1
|
Arif I, Rasheed A, Nazeer S, Shahid F. Physiological and morphological impact of physical activity and nutritional interventions to offset disuse-induced skeletal muscle atrophy. Eur J Transl Myol 2025. [PMID: 40231413 DOI: 10.4081/ejtm.2025.13177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/31/2025] [Indexed: 04/16/2025] Open
Abstract
Skeletal muscle tissue acts as a functional unit for physical movements, energy metabolism, thermogenesis, and metabolic homeostasis. In this literature review, the underlying mechanisms of skeletal muscle atrophy and the prevention strategies, including vigorous training and nutritional modifications are focused. Furthermore, the comparative analysis of multiple interventions is briefly explained. Ageing is an inevitable process often associated with cognitive impairment and physical decline due to muscular atrophy. Skeletal muscle atrophy is characterized by low muscle mass due to multiple underlying factors, i.e., genetic predisposition, ageing, inflammation, and trauma. The structural alterations include myofiber shrinkage, changes in myosin isoforms, decrease in myofiber diameter, and total protein loss. Furthermore, there is an imbalance in protein anabolic and catabolic reactions. This may be due to changes in multiple signal transduction pathways of protein degradation (i.e., caspase, calpain, ubiquitin protein degradation system, autophagy) and protein anabolism via the mTOR pathway. Consequently, certain pathophysiological factors associated with health disparities may reduce mobility and functional capacity to perform ADLs. To tackle this issue, novel strategies linked to physical movement, and dietary intake must be incorporated in life. Exercise poses multiple health benefits, including improved muscle mass and mobility. Diet diversification [particularly protein-rich meals] and the "whole food" approach (based on non-protein nutrients) may enhance intramuscular anabolic signaling and tissue remodeling. However, there is a pressing need to fund large-scale evidence-based trials based on modern machine learning techniques (AI-driven nutrition). Additionally, entrepreneurial platforms for commercialization of consumer-friendly food products must be initiated in future.
Collapse
Affiliation(s)
- Irfan Arif
- Department of Health and Medical Sciences, University of Southern Queensland, Toowoomba.
| | - Ayesha Rasheed
- Department of Medical and Dental Sciences, University of Birmingham, Birmingham.
| | - Sadia Nazeer
- Department of Food Science and Technology, Government College University Faisalabad, Faisalabad.
| | - Fareeha Shahid
- Department of National Institute of Food Science and Technology, University of Agriculture Faisalabad, Faisalabad.
| |
Collapse
|
2
|
Zhou X, Li S, Wang L, Wang J, Zhang P, Chen X. The emerging role of exercise preconditioning in preventing skeletal muscle atrophy. Front Physiol 2025; 16:1559594. [PMID: 40206380 PMCID: PMC11979144 DOI: 10.3389/fphys.2025.1559594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Skeletal muscle atrophy, characterized by the loss of muscle mass and function, can result from disuse, aging, disease, drug. Exercise preconditioning-a form of exercise training performed before these harmful threats-induces notable remodeling and extensive biochemical adaptations in skeletal muscle, creating a protective phenotype in muscle fibers, and thus serving as an effective intervention for preventing skeletal muscle atrophy. Here, we review the current understanding relating to how exercise preconditioning protects skeletal muscle from damage caused by inactivity, sarcopenia, disease, or pharmacological intervention, with an emphasis on the cellular mechanisms involved. Key mechanisms highlighted as making a significant contribution to the protective effects of exercise on skeletal muscle fibers include mitochondria; the expression of cytoprotective proteins such as HSP72, SOD2, SESN2, PGC-1α and AMPK; and the regulation of oxidative stress. These findings underscore the potential of exercise preconditioning as a non-pharmacological intervention for preserving muscle mass and function, as well as preventing muscular atrophy, ultimately improving the quality of life for at-risk populations.
Collapse
Affiliation(s)
- Xu Zhou
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Shiming Li
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Lu Wang
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Jun Wang
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Peng Zhang
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoping Chen
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
3
|
Zhang C, Chang X, Zhao D, He Y, Dong G, Gao L. Mitochondria and myocardial ischemia/reperfusion injury: Effects of Chinese herbal medicine and the underlying mechanisms. J Pharm Anal 2025; 15:101051. [PMID: 39931135 PMCID: PMC11808734 DOI: 10.1016/j.jpha.2024.101051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 02/03/2025] Open
Abstract
Ischemic heart disease (IHD) is associated with high morbidity and mortality rates. Reperfusion therapy is the best treatment option for this condition. However, reperfusion can aggravate myocardial damage through a phenomenon known as myocardial ischemia/reperfusion (I/R) injury, which has recently gained the attention of researchers. Several studies have shown that Chinese herbal medicines and their natural monomeric components exert therapeutic effects against I/R injury. This review outlines the current knowledge on the pathological mechanisms through which mitochondria participate in I/R injury, focusing on the issues related to energy metabolism, mitochondrial quality control disorders, oxidative stress, and calcium. The mechanisms by which mitochondria mediate cell death have also been discussed. To develop a resource for the prevention and management of clinical myocardial I/R damage, we compiled the most recent research on the effects of Chinese herbal remedies and their monomer components.
Collapse
Affiliation(s)
- Chuxin Zhang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xing Chang
- Guang'anmen Hospital of Chinese Academy of Traditional Chinese Medicine, Beijing, 100053, China
| | - Dandan Zhao
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu He
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Guangtong Dong
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Lin Gao
- Beijing University of Chinese Medicine, Beijing, 100029, China
| |
Collapse
|
4
|
Powers SK. Ventilator-induced diaphragm dysfunction: phenomenology and mechanism(s) of pathogenesis. J Physiol 2024; 602:4729-4752. [PMID: 39216087 DOI: 10.1113/jp283860] [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: 03/03/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Mechanical ventilation (MV) is used to support ventilation and pulmonary gas exchange in patients during critical illness and surgery. Although MV is a life-saving intervention for patients in respiratory failure, an unintended side-effect of MV is the rapid development of diaphragmatic atrophy and contractile dysfunction. This MV-induced diaphragmatic weakness is labelled as 'ventilator-induced diaphragm dysfunction' (VIDD). VIDD is an important clinical problem because diaphragmatic weakness is a risk factor for the failure to wean patients from MV. Indeed, the inability to remove patients from ventilator support results in prolonged hospitalization and increased morbidity and mortality. The pathogenesis of VIDD has been extensively investigated, revealing that increased mitochondrial production of reactive oxygen species within diaphragm muscle fibres promotes a cascade of redox-regulated signalling events leading to both accelerated proteolysis and depressed protein synthesis. Together, these events promote the rapid development of diaphragmatic atrophy and contractile dysfunction. This review highlights the MV-induced changes in the structure/function of diaphragm muscle and discusses the cell-signalling mechanisms responsible for the pathogenesis of VIDD. This report concludes with a discussion of potential therapeutic opportunities to prevent VIDD and suggestions for future research in this exciting field.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
5
|
Zhang J, Tu R, Guan F, Feng J, Jia J, Zhou J, Wang X, Liu L. Irisin attenuates ventilator-induced diaphragmatic dysfunction by inhibiting endoplasmic reticulum stress through activation of AMPK. J Cell Mol Med 2024; 28:e18259. [PMID: 38676364 PMCID: PMC11053354 DOI: 10.1111/jcmm.18259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 11/25/2023] [Accepted: 01/12/2024] [Indexed: 04/28/2024] Open
Abstract
Mechanical ventilation (MV) is an essential life-saving technique, but prolonged MV can cause significant diaphragmatic dysfunction due to atrophy and decreased contractility of the diaphragm fibres, called ventilator-induced diaphragmatic dysfunction (VIDD). It is not clear about the mechanism of occurrence and prevention measures of VIDD. Irisin is a newly discovered muscle factor that regulates energy metabolism. Studies have shown that irisin can exhibit protective effects by downregulating endoplasmic reticulum (ER) stress in a variety of diseases; whether irisin plays a protective role in VIDD has not been reported. Sprague-Dawley rats were mechanically ventilated to construct a VIDD model, and intervention was performed by intravenous administration of irisin. Diaphragm contractility, degree of atrophy, cross-sectional areas (CSAs), ER stress markers, AMPK protein expression, oxidative stress indicators and apoptotic cell levels were measured at the end of the experiment.Our findings showed that as the duration of ventilation increased, the more severe the VIDD was, the degree of ER stress increased, and the expression of irisin decreased.ER stress may be one of the causes of VIDD. Intervention with irisin ameliorated VIDD by reducing the degree of ER stress, attenuating oxidative stress, and decreasing the apoptotic index. MV decreases the expression of phosphorylated AMPK in the diaphragm, whereas the use of irisin increases the expression of phosphorylated AMPK. Irisin may exert its protective effect by activating the phosphorylated AMPK pathway.
