1
|
Lombardo M, Aiello G, Fratantonio D, Karav S, Baldelli S. Functional Role of Extracellular Vesicles in Skeletal Muscle Physiology and Sarcopenia: The Importance of Physical Exercise and Nutrition. Nutrients 2024; 16:3097. [PMID: 39339697 PMCID: PMC11435357 DOI: 10.3390/nu16183097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVES Extracellular vesicles (EVs) play a key role in intercellular communication by transferring miRNAs and other macromolecules between cells. Understanding how diet and exercise modulate the release and content of skeletal muscle (SM)-derived EVs could lead to novel therapeutic strategies to prevent age-related muscle decline and other chronic diseases, such as sarcopenia. This review aims to provide an overview of the role of EVs in muscle function and to explore how nutritional and physical interventions can optimise their release and function. METHODS A literature review of studies examining the impact of exercise and nutritional interventions on MS-derived EVs was conducted. Major scientific databases, including PubMed, Scopus and Web of Science, were searched using keywords such as 'extracellular vesicles', 'muscle', 'exercise', 'nutrition' and 'sarcopenia'. The selected studies included randomised controlled trials (RCTs), clinical trials and cohort studies. Data from these studies were synthesised to identify key findings related to the release of EVs, their composition and their potential role as therapeutic targets. RESULTS Dietary patterns, specific foods and supplements were found to significantly modulate EV release and composition, affecting muscle health and metabolism. Exercise-induced changes in EV content were observed after both acute and chronic interventions, with a marked impact on miRNAs and proteins related to muscle growth and inflammation. Nutritional interventions, such as the Mediterranean diet and omega-3 fatty acids, have also shown the ability to alter EV profiles, suggesting their potential to improve cardiovascular health and reduce inflammation. CONCLUSIONS EVs are emerging as critical mediators of the beneficial effects of diet and exercise on muscle health. Both exercise and nutritional interventions can modulate the release and content of MS-derived EVs, offering promising avenues for the development of novel therapeutic strategies targeting sarcopenia and other muscle diseases. Future research should focus on large-scale RCT studies with standardised methodologies to better understand the role of EVs as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Mauro Lombardo
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy
| | - Gilda Aiello
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy
| | - Deborah Fratantonio
- Department of Medicine and Surgery, LUM University, S.S. 100 Km 18, 70100 Casamassima, Italy
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Canakkale 17000, Türkiye
| | - Sara Baldelli
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy
- IRCCS San Raffaele Roma, 00166 Rome, Italy
| |
Collapse
|
2
|
Al-Rawaf HA, Gabr SA, Iqbal A, Alghadir AH. Circulating microRNAs as potential biomarkers of physical activity in geriatric patients with HCV. BMC Mol Cell Biol 2024; 25:18. [PMID: 39030480 PMCID: PMC11264506 DOI: 10.1186/s12860-024-00514-8] [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: 01/22/2024] [Accepted: 07/09/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Circulating microRNAs have been implicated in a diverse array of biological and pathological phenomena. Their potential utility as noninvasive biomarkers for screening and diagnosing various diseases has been proposed. OBJECTIVE This study aimed to explore the potential role of the miRNAs miR-122 and miR-486 as molecular biomarkers in the pathogenesis of hepatitis C virus (HCV) infection. Thus, miR-122 and miR-486 were detected in the serum of HCV patients and healthy controls. Moreover, the potential correlations of miR-122 and miR-486 with viral complications, such as physical activity, pain, muscle fatigue, and HCV infection, were identified. METHODS A total of 150 subjects aged 30 to 66 years were included in this study. The patients were classified as patients with chronic hepatitis C virus (CHC) (n = 110) or healthy controls (n = 40). Real-time polymerase chain reaction (PCR) analyses were performed to determine miR-122 and miR-486 expression. Physical activity (PA), pain score, HCV genotyping, viral overload, aspartate transaminase (AST), alanine transaminase (ALT), lactic acid dehydrogenase (LDH), creatine kinase (CK), and antioxidant status were also estimated by using prevalidated questionnaires, PCR, and spectrophotometric analyses. RESULTS Compared with those in normal controls, significant increases in the serum levels of miR-122 and miR-486 were reported in patients with CHC. In physically active CHC patients, there was a significant correlation between the expression of miRNAs and increased alanine transaminase (ALT), aspartate transaminase (AST), fibrosis scores, and inflammation activity, but no association was reported for hepatitis C virus (HCV) RNA or viral load. Additionally, significant decreases in LDH, CK, GSSG, and pain scores and increases in TAC, GSH, and the GSH/GSSG ratio were reported. Moreover, the expression of miR-122 and miR-486 was positively correlated with changes in body mass index (BMI) and liver fibrosis stage, as well as negatively correlated with sex, PA, TAC, GSH, GSSG, and the GSH/GSSG ratio. CONCLUSION MiR-122 and miR-486 expression levels were strongly correlated with physical activity, pain perception, and muscle fatigue biomarkers in HCV-infected patients. These miRNA levels were associated with elevated AST, ALT, fibrosis scores, LDH, CK, and antioxidant status, thus suggesting their potential as biomarkers for disease severity and oxidative stress. However, no correlation was observed with viral load or HCV-RNA expression, thus implying that these miRNAs may impact disease progression and symptoms through host factors, rather than directly affecting viral replication. In summary, the results demonstrated that molecular studies of miR-22 and miR-468 and their associations with PA, pain, adiposity, sex differences, and muscle fatigue, as well as routine biomarkers, could be useful as prognostic nanoninvasive biomarkers, thus providing novel therapeutic targets for CHC infection.
Collapse
Affiliation(s)
- Hadeel A Al-Rawaf
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Sami A Gabr
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Amir Iqbal
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia.
| | - Ahmad H Alghadir
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| |
Collapse
|
3
|
Lee S. Cardiovascular Disease and miRNAs: Possible Oxidative Stress-Regulating Roles of miRNAs. Antioxidants (Basel) 2024; 13:656. [PMID: 38929095 PMCID: PMC11200533 DOI: 10.3390/antiox13060656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
MicroRNAs (miRNAs) have been highlighted as key players in numerous diseases, and accumulating evidence indicates that pathological expressions of miRNAs contribute to both the development and progression of cardiovascular diseases (CVD), as well. Another important factor affecting the development and progression of CVD is reactive oxygen species (ROS), as well as the oxidative stress they may impose on the cells. Considering miRNAs are involved in virtually every biological process, it is not unreasonable to assume that miRNAs also play critical roles in the regulation of oxidative stress. This narrative review aims to provide mechanistic insights on possible oxidative stress-regulating roles of miRNAs in cardiovascular diseases based on differentially expressed miRNAs reported in various cardiovascular diseases and their empirically validated targets that have been implicated in the regulation of oxidative stress.
Collapse
Affiliation(s)
- Seahyoung Lee
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| |
Collapse
|
4
|
Qian L, Zhu Y, Deng C, Liang Z, Chen J, Chen Y, Wang X, Liu Y, Tian Y, Yang Y. Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family in physiological and pathophysiological process and diseases. Signal Transduct Target Ther 2024; 9:50. [PMID: 38424050 PMCID: PMC10904817 DOI: 10.1038/s41392-024-01756-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family (PGC-1s), consisting of three members encompassing PGC-1α, PGC-1β, and PGC-1-related coactivator (PRC), was discovered more than a quarter-century ago. PGC-1s are essential coordinators of many vital cellular events, including mitochondrial functions, oxidative stress, endoplasmic reticulum homeostasis, and inflammation. Accumulating evidence has shown that PGC-1s are implicated in many diseases, such as cancers, cardiac diseases and cardiovascular diseases, neurological disorders, kidney diseases, motor system diseases, and metabolic disorders. Examining the upstream modulators and co-activated partners of PGC-1s and identifying critical biological events modulated by downstream effectors of PGC-1s contribute to the presentation of the elaborate network of PGC-1s. Furthermore, discussing the correlation between PGC-1s and diseases as well as summarizing the therapy targeting PGC-1s helps make individualized and precise intervention methods. In this review, we summarize basic knowledge regarding the PGC-1s family as well as the molecular regulatory network, discuss the physio-pathological roles of PGC-1s in human diseases, review the application of PGC-1s, including the diagnostic and prognostic value of PGC-1s and several therapies in pre-clinical studies, and suggest several directions for future investigations. This review presents the immense potential of targeting PGC-1s in the treatment of diseases and hopefully facilitates the promotion of PGC-1s as new therapeutic targets.
Collapse
Affiliation(s)
- Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, 450052, China
| | - Junmin Chen
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xue Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yanqing Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ye Tian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China.
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
5
|
Han L, Li P, He Q, Yang C, Jiang M, Wang Y, Cao Y, Han X, Liu X, Wu W. Revisiting Skeletal Muscle Dysfunction and Exercise in Chronic Obstructive Pulmonary Disease: Emerging Significance of Myokines. Aging Dis 2023; 15:2453-2469. [PMID: 38270119 PMCID: PMC11567253 DOI: 10.14336/ad.2023.1125] [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: 09/13/2023] [Accepted: 11/25/2023] [Indexed: 01/26/2024] Open
Abstract
Skeletal muscle dysfunction (SMD) is the most significant extrapulmonary complication and an independent prognostic indicator in patients with chronic obstructive pulmonary disease (COPD). Myokines, such as interleukin (IL)-6, IL-15, myostatin, irisin, and insulin-like growth factor (IGF)-1, play important roles in skeletal muscle mitochondrial function, protein synthesis and breakdown balance, and regeneration of skeletal muscles in COPD. As the main component of pulmonary rehabilitation, exercise can improve muscle strength, muscle endurance, and exercise capacity in patients with COPD, as well as improve the prognosis of SMD and COPD by regulating the expression levels of myokines. The mechanisms by which exercise regulates myokine levels are related to microRNAs. IGF-1 expression is upregulated by decreasing the expression of miR-1 or miR-29b. Myostatin downregulation and irisin upregulation are associated with increased miR-27a expression and decreased miR-696 expression, respectively. These findings suggest that myokines are potential targets for the prevention and treatment of SMD in COPD. A comprehensive analysis of the role and regulatory mechanisms of myokines can facilitate the development of new exercise-based therapeutic approaches for patients with COPD.
Collapse
Affiliation(s)
- Lihua Han
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| | - Peijun Li
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Qinglan He
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| | - Chen Yang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| | - Meiling Jiang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| | - Yingqi Wang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yuanyuan Cao
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| | - Xiaoyu Han
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Weibing Wu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
6
|
Long YF, Chow SKH, Cui C, Wong RMY, Zhang N, Qin L, Law SW, Cheung WH. Does exercise influence skeletal muscle by modulating mitochondrial functions via regulating MicroRNAs? A systematic review. Ageing Res Rev 2023; 91:102048. [PMID: 37652311 DOI: 10.1016/j.arr.2023.102048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/31/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Sarcopenia is the accelerated loss of muscle mass, strength and function. Mitochondrial dysfunction was related to the progression of sarcopenia; meanwhile, microRNAs were regarded as core roles in regulating mitochondrial function. Physical exercise is a well-accepted approach to attenuate sarcopenia, yet very few studies depict the molecular mechanisms. The aim of this systematic review is to explore the potential relationships among physical exercise, mitochondrial function, and microRNAs, which may give new insight for retarding sarcopenia. METHODS A systematic literature search was performed in PubMed, Embase and Web of Science. The keywords were combined as "(microRNA OR miR) AND mitochondri* AND muscle AND exercise" and searched in all fields. PRISMA guidelines were followed. Information was extracted from the included studies for review. RESULTS In this review, 18 preclinical studies and 5 clinical studies were included. Most of the included studies suggested that effective physical exercise had positive effects on mitochondrial functions by regulating microRNAs. The results showed that 12 microRNAs improved mitochondrial functions, while 18 microRNAs suppressed them. Meanwhile, the results showed that 5 microRNAs improved muscle performance. CONCLUSIONS This systematic review provides an up-to-date sequential overview and highlights the potential relationship among exercise, mitochondrial function, and microRNAs in muscle. Meanwhile, evidence revealed that physical exercise can improve muscle performance by up-regulating mitochondrial functions, especially mitochondrial biogenesis, through modulating microRNAs.
