) and age-matched non-diabetic controls (n =12). The muscle fibre phenotype of the soleus, gluteus maximus, and gastrocnemius muscles were characterized based on the expression of MyHC isoforms, while capillaries of the gluteus maximus were assessed with immunofluorescence staining, confocal laser microscopy and 3D image analysis. STZ-induced diabetic mice exhibited elevated glucose levels, reduced body weight, and prolonged thermal latency, verifying the T1DM phenotype. In both T1DM and non-diabetic mice, the gluteus maximus and gastrocnemius muscles predominantly expressed fast-twitch type 2b fibers, with no significant differences noted. However, the soleus muscle in non-diabetic mice had a greater proportion of type 2a fibers and comparable type 1 fiber densities (26.2 ± 14.6% vs 21.9 ± 13.5%) relative to diabetic mice. T1DM mice showed reduced fiber diameters (P = 0.026), and the 3D capillary network analysis indicated a higher capillary length per muscle volume in the gluteus maximus of diabetic mice compared to controls (P < 0.05). Overall, T1DM induced significant changes in the skeletal muscle, including shifts in MyHC fibre types, decreased fibre diameters, and increased relative capillarisation, possibly due to muscle fibre atrophy. Our findings emphasize the superior detail provided by the 3D analytical method for characterizing skeletal muscle capillary architecture, highlighting caution in interpreting 2D data for capillary changes in T1DM.
Collapse
Affiliation(s)
- Nejc Umek
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Luka Pušnik
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | - Žiga Šink
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Simon Horvat
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Jiří Janáček
- Laboratory of Biomathematics, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Erika Cvetko
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
22
|
Özen SD, Kir S. Ectodysplasin A2 receptor signaling in skeletal muscle pathophysiology. Trends Mol Med 2024; 30:471-483. [PMID: 38443222 DOI: 10.1016/j.molmed.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 03/07/2024]
Abstract
Skeletal muscle is essential in generating mechanical force and regulating energy metabolism and body temperature. Pathologies associated with muscle tissue often lead to impaired physical activity and imbalanced metabolism. Recently, ectodysplasin A2 receptor (EDA2R) signaling has been shown to promote muscle loss and glucose intolerance. Upregulated EDA2R expression in muscle tissue was associated with aging, denervation, cancer cachexia, and muscular dystrophies. Here, we describe the roles of EDA2R signaling in muscle pathophysiology, including muscle atrophy, insulin resistance, and aging-related sarcopenia. We also discuss the EDA2R pathway, which involves EDA-A2 as the ligand and nuclear factor (NF)κB-inducing kinase (NIK) as a downstream mediator, and the therapeutic potential of targeting these proteins in the treatment of muscle wasting and metabolic dysfunction.
Collapse
Affiliation(s)
- Sevgi Döndü Özen
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Serkan Kir
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey.
| |
Collapse
|
23
|
Ji Y, Lin J, Liu R, Wang K, Chang M, Gao Z, Liu B, Shen Y, Zhu J, Yao X, Qi L, Sun H. Celecoxib attenuates hindlimb unloading-induced muscle atrophy via suppressing inflammation, oxidative stress and ER stress by inhibiting STAT3. Inflammopharmacology 2024; 32:1633-1646. [PMID: 38451396 DOI: 10.1007/s10787-024-01454-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024]
Abstract
Improving inflammation may serve as useful therapeutic interventions for the hindlimb unloading-induced disuse muscle atrophy. Celecoxib is a selective non-steroidal anti-inflammatory drug. We aimed to determine the role and mechanism of celecoxib in hindlimb unloading-induced disuse muscle atrophy. Celecoxib significantly attenuated the decrease in soleus muscle mass, hindlimb muscle function and the shift from slow- to fast-twitch muscle fibers caused by hindlimb unloading in rats. Importantly, celecoxib inhibited the increased expression of inflammatory factors, macrophage infiltration in damaged soleus muscle. Mechanistically, Celecoxib could significantly reduce oxidative stress and endoplasmic reticulum stress in soleus muscle of unloaded rats. Furthermore, celecoxib inhibited muscle proteolysis by reducing the levels of MAFbx, MuRF1, and autophagy related proteins maybe by inhibiting the activation of pro-inflammatory STAT3 pathway in vivo and in vitro. This study is the first to demonstrate that celecoxib can attenuate disuse muscle atrophy caused by hindlimb unloading via suppressing inflammation, oxidative stress and endoplasmic reticulum stress probably, improving target muscle function and reversing the shift of muscle fiber types by inhibiting STAT3 pathways-mediated inflammatory cascade. This study not only enriches the potential molecular regulatory mechanisms, but also provides new potential therapeutic targets for disuse muscle atrophy.
Collapse
Affiliation(s)
- Yanan Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, Nantong, Jiangsu Province, 226001, People's Republic of China
| | - Junfei Lin
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, People's Republic of China
| | - Ruiqi Liu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, 226001, 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, Medical School of Nantong University, Nantong University, Nantong, Jiangsu Province, 226001, People's Republic of China
| | - Mengyuan Chang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, Nantong, Jiangsu Province, 226001, People's Republic of China
| | - Zihui Gao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, Nantong, Jiangsu Province, 226001, People's Republic of China
| | - Boya 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, Medical School of Nantong University, Nantong University, Nantong, Jiangsu Province, 226001, People's Republic of China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, Nantong, Jiangsu Province, 226001, People's Republic of China
| | - Jianwei Zhu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, People's Republic of China
| | - Xinlei Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, Nantong, Jiangsu Province, 226001, People's Republic of China.
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, People's Republic of China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, Nantong, Jiangsu Province, 226001, People's Republic of China.
| |
Collapse
|
24
|
Wan R, Liu S, Feng X, Luo W, Zhang H, Wu Y, Chen S, Shang X. The Revolution of exosomes: From biological functions to therapeutic applications in skeletal muscle diseases. J Orthop Translat 2024; 45:132-139. [PMID: 38544740 PMCID: PMC10966453 DOI: 10.1016/j.jot.2024.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 11/11/2024] Open
Abstract
Skeletal muscle diseases, a broad category encompassing a myriad of afflictions such as acute muscle injury and muscular dystrophies, pose a significant health burden globally. These conditions often lead to muscle weakness, compromised mobility, and a diminished quality of life. In light of this, innovative and effective therapeutic strategies are fervently sought after. Exosomes, naturally extracellular vesicles with a diameter of 30-150 nm, pervade biological fluids. These microscopic entities harbor a host of biological molecules, including proteins, nucleic acids, and lipids, bearing a significant resemblance to their parent cells. The roles they play in the biological theater are manifold, influencing crucial physiological and pathological processes within the organism. In the context of skeletal muscle diseases, their potential extends beyond these roles, as they present a promising therapeutic target and a vehicle for targeted drug delivery. This potentially paves the way for significant clinical applications. This review aims to elucidate the mechanisms underpinning exosome action, their myriad biological functions, and the strides made in exosome research and application. A comprehensive exploration of the part played by exosomes in skeletal muscle repair and regeneration is undertaken. In addition, we delve into the use of exosomes in the therapeutic landscape of skeletal muscle diseases, providing a valuable reference for a deeper understanding of exosome applications in this realm. The concluding section encapsulates the prospective avenues for exosome research and the promising future they hold, underscoring the tremendous potential these diminutive vesicles possess in the field of skeletal muscle diseases. The Translational Potential of this Article. The comprehensive exploration of exosome's diverse biological functions and translational potential in the context of skeletal muscle diseases presented in this review underscores their promising future as a therapeutic target with significant clinical applications, thus paving the way for innovative and effective therapeutic strategies in this realm.
