1
|
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
|
2
|
Wu H, Chasteen B. Rapid review of ventilator-induced diaphragm dysfunction. Respir Med 2024; 223:107541. [PMID: 38290603 DOI: 10.1016/j.rmed.2024.107541] [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: 10/01/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
Ventilator-induced diaphragm dysfunction is gaining increased recognition. Evidence of diaphragm weakness can manifest within 12 h to a few days after the initiation of mechanical ventilation. Various noninvasive and invasive methods have been developed to assess diaphragm function. The implementation of diaphragm-protective ventilation strategies is crucial for preventing diaphragm injuries. Furthermore, diaphragm neurostimulation emerges as a promising and novel treatment option. In this rapid review, our objective is to discuss the current understanding of ventilator-induced diaphragm dysfunction, diagnostic approaches, and updates on strategies for prevention and management.
Collapse
Affiliation(s)
- Huimin Wu
- Pulmonary, Critical Care and Sleep Medicine Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, United States; Department of Adult Respiratory Care, University of Oklahoma Medical Center, Oklahoma City, OK, 73104, United States.
| | - Bobby Chasteen
- Department of Adult Respiratory Care, University of Oklahoma Medical Center, Oklahoma City, OK, 73104, United States.
| |
Collapse
|
3
|
Liu K, Tronstad O, Flaws D, Churchill L, Jones AYM, Nakamura K, Fraser JF. From bedside to recovery: exercise therapy for prevention of post-intensive care syndrome. J Intensive Care 2024; 12:11. [PMID: 38424645 PMCID: PMC10902959 DOI: 10.1186/s40560-024-00724-4] [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: 10/18/2023] [Accepted: 02/17/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND As advancements in critical care medicine continue to improve Intensive Care Unit (ICU) survival rates, clinical and research attention is urgently shifting toward improving the quality of survival. Post-Intensive Care Syndrome (PICS) is a complex constellation of physical, cognitive, and mental dysfunctions that severely impact patients' lives after hospital discharge. This review provides a comprehensive and multi-dimensional summary of the current evidence and practice of exercise therapy (ET) during and after an ICU admission to prevent and manage the various domains of PICS. The review aims to elucidate the evidence of the mechanisms and effects of ET in ICU rehabilitation and highlight that suboptimal clinical and functional outcomes of ICU patients is a growing public health concern that needs to be urgently addressed. MAIN BODY This review commences with a brief overview of the current relationship between PICS and ET, describing the latest research on this topic. It subsequently summarises the use of ET in ICU, hospital wards, and post-hospital discharge, illuminating the problematic transition between these settings. The following chapters focus on the effects of ET on physical, cognitive, and mental function, detailing the multi-faceted biological and pathophysiological mechanisms of dysfunctions and the benefits of ET in all three domains. This is followed by a chapter focusing on co-interventions and how to maximise and enhance the effect of ET, outlining practical strategies for how to optimise the effectiveness of ET. The review next describes several emerging technologies that have been introduced/suggested to augment and support the provision of ET during and after ICU admission. Lastly, the review discusses future research directions. CONCLUSION PICS is a growing global healthcare concern. This review aims to guide clinicians, researchers, policymakers, and healthcare providers in utilising ET as a therapeutic and preventive measure for patients during and after an ICU admission to address this problem. An improved understanding of the effectiveness of ET and the clinical and research gaps that needs to be urgently addressed will greatly assist clinicians in their efforts to rehabilitate ICU survivors, improving patients' quality of survival and helping them return to their normal lives after hospital discharge.
Collapse
Affiliation(s)
- Keibun Liu
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Chermside, QLD, 4032, Australia.
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.