Collapse
Affiliation(s)
- Yu-Feng Long
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Simon Kwoon-Ho Chow
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China; Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA.
| | - Can Cui
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Ronald Man Yeung Wong
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Ning Zhang
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China; Bone Quality and Health Centre, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Sheung-Wai Law
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China; Bone Quality and Health Centre, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing-Hoi Cheung
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China; Bone Quality and Health Centre, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
7
|
Hou J, Liang WY, Xiong S, Long P, Yue T, Wen X, Wang T, Deng H. Identification of hub genes and potential ceRNA networks of diabetic cardiomyopathy. Sci Rep 2023; 13:10258. [PMID: 37355664 PMCID: PMC10290640 DOI: 10.1038/s41598-023-37378-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023] Open
Abstract
Diabetic cardiomyopathy (DCM), a common complication of diabetes, is defined as ventricular dysfunction in the absence of underlying heart disease. Noncoding RNAs (ncRNAs), including long noncoding RNAs (lncRNAs) and microRNAs (miRNAs), play a crucial role in the development of DCM. Weighted Gene Co-Expression Network Analysis (WGCNA) was used to identify key modules in DCM-related pathways. DCM-related miRNA-mRNA network and DCM-related ceRNA network were constructed by miRNA-seq to identify hub genes in these modules. We identified five hub genes that are associated with the onset of DCM, including Troponin C1 (Tnnc1), Phospholamban (Pln), Fatty acid binding proteins 3 (Fabp3), Popeye domain containing 2 (Popdc2), and Tripartite Motif-containing Protein 63 (Trim63). miRNAs that target the hub genes were mainly involved in TGF-β and Wnt signaling pathways. GO BP enrichment analysis found these miRNAs were involved in the signaling of TGF-β and glucose homeostasis. Q-PCR results found the gene expressions of Pln, Fabp3, Trim63, Tnnc1, and Popdc2 were significantly increased in DCM. Our study identified five hub genes (Tnnc1, Pln, Fabp3, Popdc2, Trim63) whose associated ceRNA networks are responsible for the onset of DCM.
Collapse
Affiliation(s)
- Jun Hou
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Wan Yi Liang
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, BC, Canada
| | - Shiqiang Xiong
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Pan Long
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Tian Yue
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Xudong Wen
- Department of Gastroenterology and Hepatology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Tianchen Wang
- Alfred E. Mann Department of Biomedical Engineering, University of South California, Los Angeles, CA, USA
| | - Haoyu Deng
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada.
| |
Collapse
|
8
|
Skouras AZ, Antonakis-Karamintzas D, Tsantes AG, Triantafyllou A, Papagiannis G, Tsolakis C, Koulouvaris P. The Acute and Chronic Effects of Resistance and Aerobic Exercise in Hemostatic Balance: A Brief Review. Sports (Basel) 2023; 11:sports11040074. [PMID: 37104148 PMCID: PMC10143125 DOI: 10.3390/sports11040074] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Hemostatic balance refers to the dynamic balance between blood clot formation (coagulation), blood clot dissolution (fibrinolysis), anticoagulation, and innate immunity. Although regular habitual exercise may lower the incidence of cardiovascular diseases (CVD) by improving an individual’s hemostatic profile at rest and during exertion, vigorous exercise may increase the risk of sudden cardiac death and venous thromboembolism (VTE). This literature review aims to investigate the hemostatic system’s acute and chronic adaptive responses to different types of exercise in healthy and patient populations. Compared to athletes, sedentary healthy individuals demonstrate similar post-exercise responses in platelet function and coagulatory and fibrinolytic potential. However, hemostatic adaptations of patients with chronic diseases in regular training is a promising field. Despite the increased risk of thrombotic events during an acute bout of vigorous exercise, regular exposure to high-intensity exercise might desensitize exercise-induced platelet aggregation, moderate coagulatory parameters, and up-regulate fibrinolytic potential via increasing tissue plasminogen activator (tPA) and decreasing plasminogen activator inhibitor (PAI-1) response. Future research might focus on combining different types of exercise, manipulating each training characteristic (frequency, intensity, time, and volume), or investigating the minimal exercise dosage required to maintain hemostatic balance, especially in patients with various health conditions.
Collapse
|
9
|
Aoi W, Inoue R, Mizushima K, Honda A, Björnholm M, Takagi T, Naito Y. Exercise-acclimated microbiota improves skeletal muscle metabolism via circulating bile acid deconjugation. iScience 2023; 26:106251. [PMID: 36915683 PMCID: PMC10005909 DOI: 10.1016/j.isci.2023.106251] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/31/2022] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
Habitual exercise alters the intestinal microbiota composition, which may mediate its systemic benefits. We examined whether transplanting fecal microbiota from trained mice improved skeletal muscle metabolism in high-fat diet (HFD)-fed mice. Fecal samples from sedentary and exercise-trained mice were gavage-fed to germ-free mice. After receiving fecal samples from trained donor mice for 1 week, recipient mice had elevated levels of AMP-activated protein kinase (AMPK) and insulin growth factor-1 in skeletal muscle. In plasma, bile acid (BA) deconjugation was found to be promoted in recipients transplanted with feces from trained donor mice; free-form BAs also induced more AMPK signaling and glucose uptake than tauro-conjugated BAs. The transplantation of exercise-acclimated fecal microbiota improved glucose tolerance after 8 weeks of HFD administration. Intestinal microbiota may mediate exercise-induced metabolic improvements in mice by modifying circulating BAs. Our findings provide insights into the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Wataru Aoi
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 6068522, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, Osaka 5730101, Japan
| | - Katsura Mizushima
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 6028566, Japan
| | - Akira Honda
- Gastroenterology, Tokyo Medical University Ibaraki Medical Center, Ibaraki 3000395, Japan
| | - Marie Björnholm
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 17176, Sweden
| | - Tomohisa Takagi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 6028566, Japan.,Department for Medical Innovation and Translational Medical Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 6028566, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 6028566, Japan
| |
Collapse
|
10
|
Benite-Ribeiro SA, Barbosa HC, Ramadan W, dos Santos JM. Exercise-mediated increase in PGC1α and MEF2 expression in type 2 diabetes mellitus. GENE REPORTS 2023. [DOI: 10.1016/j.genrep.2023.101758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
11
|
Gambardella J, Fiordelisi A, Sorriento D, Cerasuolo F, Buonaiuto A, Avvisato R, Pisani A, Varzideh F, Riccio E, Santulli G, Iaccarino G. Mitochondrial microRNAs Are Dysregulated in Patients with Fabry Disease. J Pharmacol Exp Ther 2023; 384:72-78. [PMID: 35764328 PMCID: PMC9827504 DOI: 10.1124/jpet.122.001250] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 01/13/2023] Open
Abstract
Fabry disease (FD) is a lysosomal storage disorder caused by mutations in the gene for α-galactosidase A, inducing a progressive accumulation of globotriaosylceramide (GB3) and its metabolites in different organs and tissues. GB3 deposition does not fully explain the clinical manifestations of FD, and other pathogenetic mechanisms have been proposed, requiring the identification of new biomarkers for monitoring FD patients. Emerging evidence suggests the involvement of mitochondrial alterations in FD. Here, we propose mitochondrial-related microRNAs (miRs) as potential biomarkers of mitochondrial involvement in FD. Indeed, we demonstate that miRs regulating different aspects of mitochondrial homeostasis including expression and assembly of respiratory chain, mitogenesis, antioxidant capacity, and apoptosis are consistently dysregulated in FD patients. Our data unveil a novel noncoding RNA signature of FD patients, indicating mitochondrial-related miRs as new potential pathogenic players and biomarkers in FD. SIGNIFICANCE STATEMENT: This study demonstrates for the first time that a specific signature of circulating mitochondrial miRs (mitomiRs) is dysregulated in FD patients. MitomiRs regulating fundamental aspects of mitochondrial homeostasis and fitness, including expression and assembly of the respiratory chain, mitogenesis, antioxidant capacity, and apoptosis are significantly dysregulated in FD patients. Taken together, these new findings introduce mitomiRs as unprecedented biomarkers of FD and point at mitochondrial dysfunction as a novel potential mechanistic target for therapeutic approaches.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Advanced Biomedical Sciences (J.G., A.F., D.S., F.C., A.B., R.A., G.I.); Interdepartmental Center of Research on Hypertension and Related Conditions (J.G., G.I.), and Department of Public Health (A.P., E.R.); Federico II University, Naples, Italy; and Departments of Medicine (Cardiology) and Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York City, New York (J.G., F.V., G.S.)
| | - Antonella Fiordelisi
- Department of Advanced Biomedical Sciences (J.G., A.F., D.S., F.C., A.B., R.A., G.I.); Interdepartmental Center of Research on Hypertension and Related Conditions (J.G., G.I.), and Department of Public Health (A.P., E.R.); Federico II University, Naples, Italy; and Departments of Medicine (Cardiology) and Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York City, New York (J.G., F.V., G.S.)
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences (J.G., A.F., D.S., F.C., A.B., R.A., G.I.); Interdepartmental Center of Research on Hypertension and Related Conditions (J.G., G.I.), and Department of Public Health (A.P., E.R.); Federico II University, Naples, Italy; and Departments of Medicine (Cardiology) and Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York City, New York (J.G., F.V., G.S.)
| | - Federica Cerasuolo
- Department of Advanced Biomedical Sciences (J.G., A.F., D.S., F.C., A.B., R.A., G.I.); Interdepartmental Center of Research on Hypertension and Related Conditions (J.G., G.I.), and Department of Public Health (A.P., E.R.); Federico II University, Naples, Italy; and Departments of Medicine (Cardiology) and Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York City, New York (J.G., F.V., G.S.)
| | - Antonietta Buonaiuto
- Department of Advanced Biomedical Sciences (J.G., A.F., D.S., F.C., A.B., R.A., G.I.); Interdepartmental Center of Research on Hypertension and Related Conditions (J.G., G.I.), and Department of Public Health (A.P., E.R.); Federico II University, Naples, Italy; and Departments of Medicine (Cardiology) and Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York City, New York (J.G., F.V., G.S.)
| | - Roberta Avvisato
- Department of Advanced Biomedical Sciences (J.G., A.F., D.S., F.C., A.B., R.A., G.I.); Interdepartmental Center of Research on Hypertension and Related Conditions (J.G., G.I.), and Department of Public Health (A.P., E.R.); Federico II University, Naples, Italy; and Departments of Medicine (Cardiology) and Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York City, New York (J.G., F.V., G.S.)
| | - Antonio Pisani
- Department of Advanced Biomedical Sciences (J.G., A.F., D.S., F.C., A.B., R.A., G.I.); Interdepartmental Center of Research on Hypertension and Related Conditions (J.G., G.I.), and Department of Public Health (A.P., E.R.); Federico II University, Naples, Italy; and Departments of Medicine (Cardiology) and Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York City, New York (J.G., F.V., G.S.)
| | - Fahimeh Varzideh
- Department of Advanced Biomedical Sciences (J.G., A.F., D.S., F.C., A.B., R.A., G.I.); Interdepartmental Center of Research on Hypertension and Related Conditions (J.G., G.I.), and Department of Public Health (A.P., E.R.); Federico II University, Naples, Italy; and Departments of Medicine (Cardiology) and Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York City, New York (J.G., F.V., G.S.)
| | - Eleonora Riccio
- Department of Advanced Biomedical Sciences (J.G., A.F., D.S., F.C., A.B., R.A., G.I.); Interdepartmental Center of Research on Hypertension and Related Conditions (J.G., G.I.), and Department of Public Health (A.P., E.R.); Federico II University, Naples, Italy; and Departments of Medicine (Cardiology) and Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York City, New York (J.G., F.V., G.S.)
| | - Gaetano Santulli
- Department of Advanced Biomedical Sciences (J.G., A.F., D.S., F.C., A.B., R.A., G.I.); Interdepartmental Center of Research on Hypertension and Related Conditions (J.G., G.I.), and Department of Public Health (A.P., E.R.); Federico II University, Naples, Italy; and Departments of Medicine (Cardiology) and Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York City, New York (J.G., F.V., G.S.)