Collapse
Affiliation(s)
- Renwen Wan
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shan Liu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xinting Feng
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wei Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Hanli Zhang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yang Wu
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiliang Shang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|
25
|
Yan X, Fu P, Zhang Y, Ling D, Reynolds L, Hua W, Wang Z, Ma F, Li B, Yu J, Liu Y, Gong L, Zhang E. MCC950 Ameliorates Diabetic Muscle Atrophy in Mice by Inhibition of Pyroptosis and Its Synergistic Effect with Aerobic Exercise. Molecules 2024; 29:712. [PMID: 38338456 PMCID: PMC10856337 DOI: 10.3390/molecules29030712] [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: 12/22/2023] [Revised: 01/17/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Diabetic muscle atrophy is an inflammation-related complication of type-2 diabetes mellitus (T2DM). Even though regular exercise prevents further deterioration of atrophic status, there is no effective mediator available for treatment and the underlying cellular mechanisms are less explored. In this study, we investigated the therapeutic potential of MCC950, a specific, small-molecule inhibitor of NLRP3, to treat pyroptosis and diabetic muscle atrophy in mice. Furthermore, we used MCC950 to intervene in the protective effects of aerobic exercise against muscle atrophy in diabetic mice. Blood and gastrocnemius muscle (GAS) samples were collected after 12 weeks of intervention and the atrophic state was assessed. We initially corroborated a diabetic muscle atrophy phenotype in db/db mice (D) by comparison with control m/m mice (W) by examining parameters such as fasting blood glucose (D vs. W: 24.47 ± 0.45 mmol L-1 vs. 4.26 ± 0.6 mmol L-1, p < 0.05), grip strength (D vs. W: 166.87 ± 15.19 g vs. 191.76 ± 14.13 g, p < 0.05), exercise time (D vs. W: 1082.38 ± 104.67 s vs. 1716 ± 168.55 s, p < 0.05) and exercise speed to exhaustion (D vs. W: 24.25 ± 2.12 m min-1 vs. 34.75 ± 2.66 m min-1, p < 0.05), GAS wet weight (D vs. W: 0.07 ± 0.01 g vs. 0.13 ± 0.01 g, p < 0.05), the ratio of GAS wet weight to body weight (D vs. W: 0.18 ± 0.01% vs. 0.54 ± 0.02%, p < 0.05), and muscle fiber cross-sectional area (FCSA) (D vs. W: 1875 ± 368.19 µm2 vs. 2747.83 ± 406.44 µm2, p < 0.05). We found that both MCC950 (10 mg kg-1) treatment and exercise improved the atrophic parameters that had deteriorated in the db/db mice, inhibited serum inflammatory markers and significantly attenuated pyroptosis in atrophic GAS. In addition, a combined MCC950 treatment with exercise (DEI) exhibited a further improvement in glucose uptake capacity and muscle performance. This combined treatment also improved the FCSA of GAS muscle indicated by Laminin immunofluorescence compared to the group with the inhibitor treatment alone (DI) (DEI vs. DI: 2597 ± 310.97 vs. 1974.67 ± 326.15 µm2, p < 0.05) or exercise only (DE) (DEI vs. DE: 2597 ± 310.97 vs. 2006.33 ± 263.468 µm2, p < 0.05). Intriguingly, the combination of MCC950 treatment and exercise significantly reduced NLRP3-mediated inflammatory factors such as cleaved-Caspase-1, GSDMD-N and prevented apoptosis and pyroptosis in atrophic GAS. These findings for the first time demonstrate that targeting NLRP3-mediated pyroptosis with MCC950 improves diabetic muscle homeostasis and muscle function. We also report that inhibiting pyroptosis by MCC950 can enhance the beneficial effects of aerobic exercise on diabetic muscle atrophy. Since T2DM and muscle atrophy are age-related diseases, the young mice used in the current study do not seem to fully reflect the characteristics of diabetic muscle atrophy. Considering the fragile nature of db/db mice and for the complete implementation of the exercise intervention, we used relatively young db/db mice and the atrophic state in the mice was thoroughly confirmed. Taken together, the current study comprehensively investigated the therapeutic effect of NLRP3-mediated pyroptosis inhibited by MCC950 on diabetic muscle mass, strength and exercise performance, as well as the synergistic effects of MCC950 and exercise intervention, therefore providing a novel strategy for the treatment of the disease.
Collapse
Affiliation(s)
- Xiaoyu Yan
- Key Laboratory of Exercise and Physical Fitness of Ministry of Education, Beijing Sport University, Beijing 100084, China; (X.Y.); (J.Y.)
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
| | - Pengyu Fu
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
- Department of Physical Education, Northwestern Polytechnical University, Xi’an 710072, China
| | - Yimin Zhang
- Key Laboratory of Exercise and Physical Fitness of Ministry of Education, Beijing Sport University, Beijing 100084, China; (X.Y.); (J.Y.)
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
| | - Dongmei Ling
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
| | - Lewis Reynolds
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, 21428 Malmö, Sweden (E.Z.)
- NanoLund Center for NanoScience, Lund University, 22100 Lund, Sweden
| | - Weicheng Hua
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
| | - Zhiyuan Wang
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
| | - Fangyuan Ma
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
- School of Life Sciences, Nankai University, Tianjin 300071, China
| | - Boxuan Li
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Jingjing Yu
- Key Laboratory of Exercise and Physical Fitness of Ministry of Education, Beijing Sport University, Beijing 100084, China; (X.Y.); (J.Y.)
| | - Yujia Liu
- School of Sport Science, Beijing Sport University, Beijing 100084, China; (P.F.); (D.L.); (W.H.); (Z.W.); (F.M.); (B.L.); (Y.L.)
- Institute of Physical Education, Jiangsu Normal University, Xuzhou 221116, China
| | - Lijing Gong
- Key Laboratory of Exercise and Physical Fitness of Ministry of Education, Beijing Sport University, Beijing 100084, China; (X.Y.); (J.Y.)
| | - Enming Zhang
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, 21428 Malmö, Sweden (E.Z.)
- NanoLund Center for NanoScience, Lund University, 22100 Lund, Sweden
| |
Collapse
|
26
|
Jiang H, Liu B, Lin J, Xue T, Han Y, Lu C, Zhou S, Gu Y, Xu F, Shen Y, Xu L, Sun H. MuSCs and IPCs: roles in skeletal muscle homeostasis, aging and injury. Cell Mol Life Sci 2024; 81:67. [PMID: 38289345 PMCID: PMC10828015 DOI: 10.1007/s00018-023-05096-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/01/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024]
Abstract
Skeletal muscle is a highly specialized tissue composed of myofibres that performs crucial functions in movement and metabolism. In response to external stimuli and injuries, a range of stem/progenitor cells, with muscle stem cells or satellite cells (MuSCs) being the predominant cell type, are rapidly activated to repair and regenerate skeletal muscle within weeks. Under normal conditions, MuSCs remain in a quiescent state, but become proliferative and differentiate into new myofibres in response to injury. In addition to MuSCs, some interstitial progenitor cells (IPCs) such as fibro-adipogenic progenitors (FAPs), pericytes, interstitial stem cells expressing PW1 and negative for Pax7 (PICs), muscle side population cells (SPCs), CD133-positive cells and Twist2-positive cells have been identified as playing direct or indirect roles in regenerating muscle tissue. Here, we highlight the heterogeneity, molecular markers, and functional properties of these interstitial progenitor cells, and explore the role of muscle stem/progenitor cells in skeletal muscle homeostasis, aging, and muscle-related diseases. This review provides critical insights for future stem cell therapies aimed at treating muscle-related diseases.
Collapse
Affiliation(s)
- Haiyan Jiang
- 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, 226001, Jiangsu, People's Republic of China
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Boya 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, 226001, Jiangsu, People's Republic of China
| | - Junfei Lin
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Tong Xue
- Department of Paediatrics, Medical School of Nantong University, Nantong University, Nantong, 226001, People's Republic of China
| | - Yimin Han
- Department of Paediatrics, Medical School of Nantong University, Nantong University, Nantong, 226001, People's Republic of China
| | - Chunfeng Lu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu, People's Republic of China
| | - Songlin Zhou
- 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, 226001, Jiangsu, People's Republic of China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu, People's Republic of 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, 226001, Jiangsu, People's Republic of China.