- Non-Profit Organization ICU Collaboration Network (ICON), Tokyo, Japan.
| | - Oystein Tronstad
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Chermside, QLD, 4032, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- Physiotherapy Department, The Prince Charles Hospital, Brisbane, Australia
| | - Dylan Flaws
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Chermside, QLD, 4032, Australia
- Metro North Mental Health, Caboolture Hospital, Caboolture, Australia
- School of Clinical Science, Queensland University of Technology, Brisbane, Australia
| | - Luke Churchill
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Chermside, QLD, 4032, Australia
- Physiotherapy Department, The Prince Charles Hospital, Brisbane, Australia
- School of Health & Rehabilitation Sciences, The University of Queensland, Brisbane, Australia
| | - Alice Y M Jones
- School of Health & Rehabilitation Sciences, The University of Queensland, Brisbane, Australia
| | - Kensuke Nakamura
- Department of Critical Care Medicine, Yokohama City University Hospital, Kanagawa, Japan
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Chermside, QLD, 4032, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- Queensland University of Technology, Brisbane, Australia
- St. Andrews War Memorial Hospital, Brisbane, Australia
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Zhang D, Hao W, Niu Q, Xu D, Duan X. Identification of the co-differentially expressed hub genes involved in the endogenous protective mechanism against ventilator-induced diaphragm dysfunction. Skelet Muscle 2022; 12:21. [PMID: 36085166 PMCID: PMC9461262 DOI: 10.1186/s13395-022-00304-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In intensive care units (ICU), mechanical ventilation (MV) is commonly applied to save patients' lives. However, ventilator-induced diaphragm dysfunction (VIDD) can complicate treatment by hindering weaning in critically ill patients and worsening outcomes. The goal of this study was to identify potential genes involved in the endogenous protective mechanism against VIDD. METHODS Twelve adult male rabbits were assigned to either an MV group or a control group under the same anesthetic conditions. Immunostaining and quantitative morphometry were used to assess diaphragm atrophy, while RNA-seq was used to investigate molecular differences between the groups. Additionally, core module and hub genes were analyzed using WGCNA, and co-differentially expressed hub genes were subsequently discovered by overlapping the differentially expressed genes (DEGs) with the hub genes from WGCNA. The identified genes were validated by western blotting (WB) and quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS After a VIDD model was successfully built, 1276 DEGs were found between the MV and control groups. The turquoise and yellow modules were identified as the core modules, and Trim63, Fbxo32, Uchl1, Tmprss13, and Cst3 were identified as the five co-differentially expressed hub genes. After the two atrophy-related genes (Trim63 and Fbxo32) were excluded, the levels of the remaining three genes/proteins (Uchl1/UCHL1, Tmprss13/TMPRSS13, and Cst3/CST3) were found to be significantly elevated in the MV group (P < 0.05), suggesting the existence of a potential antiproteasomal, antiapoptotic, and antiautophagic mechanism against diaphragm dysfunction. CONCLUSION The current research helps to reveal a potentially important endogenous protective mechanism that could serve as a novel therapeutic target against VIDD.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Critical Care Medicine, Heping Hospital Affiliated to Changzhi Medical College, 110 South Yan'an Road, Luzhou District, Changzhi, 046012, China.
| | - Wenyan Hao
- Department of Biomedical Engineering, Changzhi Medical College, Changzhi, 046012, China
| | - Qi Niu
- Department of Critical Care Medicine, Heping Hospital Affiliated to Changzhi Medical College, 110 South Yan'an Road, Luzhou District, Changzhi, 046012, China
| | - Dongdong Xu
- Department of Critical Care Medicine, Heping Hospital Affiliated to Changzhi Medical College, 110 South Yan'an Road, Luzhou District, Changzhi, 046012, China
| | - Xuejiao Duan
- Department of Critical Care Medicine, Heping Hospital Affiliated to Changzhi Medical College, 110 South Yan'an Road, Luzhou District, Changzhi, 046012, China
| |
Collapse
|
7
|
Powers SK, Schrager M. Redox signaling regulates skeletal muscle remodeling in response to exercise and prolonged inactivity. Redox Biol 2022; 54:102374. [PMID: 35738088 PMCID: PMC9233275 DOI: 10.1016/j.redox.2022.102374] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/23/2022] Open
Abstract
Skeletal muscle fibers are malleable and undergo rapid remodeling in response to increased contractile activity (i.e., exercise) or prolonged periods of muscle inactivity (e.g., prolonged bedrest). Exploration of the cell signaling pathways regulating these skeletal muscle adaptations reveal that redox signaling pathways play a key role in the control of muscle remodeling during both exercise and prolonged muscle inactivity. In this regard, muscular exercise results in an acute increase in the production of reactive oxygen species (ROS) in the contracting fibers; however, this contraction-induced rise in ROS production rapidly declines when contractions cease. In contrast, prolonged muscle disuse results in a chronic elevation in ROS production within the inactive fibers. This difference in the temporal pattern of ROS production in muscle during exercise and muscle inactivity stimulates divergent cell-signaling pathways that activate both genomic and nongenomic mechanisms to promote muscle remodeling. This review examines the role that redox signaling plays in skeletal muscle adaptation in response to both prolonged muscle inactivity and endurance exercise training. We begin with a summary of the sites of ROS production in muscle fibers followed by a review of the cellular antioxidants that are responsible for regulation of ROS levels in the cell. We then discuss the specific redox-sensitive signaling pathways that promote skeletal muscle adaptation in response to both prolonged muscle inactivity and exercise. To stimulate future research, we close with a discussion of unanswered questions in this exciting field.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Health Sciences, Stetson University, Deland, FL, 32723, USA.