Collapse
Affiliation(s)
- Jumei Zhang
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Anesthesiology and Critical Care Medicine Key Laboratory of LuzhouSouthwest Medical UniversityLuzhouChina
| | - Rui Tu
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Anesthesiology and Critical Care Medicine Key Laboratory of LuzhouSouthwest Medical UniversityLuzhouChina
| | - Fasheng Guan
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Anesthesiology and Critical Care Medicine Key Laboratory of LuzhouSouthwest Medical UniversityLuzhouChina
| | - Jianguo Feng
- Anesthesiology and Critical Care Medicine Key Laboratory of LuzhouSouthwest Medical UniversityLuzhouChina
| | - Jing Jia
- Anesthesiology and Critical Care Medicine Key Laboratory of LuzhouSouthwest Medical UniversityLuzhouChina
| | - Jun Zhou
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Xiaobin Wang
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Li Liu
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
6
|
Henriquez-Olguin C, Meneses-Valdes R, Kritsiligkou P, Fuentes-Lemus E. From workout to molecular switches: How does skeletal muscle produce, sense, and transduce subcellular redox signals? Free Radic Biol Med 2023; 209:355-365. [PMID: 37923089 DOI: 10.1016/j.freeradbiomed.2023.10.404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
Skeletal muscle is crucial for maintaining human health and overall quality of life. Acute exercise introduces a multifaceted intracellular stress, with numerous post-translational modifications believed to underpin the health benefits of sustained exercise training. Reactive oxygen species (ROS) are posited to serve as second messengers, triggering cytoprotective adaptations such as the upregulation of enzymatic scavenger systems. However, a significant knowledge gap exists between the generation of oxidants in muscle and the exact mechanisms driving muscle adaptations. This review delves into the current research on subcellular redox biochemistry and its role in the physiological adaptations to exercise. We propose that the subcellular regulation of specific redox modifications is key to ensuring specificity in the intracellular response.
Collapse
Affiliation(s)
- Carlos Henriquez-Olguin
- The August Krogh Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, 2100, Denmark; Exercise Science Laboratory, Faculty of Medicine, Universidad Finis Terrae, Av. Pedro de Valdivia 1509, Santiago, Chile.
| | - Roberto Meneses-Valdes
- The August Krogh Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, 2100, Denmark
| | | | - Eduardo Fuentes-Lemus
- Department of Biomedical Sciences, Panum Institute, Blegdamsvej 3, University of Copenhagen, Copenhagen, 2200, Denmark
| |
Collapse
|
7
|
Nakai H, Hirata Y, Furue H, Oku Y. Electrical stimulation mitigates muscle degradation shift in gene expressions during 12-h mechanical ventilation. Sci Rep 2023; 13:20136. [PMID: 37978221 PMCID: PMC10656540 DOI: 10.1038/s41598-023-47093-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023] Open
Abstract
Ventilator-induced diaphragm dysfunction (VIDD), a dysfunction of the diaphragm muscle caused by prolonged mechanical ventilation (MV), is an important factor that hinders successful weaning from ventilation. We evaluated the effects of electrical stimulation of the diaphragm muscle (pulsed current with off-time intervals) on genetic changes during 12 h of MV (E-V12). Rats were divided into four groups: control, 12-h MV, sham operation, and E-V12 groups. Transcriptome analysis using an RNA microarray revealed that 12-h MV caused upregulation of genes promoting muscle atrophy and downregulation of genes facilitating muscle synthesis, suggesting that 12-h MV is a reasonable method for establishing a VIDD rat model. Of the genes upregulated by 12-h MV, 18 genes were not affected by the sham operation but were downregulated by E-V12. These included genes related to catabolic processes, inflammatory cytokines, and skeletal muscle homeostasis. Of the genes downregulated by 12-h MV, 6 genes were not affected by the sham operation but were upregulated by E-V12. These included genes related to oxygen transport and mitochondrial respiration. These results suggested that 12-h MV shifted gene expression in the diaphragm muscle toward muscle degradation and that electrical stimulation counteracted this shift by suppressing catabolic processes and increasing mitochondrial respiration.
Collapse
Affiliation(s)
- Hideki Nakai
- Physiome, Department of Physiology, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
- Department of Rehabilitation, Hyogo Prefectural Nishinomiya Hospital, 13-9, Rokutanji, Nishinomiya, Hyogo, 662-0918, Japan
| | - Yutaka Hirata
- Physiome, Department of Physiology, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.
| | - Hidemasa Furue
- Department of Neurophysiology, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Yoshitaka Oku
- Physiome, Department of Physiology, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| |
Collapse
|
8
|
Zhang J, Feng J, Jia J, Wang X, Zhou J, Liu L. Research progress on the pathogenesis and treatment of ventilator-induced diaphragm dysfunction. Heliyon 2023; 9:e22317. [PMID: 38053869 PMCID: PMC10694316 DOI: 10.1016/j.heliyon.2023.e22317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 12/07/2023] Open
Abstract
Prolonged controlled mechanical ventilation (CMV) can cause diaphragm fiber atrophy and inspiratory muscle weakness, resulting in diaphragmatic contractile dysfunction, called ventilator-induced diaphragm dysfunction (VIDD). VIDD is associated with higher rates of in-hospital deaths, nosocomial pneumonia, difficulty weaning from ventilators, and increased costs. Currently, appropriate clinical strategies to prevent and treat VIDD are unavailable, necessitating the importance of exploring the mechanisms of VIDD and suitable treatment options to reduce the healthcare burden. Numerous animal studies have demonstrated that ventilator-induced diaphragm dysfunction is associated with oxidative stress, increased protein hydrolysis, disuse atrophy, and calcium ion disorders. Therefore, this article summarizes the molecular pathogenesis and treatment of ventilator-induced diaphragm dysfunction in recent years so that it can be better served clinically and is essential to reduce the duration of mechanical ventilation use, intensive care unit (ICU) length of stay, and the medical burden.
Collapse
Affiliation(s)
- Jumei Zhang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Jianguo Feng
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Jing Jia
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Xiaobin Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| |
Collapse
|
9
|
Kubat GB, Bouhamida E, Ulger O, Turkel I, Pedriali G, Ramaccini D, Ekinci O, Ozerklig B, Atalay O, Patergnani S, Nur Sahin B, Morciano G, Tuncer M, Tremoli E, Pinton P. Mitochondrial dysfunction and skeletal muscle atrophy: Causes, mechanisms, and treatment strategies. Mitochondrion 2023; 72:33-58. [PMID: 37451353 DOI: 10.1016/j.mito.2023.07.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Skeletal muscle, which accounts for approximately 40% of total body weight, is one of the most dynamic and plastic tissues in the human body and plays a vital role in movement, posture and force production. More than just a component of the locomotor system, skeletal muscle functions as an endocrine organ capable of producing and secreting hundreds of bioactive molecules. Therefore, maintaining healthy skeletal muscles is crucial for supporting overall body health. Various pathological conditions, such as prolonged immobilization, cachexia, aging, drug-induced toxicity, and cardiovascular diseases (CVDs), can disrupt the balance between muscle protein synthesis and degradation, leading to skeletal muscle atrophy. Mitochondrial dysfunction is a major contributing mechanism to skeletal muscle atrophy, as it plays crucial roles in various biological processes, including energy production, metabolic flexibility, maintenance of redox homeostasis, and regulation of apoptosis. In this review, we critically examine recent knowledge regarding the causes of muscle atrophy (disuse, cachexia, aging, etc.) and its contribution to CVDs. Additionally, we highlight the mitochondrial signaling pathways involvement to skeletal muscle atrophy, such as the ubiquitin-proteasome system, autophagy and mitophagy, mitochondrial fission-fusion, and mitochondrial biogenesis. Furthermore, we discuss current strategies, including exercise, mitochondria-targeted antioxidants, in vivo transfection of PGC-1α, and the potential use of mitochondrial transplantation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey.
| | - Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Ozgur Ekinci
- Department of Pathology, Gazi University, 06500 Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Ozbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Beyza Nur Sahin
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
10
|
Yang S, Zhang Z, Liu Y, Liu E, Luo Z. The Effects of Combined Respiratory Muscle and Exercise Training in Children with Bronchial Asthma: A Randomised Controlled Study. J Asthma Allergy 2023; 16:293-303. [PMID: 36945322 PMCID: PMC10024882 DOI: 10.2147/jaa.s398108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/26/2023] [Indexed: 03/15/2023] Open
Abstract
Objective To investigate the effects of combined respiratory muscle and exercise training on inspiratory muscle strength, exercise capacity, spirometry measurements, asthma control the quality-of-life in children with asthma. Methods Fifty children with asthma, who were treated in children's hospital of Chongqing medical university in Chongqing between May and December 2021, were selected and randomly divided into a rehabilitation group and a control group by using a random number table. The control group was given routine drug treatment and health education while the rehabilitation group received a combination of respiratory muscle and exercise training on the basis of control group. Results After three months of treatment, the maximum inspiratory pressure, level of asthma control and quality-of-life in the rehabilitation group were significantly improved when compared with those in the control group (P<0.05); there were no significant differences in the 6-minute walking test and spirometry measurements (P>0.05). After three months of treatment, all outcome indicators in the rehabilitation group were significantly improved when compared to those before treatment (P<0.05). The mean value of maximum inspiratory pressure and some indices of spirometry measurements in the control group were significantly improved when compared to those before treatment (P<0.05). Conclusion Combining respiratory muscle and exercise training on the basis of the routine drug treatment and health education significantly improved inspiratory muscle strength, the level of asthma control and the quality-of-life in children with asthma. More research is needed to explore its role in asthma in the future.
Collapse
Affiliation(s)
- Shuhui Yang
- Department of Respiratory Disease, Children’s Hospital of Chongqing Medical University/ National Clinical Research Center for Child Health and Disorders/ Ministry of Education Key Laboratory of Child Development and Disorders/ Chongqing Key Laboratory of Pediatrics, Chongqing, People’s Republic of China
| | - Zhanmei Zhang
- Department of Respiratory Disease, Children’s Hospital of Chongqing Medical University/ National Clinical Research Center for Child Health and Disorders/ Ministry of Education Key Laboratory of Child Development and Disorders/ Chongqing Key Laboratory of Pediatrics, Chongqing, People’s Republic of China
| | - Yulin Liu
- Department of Respiratory Disease, Children’s Hospital of Chongqing Medical University/ National Clinical Research Center for Child Health and Disorders/ Ministry of Education Key Laboratory of Child Development and Disorders/ Chongqing Key Laboratory of Pediatrics, Chongqing, People’s Republic of China
- Correspondence: Yulin Liu, Email
| | - Enmei Liu
- Department of Respiratory Disease, Children’s Hospital of Chongqing Medical University/ National Clinical Research Center for Child Health and Disorders/ Ministry of Education Key Laboratory of Child Development and Disorders/ Chongqing Key Laboratory of Pediatrics, Chongqing, People’s Republic of China
| | - Zhengxiu Luo
- Department of Respiratory Disease, Children’s Hospital of Chongqing Medical University/ National Clinical Research Center for Child Health and Disorders/ Ministry of Education Key Laboratory of Child Development and Disorders/ Chongqing Key Laboratory of Pediatrics, Chongqing, People’s Republic of China
| |
Collapse
|
11
|
Powers SK, Goldstein E, Schrager M, Ji LL. Exercise Training and Skeletal Muscle Antioxidant Enzymes: An Update. Antioxidants (Basel) 2022; 12:antiox12010039. [PMID: 36670901 PMCID: PMC9854578 DOI: 10.3390/antiox12010039] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
The pivotal observation that muscular exercise is associated with oxidative stress in humans was first reported over 45 years ago. Soon after this landmark finding, it was discovered that contracting skeletal muscles produce oxygen radicals and other reactive species capable of oxidizing cellular biomolecules. Importantly, the failure to eliminate these oxidant molecules during exercise results in oxidation of cellular proteins and lipids. Fortuitously, muscle fibers and other cells contain endogenous antioxidant enzymes capable of eliminating oxidants. Moreover, it is now established that several modes of exercise training (e.g., resistance exercise and endurance exercise) increase the expression of numerous antioxidant enzymes that protect myocytes against exercise-induced oxidative damage. This review concisely summarizes the impact of endurance, high-intensity interval, and resistance exercise training on the activities of enzymatic antioxidants within skeletal muscles in humans and other mammals. We also discuss the evidence that exercise-induced up-regulation of cellular antioxidants reduces contraction-induced oxidative damage in skeletal muscles and has the potential to delay muscle fatigue and improve exercise performance. Finally, in hopes of stimulating further research, we also discuss gaps in our knowledge of exercise-induced changes in muscle antioxidant capacity.
Collapse
Affiliation(s)
- Scott K. Powers
- Department of Health Sciences, Stetson University, Deland, FL 32723, USA
- Correspondence:
| | - Erica Goldstein
- Department of Health Sciences, Stetson University, Deland, FL 32723, USA
| | - Matthew Schrager
- Department of Health Sciences, Stetson University, Deland, FL 32723, USA
| | - Li Li Ji
- Department of Kinesiology, University of Minnesota, St Paul, MN 55455, USA
| |
Collapse
|
12
|
Severin R, Franz CK, Farr E, Meirelles C, Arena R, Phillips SA, Bond S, Ferraro F, Faghy M. The effects of COVID-19 on respiratory muscle performance: making the case for respiratory muscle testing and training. Eur Respir Rev 2022; 31:31/166/220006. [PMID: 36198415 PMCID: PMC9724806 DOI: 10.1183/16000617.0006-2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 07/22/2022] [Indexed: 01/28/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection results in multiorgan damage primarily mediated by viral infiltration via angiotensin-converting enzyme-2 receptors on the surface of cells. A primary symptom for many patients is exertional dyspnoea which may persist even beyond recovery from the viral infection. Respiratory muscle (RM) performance was hypothesised as a contributing factor to the severity of coronavirus disease 2019 (COVID-19) symptoms, such as dyspnoea, and outcomes. This was attributed to similarities between patient populations at elevated risk for severe COVID-19 symptoms and those with a greater likelihood of baseline RM weakness and the effects of prolonged mechanical ventilation. More recent evidence suggests that SARS-CoV-2 infection itself may cause damage to the RM, and many patients who have recovered report persistent dyspnoea despite having mild cases, normal lung function or undamaged lung parenchyma. These more recent findings suggest that the role of RM in the persistent dyspnoea due to COVID-19 may be more substantial than originally hypothesised. Therefore, screening for RM weakness and providing interventions to improve RM performance appears to be important for patients with COVID-19. This article will review the impact of SARS-CoV-2 infection on RM performance and provide clinical recommendations for screening RM performance and treatment interventions.
Collapse
Affiliation(s)
- Richard Severin
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA,Department of Physical Therapy, Robbins College of Health and Human Sciences, Baylor University, Waco, TX, USA,Healthy Living for Pandemic Event Protection (HL-PIVOT) Network, Chicago, IL, USA,Corresponding author: Richard Severin ()
| | - Colin K. Franz
- The Shirley Ryan Ability Lab, Chicago, IL, USA,Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ellen Farr
- The Shirley Ryan Ability Lab, Chicago, IL, USA,Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Cristiane Meirelles
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, USA
| | - Ross Arena
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA,Healthy Living for Pandemic Event Protection (HL-PIVOT) Network, Chicago, IL, USA
| | - Shane A. Phillips
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA,Healthy Living for Pandemic Event Protection (HL-PIVOT) Network, Chicago, IL, USA
| | - Sam Bond
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA,Healthy Living for Pandemic Event Protection (HL-PIVOT) Network, Chicago, IL, USA
| | | | - Mark Faghy
- Healthy Living for Pandemic Event Protection (HL-PIVOT) Network, Chicago, IL, USA,Human Sciences Research Centre, University of Derby, Derby, UK
| | | |
Collapse
|
13
|
Karageorgos V, Proklou A, Vaporidi K. Lung and diaphragm protective ventilation: a synthesis of recent data. Expert Rev Respir Med 2022; 16:375-390. [PMID: 35354361 DOI: 10.1080/17476348.2022.2060824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION : To adhere to the Hippocratic Oath, to "first, do no harm", we need to make every effort to minimize the adverse effects of mechanical ventilation. Our understanding of the mechanisms of ventilator-induced lung injury (VILI) and ventilator-induced diaphragm dysfunction (VIDD) has increased in recent years. Research focuses now on methods to monitor lung stress and inhomogeneity and targets we should aim for when setting the ventilator. In parallel, efforts to promote early assisted ventilation to prevent VIDD have revealed new challenges, such as titrating inspiratory effort and synchronizing the mechanical with the patients' spontaneous breaths, while at the same time adhering to lung-protective targets. AREAS COVERED This is a narrative review of the key mechanisms contributing to VILI and VIDD and the methods currently available to evaluate and mitigate the risk of lung and diaphragm injury. EXPERT OPINION Implementing lung and diaphragm protective ventilation requires individualizing the ventilator settings, and this can only be accomplished by exploiting in everyday clinical practice the tools available to monitor lung stress and inhomogeneity, inspiratory effort, and patient-ventilator interaction.