| | - Lingchi Xu
- 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, 226001, Jiangsu, People's Republic of 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, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
27
|
Deng C, Lu C, Wang K, Chang M, Shen Y, Yang X, Sun H, Yao X, Qiu C, Xu F. Celecoxib ameliorates diabetic sarcopenia by inhibiting inflammation, stress response, mitochondrial dysfunction, and subsequent activation of the protein degradation systems. Front Pharmacol 2024; 15:1344276. [PMID: 38313305 PMCID: PMC10834620 DOI: 10.3389/fphar.2024.1344276] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 01/05/2024] [Indexed: 02/06/2024] Open
Abstract
Aim: Diabetic sarcopenia leads to disability and seriously affects the quality of life. Currently, there are no effective therapeutic strategies for diabetic sarcopenia. Our previous studies have shown that inflammation plays a critical role in skeletal muscle atrophy. Interestingly, the connection between chronic inflammation and diabetic complications has been revealed. However, the effects of non-steroidal anti-inflammatory drug celecoxib on diabetic sarcopenia remains unclear. Materials and Methods: The streptozotocin (streptozotocin)-induced diabetic sarcopenia model was established. Rotarod test and grip strength test were used to assess skeletal muscle function. Hematoxylin and eosin and immunofluorescence staining were performed to evaluate inflammatory infiltration and the morphology of motor endplates in skeletal muscles. Succinate dehydrogenase (SDH) staining was used to determine the number of succinate dehydrogenase-positive muscle fibers. Dihydroethidium staining was performed to assess the levels of reactive oxygen species (ROS). Western blot was used to measure the levels of proteins involved in inflammation, oxidative stress, endoplasmic reticulum stress, ubiquitination, and autophagic-lysosomal pathway. Transmission electron microscopy was used to evaluate mitophagy. Results: Celecoxib significantly ameliorated skeletal muscle atrophy, improving skeletal muscle function and preserving motor endplates in diabetic mice. Celecoxib also decreased infiltration of inflammatory cell, reduced the levels of IL-6 and TNF-α, and suppressed the activation of NF-κB, Stat3, and NLRP3 inflammasome pathways in diabetic skeletal muscles. Celecoxib decreased reactive oxygen species levels, downregulated the levels of Nox2 and Nox4, upregulated the levels of GPX1 and Nrf2, and further suppressed endoplasmic reticulum stress by inhibiting the activation of the Perk-EIF-2α-ATF4-Chop in diabetic skeletal muscles. Celecoxib also inhibited the levels of Foxo3a, Fbx32 and MuRF1 in the ubiquitin-proteasome system, as well as the levels of BNIP3, Beclin1, ATG7, and LC3Ⅱ in the autophagic-lysosomal system, and celecoxib protected mitochondria and promoted mitochondrial biogenesis by elevating the levels of SIRT1 and PGC1-α, increased the number of SDH-positive fibers in diabetic skeletal muscles. Conclusion: Celecoxib improved diabetic sarcopenia by inhibiting inflammation, oxidative stress, endoplasmic reticulum stress, and protecting mitochondria, and subsequently suppressing proteolytic systems. Our study provides evidences for the molecular mechanism and treatment of diabetic sarcopenia, and broaden the way for the new use of celecoxib in diabetic sarcopenia.
Collapse
Affiliation(s)
- Chunyan Deng
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People’s Hospital of Nantong City, Nantong, China
- 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, China
| | - Chunfeng Lu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People’s Hospital of Nantong City, Nantong, 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, China
| | - Mengyuan Chang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 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, China
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 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, 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, China
| | - Chunjian Qiu
- Department of Endocrinology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People’s Hospital of Nantong City, Nantong, China
| |
Collapse
|
28
|
Lopez-Pedrosa JM, Camprubi-Robles M, Guzman-Rolo G, Lopez-Gonzalez A, Garcia-Almeida JM, Sanz-Paris A, Rueda R. The Vicious Cycle of Type 2 Diabetes Mellitus and Skeletal Muscle Atrophy: Clinical, Biochemical, and Nutritional Bases. Nutrients 2024; 16:172. [PMID: 38202001 PMCID: PMC10780454 DOI: 10.3390/nu16010172] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/30/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024] Open
Abstract
Today, type 2 diabetes mellitus (T2DM) and skeletal muscle atrophy (SMA) have become increasingly common occurrences. Whether the onset of T2DM increases the risk of SMA or vice versa has long been under investigation. Both conditions are associated with negative changes in skeletal muscle health, which can, in turn, lead to impaired physical function, a lowered quality of life, and an increased risk of mortality. Poor nutrition can exacerbate both T2DM and SMA. T2DM and SMA are linked by a vicious cycle of events that reinforce and worsen each other. Muscle insulin resistance appears to be the pathophysiological link between T2DM and SMA. To explore this association, our review (i) compiles evidence on the clinical association between T2DM and SMA, (ii) reviews mechanisms underlying biochemical changes in the muscles of people with or at risk of T2DM and SMA, and (iii) examines how nutritional therapy and increased physical activity as muscle-targeted treatments benefit this population. Based on the evidence, we conclude that effective treatment of patients with T2DM-SMA depends on the restoration and maintenance of muscle mass. We thus propose that regular intake of key functional nutrients, along with guidance for physical activity, can help maintain euglycemia and improve muscle status in all patients with T2DM and SMA.
Collapse
Affiliation(s)
| | | | | | | | - Jose Manuel Garcia-Almeida
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital (IBIMA), Malaga University, 29010 Malaga, Spain;
| | - Alejandro Sanz-Paris
- Nutrition Unit, Universitary Hospital Miguel Servet, Isabel the Catholic 1-3, 50009 Zaragoza, Spain;
| | - Ricardo Rueda
- Abbott Nutrition R&D, 18004 Granada, Spain; (M.C.-R.); (A.L.-G.); (R.R.)
| |
Collapse
|
29
|
Appell CR, Jiwan NC, Wang R, Shen CL, Luk HY. Ginger Supplementation Attenuated Mitochondrial Fusion and Improved Skeletal Muscle Size in Type 2 Diabetic Rats. In Vivo 2024; 38:73-81. [PMID: 38148056 PMCID: PMC10756480 DOI: 10.21873/invivo.13412] [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: 09/28/2023] [Revised: 10/24/2023] [Accepted: 11/06/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND/AIM Oxidative stress, regulated by SOD2 and mitochondrial dynamics, contributes to muscle atrophy in diabetes. Ginger root extract (GRE) reduces oxidative stress. However, its effect on oxidative stress, mitochondrial dynamics, and muscle atrophy is not known in the diabetic muscle. This study examined the effect of GRE on intramuscular oxidative stress, mitochondrial dynamics, and muscle size in diabetic rats. MATERIALS AND METHODS Twenty-six male Sprague-Dawley rats were randomly divided into control diet (CON; n=10), high-fat diet with one dose of 35 mg/kg streptozotocin (HFD; n=9), and high-fat diet with one dose of 35 mg/kg streptozotocin and 0.75% w/w GRE (GRE; n=7) fed for seven weeks. Subsequently, the muscle was analyzed for cross-sectional area (CSA), H2O2 concentration, and DRP-1, MFN2, Parkin, PINK1, SOD2 mRNA. Additionally, the protein levels of SOD2, DRP-1, DRP-1ser616, LC3AB, MFN2, OPA1, Parkin, and PINK1 were analyzed. CSA, H2O2 concentration, and gene and protein expression levels were analyzed using a one-way ANOVA. Correlations among intramuscular H2O2, CSA, and SOD2 protein were assessed using Pearson's bivariate correlation test. RESULTS In the soleus, the GRE group had a greater CSA and lower intramuscular H2O2 concentration compared to the HFD group. Compared to the HFD group, the GRE group had higher SOD2 and DRP-1 mRNA levels and lower MFN2 and total OPA1 protein levels. H2O2 concentration was negatively correlated with CSA and positively correlated with SOD2. CONCLUSION GRE attenuated intramuscular H2O2, mitochondrial fusion, and muscle size loss. These findings suggest that GRE supplementation in diabetic rats reduces oxidative stress, which may contribute to muscle size preservation.
Collapse
Affiliation(s)
- Casey R Appell
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, U.S.A
| | - Nigel C Jiwan
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, U.S.A
| | - Rui Wang
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX, U.S.A
| | - Chwan-Li Shen
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX, U.S.A
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX, U.S.A
| | - Hui-Ying Luk
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, U.S.A.;
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX, U.S.A
| |
Collapse
|
30
|
Chen J, Feng X, Zhou X, Li Y. Role of the tripartite motif-containing (TRIM) family of proteins in insulin resistance and related disorders. Diabetes Obes Metab 2024; 26:3-15. [PMID: 37726973 DOI: 10.1111/dom.15294] [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: 06/13/2023] [Revised: 08/27/2023] [Accepted: 09/05/2023] [Indexed: 09/21/2023]
Abstract
Emerging evidence suggests that the ubiquitin-mediated degradation of insulin-signalling-related proteins may be involved in the development of insulin resistance and its related disorders. Tripartite motif-containing (TRIM) proteins, a superfamily belonging to the E3 ubiquitin ligases, are capable of controlling protein levels and function by ubiquitination, which is essential for the modulation of insulin sensitivity. Recent research has indicated that some of these TRIMs act as key regulatory factors of metabolic disorders such as type 2 diabetes mellitus, obesity, nonalcoholic fatty liver disease, and atherosclerosis. This review provides a comprehensive overview of the latest evidence linking TRIMs to the regulation of insulin resistance and its related disorders, their roles in regulating multiple signalling pathways or cellular processes, such as insulin signalling pathways, peroxisome proliferator-activated receptor signalling pathways, glucose and lipid metabolism, the inflammatory response, and cell cycle control, as well as recent advances in the development of TRIM-targeted drugs.