| | - Matthew Schrager
- Department of Health Sciences, Stetson University, Deland, FL, 32723, USA
| |
Collapse
|
8
|
Cao Y, Li P, Wang Y, Liu X, Wu W. Diaphragm Dysfunction and Rehabilitation Strategy in Patients With Chronic Obstructive Pulmonary Disease. Front Physiol 2022; 13:872277. [PMID: 35586711 PMCID: PMC9108326 DOI: 10.3389/fphys.2022.872277] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/18/2022] [Indexed: 12/03/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) affects the whole body and causes many extrapulmonary adverse effects, amongst which diaphragm dysfunction is one of the prominent manifestations. Diaphragm dysfunction in patients with COPD is manifested as structural changes, such as diaphragm atrophy, single-fibre dysfunction, sarcomere injury and fibre type transformation, and functional changes such as muscle strength decline, endurance change, diaphragm fatigue, decreased diaphragm mobility, etc. Diaphragm dysfunction directly affects the respiratory efficiency of patients and is one of the important pathological mechanisms leading to progressive exacerbation of COPD and respiratory failure, which is closely related to disease mortality. At present, the possible mechanisms of diaphragm dysfunction in patients with COPD include systemic inflammation, oxidative stress, hyperinflation, chronic hypoxia and malnutrition. However, the specific mechanism of diaphragm dysfunction in COPD is still unclear, which, to some extent, increases the difficulty of treatment and rehabilitation. Therefore, on the basis of the review of changes in the structure and function of COPD diaphragm, the potential mechanism of diaphragm dysfunction in COPD was discussed, the current effective rehabilitation methods were also summarised in this paper. In order to provide direction reference and new ideas for the mechanism research and rehabilitation treatment of diaphragm dysfunction in COPD.
Collapse
Affiliation(s)
- Yuanyuan Cao
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Peijun Li
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Yingqi Wang
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Xiaodan Liu
- School of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weibing Wu
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
9
|
The Role of Oxidative Stress in Skeletal Muscle Myogenesis and Muscle Disease. Antioxidants (Basel) 2022; 11:antiox11040755. [PMID: 35453440 PMCID: PMC9026549 DOI: 10.3390/antiox11040755] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 02/06/2023] Open
Abstract
The contractile activity, high oxygen consumption and metabolic rate of skeletal muscle cause it to continuously produce moderate levels of oxidant species, such as reactive oxygen species (ROS) and reactive nitrogen species (RNS). Under normal physiological conditions, there is a dynamic balance between the production and elimination of ROS/RNS. However, when the oxidation products exceed the antioxidant defense capacity, the body enters a state of oxidative stress. Myogenesis is an important process to maintain muscle homeostasis and the physiological function of skeletal muscle. Accumulating evidence suggests that oxidative stress plays a key role in myogenesis and skeletal muscle physiology and pathology. In this review, we summarize the sources of reactive oxygen species in skeletal muscle and the causes of oxidative stress and analyze the key role of oxidative stress in myogenesis. Then, we discuss the relationship between oxidative stress and muscle homeostasis and physiopathology. This work systematically summarizes the role of oxidative stress in myogenesis and muscle diseases and provides targets for subsequent antioxidant therapy and repair of inflammatory damage in noninflammatory muscle diseases.