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences (J.G., A.F., D.S., F.C., A.B., R.A., G.I.); Interdepartmental Center of Research on Hypertension and Related Conditions (J.G., G.I.), and Department of Public Health (A.P., E.R.); Federico II University, Naples, Italy; and Departments of Medicine (Cardiology) and Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Institute for Neuroimmunology and Inflammation, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York City, New York (J.G., F.V., G.S.)
| |
Collapse
|
12
|
Sumaiya K, Ponnusamy T, Natarajaseenivasan K, Shanmughapriya S. Cardiac Metabolism and MiRNA Interference. Int J Mol Sci 2022; 24:50. [PMID: 36613495 PMCID: PMC9820363 DOI: 10.3390/ijms24010050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
The aberrant increase in cardio-metabolic diseases over the past couple of decades has drawn researchers' attention to explore and unveil the novel mechanisms implicated in cardiometabolic diseases. Recent evidence disclosed that the derangement of cardiac energy substrate metabolism plays a predominant role in the development and progression of chronic cardiometabolic diseases. Hence, in-depth comprehension of the novel molecular mechanisms behind impaired cardiac metabolism-mediated diseases is crucial to expand treatment strategies. The complex and dynamic pathways of cardiac metabolism are systematically controlled by the novel executor, microRNAs (miRNAs). miRNAs regulate target gene expression by either mRNA degradation or translational repression through base pairing between miRNA and the target transcript, precisely at the 3' seed sequence and conserved heptametrical sequence in the 5' end, respectively. Multiple miRNAs are involved throughout every cardiac energy substrate metabolism and play a differential role based on the variety of target transcripts. Novel theoretical strategies have even entered the clinical phase for treating cardiometabolic diseases, but experimental evidence remains inadequate. In this review, we identify the potent miRNAs, their direct target transcripts, and discuss the remodeling of cardiac metabolism to cast light on further clinical studies and further the expansion of novel therapeutic strategies. This review is categorized into four sections which encompass (i) a review of the fundamental mechanism of cardiac metabolism, (ii) a divulgence of the regulatory role of specific miRNAs on cardiac metabolic pathways, (iii) an understanding of the association between miRNA and impaired cardiac metabolism, and (iv) summary of available miRNA targeting therapeutic approaches.
Collapse
Affiliation(s)
- Krishnamoorthi Sumaiya
- Medical Microbiology Laboratory, Department of Microbiology, Centre for Excellence in Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | - Thiruvelselvan Ponnusamy
- Department of Medicine, Department of Cellular and Molecular Physiology, Heart and Vascular Institute, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Kalimuthusamy Natarajaseenivasan
- Medical Microbiology Laboratory, Department of Microbiology, Centre for Excellence in Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Santhanam Shanmughapriya
- Department of Medicine, Department of Cellular and Molecular Physiology, Heart and Vascular Institute, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| |
Collapse
|
13
|
van Wijk N, Zohar K, Linial M. Challenging Cellular Homeostasis: Spatial and Temporal Regulation of miRNAs. Int J Mol Sci 2022; 23:16152. [PMID: 36555797 PMCID: PMC9787707 DOI: 10.3390/ijms232416152] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/11/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Mature microRNAs (miRNAs) are single-stranded non-coding RNA (ncRNA) molecules that act in post-transcriptional regulation in animals and plants. A mature miRNA is the end product of consecutive, highly regulated processing steps of the primary miRNA transcript. Following base-paring of the mature miRNA with its mRNA target, translation is inhibited, and the targeted mRNA is degraded. There are hundreds of miRNAs in each cell that work together to regulate cellular key processes, including development, differentiation, cell cycle, apoptosis, inflammation, viral infection, and more. In this review, we present an overlooked layer of cellular regulation that addresses cell dynamics affecting miRNA accessibility. We discuss the regulation of miRNA local storage and translocation among cell compartments. The local amounts of the miRNAs and their targets dictate their actual availability, which determines the ability to fine-tune cell responses to abrupt or chronic changes. We emphasize that changes in miRNA storage and compactization occur under induced stress and changing conditions. Furthermore, we demonstrate shared principles on cell physiology, governed by miRNA under oxidative stress, tumorigenesis, viral infection, or synaptic plasticity. The evidence presented in this review article highlights the importance of spatial and temporal miRNA regulation for cell physiology. We argue that limiting the research to mature miRNAs within the cytosol undermines our understanding of the efficacy of miRNAs to regulate cell fate under stress conditions.
Collapse
Affiliation(s)
| | | | - Michal Linial
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Faculty of Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
14
|
Inflammatory and Prothrombotic Biomarkers, DNA Polymorphisms, MicroRNAs and Personalized Medicine for Patients with Peripheral Arterial Disease. Int J Mol Sci 2022; 23:ijms231912054. [PMID: 36233355 PMCID: PMC9569699 DOI: 10.3390/ijms231912054] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 09/26/2022] [Accepted: 10/02/2022] [Indexed: 12/24/2022] Open
Abstract
Classical risk factors play a major role in the initiation and development of atherosclerosis. However, the estimation of risk for cardiovascular events based only on risk factors is often insufficient. Efforts have been made to identify biomarkers that indicate ongoing atherosclerosis. Among important circulating biomarkers associated with peripheral arterial disease (PAD) are inflammatory markers which are determined by the expression of different genes and epigenetic processes. Among these proinflammatory molecules, interleukin-6, C-reactive protein, several adhesion molecules, CD40 ligand, osteoprotegerin and others are associated with the presence and progression of PAD. Additionally, several circulating prothrombotic markers have a predictive value in PAD. Genetic polymorphisms significantly, albeit moderately, affect risk factors for PAD via altered lipoprotein metabolism, diabetes, arterial hypertension, smoking, inflammation and thrombosis. However, most of the risk variants for PAD are located in noncoding regions of the genome and their influence on gene expression remains to be explored. MicroRNAs (miRNAs) are single-stranded, noncoding RNAs that modulate gene expression at the post-transcriptional level. Patterns of miRNA expression, to some extent, vary in different atherosclerotic cardiovascular diseases. miRNAs appear to be useful in the detection of PAD and the prediction of progression and revascularization outcomes. In conclusion, taking into account one’s predisposition to PAD, i.e., DNA polymorphisms and miRNAs, together with circulating inflammatory and coagulation markers, holds promise for more accurate prediction models and personalized therapeutic options.
Collapse
|
15
|
Yin A, Yuan R, Xiao Q, Zhang W, Xu K, Yang X, Yang W, Xu L, Wang X, Zhuang F, Li Y, Cai Z, Sun Z, Zhou B, He B, Shen L. Exercise-derived peptide protects against pathological cardiac remodeling. EBioMedicine 2022; 82:104164. [PMID: 35843176 PMCID: PMC9297110 DOI: 10.1016/j.ebiom.2022.104164] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 11/22/2022] Open
Abstract
Background Exercise training protects the heart against pathological cardiac remodeling and confers cardioprotection from heart failure. However, the underlying mechanism is still elusive. Methods An integrative analysis of multi-omics data of the skeletal muscle in response to exercise is performed to search for potential exerkine. Then, CCDC80tide is examined in humans after acute exercise. The role of CCDC80tide is assessed in a mouse model of hypertensive cardiac remodeling and in hypertension-mediated cell injury models. The transcriptomic analysis and immunoprecipitation assay are conducted to explore the mechanism. Findings The coiled-coil domain-containing protein 80 (CCDC80) is found strongly positively associated with exercise. Interestingly, exercise stimuli induce the secretion of C-terminal CCDC80 (referred as CCDC80tide hereafter) via EVs-encapsulated CCDC80tide into the circulation. Importantly, cardiac-specific expression of CCDC80tide protects against angiotensin II (Ang II)-induced cardiac hypertrophy and fibrosis in mice. In in vitro studies, the expression of CCDC80tide reduces Ang II-induced cardiomyocyte hypertrophy, cardiac microvascular endothelial cell (CMEC) inflammation, and mitigated vascular smooth muscle cell (VSMC) proliferation and collagen formation. To understand the cardioprotective effect of CCDC80tide, a transcriptomic analysis reveals a dramatic inhibition of the STAT3 (Signal transducer and activator of transcription 3) signaling pathway in CCDC80tide overexpressing cells. Mechanistically, CCDC80tide selectively interacts with the kinase-active form of JAK2 (Janus kinase 2) and consequently inhibits its kinase activity to phosphorylate and activate STAT3. Interpretation The results provide new insights into exercise-afforded cardioprotection in pathological cardiac remodeling and highlight the therapeutic potential of CCDC80tide in heart failure treatment. Funding This work was supported by the National Natural Science Foundation of China [Grant/Award Numbers: 81770428, 81830010, 82130012, 81900438, 82100447); Shanghai Science and Technology Committee [Grant/Award Numbers: 21S11903000, 19JC1415702]; Emerging and Advanced Technology Programs of Hospital Development Center of Shanghai [Grant/Award Number: SHDC12018129]; China Postdoctoral Science Foundation [2021M692108]; and China National Postdoctoral Program for Innovative Talents [BX20200211].
Collapse
Affiliation(s)
- Anwen Yin
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ruosen Yuan
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Qingqing Xiao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Weifeng Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ke Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xiaoxiao Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Wentao Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Lei Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xia Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Fei Zhuang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhe Sun
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Bin Zhou
- Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
16
|
Dos Santos JAC, Veras ASC, Batista VRG, Tavares MEA, Correia RR, Suggett CB, Teixeira GR. Physical exercise and the functions of microRNAs. Life Sci 2022; 304:120723. [PMID: 35718233 DOI: 10.1016/j.lfs.2022.120723] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 10/18/2022]
Abstract
MicroRNAs (miRNAs) control RNA translation and are a class of small, tissue-specific, non-protein-coding RNAs that maintain cellular homeostasis through negative gene regulation. Maintenance of the physiological environment depends on the proper control of miRNA expression, as these molecules influence almost all genetic pathways, from the cell cycle checkpoint to cell proliferation and apoptosis, with a wide range of target genes. Dysregulation of the expression of miRNAs is correlated with several types of diseases, acting as regulators of cardiovascular functions, myogenesis, adipogenesis, osteogenesis, hepatic lipogenesis, and important brain functions. miRNAs can be modulated by environmental factors or external stimuli, such as physical exercise, and can eventually induce specific and adjusted changes in the transcriptional response. Physical exercise is used as a preventive and non-pharmacological treatment for many diseases. It is well established that physical exercise promotes various benefits in the human body such as muscle hypertrophy, mental health improvement, cellular apoptosis, weight loss, and inhibition of cell proliferation. This review highlights the current knowledge on the main miRNAs altered by exercise in the skeletal muscle, cardiac muscle, bone, adipose tissue, liver, brain, and body fluids. In addition, knowing the modifications induced by miRNAs and relating them to the results of prescribed physical exercise with different protocols and intensities can serve as markers of physical adaptation to training and responses to the effects of physical exercise for some types of chronic diseases. This narrative review consists of randomized exercise training experiments with humans and/or animals, combined with analyses of miRNA modulation.
Collapse
Affiliation(s)
| | - Allice Santos Cruz Veras
- Multicenter Graduate Program in Physiological Sciences, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | | | - Maria Eduarda Almeida Tavares
- Multicenter Graduate Program in Physiological Sciences, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Rafael Ribeiro Correia
- Department of Physical Education, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil; Multicenter Graduate Program in Physiological Sciences, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Cara Beth Suggett
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Giovana Rampazzo Teixeira
- Department of Physical Education, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil; Multicenter Graduate Program in Physiological Sciences, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil.
| |
Collapse
|
17
|
Widmann M, Mattioni Maturana F, Burgstahler C, Erz G, Schellhorn P, Fragasso A, Schmitt A, Nieß AM, Munz B. miRNAs as markers for the development of individualized training regimens: A pilot study. Physiol Rep 2022; 10:e15217. [PMID: 35274816 PMCID: PMC8915711 DOI: 10.14814/phy2.15217] [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: 10/20/2021] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 11/24/2022] Open
Abstract
Small, non‐coding RNAs (microRNAs) have been shown to regulate gene expression in response to exercise in various tissues and organs, thus possibly coordinating their adaptive response. Thus, it is likely that differential microRNA expression might be one of the factors that are responsible for different training responses of different individuals. Consequently, determining microRNA patterns might be a promising approach toward the development of individualized training strategies. However, little is known on (1) microRNA patterns and their regulation by different exercise regimens and (2) possible correlations between these patterns and individual training adaptation. Here, we present microarray data on skeletal muscle microRNA patterns in six young, female subjects before and after six weeks of either moderate‐intensity continuous or high‐intensity interval training on a bicycle ergometer. Our data show that n = 36 different microRNA species were regulated more than twofold in this cohort (n = 28 upregulated and n = 8 downregulated). In addition, we correlated baseline microRNA patterns with individual changes in VO2max and identified some specific microRNAs that might be promising candidates for further testing and evaluation in the future, which might eventually lead to the establishment of microRNA marker panels that will allow individual recommendations for specific exercise regimens.