Collapse
Affiliation(s)
- Jianrong Chen
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Clinical Research Centre for Endocrine and Metabolic disease, Nanchang, China
- Jiangxi Branch of National Clinical Research Centre for Metabolic disease, Nanchang, China
| | - Xianjie Feng
- Evidence-based Medicine Research Centre, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Xu Zhou
- Evidence-based Medicine Research Centre, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yong Li
- Department of Anaesthesiology, Medical Centre of Anaesthesiology and Pain, First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
31
|
Yuan X, Xue F, Yu Y, Cao X, Han Y, Wang F, Zhong L. The molecular mechanism of sepsis-induced diaphragm dysfunction. J Thorac Dis 2023; 15:6831-6847. [PMID: 38249924 PMCID: PMC10797340 DOI: 10.21037/jtd-23-1680] [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: 11/02/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024]
Abstract
Background No effective drugs for the treatment of sepsis-induced diaphragm dysfunction are currently available. Therefore, it is particularly important to clarify the molecular regulatory mechanism of this condition and subsequently implement effective treatment and prevention of sepsis-induced diaphragm dysfunction. Methods A mouse model of diaphragm dysfunction was established via injection of lipopolysaccharide (LPS). An RNA-sequencing (RNA-seq) technique was used to detect the differentially expressed genes (DEGs) in the diaphragms of mice. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed for functional analysis of DEGs. The protein-protein interaction network obtained from the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) website was imported into Cytoscape, the key molecular regulatory network was constructed with CytoNCA, the ClueGo plugin was further used to analyze the core regulatory pathways of key molecular, and finally, the iRegulon plugin was used to the identify key transcription factors. Results The genes upregulated after LPS treatment were involved in biological processes and pathways related to immune response; the genes downregulated after LPS treatment were mainly correlated with the muscle contraction. The expressions of several inflammation-related genes were upregulated after LPS treatment, of which tumor necrosis factor (Tnf), interleukin (Il)-1β, and Il-6 assumed a core regulatory role in the network; meanwhile, the downregulated key genes included Col1a1, Uqcrfs1, Sdhb, and ATP5a1, among others. These key regulatory factors participated in the activation of Toll-like receptor (TLR) signaling pathway, nuclear factor (NF)-κB signaling pathway, and TNF signaling pathway as well as the inhibition of oxidative phosphorylation pathway, cardiac muscle contraction pathway, and citrate cycle pathway. Finally, RelA, IRF1, and STAT3, were identified as the key regulators in the early stage of diaphragmatic inflammatory response. Conclusions Sepsis-induced diaphragm dysfunction in mice is closely correlated with the activation of TLR signaling pathway, NF-κB signaling pathway, and TNF signaling pathway and the inhibition of oxidative phosphorylation pathway, cardiac muscle contraction pathway, and citrate cycle pathway. Our findings provide insight into the molecular mechanism of sepsis-induced diaphragm dysfunction in mice and provide a promising new strategy for targeted treatment of diaphragm dysfunction.
Collapse
Affiliation(s)
- Xiaosa Yuan
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Fangsu Xue
- Department of Respiration, Binhai County People’s Hospital, Yancheng, China
| | - Yunchi Yu
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaowen Cao
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yimin Han
- Department of Pediatrics, Medical College, Nantong University, Nantong, China
| | - Fei Wang
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Lou Zhong
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
32
|
Zhong Y, Liu X, Lin T. The Relationship between Muscle Ultrasound Parameters and Diabetic Peripheral Neuropathy among Patients with Type 2 Diabetes Mellitus: A Cross-Sectional Study. J Diabetes Res 2023; 2023:8897065. [PMID: 38148834 PMCID: PMC10751167 DOI: 10.1155/2023/8897065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 12/28/2023] Open
Abstract
Background Muscle dysfunction is an early complication of diabetic peripheral neuropathy (DPN). As a convenient and low-cost tool, muscle ultrasound has been used to assess muscle quality and muscle mass. However, the relationship between different muscle ultrasound parameters and DPN is unclear. Objectives This study was designed to investigate the relationship between ultrasound parameters of different muscles and DPN among patients with type 2 diabetes mellitus, including the rectus femoris (RF), tibialis anterior (TA), and medial head of gastrocnemius (MG). Materials and Methods The research enrolled 90 patients with type 2 diabetes mellitus (T2DM). All images were attained from both sides. Muscle measurements contained muscle thickness (MT), cross-sectional area (CSA), echo intensity (EI), and corrected EI. The binary logistic regression and multiple linear regression were used to investigate the association between muscle ultrasound parameters and DPN or vibration perception threshold (VPT). Results EI, corrected EI, MT of MG, and EI of TA were associated with DPN separately after adjusting other clinical variates. Among these muscle parameters, the EI of MG had a better predictive value (OR: 1.114, 95% CI: 1.039, 1.196) of DPN. Combined with CSA of RF, peripheral artery disease (PAD), and sex, the corrected EI of MG was associated with the vibration perception threshold (VPT) (standard β = 0.242, p < 0.001), better than the EI of MG (standard β = 0.215, p = 0.002). Conclusions MG (MT, EI, and corrected EI) and TA (EI) were associated with DPN, respectively. CSA of RF and corrected EI or EI of MG combined with PAD and sex were associated with VPT significantly, which supported that muscle ultrasound might be a substantial quantitative tool for detecting the exercise benefits for DPN.
Collapse
Affiliation(s)
- Yanling Zhong
- Department of Ultrasound, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China
- Shantou University Medical College, Shantou, Guangdong 515000, China
| | - Xiaojia Liu
- Department of Ultrasound, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China
| | - Teng Lin
- Department of Ultrasound, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China
| |
Collapse
|
33
|
Lin J, Cai Y, Wang J, Liu R, Qiu C, Huang Y, Liu B, Yang X, Zhou S, Shen Y, Wang W, Zhu J. Transcriptome sequencing promotes insights on the molecular mechanism of SKP-SC-EVs mitigating denervation-induced muscle atrophy. Mol Biol Rep 2023; 51:9. [PMID: 38085347 DOI: 10.1007/s11033-023-08952-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: 08/28/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND Complex pathophysiological changes accompany denervation-induced skeletal muscle atrophy, but no effective treatment strategies exist. Our previous study indicated that extracellular vesicles derived from skin-derived precursors-derived Schwann cells (SKP-SC-EVs) can effectively mitigate denervation-induced muscle atrophy. However, the specific molecular mechanism remains unclear. METHODS AND RESULTS In this study, we used bioinformatics methods to scrutinize the impact of SKP-SC-EVs on gene expression in denervation-induced skeletal muscle atrophy. We found that SKP-SC-EVs altered the expression of 358 genes in denervated skeletal muscles. The differentially expressed genes were predominantly participated in biological processes, including cell cycle, inflammation, immunity, and adhesion, and signaling pathways, such as FoxO and PI3K.Using the Molecular Complex Detection (MCODE) plugin, we identified the two clusters with the highest score: cluster 1 comprised 37 genes, and Cluster 2 consisted of 24 genes. Then, fifty hub genes were identified using CytoHubba. The intersection of Hub genes and genes obtained by MCODE showed that all 23 genes related to the cell cycle in Cluster 1 were hub genes, and 5 genes in Cluster 2 were hub genes and associated with inflammation. CONCLUSIONS Overall, the differentially expressed genes in denervated skeletal muscle following SKP-SC-EVs treatment are primarily linked to the cell cycle and inflammation. Consequently, promoting proliferation and inhibiting inflammation may be the critical process in which SKP-SC-EVs delay denervation-induced muscle atrophy. Our findings contribute to a better understanding of the molecular mechanism of SKP-SC-EVs delaying denervation-induced muscle atrophy, offering a promising new avenue for muscle atrophy treatment.
Collapse
Affiliation(s)
- Junfei Lin
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Yong Cai
- Department of Neurology, Binhai County People's Hospital, Yancheng, Jiangsu Province, 224500, P. R. China
| | - Jian Wang
- Department of Clinical Laboratory, Nantong Third Hospital Affiliated to Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Ruiqi Liu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, 226001, Jiangsu Province, P. R. China
| | - Chong Qiu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, 226001, Jiangsu Province, P. R. China
| | - Yan Huang
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, 226001, Jiangsu Province, P. R. China
| | - Boya 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
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Songlin Zhou
- 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.
| | - Wei 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.