Collapse
|
10
|
Rosales-Antequera C, Viscor G, Araneda OF. Inflammation and Oxidative Stress as Common Mechanisms of Pulmonary, Autonomic and Musculoskeletal Dysfunction after Spinal Cord Injury. BIOLOGY 2022; 11:biology11040550. [PMID: 35453749 PMCID: PMC9032591 DOI: 10.3390/biology11040550] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/19/2022] [Accepted: 03/28/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary When a spinal cord injury occurs, the neurons that regulate our voluntary movements, those involved in environment and somatic perception and those that regulate vegetative functions are affected. Once neuronal damage is established, the cells of other tissues are also affected in their functions, altering the interaction between organs and altering the proper functioning of the organism. Multiple studies in animal models, as well as in humans, have recognized as factors involved in organ damage the imbalance between the formation of highly reactive molecules called pro-oxidants and defensive mechanisms called antioxidants. Closely associated with this phenomenon, the inflammatory response is also pathologically activated. In this narrative review, we have analyzed the information involving these pathological processes at the level of the lung, the autonomic nervous system and the skeletal musculature after spinal cord injury. Knowing the abnormal functioning mechanisms that occur after a spinal cord injury not only offers a better understanding of the organic events but also offers future possibilities for therapeutic interventions that may benefit the thousands of patients suffering this pathology. Abstract One of the etiopathogenic factors frequently associated with generalized organ damage after spinal cord injury corresponds to the imbalance of the redox state and inflammation, particularly of the respiratory, autonomic and musculoskeletal systems. Our goal in this review was to gain a better understanding of this phenomenon by reviewing both animal and human studies. At the respiratory level, the presence of tissue damage is notable in situations that require increased ventilation due to lower thoracic distensibility and alveolar inflammation caused by higher levels of leptin as a result of increased fatty tissue. Increased airway reactivity, due to loss of sympathetic innervation, and levels of nitric oxide in exhaled air that are similar to those seen in asthmatic patients have also been reported. In addition, the loss of autonomic control efficiency leads to an uncontrolled release of catecholamines and glucocorticoids that induce immunosuppression, as well as a predisposition to autoimmune reactions. Simultaneously, blood pressure regulation is altered with vascular damage and atherogenesis associated with oxidative damage. At the muscular level, chronically elevated levels of prooxidants and lipoperoxidation associated with myofibrillar atrophy are described, with no reduction or reversibility of this process through antioxidant supplementation.
Collapse
Affiliation(s)
- Cristián Rosales-Antequera
- Physical Medicine and Rehabilitation Unit, Clínica Universidad de los Andes, Santiago 8320000, Chile;
- Integrative Laboratory of Biomechanics and Physiology of Effort, LIBFE, School of Kinesiology, Faculty of Medicine, Universidad de los Andes, Santiago 8320000, Chile
| | - Ginés Viscor
- Physiology Section, Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain;
| | - Oscar F. Araneda
- Integrative Laboratory of Biomechanics and Physiology of Effort, LIBFE, School of Kinesiology, Faculty of Medicine, Universidad de los Andes, Santiago 8320000, Chile
- Correspondence:
| |
Collapse
|
11
|
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
|
12
|
Arancibia-Hernández YL, Aranda-Rivera AK, Cruz-Gregorio A, Pedraza-Chaverri J. Antioxidant/anti-inflammatory effect of Mg 2+ in coronavirus disease 2019 (COVID-19). Rev Med Virol 2022; 32:e2348. [PMID: 35357063 PMCID: PMC9111052 DOI: 10.1002/rmv.2348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/09/2022] [Accepted: 03/17/2022] [Indexed: 12/26/2022]
Abstract
Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), characterised by high levels of inflammation and oxidative stress (OS). Oxidative stress induces oxidative damage to lipids, proteins, and DNA, causing tissue damage. Both inflammation and OS contribute to multi-organ failure in severe cases. Magnesium (Mg2+ ) regulates many processes, including antioxidant and anti-inflammatory responses, as well as the proper functioning of other micronutrients such as vitamin D. In addition, Mg2+ participates as a second signalling messenger in the activation of T cells. Therefore, Mg2+ deficiency can cause immunodeficiency, exaggerated acute inflammatory response, decreased antioxidant response, and OS. Supplementation with Mg2+ has an anti-inflammatory response by reducing the levels of nuclear factor kappa B (NF-κB), interleukin (IL) -6, and tumor necrosis factor alpha. Furthermore, Mg2+ supplementation improves mitochondrial function and increases the antioxidant glutathione (GSH) content, reducing OS. Therefore, Mg2+ supplementation is a potential way to reduce inflammation and OS, strengthening the immune system to manage COVID-19. This narrative review will address Mg2+ deficiency associated with a worse disease prognosis, Mg2+ supplementation as a potent antioxidant and anti-inflammatory therapy during and after COVID-19 disease, and suggest that randomised controlled trials are indicated.