Collapse
Affiliation(s)
- Manuel Widmann
- Department of Sports Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Research Institute for Sports and Physical Activity, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Felipe Mattioni Maturana
- Department of Sports Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Research Institute for Sports and Physical Activity, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Christof Burgstahler
- Department of Sports Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Research Institute for Sports and Physical Activity, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Gunnar Erz
- Department of Sports Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Research Institute for Sports and Physical Activity, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Philipp Schellhorn
- Department of Sports Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Research Institute for Sports and Physical Activity, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Annunziata Fragasso
- Department of Sports Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Research Institute for Sports and Physical Activity, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Angelika Schmitt
- Department of Sports Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Research Institute for Sports and Physical Activity, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Andreas M Nieß
- Department of Sports Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Research Institute for Sports and Physical Activity, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Barbara Munz
- Department of Sports Medicine, University Hospital Tübingen, Tübingen, Germany.,Interfaculty Research Institute for Sports and Physical Activity, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
18
|
Stevanović-Silva J, Beleza J, Coxito P, Costa RC, Ascensão A, Magalhães J. Fit mothers for a healthy future: Breaking the intergenerational cycle of non-alcoholic fatty liver disease with maternal exercise. Eur J Clin Invest 2022; 52:e13596. [PMID: 34120338 DOI: 10.1111/eci.13596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/01/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022]
Abstract
UNLABELLED SPECIAL ISSUE: 'FOIEGRAS-Bioenergetic Remodelling in the Pathophysiology and Treatment of Non-Alcoholic Fatty Liver Disease'. BACKGROUND Non-alcoholic fatty liver disease (NAFLD) emerges as significant health burden worldwide. Lifestyle changes, unhealthy dietary habits and physical inactivity, can trigger NAFLD development. Persisting on these habits during pregnancy affects in utero environment and prompts a specific metabolic response in foetus resulting in offspring metabolic maladjustments potentially critical for developing NAFLD later in life. The increasing prevalence of NAFLD, particularly in children, has shifted the research focus towards preventive and therapeutic strategies. Yet, designing effective approaches that can break the NAFLD intergenerational cycle becomes even more complicated. Regular physical exercise (PE) is a powerful non-pharmacological strategy known to counteract deleterious metabolic outcomes. In this narrative review, we aimed to briefly describe NAFLD pathogenesis focusing on maternal nutritional challenge and foetal programming, and to provide potential mechanisms behind the putative intergenerational effect of PE against metabolic diseases, including liver diseases. METHODS Following detailed electronic database search, recent existing evidence about NAFLD development, intergenerational programming and gestational exercise effects was critically analysed and discussed. RESULTS PE during pregnancy could have a great potential to counteract intergenerational transmission of metabolic burden. The interplay between different PE roles-metabolic, endocrine and epigenetic-could offer a more stable in utero environment to the foetus, thus rescuing offspring vulnerability to metabolic disturbances. CONCLUSIONS The better understanding of maternal PE beneficial consequences on offspring metabolism could reinforce the importance of PE during pregnancy as an indispensable strategy in improving offspring health.
Collapse
Affiliation(s)
- Jelena Stevanović-Silva
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - Jorge Beleza
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - Rui Carlos Costa
- Department of Communication and Art, Research Institute for Design, Media and Culture (ID+), Aveiro University, Aveiro, Portugal
| | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| |
Collapse
|
19
|
Shaker M, Khamisipour G, Sadeghipour H, Zar A, Naeimi B, Akbarzadeh S. Effect of resistance training and garlic extract on insulin sensitivity/resistance and biochemical parameters in diabetic rats. COMPARATIVE EXERCISE PHYSIOLOGY 2022. [DOI: 10.3920/cep210031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Diabetes is one of the most important endocrine diseases in the world and obesity is one of the risk factors for this disease. The aim of this study was to evaluate the effect of a resistance exercise and garlic extract on insulin sensitivity/resistance and signal pathway of white adipose tissue to brown factors in diabetic rats. A total of 48 male Wistar rats weighing 180 to 250 g were divided into six groups (n=8): healthy control (C), diabetic control (D), diabetic with garlic extract at a dose of 50 mg/kg body weight (bw) (D+50), diabetic with garlic extract dose of 200 mg/kg bw (D+200), diabetic resistance training (D+Ex), and diabetic resistance training with garlic extract dose of 200 mg/kg bw (D+Ex+200). Plasma irisin levels in the D+200 and D+Ex groups, as well as the D+Ex+200 group showed a significant increase compared to the D group (P<0.001), while in the D+50 group no significant change was observed. Compared with group D, the expression of peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1α gene was significantly increased in groups D+200 and D+Ex, as well as group D+Ex+200 (P<0.001). It can be said that resistance exercise with garlic extract is effective in controlling diabetes and reducing its complications. It also increases the expression of PGC-1α and uncoupling protein 1 genes in white adipose tissue and therefore has a positive effect on beta cell function by irisin.
Collapse
Affiliation(s)
- M. Shaker
- Faculty of Science, Agriculture and New Technologies Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - G.R. Khamisipour
- Department of Laboratory Sciences, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - H. Sadeghipour
- Department of Sport Science, School of Literature and Humanities, Persian Gulf University, Bushehr, Iran
| | - A. Zar
- Department of Sport Science, School of Literature and Humanities, Persian Gulf University, Bushehr, Iran
| | - B. Naeimi
- Department of Laboratory Sciences, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - S. Akbarzadeh
- Department of Biochemistry, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
20
|
Anderson JE. Key concepts in muscle regeneration: muscle "cellular ecology" integrates a gestalt of cellular cross-talk, motility, and activity to remodel structure and restore function. Eur J Appl Physiol 2022; 122:273-300. [PMID: 34928395 PMCID: PMC8685813 DOI: 10.1007/s00421-021-04865-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/10/2021] [Indexed: 12/21/2022]
Abstract
This review identifies some key concepts of muscle regeneration, viewed from perspectives of classical and modern research. Early insights noted the pattern and sequence of regeneration across species was similar, regardless of the type of injury, and differed from epimorphic limb regeneration. While potential benefits of exercise for tissue repair was debated, regeneration was not presumed to deliver functional restoration, especially after ischemia-reperfusion injury; muscle could develop fibrosis and ectopic bone and fat. Standard protocols and tools were identified as necessary for tracking injury and outcomes. Current concepts vastly extend early insights. Myogenic regeneration occurs within the environment of muscle tissue. Intercellular cross-talk generates an interactive system of cellular networks that with the extracellular matrix and local, regional, and systemic influences, forms the larger gestalt of the satellite cell niche. Regenerative potential and adaptive plasticity are overlain by epigenetically regionalized responsiveness and contributions by myogenic, endothelial, and fibroadipogenic progenitors and inflammatory and metabolic processes. Muscle architecture is a living portrait of functional regulatory hierarchies, while cellular dynamics, physical activity, and muscle-tendon-bone biomechanics arbitrate regeneration. The scope of ongoing research-from molecules and exosomes to morphology and physiology-reveals compelling new concepts in muscle regeneration that will guide future discoveries for use in application to fitness, rehabilitation, and disease prevention and treatment.
Collapse
Affiliation(s)
- Judy E Anderson
- Department of Biological Sciences, Faculty of Science, University of Manitoba, 50 Sifton Road, Winnipeg, MB, R3T 2N2, Canada.
| |
Collapse
|
21
|
miR-27b-3p Attenuates Muscle Atrophy by Targeting Cbl-b in Skeletal Muscles. Biomolecules 2022; 12:biom12020191. [PMID: 35204692 PMCID: PMC8961554 DOI: 10.3390/biom12020191] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/08/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
As it is well known, muscle atrophy is a process in which protein degradation increases and protein synthesis decreases. This process is regulated by a variety of links. Among them, microRNAs play an essential role in this process, which has attracted widespread attention. In this paper, we find that miR-27b-3p and Cbl-b genes are significantly differentially expressed in the induced atrophy model. The dual-luciferase experiment and Western blot analysis confirmed that miR-27b-3p could regulate the expression of Cbl-b. In C2C12-differentiated myotubes, the overexpression of the Cbl-b gene showed that Cbl-b could upregulate the expression of MuRF-1 and Atrogin-1, which are related marker genes of muscle atrophy, at both the mRNA and protein levels, indicating that the Cbl-b gene can specifically affect muscle atrophy. The knockdown of the Cbl-b gene after C2C12-differentiated myotubes induced atrophy treatment can downregulate the expression of muscle-atrophy-related genes, indicating that manual intervention to downregulate the expression of Cbl-b has a certain alleviating effect on muscle atrophy. These data suggest that miR-27b-3p can regulate the expression of the Cbl-b gene and then exert a particular influence on muscle atrophy through the Cbl-b gene.
Collapse
|
22
|
Evaluation of muscle-specific and metabolism regulating microRNAs in a chronic swimming rat model. J Muscle Res Cell Motil 2021; 43:21-33. [PMID: 34893938 PMCID: PMC8897377 DOI: 10.1007/s10974-021-09612-y] [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: 08/17/2021] [Accepted: 11/25/2021] [Indexed: 10/27/2022]
Abstract
Making benefit from the epigenetic effects of environmental factors such as physical activity may result in a considerable improvement in the prevention of chronic civilization diseases. In our chronic swimming rat model, the expression levels of such microRNAs were characterized, that are involved in skeletal muscle differentiation, hypertrophy and fine-tuning of metabolism, which processes are influenced by chronic endurance training, contributing to the metabolic adaptation of skeletal muscle during physical activity. After chronic swimming, the level of miR-128a increased significantly in EDL muscles, which may influence metabolic adaptation and stress response as well. In SOL, the expression level of miR-15b and miR-451 decreased significantly after chronic swimming, which changes are opposite to their previously described increment in insulin resistant skeletal muscle. MiR-451 also targets PGC-1α mRNA, whiches expression level significantly increased in SOL muscles, resulting in enhanced biogenesis and oxidative capacity of mitochondria. In summary, the microRNA expression changes that were observed during our experiments suggest that chronic swim training contributes to a beneficial metabolic profile of skeletal muscle.
Collapse
|
23
|
Beleza J, Stevanović-Silva J, Coxito P, Costa RC, Ascensão A, Torrella JR, Magalhães J. Building-up fit muscles for the future: Transgenerational programming of skeletal muscle through physical exercise. Eur J Clin Invest 2021; 51:e13515. [PMID: 33580562 DOI: 10.1111/eci.13515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/31/2021] [Accepted: 02/03/2021] [Indexed: 12/19/2022]
Abstract
'Special issue - In Utero and Early Life Programming of Aging and Disease'. Skeletal muscle (SM) adaptations to physical exercise (PE) have been extensively studied due, not only to the relevance of its in situ plasticity, but also to the SM endocrine-like effects in noncontractile tissues, such as brain, liver or adipocytes. Regular PE has been considered a pleiotropic nonpharmacological strategy to prevent and counteract the deleterious consequences of several metabolic, cardiovascular, oncological and neurodegenerative disorders. Additionally, PE performed by parents seems to have a direct impact in the offspring through the transgenerational programming of different tissues, such as SM. In fact, SM offspring programming mechanisms seems to be orchestrated, at least in part, by epigenetic machinery conditioning transcriptional or post-transcriptional processes. Ultimately, PE performed in the early in life is also a critical window of opportunity to positively modulate the juvenile and adult phenotype. Parental PE has a positive impact in several health-related offspring outcomes, such as SM metabolism, differentiation, morphology and ultimately in offspring exercise volition and endurance. Also, early-life PE counteracts conceptional-related adverse effects and induces long-lasting healthy benefits throughout adulthood. Additionally, epigenetics mechanisms seem to play a key role in the PE-induced SM adaptations. Despite the undoubtedly positive role of parental and early-life PE on SM phenotype, a strong research effort is still needed to better understand the mechanisms that positively regulate PE-induced SM programming.