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, P. R. China.
| | - Jianwei Zhu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, P. R. China.
| |
Collapse
|
34
|
Sun J, Zhou H, Chen Z, Zhang H, Cao Y, Yao X, Chen X, Liu B, Gao Z, Shen Y, Qi L, Sun H. Altered m6A RNA methylation governs denervation-induced muscle atrophy by regulating ubiquitin proteasome pathway. J Transl Med 2023; 21:845. [PMID: 37996930 PMCID: PMC10668433 DOI: 10.1186/s12967-023-04694-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Denervation-induced muscle atrophy is complex disease involving multiple biological processes with unknown mechanisms. N6-methyladenosine (m6A) participates in skeletal muscle physiology by regulating multiple levels of RNA metabolism, but its impact on denervation-induced muscle atrophy is still unclear. Here, we aimed to explore the changes, functions, and molecular mechanisms of m6A RNA methylation during denervation-induced muscle atrophy. METHODS During denervation-induced muscle atrophy, the m6A immunoprecipitation sequencing (MeRIP-seq) as well as enzyme-linked immunosorbent assay analysis were used to detect the changes of m6A modified RNAs and the involved biological processes. 3-deazidenosine (Daa) and R-2-hydroxyglutarate (R-2HG) were used to verify the roles of m6A RNA methylation. Through bioinformatics analysis combined with experimental verification, the regulatory roles and mechanisms of m6A RNA methylation had been explored. RESULTS There were many m6A modified RNAs with differences during denervation-induced muscle atrophy, and overall, they were mainly downregulated. After 72 h of denervation, the biological processes involved in the altered mRNA with m6A modification were mainly related to zinc ion binding, ubiquitin protein ligase activity, ATP binding and sequence-specific DNA binding and transcription coactivator activity. Daa reduced overall m6A levels in healthy skeletal muscles, which reduced skeletal muscle mass. On the contrary, the increase in m6A levels mediated by R-2HG alleviated denervation induced muscle atrophy. The m6A RNA methylation regulated skeletal muscle mass through ubiquitin-proteasome pathway. CONCLUSION This study indicated that decrease in m6A RNA methylation was a new symptom of denervation-induced muscle atrophy, and confirmed that targeting m6A alleviated denervation-induced muscle atrophy.
Collapse
Affiliation(s)
- Junjie 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, 226001, Jiangsu, People's Republic of China
| | - Hai Zhou
- Department of Neurosurgery, Binhai County People's Hospital, Yancheng, 224500, Jiangsu, People's Republic of China
| | - Zehao Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Han Zhang
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, 226001, China
| | - Yanzhe Cao
- 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, 226001, Jiangsu, People's Republic of 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, 226001, Jiangsu, People's Republic of China
| | - Xin Chen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Boya 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, 226001, Jiangsu, People's Republic of China
| | - Zihui Gao
- 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, 226001, Jiangsu, People's Republic of 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, 226001, Jiangsu, People's Republic of China.
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of 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, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
35
|
Kumar A, Prajapati P, Raj V, Kim SC, Mishra V, Raorane CJ, Raj R, Kumar D, Kushwaha S. Salbutamol ameliorates skeletal muscle wasting and inflammatory markers in streptozotocin (STZ)-induced diabetic rats. Int Immunopharmacol 2023; 124:110883. [PMID: 37666067 DOI: 10.1016/j.intimp.2023.110883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/13/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023]
Abstract
Diabetes accelerates muscle atrophy, leading to the deterioration of skeletal muscles. This study aimed to assess the potential of the β2-adrenoceptor agonist, salbutamol (SLB), to alleviate muscle atrophy in streptozotocin (STZ)-induced diabetic rats. Male Sprague Dawley rats were randomized into four groups (n=6): control, SLB, STZ (55 mg/kg, single i.p.), and STZ + SLB (6 mg/kg, orally for 4 weeks). After the final SLB dose, animals underwent tests to evaluate muscle strength and coordination, including forelimb grip strength, wire-hanging, actophotometer, rotarod, and footprint assessments. Rats were then sacrificed, and serum and gastrocnemius (GN) muscles were collected for further analysis. Serum evaluations included proinflammatory markers (tumor necrosis factor α, interleukin-1β, interleukin-6), muscle markers (creatine kinase, myostatin), testosterone, and lipidemic markers. Muscle oxidative stress (malonaldehyde, protein carbonyl), antioxidants (glutathione, catalase, superoxide dismutase), and histology were also performed. Additionally, 1H nuclear magnetic resonance serum profiling was conducted. SLB notably enhanced muscle grip strength, coordination, and antioxidant levels, while reduced proinflammatory markers and oxidative stress in STZ-induced diabetic rats. Reduced serum muscle biomarkers, increased testosterone, restored lipidemic levels, and improved muscle cellular architecture indicated SLB's positive effect on muscle condition in diabetic rats. Metabolomics profiling revealed that the STZ group significantly increased the phenylalanine-to-tyrosine ratio (PTR), lactate-to-pyruvate ratio (LPR), acetate, succinate, isobutyrate, and histidine. SLB administration restored these perturbed serum metabolites in the STZ-induced diabetic group. In conclusion, salbutamol significantly protected against skeletal muscle wasting in STZ-induced diabetic rats.
Collapse
Affiliation(s)
- Anand Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Priyanka Prajapati
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Vinit Raj
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Seong-Cheol Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India.
| | | | - Ritu Raj
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow 226014, India
| | - Dinesh Kumar
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow 226014, India
| | - Sapana Kushwaha
- National Institutes of Pharmaceutical Education and Research (NIPER), Raebareli, Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow 226002, India.
| |
Collapse
|
36
|
Muvhulawa N, Mazibuko-Mbeje SE, Ndwandwe D, Silvestri S, Ziqubu K, Moetlediwa MT, Mthembu SXH, Marnewick JL, Van der Westhuizen FH, Nkambule BB, Basson AK, Tiano L, Dludla PV. Sarcopenia in a type 2 diabetic state: Reviewing literature on the pathological consequences of oxidative stress and inflammation beyond the neutralizing effect of intracellular antioxidants. Life Sci 2023; 332:122125. [PMID: 37769808 DOI: 10.1016/j.lfs.2023.122125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/15/2023] [Accepted: 09/23/2023] [Indexed: 10/03/2023]
Abstract
Sarcopenia remains one of the major pathological features of type 2 diabetes (T2D), especially in older individuals. This condition describes gradual loss of muscle mass, strength, and function that reduces the overall vitality and fitness, leading to increased hospitalizations and even fatalities to those affected. Preclinical evidence indicates that dysregulated mitochondrial dynamics, together with impaired activity of the NADPH oxidase system, are the major sources of oxidative stress that drive skeletal muscle damage in T2D. While patients with T2D also display relatively higher levels of circulating inflammatory markers in the serum, including high sensitivity-C-reactive protein, interleukin-6, and tumor necrosis factor-α that are independently linked with the deterioration of muscle function and sarcopenia in T2D. In fact, beyond reporting on the pathological consequences of both oxidative stress and inflammation, the current review highlights the importance of strengthening intracellular antioxidant systems to preserve muscle mass, strength, and function in individuals with T2D.
Collapse
Affiliation(s)
- Ndivhuwo Muvhulawa
- Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho 2735, South Africa
| | | | - Duduzile Ndwandwe
- Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Khanyisani Ziqubu
- Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho 2735, South Africa
| | - Marakiya T Moetlediwa
- Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho 2735, South Africa
| | | | - Jeanine L Marnewick
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Bellville 7535, South Africa
| | | | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Albertus K Basson
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Phiwayinkosi V Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa.