Collapse
Affiliation(s)
- Vlasios Karageorgos
- Department of Intensive Care, University Hospital of Heraklion and University of Crete Medical School, Greece
| | - Athanasia Proklou
- Department of Intensive Care, University Hospital of Heraklion and University of Crete Medical School, Greece
| | - Katerina Vaporidi
- Department of Intensive Care, University Hospital of Heraklion and University of Crete Medical School, Greece
| |
Collapse
|
14
|
Balakrishnan R, Mareedu S, Babu GJ. Reducing sarcolipin expression improves muscle metabolism in mdx mice. Am J Physiol Cell Physiol 2022; 322:C260-C274. [PMID: 34986021 PMCID: PMC8816636 DOI: 10.1152/ajpcell.00125.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an inherited muscle wasting disease. Metabolic impairments and oxidative stress are major secondary mechanisms that severely worsen muscle function in DMD. Here, we sought to determine whether germline reduction or ablation of sarcolipin (SLN), an inhibitor of sarco/endoplasmic reticulum (SR) Ca2+ ATPase (SERCA), improves muscle metabolism and ameliorates muscle pathology in the mdx mouse model of DMD. Glucose and insulin tolerance tests show that glucose clearance rate and insulin sensitivity were improved in the SLN haploinsufficient mdx (mdx:sln+/-) and SLN-deficient mdx (mdx:sln-/-) mice. The histopathological analysis shows that fibrosis and necrosis were significantly reduced in muscles of mdx:sln+/- and mdx:sln-/- mice. SR Ca2+ uptake, mitochondrial complex protein levels, complex activities, mitochondrial Ca2+ uptake and release, and mitochondrial metabolism were significantly improved, and lipid peroxidation and protein carbonylation were reduced in the muscles of mdx:sln+/- and mdx:sln-/- mice. These data demonstrate that reduction or ablation of SLN expression can improve muscle metabolism, reduce oxidative stress, decrease muscle pathology, and protects the mdx mice from glucose intolerance.
Collapse
Affiliation(s)
- Rekha Balakrishnan
- Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, New Jersey
| | - Satvik Mareedu
- Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, New Jersey
| | - Gopal J. Babu
- Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
15
|
Togni G, Puccinelli PJ, Costa T, Seffrin A, de Lira CAB, Vancini RL, Santos DDAT, Nikolaidis PT, Knechtle B, Andrade MS. Factors Associated with Reduction in Physical Activity during the COVID-19 Pandemic in São Paulo, Brazil: An Internet-Based Survey Conducted in June 2020. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:11397. [PMID: 34769910 PMCID: PMC8583209 DOI: 10.3390/ijerph182111397] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The COVID-19 pandemic negatively affected physical activity levels. This study investigated the factors associated with the change in physical activity level in Brazilians residing in the city of São Paulo. METHODS A self-administered questionnaire, addressing personal data, restriction level, education level, family income, daily working hours, and physical activity level, was answered by 2140 volunteers, of which 1179 were excluded because the answers were either incomplete or the respondents were not from São Paulo. The total number of participants selected was 961 (581 female and 380 male). RESULTS The physical activity level adopted prior to the pandemic period (p < 0.001) and family income (p = 0.001) correlated significantly with physical activity level reduction during the pandemic. The proportion of people who reduced their physical activity was greater among those who were very active than those who were active (adjusted prevalence ratio [aPR]: 0.65 [confidence interval (CI): 0.52-0.80]) or insufficiently active [aPR: 0.39 (0.18-0.82)]. The proportion of people who reduced their physical activity was greater among those who received a salary less than minimum wage (MW) than those who received a salary between three to six times minimum wage [(aPR: 0.50 (CI 0.35-0.70)] or more than 6 MW [(aPR: 0.56 (CI 0.40-0.79)]. CONCLUSIONS A higher prevalence of Brazilians residing in the city of São Paulo reduced their physical activity who had a vigorous level of physical activity prior to the pandemic and who received less than a MW.
Collapse
Affiliation(s)
- Gabriela Togni
- Department of Physiology, Federal University of São Paulo, São Paulo 04021-001, Brazil; (G.T.); (P.J.P.); (T.C.); (A.S.); (M.S.A.)
| | - Paulo José Puccinelli
- Department of Physiology, Federal University of São Paulo, São Paulo 04021-001, Brazil; (G.T.); (P.J.P.); (T.C.); (A.S.); (M.S.A.)
| | - Taline Costa
- Department of Physiology, Federal University of São Paulo, São Paulo 04021-001, Brazil; (G.T.); (P.J.P.); (T.C.); (A.S.); (M.S.A.)
| | - Aldo Seffrin
- Department of Physiology, Federal University of São Paulo, São Paulo 04021-001, Brazil; (G.T.); (P.J.P.); (T.C.); (A.S.); (M.S.A.)
| | - Claudio Andre Barbosa de Lira
- Human and Exercise Physiology Division, Faculty of Physical Education and Dance, Federal University of Goiás, Goiânia 74690-900, Brazil
| | - Rodrigo Luiz Vancini
- Center for Physical Education and Sports, Federal University of Espírito Santo, Vitória 29075-910, Brazil;
| | | | | | - Beat Knechtle
- Institute of Primary Care, University of Zurich, 8091 Zurich, Switzerland;
| | - Marilia Santos Andrade
- Department of Physiology, Federal University of São Paulo, São Paulo 04021-001, Brazil; (G.T.); (P.J.P.); (T.C.); (A.S.); (M.S.A.)
| |
Collapse
|
16
|
Memme JM, Slavin M, Moradi N, Hood DA. Mitochondrial Bioenergetics and Turnover during Chronic Muscle Disuse. Int J Mol Sci 2021; 22:ijms22105179. [PMID: 34068411 PMCID: PMC8153634 DOI: 10.3390/ijms22105179] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022] Open
Abstract
Periods of muscle disuse promote marked mitochondrial alterations that contribute to the impaired metabolic health and degree of atrophy in the muscle. Thus, understanding the molecular underpinnings of muscle mitochondrial decline with prolonged inactivity is of considerable interest. There are translational applications to patients subjected to limb immobilization following injury, illness-induced bed rest, neuropathies, and even microgravity. Studies in these patients, as well as on various pre-clinical rodent models have elucidated the pathways involved in mitochondrial quality control, such as mitochondrial biogenesis, mitophagy, fission and fusion, and the corresponding mitochondrial derangements that underlie the muscle atrophy that ensues from inactivity. Defective organelles display altered respiratory function concurrent with increased accumulation of reactive oxygen species, which exacerbate myofiber atrophy via degradative pathways. The preservation of muscle quality and function is critical for maintaining mobility throughout the lifespan, and for the prevention of inactivity-related diseases. Exercise training is effective in preserving muscle mass by promoting favourable mitochondrial adaptations that offset the mitochondrial dysfunction, which contributes to the declines in muscle and whole-body metabolic health. This highlights the need for further investigation of the mechanisms in which mitochondria contribute to disuse-induced atrophy, as well as the specific molecular targets that can be exploited therapeutically.
Collapse
Affiliation(s)
| | | | | | - David A. Hood
- Correspondence: ; Tel.: +1-(416)-736-2100 (ext. 66640)
| |
Collapse
|
17
|
Abstract
Exercise stimulates the biogenesis of mitochondria in muscle. Some literature supports the use of pharmaceuticals to enhance mitochondria as a substitute for exercise. We provide evidence that exercise rejuvenates mitochondrial function, thereby augmenting muscle health with age, in disease, and in the absence of cellular regulators. This illustrates the power of exercise to act as mitochondrial medicine in muscle.