Collapse
Affiliation(s)
- Jorge Beleza
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Jelena Stevanović-Silva
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| | - Rui Carlos Costa
- Department of Communication and Art, Research Institute for Design, Media and Culture (ID+), Aveiro University, Aveiro, Portugal
| | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| | - Joan Ramon Torrella
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Faculty of Sport, Research Centre in Physical Activity, Health and Leisure (CIAFEL), University of Porto, Porto, Portugal
| |
Collapse
|
24
|
Rafiei MM, Soltani R, Kordi MR, Nouri R, Gaeini AA. Gene expression of angiogenesis and apoptotic factors in female BALB/c mice with breast cancer after eight weeks of aerobic training. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1196-1202. [PMID: 35083006 PMCID: PMC8751744 DOI: 10.22038/ijbms.2021.55582.12427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 07/31/2021] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Breast cancer is the most common cancer in women, caused by a disorder in the angiogenesis and apoptosis process. Exercise can affect the process of angiogenesis and apoptosis in the tumor tissue. Thus, the aim of the present study was to investigate the changes in angiogenesis and apoptotic factors in mice with breast cancer after 8 weeks of exercise training. MATERIALS AND METHODS Sixteen females BALB/c mice (age: 3-5 weeks and weight: 17.1 ± 0.1 g) with breast cancer were randomly divided into two groups of aerobic training and control. The aerobic training included 8 weeks and 5 sessions per week of running with an intensity of 14-20 m.min-1. HIF-1α, VEGF, miR-21 and cytochrome C, Apaf-1, caspase-9, and caspase-3 gene expressions were examined by real-time PCR. Repeated measures ANOVA, Bonferroni's post hoc test, and independent samples t-test were used to analyze the data (P<0.05). RESULTS The results showed that aerobic training reduced the growth of tumor volume and significantly reduced miR-21 gene expression. Aerobic training also significantly increased the gene expression of HIF-1α, cytochrome C, Apaf-1, caspase-9, and caspase-3, while changes in VEGF gene expression were not statistically significant. CONCLUSION It appears that aerobic exercise training reduces tumor size and ameliorates breast cancer by reducing miR-21 gene expression, suppressing the apoptosis process, and reducing angiogenesis.
Collapse
Affiliation(s)
- Mohammad Mahdi Rafiei
- Department of Sport Sciences, Kish International Campus, University of Tehran, Kish, Iran
| | - Rahele Soltani
- Department of Sport Sciences, Kish International Campus, University of Tehran, Kish, Iran
| | - Mohammad Reza Kordi
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Tehran, Tehran, Iran
| | - Reza Nouri
- Department of Sport Sciences, Kish International Campus, University of Tehran, Kish, Iran
| | - Abbas Ali Gaeini
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Tehran, Tehran, Iran
| |
Collapse
|
25
|
Effects of single bouts of different endurance exercises with different intensities on microRNA biomarkers with and without blood flow restriction: a three-arm, randomized crossover trial. Eur J Appl Physiol 2021; 121:3243-3255. [PMID: 34435273 PMCID: PMC8505326 DOI: 10.1007/s00421-021-04786-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 08/10/2021] [Indexed: 10/27/2022]
Abstract
PURPOSE Physical activity is associated with altered levels of circulating microRNAs (ci-miRNAs). Changes in miRNA expression have great potential to modulate biological pathways of skeletal muscle hypertrophy and metabolism. This study was designed to determine whether the profile of ci-miRNAs is altered after different approaches of endurance exercise. METHODS Eighteen healthy volunteers (aged 24 ± 3 years) participated this three-arm, randomized-balanced crossover study. Each arm was a single bout of treadmill-based acute endurance exercise at (1) 100% of the individual anaerobic threshold (IANS), (2) at 80% of the IANS and (3) at 80% of the IANS with blood flow restriction (BFR). Load-associated outcomes (fatigue, feeling, heart rate, and exhaustion) as well as acute effects (circulating miRNA patterns and lactate) were determined. RESULTS All training interventions increased the lactate concentration (LC) and heart rate (HR) (p < 0.001). The high-intensity intervention (HI) resulted in a higher LC than both lower intensity protocols (p < 0.001). The low-intensity blood flow restriction (LI-BFR) protocol led to a higher HR and higher LC than the low-intensity (LI) protocol without BFR (p = 0.037 and p = 0.003). The level of miR-142-5p and miR-197-3p were up-regulated in both interventions without BFR (p < 0.05). After LI exercise, the expression of miR-342-3p was up-regulated (p = 0.038). In LI-BFR, the level of miR-342-3p and miR-424-5p was confirmed to be up-regulated (p < 0.05). Three miRNAs and LC show a significant negative correlation (miR-99a-5p, p = 0.011, r = - 0.343/miR-199a-3p, p = 0.045, r = - 0.274/miR-125b-5p, p = 0.026, r = - 0.302). Two partial correlations (intervention partialized) showed a systematic impact of the type of exercise (LI-BFR vs. HI) (miR-99a-59: r = - 0.280/miR-199a-3p: r = - 0.293). CONCLUSION MiRNA expression patterns differ according to type of activity. We concluded that not only the intensity of the exercise (LC) is decisive for the release of circulating miRNAs-as essential is the type of training and the oxygen supply.
Collapse
|
26
|
MicroRNA-128 inhibits mitochondrial biogenesis and function via targeting PGC1α and NDUFS4. Mitochondrion 2021; 60:160-169. [PMID: 34384932 DOI: 10.1016/j.mito.2021.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 07/28/2021] [Accepted: 08/05/2021] [Indexed: 01/25/2023]
Abstract
The size and morphology of mitochondria are very heterogeneous and correlates well with their healthy functioning. In many pathological conditions, mitochondrial morphology is altered due to impaired mitochondrial dynamics (a collective term for mitochondrial fusion and fission) and dysfunction. The current study aimed at identifying the role of microRNA-128 (miR-128) in regulating mitochondrial biogenesis. Previously, peroxisome proliferator activator receptor γ coactivator 1α (PGC1α) has been shown to co-activate key intermediates of mitochondrial biogenesis, function, and dynamics; however, the upstream regulatory network remains largely unknown. We, herein using in silico analysis followed by in vitro experiments in C2C12 myoblasts, showed that miR-128 reduces mitochondrial biogenesis by directly targeting PGC1α. The expression of downstream genes, nuclear respiratory factors 1 and 2 (NRF1 and NRF2, respectively), and mitochondrial transcription factor A (TFAM) were decreased in C2C12 myoblasts upon overexpression of miR-128. Also, miR-128 is shown to promote mitochondrial dysfunction by directly targeting NADH Dehydrogenase (Ubiquinone) Fe-S Protein 4 (NDUFS4). The mitochondrial dynamics and morphology were impaired post miR-128 overexpression, as revealed by downregulation of fusion proteins (mitofusin1 and 2, i.e., MFN1 and MFN2, respectively) and upregulation of fission protein (dynamin-related protein 1, i.e., DRP1). Conversely, inhibition of miR-128 expression improved mitochondrial biogenesis, function, and dynamics, as evidenced by increased mitochondrial mass and ATP production after antimiR-128 treatment. Our findings reveal that inhibition of miR-128 can be a new potential target for reversing the effects of metabolic disorders of skeletal muscle as observed during many pathophysiological conditions such as obesity and type II diabetes.
Collapse
|
27
|
The Glitazars Paradox: Cardiotoxicity of the Metabolically Beneficial Dual PPARα and PPARγ Activation. J Cardiovasc Pharmacol 2021; 76:514-526. [PMID: 33165133 DOI: 10.1097/fjc.0000000000000891] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The most common complications in patients with type-2 diabetes are hyperglycemia and hyperlipidemia that can lead to cardiovascular disease. Alleviation of these complications constitutes the major therapeutic approach for the treatment of diabetes mellitus. Agonists of peroxisome proliferator-activated receptor (PPAR) alpha and PPARγ are used for the treatment of hyperlipidemia and hyperglycemia, respectively. PPARs belong to the nuclear receptors superfamily and regulate fatty acid metabolism. PPARα ligands, such as fibrates, reduce circulating triglyceride levels, and PPARγ agonists, such as thiazolidinediones, improve insulin sensitivity. Dual-PPARα/γ agonists (glitazars) were developed to combine the beneficial effects of PPARα and PPARγ agonism. Although they improved metabolic parameters, they paradoxically aggravated congestive heart failure in patients with type-2 diabetes via mechanisms that remain elusive. Many of the glitazars, such as muraglitazar, tesaglitazar, and aleglitazar, were abandoned in phase-III clinical trials. The objective of this review article pertains to the understanding of how combined PPARα and PPARγ activation, which successfully targets the major complications of diabetes, causes cardiac dysfunction. Furthermore, it aims to suggest interventions that will maintain the beneficial effects of dual PPARα/γ agonism and alleviate adverse cardiac outcomes in diabetes.
Collapse
|
28
|
Multi-Omics Analysis of Key microRNA-mRNA Metabolic Regulatory Networks in Skeletal Muscle of Obese Rabbits. Int J Mol Sci 2021; 22:ijms22084204. [PMID: 33921578 PMCID: PMC8072691 DOI: 10.3390/ijms22084204] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/10/2021] [Accepted: 04/15/2021] [Indexed: 12/25/2022] Open
Abstract
microRNAs (miRNAs), small non-coding RNA with a length of about 22 nucleotides, are involved in the energy metabolism of skeletal muscle cells. However, their molecular mechanism of metabolism in rabbit skeletal muscle is still unclear. In this study, 16 rabbits, 8 in the control group (CON-G) and 8 in the experimental group (HFD-G), were chosen to construct an obese model induced by a high-fat diet fed from 35 to 70 days of age. Subsequently, 54 differentially expressed miRNAs, 248 differentially expressed mRNAs, and 108 differentially expressed proteins related to the metabolism of skeletal muscle were detected and analyzed with three sequencing techniques (small RNA sequencing, transcriptome sequencing, and tandem mass tab (TMT) protein technology). It was found that 12 miRNAs and 12 core genes (e.g., CRYL1, VDAC3 and APIP) were significantly different in skeletal muscle from rabbits in the two groups. The network analysis showed that seven miRNA-mRNA pairs were involved in metabolism. Importantly, two miRNAs (miR-92a-3p and miR-30a/c/d-5p) regulated three transcription factors (MYBL2, STAT1 and IKZF1) that may be essential for lipid metabolism. These results enhance our understanding of molecular mechanisms associated with rabbit skeletal muscle metabolism and provide a basis for future studies in the metabolic diseases of human obesity.