| |
Collapse
|
37
|
Valentin B, Maes-Festen D, Schoufour J, Oppewal A. Sarcopenia predicts 5-year mortality in older adults with intellectual disabilities. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2023; 67:1161-1173. [PMID: 37608512 DOI: 10.1111/jir.13078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 06/06/2023] [Accepted: 07/25/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND People with intellectual disabilities (ID) have a lower life expectancy than their peers without ID. A contributing factor to the lower life expectancy and early mortality could be sarcopenia: low muscle mass and low muscle function. In the general population, sarcopenia strongly predicts early mortality, but this association is unknown in people with ID. Therefore, this study aims to explore the association between sarcopenia and 5-year mortality in older adults with ID. METHODS In the Healthy Ageing and Intellectual Disabilities (HA-ID) study, the prevalence of sarcopenia was measured at baseline among 884 older adults (≥50 years) with ID. All-cause mortality was measured over a 5-year follow-up period. Univariable and multivariable Cox proportional hazard models were applied to determine the association between sarcopenia (no sarcopenia, pre-sarcopenia, sarcopenia, severe sarcopenia) and early mortality, adjusted for age, sex, level of ID, presence of Down syndrome, and co-morbidity (chronic obstructive pulmonary disease, diabetes type 2 and metabolic syndrome). RESULTS The unadjusted hazard ratio (HR) for sarcopenia was 2.28 [95% confidence interval (CI) 1.48-3.42], P < 0.001), and 2.40 (95% CI 1.40-4.10, P = 0.001) for severe sarcopenia. When adjusted for age, sex, level of ID, and Down syndrome, sarcopenia (HR = 1.72, 95% CI 1.08-2.75, P = 0.022) and severe sarcopenia (HR = 1.86, 95% CI 1.07-3.23, P = 0.028) were significantly associated with early mortality. When additionally adjusted for co-morbidity, the adjusted HR decreased to 1.62 (95% CI 1.02-2.59, P = 0.043) and 1.81 (95% CI 1.04-3.15, P = 0.035) for sarcopenia and severe sarcopenia, respectively. CONCLUSION Sarcopenia is an independent risk factor for early mortality in older adults with ID over a 5-year follow-up period. Our results stress the need to delay the incidence and development of sarcopenia in older adults with ID.
Collapse
Affiliation(s)
- B Valentin
- Department of General Practice, Intellectual Disability Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Faculty of Sports and Nutrition, Faculty of Health, Center of Expertise Urban Vitality, Amsterdam University of Applied Sciences, Amsterdam, The Netherlands
| | - D Maes-Festen
- Department of General Practice, Intellectual Disability Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - J Schoufour
- Faculty of Sports and Nutrition, Faculty of Health, Center of Expertise Urban Vitality, Amsterdam University of Applied Sciences, Amsterdam, The Netherlands
| | - A Oppewal
- Department of General Practice, Intellectual Disability Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
38
|
Hong SM, Lee EY, Park J, Kim J, Kim SY. Aerobic Exercise Ameliorates Muscle Atrophy Induced by Methylglyoxal via Increasing Gastrocnemius and Extensor Digitorum Longus Muscle Sensitivity. Biomol Ther (Seoul) 2023; 31:573-582. [PMID: 37562979 PMCID: PMC10468420 DOI: 10.4062/biomolther.2023.130] [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: 07/17/2023] [Revised: 07/28/2023] [Accepted: 07/30/2023] [Indexed: 08/12/2023] Open
Abstract
Muscle atrophy is characterized by the loss of muscle function. Many efforts are being made to prevent muscle atrophy, and exercise is an important alternative. Methylglyoxal is a well-known causative agent of metabolic diseases and diabetic complications. This study aimed to evaluate whether methylglyoxal induces muscle atrophy and to evaluate the ameliorative effect of moderate-intensity aerobic exercise in a methylglyoxal-induced muscle atrophy animal model. Each mouse was randomly divided into three groups: control, methylglyoxal-treated, and methylglyoxal-treated within aerobic exercise. In the exercise group, each mouse was trained on a treadmill for 2 weeks. On the last day, all groups were evaluated for several atrophic behaviors and skeletal muscles, including the soleus, plantaris, gastrocnemius, and extensor digitorum longus were analyzed. In the exercise group, muscle mass was restored, causing in attenuation of muscle atrophy. The gastrocnemius and extensor digitorum longus muscles showed improved fiber cross-sectional area and reduced myofibrils. Further, they produced regulated atrophy-related proteins (i.e., muscle atrophy F-box, muscle RING-finger protein-1, and myosin heavy chain), indicating that aerobic exercise stimulated their muscle sensitivity to reverse skeletal muscle atrophy. In conclusion, shortness of the gastrocnemius caused by methylglyoxal may induce the dynamic imbalance of skeletal muscle atrophy, thus methylglyoxal may be a key target for treating skeletal muscle atrophy. To this end, aerobic exercise may be a powerful tool for regulating methylglyoxal-induced skeletal muscle atrophy.
Collapse
Affiliation(s)
- Seong-Min Hong
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Eun Yoo Lee
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Jinho Park
- Department of Exercise Rehabilitation, Gachon University, Incheon 21936, Republic of Korea
| | - Jiyoun Kim
- Department of Exercise Rehabilitation, Gachon University, Incheon 21936, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
- Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Republic of Korea
| |
Collapse
|
39
|
Liu SH, Lin WH, Tzeng HP, Chiang MT. Attenuation of diabetes-mediated muscle atrophy in rats by fish oil enriched omega-3 polyunsaturated fatty acids supplementation. J Food Drug Anal 2023; 31:458-472. [PMID: 39666277 PMCID: PMC10629917 DOI: 10.38212/2224-6614.3468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/12/2023] [Indexed: 12/13/2024] Open
Abstract
Diabetes is associated with an increased risk of muscle wasting/atrophy, which adversely affects quality of life. We hypothesized that long term supplementation of fish oil may have protective effects against sarcopenia or muscle atrophy in streptozotocin (STZ) and high-fat (HF) diet-induced diabetic rat model. Wistar rats at age of 7 weeks were injected with saline or STZ to induce hyperglycemia. After one week, they were fed on a normal control diet or HF diet with/without supplementation of fish oil for 18 weeks. Feeding diabetic rats with a fish oil-enriched diet alleviated body weight loss and the impaired glucose tolerance using OGTT test. Although fish oil did not improve the decreased muscle mass, the muscle atrophy induced by diabetes was attenuated by fish oil in gastrocnemius, soleus, tibialis anterior, and extensor digitorum longus muscles. Fish oil supplementation reversed the decreased expression of phospho (p)-AKT, pmTOR, and p-p70s6k, which are molecules related to protein synthesis. Besides, protein degradation-related signaling pathways were inhibited by fish oil, such as increasing p-FoxO1 and decreasing Atrogin-1 and MURF1 protein expression. Fish oil down-regulated the expression of autophagy-related molecules including ATG5, p62, and LC3B II/I ratio, which may result in less muscle atrophy. Inflammation-related signaling regulators including TNF-α, NF-κB, AGEs, and RAGE were suppressed by fish oil supplementation as well. Moreover, the down-regulated p-AMPKα, SIRT1, and PGC-1 in diabetic rats were counteracted by fish oil, which may improve mitochondrial function and further block FoxO action. These data suggest that long-term fish oil supplementation exerts protective effects against diabetes-induced muscle atrophy, which may in turn ameliorate insulin resistance and impaired glucose tolerance.
Collapse
Affiliation(s)
- Shing-Hwa Liu
- Institute of Toxicology, National Taiwan University, Taipei,
Taiwan
- Department of Pediatrics, College of Medicine and Hospital, National Taiwan University, Taipei,
Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung,
Taiwan
| | - Wei-Hsuan Lin
- Department of Food Science, National Taiwan Ocean University, Keelung,
Taiwan
| | - Huei-Ping Tzeng
- Institute of Toxicology, National Taiwan University, Taipei,
Taiwan
| | - Meng-Tsan Chiang
- Department of Food Science, National Taiwan Ocean University, Keelung,
Taiwan
| |
Collapse
|
40
|
Paez HG, Ferrandi PJ, Pitzer CR, Mohamed JS, Alway SE. Loss of NOR-1 represses muscle metabolism through mTORC1-mediated signaling and mitochondrial gene expression in C2C12 myotubes. FASEB J 2023; 37:e23050. [PMID: 37389860 DOI: 10.1096/fj.202202029r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 05/26/2023] [Accepted: 06/08/2023] [Indexed: 07/01/2023]
Abstract
Gene expression of the NR4A nuclear orphan receptor NOR-1 is reduced in obesity and in human skeletal muscle during disuse. It has been well established that NOR-1 is highly responsive to both aerobic and resistance exercise and NOR-1 overexpression is coincident with a plethora of metabolic benefits. However, it is unclear whether loss of NOR-1 contributes to inappropriate metabolic signaling in skeletal muscle that could lead to insulin resistance. The purpose of this study was to elucidate the impact of NOR-1 deficiency on C2C12 metabolic signaling. Changes in gene expression after siRNA-mediated NOR-1 knockdown in C2C12 myotubes were determined by qPCR and bioinformatic analysis of RNA-Seq data. Our RNA-Seq data identified several metabolic targets regulated by NOR-1 and implicates NOR-1 as a modulator of mTORC1 signaling via Akt-independent mechanisms. Furthermore, pathway analysis revealed NOR-1 knockdown perturbs the insulin resistance and insulin sensitivity pathways. Taken together, these data suggest skeletal muscle NOR-1 deficiency may contribute to altered metabolic signaling that is consistent with metabolic disease. We postulate that strategies that improve NOR-1 may be important to offset the negative impact that inactivity, obesity, and type 2 diabetes have on mitochondria and muscle metabolism.