Collapse
Affiliation(s)
- Ashley N Oliveira
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario M3J 1P3, Canada
| | | | | | | |
Collapse
|
18
|
Hermes TDA, Mizobuti DS, da Rocha GL, da Silva HNM, Covatti C, Pereira ECL, Ferretti R, Minatel E. Tempol improves redox status in mdx dystrophic diaphragm muscle. Int J Exp Pathol 2020; 101:289-297. [PMID: 33098599 DOI: 10.1111/iep.12376] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/10/2020] [Accepted: 08/23/2020] [Indexed: 12/15/2022] Open
Abstract
Oxidative stress is a critical element in relationship to the pathophysiology of Duchenne muscular dystrophy (DMD). In the mice the diaphragm (DIA) is most resembles the dystrophic human pathology. In this study we have evaluated the consequences of a synthetic antioxidant (tempol) on oxidative stress parameters in the DIA muscle of mdx mice. The mdx mice were separated into two groups: mdx, the control group receiving intraperitoneal (i.p.) injections of saline solution (100 µL), and mdxT, the treated group receiving i.p. injections of tempol (100 mg/kg). The tempol-treated group showed reduced oxidative stress markers, decreasing the dihydroethidium reaction (DHE) area; autofluorescent lipofuscin granules; and 4-hydroxynonenal (4-HNE)-protein adduct levels. DIA muscle of mdx mice. At the same time, the manganese-superoxide dismutase 2 (SOD2) levels were increased in the tempol-treated group. In addition, the tempol-treated group showed reduced levels of glutathione-disulphide reductase (GSR), glutathione peroxidase 1 (GPx1) and catalase (CAT) in immunoblots. The tempol-treated group has also shown lower relative gene expression of SOD1, CAT and GPx than the non-treated group. Our data demonstrated that tempol treatment reduced oxidant parameters and increased anti-oxidant SOD2 levels in the DIA muscle of mdx mice, which may contribute to the normalization of the redox homeostasis of dystrophic muscles.
Collapse
Affiliation(s)
- Túlio de Almeida Hermes
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Daniela Sayuri Mizobuti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Guilherme Luiz da Rocha
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Caroline Covatti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Elaine Cristina Leite Pereira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil.,Faculty of Ceilandia, University of Brasilia (UnB), Brasília, Brazil
| | - Renato Ferretti
- Department of Anatomy, Institute of Bioscience of Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Elaine Minatel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
19
|
Chen Y, Liu C, Zhou P, Li J, Zhao X, Wang Y, Chen R, Song L, Zhao H, Yan H. Liraglutide reduces coronary endothelial cells no-reflow damage through activating MAPK/ERK signaling pathway. J Recept Signal Transduct Res 2020; 41:553-557. [PMID: 33045879 DOI: 10.1080/10799893.2020.1833921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Yi Chen
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chen Liu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Peng Zhou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiannan Li
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoxiao Zhao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Wang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Runzhen Chen
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Song
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Hanjun Zhao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China
| | - Hongbing Yan
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China
| |
Collapse
|
20
|
Zou H, Liu G. Inhibition of endoplasmic reticulum stress through activation of MAPK/ERK signaling pathway attenuates hypoxia-mediated cardiomyocyte damage. J Recept Signal Transduct Res 2020; 41:532-537. [PMID: 33023351 DOI: 10.1080/10799893.2020.1831534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Huanxue Zou
- Department of Cardiology, Yuyao People’s Hospital, Yuyao, China
| | - Gang Liu
- Department of Cardiology, Yuyao People’s Hospital, Yuyao, China
| |
Collapse
|
21
|
Wang J, Zhou H. Mitochondrial quality control mechanisms as molecular targets in cardiac ischemia -reperfusion injury. Acta Pharm Sin B 2020; 10:1866-1879. [PMID: 33163341 PMCID: PMC7606115 DOI: 10.1016/j.apsb.2020.03.004] [Citation(s) in RCA: 240] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/19/2020] [Accepted: 02/27/2020] [Indexed: 12/23/2022] Open
Abstract
Mitochondrial damage is a critical contributor to cardiac ischemia/reperfusion (I/R) injury. Mitochondrial quality control (MQC) mechanisms, a series of adaptive responses that preserve mitochondrial structure and function, ensure cardiomyocyte survival and cardiac function after I/R injury. MQC includes mitochondrial fission, mitochondrial fusion, mitophagy and mitochondria-dependent cell death. The interplay among these responses is linked to pathological changes such as redox imbalance, calcium overload, energy metabolism disorder, signal transduction arrest, the mitochondrial unfolded protein response and endoplasmic reticulum stress. Excessive mitochondrial fission is an early marker of mitochondrial damage and cardiomyocyte death. Reduced mitochondrial fusion has been observed in stressed cardiomyocytes and correlates with mitochondrial dysfunction and cardiac depression. Mitophagy allows autophagosomes to selectively degrade poorly structured mitochondria, thus maintaining mitochondrial network fitness. Nevertheless, abnormal mitophagy is maladaptive and has been linked to cell death. Although mitochondria serve as the fuel source of the heart by continuously producing adenosine triphosphate, they also stimulate cardiomyocyte death by inducing apoptosis or necroptosis in the reperfused myocardium. Therefore, defects in MQC may determine the fate of cardiomyocytes. In this review, we summarize the regulatory mechanisms and pathological effects of MQC in myocardial I/R injury, highlighting potential targets for the clinical management of reperfusion.
Collapse
Affiliation(s)
- Jin Wang
- Department of Cardiology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| |
Collapse
|
22
|
Smuder AJ, Turner SM, Schuster CM, Morton AB, Hinkley JM, Fuller DD. Hyperbaric Oxygen Treatment Following Mid-Cervical Spinal Cord Injury Preserves Diaphragm Muscle Function. Int J Mol Sci 2020; 21:ijms21197219. [PMID: 33007822 PMCID: PMC7582297 DOI: 10.3390/ijms21197219] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/24/2020] [Accepted: 09/27/2020] [Indexed: 12/17/2022] Open
Abstract
Oxidative damage to the diaphragm as a result of cervical spinal cord injury (SCI) promotes muscle atrophy and weakness. Respiratory insufficiency is the leading cause of morbidity and mortality in cervical spinal cord injury (SCI) patients, emphasizing the need for strategies to maintain diaphragm function. Hyperbaric oxygen (HBO) increases the amount of oxygen dissolved into the blood, elevating the delivery of oxygen to skeletal muscle and reactive oxygen species (ROS) generation. It is proposed that enhanced ROS production due to HBO treatment stimulates adaptations to diaphragm oxidative capacity, resulting in overall reductions in oxidative stress and inflammation. Therefore, we tested the hypothesis that exposure to HBO therapy acutely following SCI would reduce oxidative damage to the diaphragm muscle, preserving muscle fiber size and contractility. Our results demonstrated that lateral contusion injury at C3/4 results in a significant reduction in diaphragm muscle-specific force production and fiber cross-sectional area, which was associated with augmented mitochondrial hydrogen peroxide emission and a reduced mitochondrial respiratory control ratio. In contrast, rats that underwent SCI followed by HBO exposure consisting of 1 h of 100% oxygen at 3 atmospheres absolute (ATA) delivered for 10 consecutive days demonstrated an improvement in diaphragm-specific force production, and an attenuation of fiber atrophy, mitochondrial dysfunction and ROS production. These beneficial adaptations in the diaphragm were related to HBO-induced increases in antioxidant capacity and a reduction in atrogene expression. These findings suggest that HBO therapy may be an effective adjunctive therapy to promote respiratory health following cervical SCI.
Collapse
Affiliation(s)
- Ashley J. Smuder
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA; (A.B.M.); (J.M.H.)
- Breathing Research and Therapeutics, University of Florida, Gainesville, FL 32610, USA;
- Correspondence:
| | - Sara M. Turner
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA; (S.M.T.); (C.M.S.)
| | - Cassandra M. Schuster
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA; (S.M.T.); (C.M.S.)
| | - Aaron B. Morton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA; (A.B.M.); (J.M.H.)
| | - J. Matthew Hinkley
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA; (A.B.M.); (J.M.H.)
| | - David D. Fuller
- Breathing Research and Therapeutics, University of Florida, Gainesville, FL 32610, USA;
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA; (S.M.T.); (C.M.S.)
- McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
23
|
Xin T, Lv W, Liu D, Jing Y, Hu F. Opa1 Reduces Hypoxia-Induced Cardiomyocyte Death by Improving Mitochondrial Quality Control. Front Cell Dev Biol 2020; 8:853. [PMID: 32984338 PMCID: PMC7483501 DOI: 10.3389/fcell.2020.00853] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction contributes to cardiovascular disorders, especially post-infarction cardiac injury, through incompletely characterized mechanisms. Among the latter, increasing evidence points to alterations in mitochondrial quality control, a range of adaptive responses regulating mitochondrial morphology and function. Optic atrophy 1 (Opa1) is a mitochondrial inner membrane GTPase known to promote mitochondrial fusion. In this study, hypoxia-mediated cardiomyocyte damage was induced to mimic post-infarction cardiac injury in vitro. Loss- and gain-of-function assays were then performed to evaluate the impact of Opa1 expression on mitochondrial quality control and cardiomyocyte survival and function. Hypoxic stress reduced cardiomyocyte viability, impaired contractile/relaxation functions, and augmented the synthesis of pro-inflammatory mediators. These effects were exacerbated by Opa1 knockdown, and significantly attenuated by Opa1 overexpression. Mitochondrial quality control was disturbed by hypoxia, as reflected by multiple mitochondrial deficits; i.e., increased fission, defective fusion, impaired mitophagy, decreased biogenesis, increased oxidative stress, and blunted respiration. By contrast, overexpression of Opa1 normalized mitochondrial quality control and sustained cardiomyocyte function. We also found that ERK, AMPK, and YAP signaling can regulate Opa1 expression. These results identify Opa1 as a novel regulator of mitochondrial quality control and highlight a key role for Opa1 in protecting cardiomyocytes against post-infarction cardiac injury.
Collapse
Affiliation(s)
- Ting Xin
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Wei Lv
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Dongmei Liu
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Yongle Jing
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Fang Hu
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| |
Collapse
|
24
|
Xie Z, He B, Jiang Z, Zhao L. Tanshinone IIA inhibits osteosarcoma growth through modulation of AMPK-Nrf2 signaling pathway. J Recept Signal Transduct Res 2020; 40:591-598. [PMID: 32496906 DOI: 10.1080/10799893.2020.1772296] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tanshinone IIA (Tan IIA) is a member of the major lipophilic components extracted from the root of Salvia miltiorrhiza Bunge. Osteosarcomas are primary malignant tumors of bone. The aim of our study is to explore the role of Tan IIA in osteosarcomas survival, migration, and proliferation. MG63 osteosarcoma cell line was cultured in vitro and treated with different concentrations of Tan IIA. Then, ELISA, immunofluorescence, qPCR, western blots, and pathway analysis were conducted to verify whether Tan II modulated osteosarcoma survival, migration, and proliferation through the AMPK/Nrf2 signaling pathway. Our results indicated that Tan IIA dose-dependently inhibited MG63 osteosarcoma cell survival, migration, and proliferation. Mechanistically, Tan IIA reduced cell viability and inhibited the transcriptions of migratory factors. In addition, the number of proliferative MG63 osteosarcoma cell was also reduced by Tan IIA. Molecular investigations demonstrated that Tan IIA treatment caused a drop in the transcriptions and activities of AMPK and Nrf2. Interestingly, knockdown of AMPK and Nrf2 markedly attenuated MG63 osteosarcoma cell survival, migration, and proliferation. Altogether, our results indicate that Tan IIA could be used as an effective anticancer drug to control osteosarcoma proliferation through affecting its survival, migration, and proliferation.
Collapse
Affiliation(s)
- Zengjun Xie
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Binbin He
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Ziyun Jiang
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Liang Zhao
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| |
Collapse
|
25
|
Woods JA, Hutchinson NT, Powers SK, Roberts WO, Gomez-Cabrera MC, Radak Z, Berkes I, Boros A, Boldogh I, Leeuwenburgh C, Coelho-Júnior HJ, Marzetti E, Cheng Y, Liu J, Durstine JL, Sun J, Ji LL. The COVID-19 pandemic and physical activity. SPORTS MEDICINE AND HEALTH SCIENCE 2020; 2:55-64. [PMID: 34189484 PMCID: PMC7261095 DOI: 10.1016/j.smhs.2020.05.006] [Citation(s) in RCA: 270] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 05/25/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
The SARS-CoV-2-caused COVID-19 pandemic has resulted in a devastating threat to human society in terms of health, economy, and lifestyle. Although the virus usually first invades and infects the lung and respiratory track tissue, in extreme cases, almost all major organs in the body are now known to be negatively impacted often leading to severe systemic failure in some people. Unfortunately, there is currently no effective treatment for this disease. Pre-existing pathological conditions or comorbidities such as age are a major reason for premature death and increased morbidity and mortality. The immobilization due to hospitalization and bed rest and the physical inactivity due to sustained quarantine and social distancing can downregulate the ability of organs systems to resist to viral infection and increase the risk of damage to the immune, respiratory, cardiovascular, musculoskeletal systems and the brain. The cellular mechanisms and danger of this "second wave" effect of COVID-19 to the human body, along with the effects of aging, proper nutrition, and regular physical activity, are reviewed in this article.
Collapse
Affiliation(s)
- Jeffrey A. Woods
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, USA
| | - Noah T. Hutchinson
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, USA
| | - Scott K. Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, USA
| | - William O. Roberts
- Department of Family Medicine and Community Health, University of Minnesota Medical School, Minneapolis, USA
| | - Mari Carmen Gomez-Cabrera
- Freshage Research Group, Department of Physiology Faculty of Medicine, University of Valencia and CIBERFES. Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Zsolt Radak
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Istvan Berkes
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Anita Boros
- National University of Public Service, Budapest, Hungary
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, USA
| | | | | | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Ying Cheng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - J. Larry Durstine
- Department of Exercise Science, Norman J. Arnold School of Public Health, University of South Carolina, Columbia, USA
| | - Junzhi Sun
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Li Li Ji
- The Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota Twin Cities, Minneapolis, USA
| |
Collapse
|
26
|
Is Mitochondrial Oxidative Stress the Key Contributor to Diaphragm Atrophy and Dysfunction in Critically Ill Patients? Crit Care Res Pract 2020; 2020:8672939. [PMID: 32377432 PMCID: PMC7191397 DOI: 10.1155/2020/8672939] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 02/08/2023] Open
Abstract
Diaphragm dysfunction is prevalent in the progress of respiratory dysfunction in various critical illnesses. Respiratory muscle weakness may result in insufficient ventilation, coughing reflection suppression, pulmonary infection, and difficulty in weaning off respirators. All of these further induce respiratory dysfunction and even threaten the patients' survival. The potential mechanisms of diaphragm atrophy and dysfunction include impairment of myofiber protein anabolism, enhancement of myofiber protein degradation, release of inflammatory mediators, imbalance of metabolic hormones, myonuclear apoptosis, autophagy, and oxidative stress. Among these contributors, mitochondrial oxidative stress is strongly implicated to play a key role in the process as it modulates diaphragm protein synthesis and degradation, induces protein oxidation and functional alteration, enhances apoptosis and autophagy, reduces mitochondrial energy supply, and is regulated by inflammatory cytokines via related signaling molecules. This review aims to provide a concise overview of pathological mechanisms of diaphragmatic dysfunction in critically ill patients, with special emphasis on the role and modulating mechanisms of mitochondrial oxidative stress.