Collapse
|
29
|
Queiroz AL, Lessard SJ, Ouchida AT, Araujo HN, Gonçalves DA, Simões Fróes Guimarães DSP, Teodoro BG, So K, Espreafico EM, Hirshman MF, Alberici LC, Kettelhut IDC, Goodyear LJ, Silveira LR. The MicroRNA miR-696 is regulated by SNARK and reduces mitochondrial activity in mouse skeletal muscle through Pgc1α inhibition. Mol Metab 2021; 51:101226. [PMID: 33812060 PMCID: PMC8121711 DOI: 10.1016/j.molmet.2021.101226] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE MicroRNAs (miRNA) are known to regulate the expression of genes involved in several physiological processes including metabolism, mitochondrial biogenesis, proliferation, differentiation, and cell death. METHODS Using "in silico" analyses, we identified 219 unique miRNAs that potentially bind to the 3'UTR region of a critical mitochondrial regulator, the peroxisome proliferator-activated receptor gamma coactivator (PGC) 1 alpha (Pgc1α). Of the 219 candidate miRNAs, miR-696 had one of the highest interactions at the 3'UTR of Pgc1α, suggesting that miR-696 may be involved in the regulation of Pgc1α. RESULTS Consistent with this hypothesis, we found that miR-696 was highly expressed in the skeletal muscle of STZ-induced diabetic mice and chronic high-fat-fed mice. C2C12 muscle cells exposed to palmitic acid also exhibited a higher expression of miR-696. This increased expression corresponded with a reduced expression of oxidative metabolism genes and reduced mitochondrial respiration. Importantly, reducing miR-696 reversed decreases in mitochondrial activity in response to palmitic acid. Using C2C12 cells treated with the AMP-activated protein kinase (AMPK) activator AICAR and skeletal muscle from AMPKα2 dominant-negative (DN) mice, we found that the signaling mechanism regulating miR-696 did not involve AMPK. In contrast, overexpression of SNF1-AMPK-related kinase (SNARK) in C2C12 cells increased miR-696 transcription while knockdown of SNARK significantly decreased miR-696. Moreover, muscle-specific transgenic mice overexpressing SNARK exhibited a lower expression of Pgc1α, elevated levels of miR-696, and reduced amounts of spontaneous activity. CONCLUSIONS Our findings demonstrate that metabolic stress increases miR-696 expression in skeletal muscle cells, which in turn inhibits Pgc1α, reducing mitochondrial function. SNARK plays a role in this process as a metabolic stress signaling molecule inducing the expression of miR-696.
Collapse
Affiliation(s)
- André L Queiroz
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil; Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sarah J Lessard
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Amanda T Ouchida
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Hygor N Araujo
- Obesity and Comorbidities Research Center, OCRC, IB, UNICAMP, Campinas, Brazil
| | - Dawit A Gonçalves
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | | | - Bruno G Teodoro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil; Department of Physics and Chemistry, Faculty of Pharmaceutical Science, USP, Ribeirão Preto, Brazil
| | - Kawai So
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Enilza M Espreafico
- Department of Cell Biology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Michael F Hirshman
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Luciane C Alberici
- Department of Physics and Chemistry, Faculty of Pharmaceutical Science, USP, Ribeirão Preto, Brazil
| | - Isis do Carmo Kettelhut
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Laurie J Goodyear
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Leonardo R Silveira
- Obesity and Comorbidities Research Center, OCRC, IB, UNICAMP, Campinas, Brazil.
| |
Collapse
|
30
|
Loussouarn C, Pers YM, Bony C, Jorgensen C, Noël D. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Regulate the Mitochondrial Metabolism via Transfer of miRNAs. Front Immunol 2021; 12:623973. [PMID: 33796099 PMCID: PMC8007981 DOI: 10.3389/fimmu.2021.623973] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are the most commonly tested adult progenitor cells in regenerative medicine. They stimulate tissue repair primarily through the secretion of immune-regulatory and pro-regenerative factors. There is increasing evidence that most of these factors are carried on extracellular vesicles (EVs) that are released by MSCs, either spontaneously or after activation. Exosomes and microvesicles are the most investigated types of EVs that act through uptake by target cells and cargo release inside the cytoplasm or through interactions with receptors expressed on target cells to stimulate downstream intracellular pathways. They convey different types of molecules, including proteins, lipids and acid nucleics among which, miRNAs are the most widely studied. The cargo of EVs can be impacted by the culture or environmental conditions that MSCs encounter and by changes in the energy metabolism that regulate the functional properties of MSCs. On the other hand, MSC-derived EVs are also reported to impact the metabolism of target cells. In the present review, we discuss the role of MSC-EVs in the regulation of the energy metabolism and oxidative stress of target cells and tissues with a focus on the role of miRNAs.
Collapse
Affiliation(s)
- Claire Loussouarn
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Yves-Marie Pers
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Department of Rheumatology, Lapeyronie University Hospital, Montpellier, France
| | - Claire Bony
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Department of Rheumatology, Lapeyronie University Hospital, Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Department of Rheumatology, Lapeyronie University Hospital, Montpellier, France
| |
Collapse
|
31
|
Choudhuri S, Chowdhury IH, Garg NJ. Mitochondrial Regulation of Macrophage Response Against Pathogens. Front Immunol 2021; 11:622602. [PMID: 33679710 PMCID: PMC7925834 DOI: 10.3389/fimmu.2020.622602] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/29/2020] [Indexed: 12/18/2022] Open
Abstract
Innate immune cells play the first line of defense against pathogens. Phagocytosis or invasion by pathogens can affect mitochondrial metabolism in macrophages by diverse mechanisms and shape the macrophage response (proinflammatory vs. immunomodulatory) against pathogens. Besides β-nicotinamide adenine dinucleotide 2'-phosphate, reduced (NADPH) oxidase, mitochondrial electron transport chain complexes release superoxide for direct killing of the pathogen. Mitochondria that are injured are removed by mitophagy, and this process can be critical for regulating macrophage activation. For example, impaired mitophagy can result in cytosolic leakage of mitochondrial DNA (mtDNA) that can lead to activation of cGAS-STING signaling pathway of macrophage proinflammatory response. In this review, we will discuss how metabolism, mtDNA, mitophagy, and cGAS-STING pathway shape the macrophage response to infectious agents.
Collapse
Affiliation(s)
- Subhadip Choudhuri
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, United States
| | - Imran Hussain Chowdhury
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, United States
| | - Nisha Jain Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, United States
- Institute for Human Infections and Immunity, UTMB, Galveston, TX, United States
| |
Collapse
|
32
|
Lee H, Kim YI, Nirmala FS, Kim JS, Seo HD, Ha TY, Jang YJ, Jung CH, Ahn J. MiR-141-3p promotes mitochondrial dysfunction in ovariectomy-induced sarcopenia via targeting Fkbp5 and Fibin. Aging (Albany NY) 2021; 13:4881-4894. [PMID: 33534778 PMCID: PMC7950230 DOI: 10.18632/aging.202617] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 12/09/2020] [Indexed: 01/12/2023]
Abstract
Post-menopausal conditions exacerbate the biological aging process and this is often accompanied by visceral adiposity with sarcopenia. Mitochondrial impairment is a hallmark of frailty and sarcopenia in the elderly. However, the exact mechanism underlying the development of obesogenic sarcopenia and the involvement of mitochondria remains unclear. This study confirmed that there is a decline in muscle mass and function as well as mitochondrial dysfunction in the quadriceps of ovariectomized (OVX) mice. To investigate the role of microRNA (miRNA) in this process, we performed miRNA and mRNA arrays and found that miR-141-3p directly targets and downregulates FK506 binding protein 5 (Fkbp5) and Fibin. Overexpression of miR-141-3p decreased mitochondrial function and inhibited myogenic differentiation in C2C12 cells. These effects were mediated by Fkbp5 and Fibin inhibition. Conversely, knockdown of miR-141-3p increased mitochondrial respiration and enhanced myogenesis. Treatment with β-estradiol effectively reversed the palmitic acid-induced upregulation of miR-141-3p and subsequent downregulation of Fkbp5 and Fibin. In conclusion, miR-141-3p is upregulated in OVX mice, and this is associated with mitochondrial dysfunction through inhibition of Fkbp5 and Fibin. These findings suggest that inhibiting miR-141-3p could be a therapeutic target for alleviating obesogenic sarcopenia.
Collapse
Affiliation(s)
- Hyunjung Lee
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, South Korea
| | - Young In Kim
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, South Korea.,Department of Food Science and Technology, Jeonbuk National University, Jeonju-Si, South Korea
| | - Farida S Nirmala
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, South Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, South Korea
| | - Ji-Sun Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Hyo-Deok Seo
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, South Korea
| | - Tae Youl Ha
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, South Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, South Korea
| | - Young-Jin Jang
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, South Korea
| | - Chang Hwa Jung
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, South Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, South Korea
| | - Jiyun Ahn
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, South Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
33
|
Xie WQ, Men C, He M, Li YS, Lv S. The Effect of MicroRNA-Mediated Exercise on Delaying Sarcopenia in Elderly Individuals. Dose Response 2020; 18:1559325820974543. [PMID: 33293908 PMCID: PMC7705785 DOI: 10.1177/1559325820974543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/30/2020] [Accepted: 10/27/2020] [Indexed: 11/24/2022] Open
Abstract
Sarcopenia is often regarded as an early sign of weakness and is the core element
of muscle weakness in elderly individuals. Sarcopenia is closely related to the
reduction of exercise, and elderly individuals often suffer from decreased
muscle mass and function due to a lack of exercise. At present, studies have
confirmed that resistance and aerobic exercise are related to muscle mass,
strength and fiber type and to the activation and proliferation of muscle stem
cells (MuSCs). Increasing evidence shows that microRNAs (miRNAs) play an
important role in exercise-related changes in the quantity, composition and
function of skeletal muscle. At the cellular level, miRNAs have been shown to
regulate the proliferation and differentiation of muscle cells. In addition,
miRNAs are related to the composition and transformation of muscle fibers and
involved in the transition of MuSCs from the resting state to the activated
state. Therefore, exercise may delay sarcopenia in elderly individuals by
regulating miRNAs in skeletal muscle. In future miRNA-focused treatment
strategies, these studies will provide valuable information for the formulation
of exercise methods and will provide useful and targeted exercise programs for
elderly individuals with sarcopenia.
Collapse
Affiliation(s)
- Wen-Qing Xie
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chen Men
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Miao He
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu-Sheng Li
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shan Lv
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
34
|
Torma F, Gombos Z, Jokai M, Berkes I, Takeda M, Mimura T, Radak Z, Gyori F. The roles of microRNA in redox metabolism and exercise-mediated adaptation. JOURNAL OF SPORT AND HEALTH SCIENCE 2020; 9:405-414. [PMID: 32780693 PMCID: PMC7498669 DOI: 10.1016/j.jshs.2020.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 01/03/2020] [Accepted: 02/10/2020] [Indexed: 05/10/2023]
Abstract
MicroRNAs (miRs) are small regulatory RNA transcripts capable of post-transcriptional silencing of mRNA messages by entering a cellular bimolecular apparatus called RNA-induced silencing complex. miRs are involved in the regulation of cellular processes producing, eliminating or repairing the damage caused by reactive oxygen species, and they are active players in redox homeostasis. Increased mitochondrial biogenesis, function and hypertrophy of skeletal muscle are important adaptive responses to regular exercise. In the present review, we highlight some of the redox-sensitive regulatory roles of miRs.
Collapse
Affiliation(s)
- Ferenc Torma
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest 1123, Hungary
| | - Zoltan Gombos
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest 1123, Hungary
| | - Matyas Jokai
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest 1123, Hungary
| | - Istvan Berkes
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest 1123, Hungary
| | - Masaki Takeda
- Faculty of Health and Sports Science, Doshisha University, Kyotanabe 610-0394, Japan
| | - Tatsuya Mimura
- Faculty of Sport and Health Sciences, Osaka Sangyo University, Osaka 573-1004, Japan
| | - Zsolt Radak
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest 1123, Hungary; Faculty of Sport Sciences, Waseda University, Tokorozawa, Saitama 359-1192, Japan; Institute of Physical Education and Sport Science, JGYPK, University of Szeged, Szeged 6726, Hungary.
| | - Ferenc Gyori
- Institute of Physical Education and Sport Science, JGYPK, University of Szeged, Szeged 6726, Hungary
| |
Collapse
|
35
|
Widmann M, Nieß AM, Munz B. Physical Exercise and Epigenetic Modifications in Skeletal Muscle. Sports Med 2020; 49:509-523. [PMID: 30778851 DOI: 10.1007/s40279-019-01070-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Physical activity and sports play major roles in the overall health status of humans. It is well known that regular exercise helps to lower the risk for a broad variety of health problems, such as cardiovascular disease, type 2 diabetes, and cancer. Being physically active induces a wide variety of molecular adaptations, for example fiber type switches or other metabolic alterations, in skeletal muscle tissue. These adaptations are based on exercise-induced changes to the skeletal muscle transcriptome. Understanding their nature is crucial to improve the development of exercise-based therapeutic strategies. Recent research indicates that specifically epigenetic mechanisms, i.e., pathways that induce changes in gene expression patterns without altering the DNA base sequence, might play a major role in controlling skeletal muscle transcriptional patterns. Epigenetic mechanisms include DNA and histone modifications, as well as expression of specific microRNAs. They can be modulated by environmental factors or external stimuli, such as exercise, and eventually induce specific and fine-tuned changes to the transcriptional response. In this review, we highlight current knowledge on epigenetic changes induced in exercising skeletal muscle, their target genes, and resulting phenotypic changes. In addition, we raise the question of whether epigenetic modifications might serve as markers for the design and management of optimized and individualized training protocols, as prognostic tools to predict training adaptation, or even as targets for the design of "exercise mimics".