Collapse
Affiliation(s)
- Hector G Paez
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Division of Regenerative and Rehabilitation Sciences, Center for Muscle, Metabolism and Neuropathology, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Peter J Ferrandi
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Division of Regenerative and Rehabilitation Sciences, Center for Muscle, Metabolism and Neuropathology, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Laboratory of Muscle and Nerve, Department of Diagnostic and Health Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Christopher R Pitzer
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Division of Regenerative and Rehabilitation Sciences, Center for Muscle, Metabolism and Neuropathology, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Junaith S Mohamed
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Division of Regenerative and Rehabilitation Sciences, Center for Muscle, Metabolism and Neuropathology, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Laboratory of Muscle and Nerve, Department of Diagnostic and Health Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Stephen E Alway
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Division of Regenerative and Rehabilitation Sciences, Center for Muscle, Metabolism and Neuropathology, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
41
|
Chen X, Ji Y, Liu R, Zhu X, Wang K, Yang X, Liu B, Gao Z, Huang Y, Shen Y, Liu H, Sun H. Mitochondrial dysfunction: roles in skeletal muscle atrophy. J Transl Med 2023; 21:503. [PMID: 37495991 PMCID: PMC10373380 DOI: 10.1186/s12967-023-04369-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/18/2023] [Indexed: 07/28/2023] Open
Abstract
Mitochondria play important roles in maintaining cellular homeostasis and skeletal muscle health, and damage to mitochondria can lead to a series of pathophysiological changes. Mitochondrial dysfunction can lead to skeletal muscle atrophy, and its molecular mechanism leading to skeletal muscle atrophy is complex. Understanding the pathogenesis of mitochondrial dysfunction is useful for the prevention and treatment of skeletal muscle atrophy, and finding drugs and methods to target and modulate mitochondrial function are urgent tasks in the prevention and treatment of skeletal muscle atrophy. In this review, we first discussed the roles of normal mitochondria in skeletal muscle. Importantly, we described the effect of mitochondrial dysfunction on skeletal muscle atrophy and the molecular mechanisms involved. Furthermore, the regulatory roles of different signaling pathways (AMPK-SIRT1-PGC-1α, IGF-1-PI3K-Akt-mTOR, FoxOs, JAK-STAT3, TGF-β-Smad2/3 and NF-κB pathways, etc.) and the roles of mitochondrial factors were investigated in mitochondrial dysfunction. Next, we analyzed the manifestations of mitochondrial dysfunction in muscle atrophy caused by different diseases. Finally, we summarized the preventive and therapeutic effects of targeted regulation of mitochondrial function on skeletal muscle atrophy, including drug therapy, exercise and diet, gene therapy, stem cell therapy and physical therapy. This review is of great significance for the holistic understanding of the important role of mitochondria in skeletal muscle, which is helpful for researchers to further understanding the molecular regulatory mechanism of skeletal muscle atrophy, and has an important inspiring role for the development of therapeutic strategies for muscle atrophy targeting mitochondria in the future.
Collapse
Affiliation(s)
- Xin Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yanan Ji
- 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, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Ruiqi Liu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Xucheng Zhu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of 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, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Boya 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, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Zihui Gao
- 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, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yan Huang
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of 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, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Hua Liu
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, 55 Ninghai Middle Road, Nantong, Jiangsu, 226600, People's Republic of 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, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
42
|
Taldaev A, Savina AD, Olicheva VV, Ivanov SV, Terekhov RP, Ilyasov IR, Zhevlakova AK, Selivanova IA. Protective Properties of Spheroidal Taxifolin Form in Streptozotocin-Induced Diabetic Rats. Int J Mol Sci 2023; 24:11962. [PMID: 37569337 PMCID: PMC10418707 DOI: 10.3390/ijms241511962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
One of the key factors in the pathogenesis of diabetes and its complications is oxidative stress. To inhibit this process, antioxidants may be helpful. Herein, we focused on the protective properties of taxifolin spheroidal form (TS) in the streptozotocin rat model of diabetes mellitus. After 4 weeks of treatment with TS, the fasting blood glucose level of the diabetic animals decreased by 12% compared with the level right after the injection of streptozotocin. While the feed intake in the untreated diabetic rats increased by 5.3% compared with the healthy group, the TS-treated group showed a pronounced 15.3% decrease. Therapeutic administration of TS has a protective effect on the pancreas and the liver against the cytotoxic action of streptozotocin. The plasma antioxidant capacity of all diabetic groups appeared to be approximately 15% lower than in healthy rats with no significant difference between the TS-treated and untreated diabetic animals. Apparently, this can be attributed to taxifolin and plasma proteins binding. These data demonstrate the potential of TS in antidiabetic therapy.
Collapse
Affiliation(s)
- Amir Taldaev
- Laboratory of Nanobiotechnology, Institute of Biomedical Chemistry, Pogodinskaya Str. 10/8, 119121 Moscow, Russia
| | - Anastasiya D. Savina
- Nelubin Institute of Pharmacy, Sechenov First Moscow State Medical University, Trubetskaya Str. 8/2, 119991 Moscow, Russia
| | - Vera V. Olicheva
- Nelubin Institute of Pharmacy, Sechenov First Moscow State Medical University, Trubetskaya Str. 8/2, 119991 Moscow, Russia
| | - Sergey V. Ivanov
- Laboratory of Psychopharmacology, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia
| | - Roman P. Terekhov
- Nelubin Institute of Pharmacy, Sechenov First Moscow State Medical University, Trubetskaya Str. 8/2, 119991 Moscow, Russia
| | - Igor R. Ilyasov
- Nelubin Institute of Pharmacy, Sechenov First Moscow State Medical University, Trubetskaya Str. 8/2, 119991 Moscow, Russia
| | - Anastasiya K. Zhevlakova
- Nelubin Institute of Pharmacy, Sechenov First Moscow State Medical University, Trubetskaya Str. 8/2, 119991 Moscow, Russia
| | - Irina A. Selivanova
- Nelubin Institute of Pharmacy, Sechenov First Moscow State Medical University, Trubetskaya Str. 8/2, 119991 Moscow, Russia
| |
Collapse
|
43
|
Kim Y, Kim W, Kim SH, Sim KS, Kim KH, Cho KH, Kwon GS, Lee JB, Kim JH. Protective Effects of Hemp ( Cannabis sativa) Root Extracts against Insulin-Deficient Diabetes Mellitus In Mice. Molecules 2023; 28:molecules28093814. [PMID: 37175224 PMCID: PMC10179809 DOI: 10.3390/molecules28093814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
The pharmacological potential of industrial hemp (Cannabis sativa) has been widely studied. However, the majority of studies have focused on cannabidiol, isolated from the inflorescence and leaf of the plant. In the present study, we evaluated the anti-diabetic potential of hemp root water (HWE) and ethanol extracts (HEE) in streptozotocin (STZ)-induced insulin-deficient diabetic mice. The administration of HWE and HEE ameliorated hyperglycemia and improved glucose homeostasis and islet function in STZ-treated mice (p < 0.05). HWE and HEE suppressed β-cell apoptosis and cytokine-induced inflammatory signaling in the pancreas (p < 0.05). Moreover, HWE and HEE normalized insulin-signaling defects in skeletal muscles and apoptotic response in the liver and kidney induced by STZ (p < 0.05). Gas chromatography-mass spectrometry analysis of HWE and HEE showed possible active compounds which might be responsible for the observed anti-diabetic potential. These findings indicate the possible mechanisms by which hemp root extracts protect mice against insulin-deficient diabetes, and support the need for further studies geared towards the application of hemp root as a novel bioactive material.