Collapse
Affiliation(s)
| | - Ana Karina Aranda-Rivera
- Facultad de Química, Departamento de Biología, Laboratorio F-315, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alfredo Cruz-Gregorio
- Facultad de Química, Departamento de Biología, Laboratorio F-315, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - José Pedraza-Chaverri
- Facultad de Química, Departamento de Biología, Laboratorio F-315, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
13
|
Hyatt HW, Ozdemir M, Bomkamp MP, Powers SK. Activation of Calpain Contributes to Mechanical Ventilation-Induced Depression of Protein Synthesis in Diaphragm Muscle. Cells 2022; 11:cells11061028. [PMID: 35326479 PMCID: PMC8947683 DOI: 10.3390/cells11061028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 11/16/2022] Open
Abstract
Mechanical ventilation (MV) is a clinical tool that provides respiratory support to patients unable to maintain adequate alveolar ventilation on their own. Although MV is often a life-saving intervention in critically ill patients, an undesired side-effect of prolonged MV is the rapid occurrence of diaphragmatic atrophy due to accelerated proteolysis and depressed protein synthesis. Investigations into the mechanism(s) responsible for MV-induced diaphragmatic atrophy reveal that activation of the calcium-activated protease, calpain, plays a key role in accelerating proteolysis in diaphragm muscle fibers. Moreover, active calpain has been reported to block signaling events that promote protein synthesis (i.e., inhibition of mammalian target of rapamycin (mTOR) activation). While this finding suggests that active calpain can depress muscle protein synthesis, this postulate has not been experimentally verified. Therefore, we tested the hypothesis that active calpain plays a key role in the MV-induced depression of both anabolic signaling events and protein synthesis in the diaphragm muscle. MV-induced activation of calpain in diaphragm muscle fibers was prevented by transgene overexpression of calpastatin, an endogenous inhibitor of calpain. Our findings indicate that overexpression of calpastatin averts MV-induced activation of calpain in diaphragm fibers and rescues the MV-induced depression of protein synthesis in the diaphragm muscle. Surprisingly, deterrence of calpain activation did not impede the MV-induced inhibition of key anabolic signaling events including mTOR activation. However, blockade of calpain activation prevented the calpain-induced cleavage of glutaminyl-tRNA synthetase in diaphragm fibers; this finding is potentially important because aminoacyl-tRNA synthetases play a central role in protein synthesis. Regardless of the mechanism(s) responsible for calpain’s depression of protein synthesis, these results provide the first evidence that active calpain plays an important role in promoting the MV-induced depression of protein synthesis within diaphragm fibers.
Collapse
Affiliation(s)
- Hayden W. Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA; (H.W.H.); (M.O.); (M.P.B.)
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21201, USA
| | - Mustafa Ozdemir
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA; (H.W.H.); (M.O.); (M.P.B.)
| | - Matthew P. Bomkamp
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA; (H.W.H.); (M.O.); (M.P.B.)
| | - Scott K. Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA; (H.W.H.); (M.O.); (M.P.B.)