Collapse
|
27
|
Garnham JO, Roberts LD, Espino-Gonzalez E, Whitehead A, Swoboda PP, Koshy A, Gierula J, Paton MF, Cubbon RM, Kearney MT, Egginton S, Bowen TS, Witte KK. Chronic heart failure with diabetes mellitus is characterized by a severe skeletal muscle pathology. J Cachexia Sarcopenia Muscle 2020; 11:394-404. [PMID: 31863644 PMCID: PMC7113493 DOI: 10.1002/jcsm.12515] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 10/07/2019] [Accepted: 10/17/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Patients with coexistent chronic heart failure (CHF) and diabetes mellitus (DM) demonstrate greater exercise limitation and worse prognosis compared with CHF patients without DM, even when corrected for cardiac dysfunction. Understanding the origins of symptoms in this subgroup may facilitate development of targeted treatments. We therefore characterized the skeletal muscle phenotype and its relationship to exercise limitation in patients with diabetic heart failure (D-HF). METHODS In one of the largest muscle sampling studies in a CHF population, pectoralis major biopsies were taken from age-matched controls (n = 25), DM (n = 10), CHF (n = 52), and D-HF (n = 28) patients. In situ mitochondrial function and reactive oxygen species, fibre morphology, capillarity, and gene expression analyses were performed and correlated to whole-body exercise capacity. RESULTS Mitochondrial respiration, content, coupling efficiency, and intrinsic function were lower in D-HF patients compared with other groups (P < 0.05). A unique mitochondrial complex I dysfunction was present in D-HF patients only (P < 0.05), which strongly correlated to exercise capacity (R2 = 0.64; P < 0.001). Mitochondrial impairments in D-HF corresponded to higher levels of mitochondrial reactive oxygen species (P < 0.05) and lower gene expression of anti-oxidative enzyme superoxide dismutase 2 (P < 0.05) and complex I subunit NDUFS1 (P < 0.05). D-HF was also associated with severe fibre atrophy (P < 0.05) and reduced local fibre capillarity (P < 0.05). CONCLUSIONS Patients with D-HF develop a specific skeletal muscle pathology, characterized by mitochondrial impairments, fibre atrophy, and derangements in the capillary network that are linked to exercise intolerance. These novel preliminary data support skeletal muscle as a potential therapeutic target for treating patients with D-HF.
Collapse
Affiliation(s)
- Jack O Garnham
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Ever Espino-Gonzalez
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Anna Whitehead
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Peter P Swoboda
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Aaron Koshy
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - John Gierula
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Maria F Paton
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Stuart Egginton
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Klaus K Witte
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
28
|
Lu K, Zhao J, Liu W. Macrophage stimulating 1-induced inflammation response promotes aortic aneurysm formation through triggering endothelial cells death and activating the NF-κB signaling pathway. J Recept Signal Transduct Res 2020; 40:374-382. [PMID: 32156191 DOI: 10.1080/10799893.2020.1738484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aortic aneurysm formation is associated with endothelial cells dysfunction through an undefined mechanism. Macrophage stimulating 1 (Mst1) and NF-κB signaling pathway have been found to be related to inflammation response in endothelial cell damage. The goal of our study is to explore the role of Mst1 in regulating endothelial cell viability with a focus on NF-κB signaling pathway and inflammation response. Endothelial cell viability and death were determined via immunofluorescence and ELISA. Agonist of NF-κB signaling pathway and siRNA against Mst1 were used. The results in our study demonstrated that Mst1 transcription and expression were significantly elevated after exposure to oxidative stress in endothelial cells. Once loss of Mst1 through transfection of siRNA (si-Mst1), endothelial cell viability and survival rate were rapidly increased in response to oxidative stress. In addition, we also found that Mst1 controlled inflammation response and mitochondrial function in endothelial cells. Re-activation of NF-κB signaling pathway was followed by an activation of inflammation response and mitochondrial dysfunction, as evidenced by increased expression of inflammation factors and decreased ATP synthesis. Altogether, our results identify Mst1 as the primary factors responsible for endothelial cells dysfunction in aneurysms formation through inducing inflammation response, endothelial apoptosis, and NF-κB signaling pathway activation.
Collapse
Affiliation(s)
- Kai Lu
- Daqing Oilfield General Hospital, Daqing, P. R. China
| | - Jianfei Zhao
- Daqing Oilfield General Hospital, Daqing, P. R. China
| | - Weili Liu
- Daqing Oilfield General Hospital, Daqing, P. R. China
| |
Collapse
|
29
|
Powers SK, Bomkamp M, Ozdemir M, Hyatt H. Mechanisms of exercise-induced preconditioning in skeletal muscles. Redox Biol 2020; 35:101462. [PMID: 32089451 PMCID: PMC7284917 DOI: 10.1016/j.redox.2020.101462] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 02/09/2020] [Indexed: 12/15/2022] Open
Abstract
Endurance exercise training promotes numerous biochemical adaptations within skeletal muscle fibers culminating into a phenotype that is safeguarded against numerous perils including doxorubicin-induced myopathy and inactivity-induced muscle atrophy. This exercise-induced protection of skeletal muscle fibers is commonly termed "exercise preconditioning". This review will discuss the biochemical mechanisms responsible for exercise-induced protection of skeletal muscle fibers against these harmful events. The first segment of this report highlights the evidence that endurance exercise training provides cytoprotection to skeletal muscle fibers against several potentially damaging insults. The second and third sections of the review will discuss the cellular adaptations responsible for exercise-induced protection of skeletal muscle fibers against doxorubicin-provoked damage and inactivity-induced fiber atrophy, respectively. Importantly, we also identify gaps in our understanding of exercise preconditioning in hopes of stimulating future research.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32611, USA
| | - Matthew Bomkamp
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32611, USA.
| | - Mustafa Ozdemir
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32611, USA
| | - Hayden Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
30
|
Zhou D, Zhang M, Min L, Jiang K, Jiang Y. Cerebral ischemia-reperfusion is modulated by macrophage-stimulating 1 through the MAPK-ERK signaling pathway. J Cell Physiol 2020; 235:7067-7080. [PMID: 32017081 DOI: 10.1002/jcp.29603] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/22/2020] [Indexed: 01/04/2023]
Abstract
Cerebral ischemia-reperfusion (IR) injury is associated with mitochondrial damage. Macrophage-stimulating 1 (MST1) reportedly stimulates mitochondrial apoptosis by suppressing BCL-2. We investigated whether MST1 promotes the progression of cerebral IR injury by inducing mitochondrial dysfunction in vivo and in vitro. Western blot analysis, quantitative polymerase chain reaction, immunofluorescence, and mitochondrial function assays were conducted in cells from wild-type and Mst1-knockout mice subjected to cerebral IR injury. MST1 expression in wild-type glial cells increased following cerebral IR injury. Cerebral IR injury reduced the mitochondrial membrane potential and mitochondrial metabolism in glial cells, while it enhanced mitochondrial reactive oxygen species generation and mitochondrial calcium levels in these cells. The deletion of Mst1 attenuated cerebral IR injury by improving mitochondrial function and reducing mitochondrial damage. The mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway was suppressed in wild-type glial cell upon cerebral IR injury but was reactivated in Mst1-knockout glial cell. Accordingly, blocking the MAPK/ERK pathway abolished the beneficial effects of Mst1 deletion during cerebral IR injury by inducing mitochondrial damage in glial cells. Our results suggest that cerebral IR injury is associated with MST1 upregulation in the brain, while the genetic ablation of Mst1 can attenuate mitochondrial damage and sustain brain function following cerebral IR injury.
Collapse
Affiliation(s)
- Dingzhou Zhou
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mingming Zhang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liu Min
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kaiyuan Jiang
- Department of Neurosurgery, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Yugang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
31
|
Morton AB, Norton CE, Jacobsen NL, Fernando CA, Cornelison DDW, Segal SS. Barium chloride injures myofibers through calcium-induced proteolysis with fragmentation of motor nerves and microvessels. Skelet Muscle 2019; 9:27. [PMID: 31694693 PMCID: PMC6833148 DOI: 10.1186/s13395-019-0213-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/30/2019] [Indexed: 12/15/2022] Open
Abstract
Background Local injection of BaCl2 is an established model of acute injury to study the regeneration of skeletal muscle. However, the mechanism by which BaCl2 causes muscle injury is unresolved. Because Ba2+ inhibits K+ channels, we hypothesized that BaCl2 induces myofiber depolarization leading to Ca2+ overload, proteolysis, and membrane disruption. While BaCl2 spares resident satellite cells, its effect on other tissue components integral to contractile function has not been defined. We therefore asked whether motor nerves and microvessels, which control and supply myofibers, are injured by BaCl2 treatment. Methods The intact extensor digitorum longus (EDL) muscle was isolated from male mice (aged 3–4 months) and irrigated with physiological salt solution (PSS) at 37 °C. Myofiber membrane potential (Vm) was recorded using sharp microelectrodes while intracellular calcium concentration ([Ca2+]i) was evaluated with Fura 2 dye. Isometric force production of EDL was measured in situ, proteolytic activity was quantified by calpain degradation of αII-spectrin, and membrane disruption was marked by nuclear staining with propidium iodide (PI). To test for effects on motor nerves and microvessels, tibialis anterior or gluteus maximus muscles were injected with 1.2% BaCl2 (50–75 μL) in vivo followed by immunostaining to evaluate the integrity of respective tissue elements post injury. Data were analyzed using Students t test and analysis of variance with P ≤ 0.05 considered statistically significant. Results Addition of 1.2% BaCl2 to PSS depolarized myofibers from − 79 ± 3 mV to − 17 ± 7 mV with a corresponding rise in [Ca2+]i; isometric force transiently increased from 7.4 ± 0.1 g to 11.1 ± 0.4 g. Following 1 h of BaCl2 exposure, 92 ± 3% of myonuclei stained with PI (vs. 8 ± 3% in controls) with enhanced cleavage of αII-spectrin. Eliminating Ca2+ from PSS prevented the rise in [Ca2+]i and ameliorated myonuclear staining with PI during BaCl2 exposure. Motor axons and capillary networks appeared fragmented within 24 h following injection of 1.2% BaCl2 and morphological integrity deteriorated through 72 h. Conclusions BaCl2 injures myofibers through depolarization of the sarcolemma, causing Ca2+ overload with transient contraction, leading to proteolysis and membrane rupture. Motor innervation and capillarity appear disrupted concomitant with myofiber damage, further compromising muscle integrity.