Collapse
Affiliation(s)
- Yujeong Kim
- Department of Food Science and Biotechnology, Andong National University, Andong 36729, Republic of Korea
| | - Wonhee Kim
- Department of Food Science and Biotechnology, Andong National University, Andong 36729, Republic of Korea
| | - Soo-Hyun Kim
- Life Science Team, Kyochon F&B Co., Ltd., Osan 18150, Republic of Korea
| | - Kyu-Sang Sim
- Life Science Team, Kyochon F&B Co., Ltd., Osan 18150, Republic of Korea
| | - Ki-Hyun Kim
- Department of Research Project, Gyeongbuk Institute for Bioindustry, Andong 36618, Republic of Korea
| | - Kiu-Hyung Cho
- Department of Research Project, Gyeongbuk Institute for Bioindustry, Andong 36618, Republic of Korea
| | - Gi-Seok Kwon
- Department of Horticulture & Medicinal Plant, Andong National University, Andong 36729, Republic of Korea
| | - Jung-Bok Lee
- Research Institute of Food & Bio, BHNBIO Co., Ltd., Jincheon-gun 27850, Republic of Korea
| | - Jun-Ho Kim
- Department of Food Science and Biotechnology, Andong National University, Andong 36729, Republic of Korea
| |
Collapse
|
44
|
Jun L, Robinson M, Geetha T, Broderick TL, Babu JR. Prevalence and Mechanisms of Skeletal Muscle Atrophy in Metabolic Conditions. Int J Mol Sci 2023; 24:ijms24032973. [PMID: 36769296 PMCID: PMC9917738 DOI: 10.3390/ijms24032973] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Skeletal muscle atrophy is prevalent in a myriad of pathological conditions, such as diabetes, denervation, long-term immobility, malnutrition, sarcopenia, obesity, Alzheimer's disease, and cachexia. This is a critically important topic that has significance in the health of the current society, particularly older adults. The most damaging effect of muscle atrophy is the decreased quality of life from functional disability, increased risk of fractures, decreased basal metabolic rate, and reduced bone mineral density. Most skeletal muscle in humans contains slow oxidative, fast oxidative, and fast glycolytic muscle fiber types. Depending on the pathological condition, either oxidative or glycolytic muscle type may be affected to a greater extent. This review article discusses the prevalence of skeletal muscle atrophy and several mechanisms, with an emphasis on high-fat, high-sugar diet patterns, obesity, and diabetes, but including other conditions such as sarcopenia, Alzheimer's disease, cancer cachexia, and heart failure.
Collapse
Affiliation(s)
- Lauren Jun
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| | - Megan Robinson
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| | - Thangiah Geetha
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
- Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA
| | - Tom L. Broderick
- Department of Physiology, Laboratory of Diabetes and Exercise Metabolism, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| | - Jeganathan Ramesh Babu
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
- Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA
- Correspondence: ; Tel.: +1-223-844-3840
| |
Collapse
|
45
|
Wang K, Liu Q, Tang M, Qi G, Qiu C, Huang Y, Yu W, Wang W, Sun H, Ni X, Shen Y, Fang X. Chronic kidney disease-induced muscle atrophy: Molecular mechanisms and promising therapies. Biochem Pharmacol 2023; 208:115407. [PMID: 36596414 DOI: 10.1016/j.bcp.2022.115407] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/02/2023]
Abstract
Chronic kidney disease (CKD) is a high-risk chronic catabolic disease due to its high morbidity and mortality. CKD is accompanied by many complications, leading to a poor quality of life, and serious complications may even threaten the life of CKD patients. Muscle atrophy is a common complication of CKD. Muscle atrophy and sarcopenia in CKD patients have complex pathways that are related to multiple mechanisms and related factors. This review not only discusses the mechanisms by which inflammation, oxidative stress, mitochondrial dysfunction promote CKD-induced muscle atrophy but also explores other CKD-related complications, such as metabolic acidosis, vitamin D deficiency, anorexia, and excess angiotensin II, as well as other related factors that play a role in CKD muscle atrophy, such as insulin resistance, hormones, hemodialysis, uremic toxins, intestinal flora imbalance, and miRNA. We highlight potential treatments and drugs that can effectively treat CKD-induced muscle atrophy in terms of complication treatment, nutritional supplementation, physical exercise, and drug intervention, thereby helping to improve the prognosis and quality of life of CKD patients.
Collapse
Affiliation(s)
- Kexin Wang
- Department of Nephrology, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, PR China; 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, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Qingyuan Liu
- Department of Endocrinology, Binhai County People's Hospital, Yancheng, Jiangsu Province 224500, PR China
| | - Mingyu Tang
- Xinglin College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Guangdong Qi
- Department of Endocrinology, Binhai County People's Hospital, Yancheng, Jiangsu Province 224500, PR China
| | - Chong Qiu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Yan Huang
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Weiran Yu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Wei 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, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, PR China; Department of Pathology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, PR 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, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Xuejun Ni
- Department of Ultrasound Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, PR 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, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, PR China.
| | - Xingxing Fang
- Department of Nephrology, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, PR China.
| |
Collapse
|
46
|
Potential Therapeutic Strategies for Skeletal Muscle Atrophy. Antioxidants (Basel) 2022; 12:antiox12010044. [PMID: 36670909 PMCID: PMC9854691 DOI: 10.3390/antiox12010044] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/13/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The maintenance of muscle homeostasis is vital for life and health. Skeletal muscle atrophy not only seriously reduces people's quality of life and increases morbidity and mortality, but also causes a huge socioeconomic burden. To date, no effective treatment has been developed for skeletal muscle atrophy owing to an incomplete understanding of its molecular mechanisms. Exercise therapy is the most effective treatment for skeletal muscle atrophy. Unfortunately, it is not suitable for all patients, such as fractured patients and bedridden patients with nerve damage. Therefore, understanding the molecular mechanism of skeletal muscle atrophy is crucial for developing new therapies for skeletal muscle atrophy. In this review, PubMed was systematically screened for articles that appeared in the past 5 years about potential therapeutic strategies for skeletal muscle atrophy. Herein, we summarize the roles of inflammation, oxidative stress, ubiquitin-proteasome system, autophagic-lysosomal pathway, caspases, and calpains in skeletal muscle atrophy and systematically expound the potential drug targets and therapeutic progress against skeletal muscle atrophy. This review focuses on current treatments and strategies for skeletal muscle atrophy, including drug treatment (active substances of traditional Chinese medicine, chemical drugs, antioxidants, enzyme and enzyme inhibitors, hormone drugs, etc.), gene therapy, stem cell and exosome therapy (muscle-derived stem cells, non-myogenic stem cells, and exosomes), cytokine therapy, physical therapy (electroacupuncture, electrical stimulation, optogenetic technology, heat therapy, and low-level laser therapy), nutrition support (protein, essential amino acids, creatine, β-hydroxy-β-methylbutyrate, and vitamin D), and other therapies (biomaterial adjuvant therapy, intestinal microbial regulation, and oxygen supplementation). Considering many treatments have been developed for skeletal muscle atrophy, we propose a combination of proper treatments for individual needs, which may yield better treatment outcomes.
Collapse
|
47
|
Inflammation: Roles in Skeletal Muscle Atrophy. Antioxidants (Basel) 2022; 11:antiox11091686. [PMID: 36139760 PMCID: PMC9495679 DOI: 10.3390/antiox11091686] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 12/03/2022] Open
Abstract
Various diseases can cause skeletal muscle atrophy, usually accompanied by inflammation, mitochondrial dysfunction, apoptosis, decreased protein synthesis, and enhanced proteolysis. The underlying mechanism of inflammation in skeletal muscle atrophy is extremely complex and has not been fully elucidated, thus hindering the development of effective therapeutic drugs and preventive measures for skeletal muscle atrophy. In this review, we elaborate on protein degradation pathways, including the ubiquitin-proteasome system (UPS), the autophagy-lysosome pathway (ALP), the calpain and caspase pathways, the insulin growth factor 1/Akt protein synthesis pathway, myostatin, and muscle satellite cells, in the process of muscle atrophy. Under an inflammatory environment, various pro-inflammatory cytokines directly act on nuclear factor-κB, p38MAPK, and JAK/STAT pathways through the corresponding receptors, and then are involved in muscle atrophy. Inflammation can also indirectly trigger skeletal muscle atrophy by changing the metabolic state of other tissues or cells. This paper explores the changes in the hypothalamic-pituitary-adrenal axis and fat metabolism under inflammatory conditions as well as their effects on skeletal muscle. Moreover, this paper also reviews various signaling pathways related to muscle atrophy under inflammatory conditions, such as cachexia, sepsis, type 2 diabetes mellitus, obesity, chronic obstructive pulmonary disease, chronic kidney disease, and nerve injury. Finally, this paper summarizes anti-amyotrophic drugs and their therapeutic targets for inflammation in recent years. Overall, inflammation is a key factor causing skeletal muscle atrophy, and anti-inflammation might be an effective strategy for the treatment of skeletal muscle atrophy. Various inflammatory factors and their downstream pathways are considered promising targets for the treatment and prevention of skeletal muscle atrophy.
Collapse
|