- Department of Health Sciences, Stetson University, Deland, FL 32720, USA
- Correspondence: author:
| |
Collapse
|
14
|
Trinity JD, Drummond MJ, Fermoyle CC, McKenzie AI, Supiano MA, Richardson RS. Cardiovasomobility: an integrative understanding of how disuse impacts cardiovascular and skeletal muscle health. J Appl Physiol (1985) 2022; 132:835-861. [PMID: 35112929 PMCID: PMC8934676 DOI: 10.1152/japplphysiol.00607.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cardiovasomobility is a novel concept that encompasses the integration of cardiovascular and skeletal muscle function in health and disease with critical modification by physical activity, or lack thereof. Compelling evidence indicates that physical activity improves health while a sedentary, or inactive, lifestyle accelerates cardiovascular and skeletal muscle dysfunction and hastens disease progression. Identifying causative factors for vascular and skeletal muscle dysfunction, especially in humans, has proven difficult due to the limitations associated with cross-sectional investigations. Therefore, experimental models of physical inactivity and disuse, which mimic hospitalization, injury, and illness, provide important insight into the mechanisms and consequences of vascular and skeletal muscle dysfunction. This review provides an overview of the experimental models of disuse and inactivity and focuses on the integrated responses of the vasculature and skeletal muscle in response to disuse/inactivity. The time course and magnitude of dysfunction evoked by various models of disuse/inactivity are discussed in detail, and evidence in support of the critical roles of mitochondrial function and oxidative stress are presented. Lastly, strategies aimed at preserving vascular and skeletal muscle dysfunction during disuse/inactivity are reviewed. Within the context of cardiovasomobility, experimental manipulation of physical activity provides valuable insight into the mechanisms responsible for vascular and skeletal muscle dysfunction that limit mobility, degrade quality of life, and hasten the onset of disease.
Collapse
Affiliation(s)
- Joel D Trinity
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Micah J Drummond
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah.,Department of Physical Therapy, University of Utah, Salt Lake City, Utah
| | - Caitlin C Fermoyle
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Alec I McKenzie
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Mark A Supiano
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Russell S Richardson
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
15
|
Ultrasound assessment of the diaphragm during the first days of mechanical ventilation compared to spontaneous respiration: a comparative study. LA TUNISIE MEDICALE 2021; 99:1055-1065. [PMID: 35288909 PMCID: PMC9390126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
INTRODUCTION In critically ill patients, the diaphragm is subject to several aggressions mainly those induced by mechanical ventilation (MV). Currently, diaphragmatic ultrasound has become the most useful bedside for the clinician to evaluate diaphragm contractility. AIM To examine the effects of MV on the diaphragm contractility during the first days of ventilation. METHODS Two groups of subjects were studied: a study group (n=30) of adults receiving MV versus a control group (n=30) of volunteers on spontaneous ventilation (SV). Using an ultrasound device, we compared the diaphragmatic thickening fraction (DTF). Secondly, we analysed the relationship between DTF and weaning. RESULTS comparatively to SV group, patients of MV group have a higher end expiratory diameter (EED) (2.09 ± 0.6 vs. 1.76 ± 0.32 mm, p=0.01) and a lower DTF (39.9 ± 12.5% vs. 49.0 ± 20.5%, p=0.043). Fourteen among the 30 ventilated patients successfully weaned. No significant correlation was shown between DTF and weaning duration (Rho= - 0.464, p=0.09). A DTF value > 33% was near to be significantly associated with weaning success (OR=2; 95% CI= [1.07-3.7], p=0.05) with a sensitivity at 85.7%. CONCLUSIONS diaphragmatic contractility was altered from the first days of MV. A DTF value >32,7% was associated to the weaning success and that may be useful to predict successful weaning with sensitivity at 85.7%.
Collapse
|
16
|
Hasanloei MAV, Zeinaly A, Rahimlou M, Houshyar H, Moonesirad S, Hashemi R. Effect of coenzyme Q10 supplementation on oxidative stress and clinical outcomes in patients with low levels of coenzyme Q10 admitted to the intensive care unit. J Nutr Sci 2021; 10:e48. [PMID: 34290862 PMCID: PMC8278158 DOI: 10.1017/jns.2021.39] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/09/2021] [Accepted: 06/08/2021] [Indexed: 12/28/2022] Open
Abstract
Today, trauma is known to be the third leading cause of death in most countries. Studies have demonstrated below-normal plasma levels of antioxidants in trauma patients. The present study aimed to assess the efficacy of Coenzyme Q10 (CoQ10) on oxidative stress, clinical outcomes and anthropometrical parameters in traumatic mechanical ventilated patients admitted to the intensive care unit. Patients were randomised to receive sublingual CoQ10 (400 mg/d) or placebo for 7 d. Primary and secondary outcomes were measured at the baseline and end of the study. We enrolled forty patients for this trial: twenty in the CoQ10 group and twenty in the placebo group. There was not any significant difference in the baseline variables (P > 0⋅05). At the end of the study, CoQ10 administration caused a considerable reduction in the Malondialdehyde (MDA) and Interleukin 6 (IL-6) concentrations (P < 0⋅001), Glasgow Coma Score (GCS; P = 0⋅02), ICU and hospital length of stay and mechanical ventilation (MV) duration (P < 0⋅001). We found that CoQ10 administration could increase Fat-Free Mass (P < 0⋅001) (FFM; P = 0⋅04), Skeletal Muscle Mass (SMM; P = 0⋅04) and Body Cell Mass (BCM) percent (P = 0⋅03). There was not any significant difference in other factors between the two groups (P > 0⋅05). CoQ10 administration has beneficial effects on patients with traumatic injury and has no side effects. However, since the possibility of the type II error was high, the outcomes on the duration of MV, ICU stay and hospital stay, and GCS may very well be false positives.