Collapse
Affiliation(s)
- Aaron B Morton
- Department of Medical Pharmacology and Physiology, University of Missouri, MA415 Medical Sciences Building, 1 Hospital Drive, Columbia, MO, 65212, USA
| | - Charles E Norton
- Department of Medical Pharmacology and Physiology, University of Missouri, MA415 Medical Sciences Building, 1 Hospital Drive, Columbia, MO, 65212, USA
| | - Nicole L Jacobsen
- Department of Medical Pharmacology and Physiology, University of Missouri, MA415 Medical Sciences Building, 1 Hospital Drive, Columbia, MO, 65212, USA
| | - Charmain A Fernando
- Department of Medical Pharmacology and Physiology, University of Missouri, MA415 Medical Sciences Building, 1 Hospital Drive, Columbia, MO, 65212, USA
| | - D D W Cornelison
- Division of Biological Sciences and Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, 65201, USA
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, MA415 Medical Sciences Building, 1 Hospital Drive, Columbia, MO, 65212, USA. .,Dalton Cardiovascular Research Center, Columbia, MO, 65211, USA.
| |
Collapse
|
32
|
Guigni BA, Fix DK, Bivona JJ, Palmer BM, Carson JA, Toth MJ. Electrical stimulation prevents doxorubicin-induced atrophy and mitochondrial loss in cultured myotubes. Am J Physiol Cell Physiol 2019; 317:C1213-C1228. [PMID: 31532714 DOI: 10.1152/ajpcell.00148.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Muscle contraction may protect against the effects of chemotherapy to cause skeletal muscle atrophy, but the mechanisms underlying these benefits are unclear. To address this question, we utilized in vitro modeling of contraction and mechanotransduction in C2C12 myotubes treated with doxorubicin (DOX; 0.2 μM for 3 days). Myotubes expressed contractile proteins and organized these into functional myofilaments, as electrical field stimulation (STIM) induced intracellular calcium (Ca2+) transients and contractions, both of which were prevented by inhibition of membrane depolarization. DOX treatment reduced myotube myosin content, protein synthesis, and Akt (S308) and forkhead box O3a (FoxO3a; S253) phosphorylation and increased muscle RING finger 1 (MuRF1) expression. STIM (1 h/day) prevented DOX-induced reductions in myotube myosin content and Akt and FoxO3a phosphorylation, as well as increases in MuRF1 expression, but did not prevent DOX-induced reductions in protein synthesis. Inhibition of myosin-actin interaction during STIM prevented contraction and the antiatrophic effects of STIM without affecting Ca2+ cycling, suggesting that the beneficial effect of STIM derives from mechanotransductive pathways. Further supporting this conclusion, mechanical stretch of myotubes recapitulated the effects of STIM to prevent DOX suppression of FoxO3a phosphorylation and upregulation of MuRF1. DOX also increased reactive oxygen species (ROS) production, which led to a decrease in mitochondrial content. Although STIM did not alter DOX-induced ROS production, peroxisome proliferator-activated receptor-γ coactivator-1α and antioxidant enzyme expression were upregulated, and mitochondrial loss was prevented. Our results suggest that the activation of mechanotransductive pathways that downregulate proteolysis and preserve mitochondrial content protects against the atrophic effects of chemotherapeutics.
Collapse
Affiliation(s)
- Blas A Guigni
- Department of Medicine, College of Medicine, University of Vermont, Burlington, Vermont.,Department of Molecular Physiology and Biophysics, College of Medicine, University of Vermont, Burlington, Vermont
| | - Dennis K Fix
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Joseph J Bivona
- Department of Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Bradley M Palmer
- Department of Molecular Physiology and Biophysics, College of Medicine, University of Vermont, Burlington, Vermont
| | - James A Carson
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina.,Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Michael J Toth
- Department of Medicine, College of Medicine, University of Vermont, Burlington, Vermont.,Department of Molecular Physiology and Biophysics, College of Medicine, University of Vermont, Burlington, Vermont.,Department of Orthopedics and Rehabilitation, College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
33
|
Smuder AJ, Morton AB, Hall SE, Wiggs MP, Ahn B, Wawrzyniak NR, Sollanek KJ, Min K, Kwon OS, Nelson WB, Powers SK. Effects of exercise preconditioning and HSP72 on diaphragm muscle function during mechanical ventilation. J Cachexia Sarcopenia Muscle 2019; 10:767-781. [PMID: 30972953 PMCID: PMC6711411 DOI: 10.1002/jcsm.12427] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 02/19/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Mechanical ventilation (MV) is a life-saving measure for patients in respiratory failure. However, prolonged MV results in significant diaphragm atrophy and contractile dysfunction, a condition referred to as ventilator-induced diaphragm dysfunction (VIDD). While there are currently no clinically approved countermeasures to prevent VIDD, increased expression of heat shock protein 72 (HSP72) has been demonstrated to attenuate inactivity-induced muscle wasting. HSP72 elicits cytoprotection via inhibition of NF-κB and FoxO transcriptional activity, which contribute to VIDD. In addition, exercise-induced prevention of VIDD is characterized by an increase in the concentration of HSP72 in the diaphragm. Therefore, we tested the hypothesis that increased HSP72 expression is required for the exercise-induced prevention of VIDD. We also determined whether increasing the abundance of HSP72 in the diaphragm, independent of exercise, is sufficient to prevent VIDD. METHODS Cause and effect was determined by inhibiting the endurance exercise-induced increase in HSP72 in the diaphragm of exercise trained animals exposed to prolonged MV via administration of an antisense oligonucleotide targeting HSP72. Additional experiments were performed to determine if increasing HSP72 in the diaphragm via genetic (rAAV-HSP72) or pharmacological (BGP-15) overexpression is sufficient to prevent VIDD. RESULTS Our results demonstrate that the exercise-induced increase in HSP72 protein abundance is required for the protective effects of exercise against VIDD. Moreover, both rAAV-HSP72 and BGP-15-induced overexpression of HSP72 were sufficient to prevent VIDD. In addition, modification of HSP72 in the diaphragm is inversely related to the expression of NF-κB and FoxO target genes. CONCLUSIONS HSP72 overexpression in the diaphragm is an effective intervention to prevent MV-induced oxidative stress and the transcriptional activity of NF-κB and FoxO. Therefore, overexpression of HSP72 in the diaphragm is a potential therapeutic target to protect against VIDD.
Collapse
Affiliation(s)
- Ashley J Smuder
- Department of Exercise Science, University of South Carolina, Columbia, USA
| | - Aaron B Morton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, USA
| | - Stephanie E Hall
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, USA
| | - Michael P Wiggs
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, USA
| | - Bumsoo Ahn
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, USA
| | - Nicholas R Wawrzyniak
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, USA
| | - Kurt J Sollanek
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, USA
| | - Kisuk Min
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, USA
| | - Oh Sung Kwon
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, USA
| | - W Bradley Nelson
- Department of Natural Sciences, Ohio Dominican University, Columbus, USA
| | - Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, USA
| |
Collapse
|