Collapse
Affiliation(s)
- Manuel Widmann
- Department of Sports Medicine, University Medicine Tübingen, Hoppe-Seyler-Str. 6, D-72076, Tübingen, Germany
| | - Andreas M Nieß
- Department of Sports Medicine, University Medicine Tübingen, Hoppe-Seyler-Str. 6, D-72076, Tübingen, Germany
| | - Barbara Munz
- Department of Sports Medicine, University Medicine Tübingen, Hoppe-Seyler-Str. 6, D-72076, Tübingen, Germany.
| |
Collapse
|
36
|
Yin J, Qian Z, Chen Y, Li Y, Zhou X. MicroRNA regulatory networks in the pathogenesis of sarcopenia. J Cell Mol Med 2020; 24:4900-4912. [PMID: 32281300 PMCID: PMC7205827 DOI: 10.1111/jcmm.15197] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/25/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
Sarcopenia is an age‐related disease characterized by disturbed homeostasis of skeletal muscle, leading to a decline in muscle mass and function. Loss of muscle mass and strength leads to falls and fracture, and is often accompanied by other geriatric diseases, including osteoporosis, frailty and cachexia, resulting in a general decrease in quality of life and an increase in mortality. Although the underlying mechanisms of sarcopenia are still not completely understood, there has been a marked improvement in the understanding of the pathophysiological changes leading to sarcopenia in recent years. The role of microRNAs (miRNAs), especially, has been clearer in skeletal muscle development and homeostasis. miRNAs form part of a gene regulatory network and have numerous activities in many biological processes. Intervention based on miRNAs may develop into an innovative treatment strategy to conquer sarcopenia. This review is divided into three sections: firstly, the latest understanding of the pathogenesis of sarcopenia is summarized; secondly, increasing evidence for the involvement of miRNAs in the regulation of muscle quantity or quality and muscle homeostasis is highlighted; and thirdly, the possibilities and limitations of miRNAs as a treatment for sarcopenia are explored.
Collapse
Affiliation(s)
- Jiayu Yin
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiyuan Qian
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuqi Chen
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Li
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiang Zhou
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
37
|
Jusic A, Devaux Y. Mitochondrial noncoding RNA-regulatory network in cardiovascular disease. Basic Res Cardiol 2020; 115:23. [PMID: 32140778 DOI: 10.1007/s00395-020-0783-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/17/2020] [Indexed: 12/22/2022]
Abstract
Mitochondrial function and integrity are vital for the maintenance of cellular homeostasis, particularly in high-energy demanding cells. Cardiomyocytes have a large number of mitochondria, which provide a continuous and bulk supply of the ATP necessary for cardiac mechanical function. More than 90% of the ATP consumed by the heart is derived from the mitochondrial oxidative metabolism. Decreased energy supply as the main consequence of mitochondrial dysfunction is closely linked to cardiovascular disease (CVD). The discovery of noncoding RNA (ncRNAs) in the mitochondrial compartment has changed the traditional view of molecular pathways involved in the regulatory network of CVD. Mitochondrial ncRNAs participate in controlling cardiovascular pathogenesis by regulating glycolysis, mitochondrial energy status, and the expression of genes involved in mitochondrial metabolism. Understanding the underlying mechanisms of the association between impaired mitochondrial function resulting from fluctuation in expression levels of ncRNAs and specific disease phenotype can aid in preventing and treating CVD. This review presents an overview of the role of mitochondrial ncRNAs in the complex regulatory network of the cardiovascular pathology. We will summarize and discuss (1) mitochondrial microRNAs (mitomiRs) and long noncoding RNAs (lncRNAs) encoded either by nuclear or mitochondrial genome which are involved in the regulation of mitochondrial metabolism; (2) the role of mitomiRs and lncRNAs in the pathogenesis of several CVD such as hypertension, cardiac hypertrophy, acute myocardial infarction and heart failure; (3) the biomarker and therapeutic potential of mitochondrial ncRNAs in CVD; (4) and the challenges inherent to their translation into clinical application.
Collapse
Affiliation(s)
- Amela Jusic
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, 1A-B rue Edison, 1445, Strassen, Luxembourg.
| | | |
Collapse
|
38
|
Gusic M, Prokisch H. ncRNAs: New Players in Mitochondrial Health and Disease? Front Genet 2020; 11:95. [PMID: 32180794 PMCID: PMC7059738 DOI: 10.3389/fgene.2020.00095] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/28/2020] [Indexed: 12/19/2022] Open
Abstract
The regulation of mitochondrial proteome is unique in that its components have origins in both mitochondria and nucleus. With the development of OMICS technologies, emerging evidence indicates an interaction between mitochondria and nucleus based not only on the proteins but also on the non-coding RNAs (ncRNAs). It is now accepted that large parts of the non‐coding genome are transcribed into various ncRNA species. Although their characterization has been a hot topic in recent years, the function of the majority remains unknown. Recently, ncRNA species microRNA (miRNA) and long-non coding RNAs (lncRNA) have been gaining attention as direct or indirect modulators of the mitochondrial proteome homeostasis. These ncRNA can impact mitochondria indirectly by affecting transcripts encoding for mitochondrial proteins in the cytoplasm. Furthermore, reports of mitochondria-localized miRNAs, termed mitomiRs, and lncRNAs directly regulating mitochondrial gene expression suggest the import of RNA to mitochondria, but also transcription from the mitochondrial genome. Interestingly, ncRNAs have been also shown to hide small open reading frames (sORFs) encoding for small functional peptides termed micropeptides, with several examples reported with a role in mitochondria. In this review, we provide a literature overview on ncRNAs and micropeptides found to be associated with mitochondrial biology in the context of both health and disease. Although reported, small study overlap and rare replications by other groups make the presence, transport, and role of ncRNA in mitochondria an attractive, but still challenging subject. Finally, we touch the topic of their potential as prognosis markers and therapeutic targets.
Collapse
Affiliation(s)
- Mirjana Gusic
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.,Institute of Human Genetics, Technical University of Munich, Munich, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Institute of Human Genetics, Technical University of Munich, Munich, Germany
| |
Collapse
|
39
|
Oka SI, Sabry AD, Cawley KM, Warren JS. Multiple Levels of PGC-1α Dysregulation in Heart Failure. Front Cardiovasc Med 2020; 7:2. [PMID: 32083094 PMCID: PMC7002390 DOI: 10.3389/fcvm.2020.00002] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/08/2020] [Indexed: 12/13/2022] Open
Abstract
Metabolic adaption is crucial for the heart to sustain its contractile activity under various physiological and pathological conditions. At the molecular level, the changes in energy demand impinge on the expression of genes encoding for metabolic enzymes. Among the major components of an intricate transcriptional circuitry, peroxisome proliferator-activated receptor γ coactivator 1 alpha (PGC-1α) plays a critical role as a metabolic sensor, which is responsible for the fine-tuning of transcriptional responses to a plethora of stimuli. Cumulative evidence suggests that energetic impairment in heart failure is largely attributed to the dysregulation of PGC-1α. In this review, we summarize recent studies revealing how PGC-1α is regulated by a multitude of mechanisms, operating at different regulatory levels, which include epigenetic regulation, the expression of variants, post-transcriptional inhibition, and post-translational modifications. We further discuss how the PGC-1α regulatory cascade can be impaired in the failing heart.
Collapse
Affiliation(s)
- Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Amira D Sabry
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Keiko M Cawley
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Junco S Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States.,Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
40
|
Mitochondrial MiRNA in Cardiovascular Function and Disease. Cells 2019; 8:cells8121475. [PMID: 31766319 PMCID: PMC6952824 DOI: 10.3390/cells8121475] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/13/2019] [Accepted: 11/18/2019] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs functioning as crucial post-transcriptional regulators of gene expression involved in cardiovascular development and health. Recently, mitochondrial miRNAs (mitomiRs) have been shown to modulate the translational activity of the mitochondrial genome and regulating mitochondrial protein expression and function. Although mitochondria have been verified to be essential for the development and as a therapeutic target for cardiovascular diseases, we are just beginning to understand the roles of mitomiRs in the regulation of crucial biological processes, including energy metabolism, oxidative stress, inflammation, and apoptosis. In this review, we summarize recent findings regarding how mitomiRs impact on mitochondrial gene expression and mitochondrial function, which may help us better understand the contribution of mitomiRs to both the regulation of cardiovascular function under physiological conditions and the pathogenesis of cardiovascular diseases.
Collapse
|
41
|
Piccinin E, Arconzo M, Graziano G, Vacca M, Peres C, Bellafante E, Villani G, Moschetta A. Hepatic microRNA Expression by PGC-1α and PGC-1β in the Mouse. Int J Mol Sci 2019; 20:ijms20225735. [PMID: 31731670 PMCID: PMC6888418 DOI: 10.3390/ijms20225735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
The fine-tuning of liver metabolism is essential to maintain the whole-body homeostasis and to prevent the onset of diseases. The peroxisome proliferator-activated receptor-γ coactivators (PGC-1s) are transcriptional key players of liver metabolism, able to regulate mitochondrial function, gluconeogenesis and lipid metabolism. Their activity is accurately modulated by post-translational modifications. Here, we showed that specific PGC-1s expression can lead to the upregulation of different microRNAs widely implicated in liver physiology and diseases development and progression, thus offering a new layer of complexity in the control of hepatic metabolism.
Collapse
Affiliation(s)
- Elena Piccinin
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy or
| | - Maria Arconzo
- INBB, National Institute for Biostuctures and Biosystems, 00136 Rome, Italy; (M.A.); (G.G.); (C.P.)
| | - Giusi Graziano
- INBB, National Institute for Biostuctures and Biosystems, 00136 Rome, Italy; (M.A.); (G.G.); (C.P.)
| | - Michele Vacca
- Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK;
| | - Claudia Peres
- INBB, National Institute for Biostuctures and Biosystems, 00136 Rome, Italy; (M.A.); (G.G.); (C.P.)
| | - Elena Bellafante
- Fondazione Mario Negri Sud, Santa Maria Imbaro, 66030 Chieti, Italy;
| | - Gaetano Villani
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, “Aldo Moro” University of Bari, 70124 Bari, Italy;
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy or
- INBB, National Institute for Biostuctures and Biosystems, 00136 Rome, Italy; (M.A.); (G.G.); (C.P.)
- Correspondence: or ; Tel.: +39-080-559-3262
| |
Collapse
|
42
|
How Epigenetic Modifications Drive the Expression and Mediate the Action of PGC-1α in the Regulation of Metabolism. Int J Mol Sci 2019; 20:ijms20215449. [PMID: 31683747 PMCID: PMC6862278 DOI: 10.3390/ijms20215449] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
Epigenetic changes are a hallmark of short- and long-term transcriptional regulation, and hence instrumental in the control of cellular identity and plasticity. Epigenetic mechanisms leading to changes in chromatin structure, accessibility for recruitment of transcriptional complexes, and interaction of enhancers and promoters all contribute to acute and chronic adaptations of cells, tissues and organs to internal and external perturbations. Similarly, the peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) is activated by stimuli that alter the cellular energetic demand, and subsequently controls complex transcriptional networks responsible for cellular plasticity. It thus is of no surprise that PGC-1α is under the control of epigenetic mechanisms, and constitutes a mediator of epigenetic changes in various tissues and contexts. In this review, we summarize the current knowledge of the link between epigenetics and PGC-1α in health and disease.
Collapse
|
43
|
Kawamura A, Aoi W, Abe R, Kobayashi Y, Wada S, Kuwahata M, Higashi A. Combined intake of astaxanthin, β-carotene, and resveratrol elevates protein synthesis during muscle hypertrophy in mice. Nutrition 2019; 69:110561. [PMID: 31539816 DOI: 10.1016/j.nut.2019.110561] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/23/2019] [Accepted: 05/12/2019] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The antioxidant factors, astaxanthin, β-carotene, and resveratrol, have a potential effect on protein synthesis in skeletal muscle and a combined intake may have a greater cumulative effect than individual intake. The aim of this study was to investigate the combined effects on skeletal muscle mass and protein metabolic signaling during the hypertrophic process from atrophy in mice. METHODS Male ICR mice were divided into five dietary groups consisting of seven animals each: normal, astaxanthin, β-carotene, resveratrol, and all three antioxidants. Equal concentrations (0.06% [w/w]) of the respective antioxidants were included in the diet of each group. In the mixed group, three antioxidants were added in equal proportion. One leg of each mouse was casted for 3 wk to induce muscle atrophy. After removal of the cast, the mice were fed each diet for 2 wk. The muscle tissues were collected, weighed, and examined for protein metabolism signaling and oxidative damage. RESULTS The weight of the soleus muscle was increased in the astaxanthin, β-carotene, and resveratrol groups to a greater extent than in the normal group; this was accelerated by intake of the mixed antioxidants (P = 0.007). Phosphorylation levels of mammalian target of rapamycin and p70 S6 K in the muscle were higher in the mixed antioxidant group than in the normal group (P = 0.025; P = 0.020). The carbonylated protein concentration was lower in the mixed antioxidant group than in the normal group (P = 0.021). CONCLUSIONS These results suggested that a combination of astaxanthin, β-carotene, and resveratrol, even in small amounts, promoted protein synthesis during the muscle hypertrophic process following atrophy.
Collapse
Affiliation(s)
- Aki Kawamura
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan; Sports Science Research Promotion Center, Nippon Sport Science University, Tokyo, Japan
| | - Wataru Aoi
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan.
| | - Ryo Abe
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan; Wakayama Medical University Hospital, Wakayama, Japan
| | - Yukiko Kobayashi
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Sayori Wada
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Masashi Kuwahata
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Akane Higashi
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| |
Collapse
|
44
|
Aoi W, Hirano N, Lassiter DG, Björnholm M, Chibalin AV, Sakuma K, Tanimura Y, Mizushima K, Takagi T, Naito Y, Zierath JR, Krook A. Secreted protein acidic and rich in cysteine (SPARC) improves glucose tolerance via AMP-activated protein kinase activation. FASEB J 2019; 33:10551-10562. [PMID: 31225998 DOI: 10.1096/fj.201900453r] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
During exercise, skeletal muscles release cytokines, peptides, and metabolites that exert autocrine, paracrine, or endocrine effects on glucose homeostasis. In this study, we investigated the effects of secreted protein acidic and rich in cysteine (SPARC), an exercise-responsive myokine, on glucose metabolism in human and mouse skeletal muscle. SPARC-knockout mice showed impaired systemic metabolism and reduced phosphorylation of AMPK and protein kinase B in skeletal muscle. Treatment of SPARC-knockout mice with recombinant SPARC improved glucose tolerance and concomitantly activated AMPK in skeletal muscle. These effects were dependent on AMPK-γ3 because SPARC treatment enhanced skeletal muscle glucose uptake in wild-type mice but not in AMPK-γ3-knockout mice. SPARC strongly interacted with the voltage-dependent calcium channel, and inhibition of calcium-dependent signaling prevented SPARC-induced AMPK phosphorylation in human and mouse myotubes. Finally, chronic SPARC treatment improved systemic glucose tolerance and AMPK signaling in skeletal muscle of high-fat diet-induced obese mice, highlighting the efficacy of SPARC treatment in the management of metabolic diseases. Thus, our findings suggest that SPARC treatment mimics the effects of exercise on glucose tolerance by enhancing AMPK-dependent glucose uptake in skeletal muscle.-Aoi, W., Hirano, N., Lassiter, D. G., Björnholm, M., Chibalin, A. V., Sakuma, K., Tanimura, Y., Mizushima, K., Takagi, T., Naito, Y., Zierath, J. R., Krook, A. Secreted protein acidic and rich in cysteine (SPARC) improves glucose tolerance via AMP-activated protein kinase activation.
Collapse
Affiliation(s)
- Wataru Aoi
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan.,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Nariyuki Hirano
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - David G Lassiter
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Marie Björnholm
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Kunihiro Sakuma
- Institute for Liberal Arts, School of Environment and Society, Tokyo Institute of Technology, Tokyo, Japan
| | - Yuko Tanimura
- Faculty of Human, Aichi-Toho University, Nagoya, Japan
| | - Katsura Mizushima
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohisa Takagi
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Naito
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
45
|
Soledad RB, Charles S, Samarjit D. The secret messages between mitochondria and nucleus in muscle cell biology. Arch Biochem Biophys 2019; 666:52-62. [PMID: 30935885 PMCID: PMC6538274 DOI: 10.1016/j.abb.2019.03.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/01/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
Over two thousand proteins are found in the mitochondrial compartment but the mitochondrial genome codes for only 13 proteins. The majority of mitochondrial proteins are products of nuclear genes and are synthesized in the cytosol, then translocated into the mitochondria. Most of the subunits of the five respiratory chain complexes in the inner mitochondrial membrane, which generate a proton gradient across the membrane and produce ATP, are encoded by nuclear genes. Therefore, it is quite clear that import of nuclear-encoded proteins into the mitochondria is essential for mitochondrial function. Nuclear to mitochondrial communication is well studied. However, there is another arm to this communication, mitochondria to nucleus retrograde signaling. This plays an important role in the maintenance of cellular homeostasis, and is less well studied. Several transcription factors, including Sp1, SIRT3 and GSP2, are activated by altered mitochondrial function. These activated transcription factors then translocate to the nucleus. Based on the mitochondrially generated molecular signal, nuclear genes are targeted, which alters transcription of nuclear genes that code for mitochondrial proteins. This review article will mainly focus on this interactive and bi-directional communication between mitochondria and nucleus, and how this communication plays a significant role in muscle cell biology.
Collapse
Affiliation(s)
| | - Steenbergen Charles
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Das Samarjit
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
46
|
Sannicandro AJ, Soriano-Arroquia A, Goljanek-Whysall K. Micro(RNA)-managing muscle wasting. J Appl Physiol (1985) 2019; 127:619-632. [PMID: 30991011 DOI: 10.1152/japplphysiol.00961.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Progressive skeletal muscle wasting is a natural consequence of aging and is common in chronic and acute diseases. Loss of skeletal muscle mass and function (strength) often leads to frailty, decreased independence, and increased risk of hospitalization. Despite progress made in our understanding of the mechanisms underlying muscle wasting, there is still no treatment available, with exercise training and dietary supplementation improving, but not restoring, muscle mass and/or function. There has been slow progress in developing novel therapies for muscle wasting, either during aging or disease, partially due to the complex nature of processes underlying muscle loss. The mechanisms of muscle wasting are multifactorial, with a combination of factors underlying age- and disease-related functional muscle decline. These factors include well-characterized changes in muscle such as changes in protein turnover and more recently described mechanisms such as autophagy or satellite cell senescence. Advances in transcriptomics and other high-throughput approaches have highlighted significant deregulation of skeletal muscle gene and protein levels during aging and disease. These changes are regulated at different levels, including posttranscriptional gene expression regulation by microRNAs. microRNAs, potent regulators of gene expression, modulate many processes in muscle, and microRNA-based interventions have been recently suggested as a promising new therapeutic strategy against alterations in muscle homeostasis. Here, we review recent developments in understanding the aging-associated mechanisms of muscle wasting and explore potential microRNA-based therapeutic avenues.
Collapse
Affiliation(s)
- Anthony J Sannicandro
- Department of Physiology, School of Medicine, National University of Ireland, Galway, Ireland
| | - Ana Soriano-Arroquia
- Institute of Ageing and Chronic Disease, University of Liverpool, United Kingdom
| | - Katarzyna Goljanek-Whysall
- Department of Physiology, School of Medicine, National University of Ireland, Galway, Ireland.,Institute of Ageing and Chronic Disease, University of Liverpool, United Kingdom
| |
Collapse
|
47
|
Kiang JG, Olabisi AO. Radiation: a poly-traumatic hit leading to multi-organ injury. Cell Biosci 2019; 9:25. [PMID: 30911370 PMCID: PMC6417034 DOI: 10.1186/s13578-019-0286-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/27/2019] [Indexed: 01/16/2023] Open
Abstract
The range of radiation threats we face today includes everything from individual radiation exposures to mass casualties resulting from a terrorist incident, and many of these exposure scenarios include the likelihood of additional traumatic injury as well. Radiation injury is defined as an ionizing radiation exposure inducing a series of organ injury within a specified time. Severity of organ injury depends on the radiation dose and the duration of radiation exposure. Organs and cells with high sensitivity to radiation injury are the skin, the hematopoietic system, the gastrointestinal (GI) tract, spermatogenic cells, and the vascular system. In general, acute radiation syndrome (ARS) includes DNA double strand breaks (DSB), hematopoietic syndrome (bone marrow cells and circulatory cells depletion), cutaneous injury, GI death, brain hemorrhage, and splenomegaly within 30 days after radiation exposure. Radiation injury sensitizes target organs and cells resulting in ARS. Among its many effects on tissue integrity at various levels, radiation exposure results in activation of the iNOS/NF-kB/NF-IL6 and p53/Bax pathways; and increases DNA single and double strand breaks, TLR signaling, cytokine concentrations, bacterial infection, cytochrome c release from mitochondria to cytoplasm, and possible PARP-dependent NAD and ATP-pool depletion. These alterations lead to apoptosis and autophagy and, as a result, increased mortality. In this review, we summarize what is known about how radiation exposure leads to the radiation response with time. We also describe current and prospective countermeasures relevant to the treatment and prevention of radiation injury.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889 USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814 USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814 USA
| | - Ayodele O. Olabisi
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889 USA
| |
Collapse
|
48
|
Loss of microRNA-23-27-24 clusters in skeletal muscle is not influential in skeletal muscle development and exercise-induced muscle adaptation. Sci Rep 2019; 9:1092. [PMID: 30705375 PMCID: PMC6355808 DOI: 10.1038/s41598-018-37765-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/06/2018] [Indexed: 01/21/2023] Open
Abstract
MicroRNAs are small regulatory noncoding RNAs that repress gene expression at the posttranscriptional level. Previous studies have reported that the expression of miR-23, miR-27, and miR-24, driven from two miR-23–27–24 clusters, is altered by various pathophysiological conditions. However, their functions in skeletal muscle have not been clarified. To define the roles of the miR-23–27–24 clusters in skeletal muscle, we generated double-knockout (dKO) mice muscle-specifically lacking the miR-23–27–24 clusters. The dKO mice were viable and showed normal growth. The contractile and metabolic features of the muscles, represented by the expression of the myosin heavy chain and the oxidative markers PGC1-α and COX IV, were not altered in the dKO mice compared with wild-type mice. The dKO mice showed increased cross-sectional areas of the oxidative fibers. However, this dKO did not induce functional changes in the muscles. The dKO mice also showed normal adaptation to voluntary wheel running for 4 weeks, including the glycolytic-to-oxidative fiber type switch, and increases in mitochondrial markers, succinate dehydrogenase activity, and angiogenesis. In conclusion, our data demonstrate that the miR-23–27–24 clusters have subtle effects on skeletal muscle development and endurance-exercise-induced muscle adaptation.
Collapse
|
49
|
Sahu B, Pani S, Swalsingh G, Bal NC. Non and Epigenetic Mechanisms in Regulation of Adaptive Thermogenesis in Skeletal Muscle. Front Endocrinol (Lausanne) 2019; 10:517. [PMID: 31456746 PMCID: PMC6700214 DOI: 10.3389/fendo.2019.00517] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 07/15/2019] [Indexed: 01/07/2023] Open
|
50
|
Exercise and the control of muscle mass in human. Pflugers Arch 2018; 471:397-411. [DOI: 10.1007/s00424-018-2217-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 12/19/2022]
|