Collapse
Affiliation(s)
- Mohammad Amin Valizade Hasanloei
- Clinical Research Development Unit, Imam Khomeini Hospital, Urmia University of Medical Sciences, Ershad Ave, 5756151818Urmia, West Azerbaijan Province, Iran
| | - Aidin Zeinaly
- Department of Anesthesiology, Urmia University of Medical Sciences, 11 km SERO Road, 5756151818Urmia, West Azerbaijan Province, Iran
| | - Mehran Rahimlou
- Department of Nutrition, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hadi Houshyar
- Department of Anesthesiology, Imam Khomeini Hospital, Faculty of Medicine, Urmia University of Medical Sciences, Imam Khomeini Avenue, 5756151818Urmia, West Azerbaijan Province, Iran
| | - Solma Moonesirad
- Urmia University of Medical Sciences, 11 km SERO Road, 5756151818Urmia, West Azerbaijan Province, Iran
| | - Reza Hashemi
- Clinical Research Development Unit, Imam Khomeini Hospital, Urmia University of Medical Sciences, Ershad Ave, 5756151818Urmia, West Azerbaijan Province, Iran
| |
Collapse
|
17
|
Zhang B, Li P, Li J, Liu X, Wu W. Effect of Oxidative Stress on Diaphragm Dysfunction and Exercise Intervention in Chronic Obstructive Pulmonary Disease. Front Physiol 2021; 12:684453. [PMID: 34163375 PMCID: PMC8215263 DOI: 10.3389/fphys.2021.684453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) can cause extrapulmonary injury such as diaphragm dysfunction. Oxidative stress is one of the main factors causing diaphragm dysfunction in COPD. Exercise plays a positive role in the prevention and treatment of diaphragm dysfunction in COPD, and the changes in diaphragm structure and function induced by exercise are closely related to the regulation of oxidative stress. Therefore, on the basis of the review of oxidative stress and the changes in diaphragm structure and function in COPD, this article analyzed the effects of exercise on oxidative stress and diaphragm dysfunction in COPD and explored the possible mechanism by which exercise improves oxidative stress. Studies have found that diaphragm dysfunction in COPD includes the decline of muscle strength, endurance, and activity. Oxidative stress mainly affects the structure and function of the diaphragm in COPD through protein oxidation, protease activation and calcium sensitivity reduction. The effects of exercise on oxidative stress level and diaphragm dysfunction may differ depending on the intensity, duration, and style of exercise. The mechanism of exercise on oxidative stress in the diaphragm of COPD may include improving antioxidant capacity, reducing oxidase activity and improving mitochondrial function.
Collapse
Affiliation(s)
- Bingzhi Zhang
- Department of Sports Medicine, Shanghai University of Sport, Shanghai, China
| | - Peijun Li
- Department of Sports Medicine, Shanghai University of Sport, Shanghai, China
| | - Jian Li
- Department of Sports Medicine, Shanghai University of Sport, Shanghai, China
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weibing Wu
- Department of Sports Medicine, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
18
|
Ichinoseki-Sekine N, Smuder AJ, Morton AB, Hinkley JM, Mor Huertas A, Powers SK. Hydrogen sulfide donor protects against mechanical ventilation-induced atrophy and contractile dysfunction in the rat diaphragm. Clin Transl Sci 2021; 14:2139-2145. [PMID: 34080307 PMCID: PMC8604213 DOI: 10.1111/cts.13081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 11/28/2022] Open
Abstract
Mechanical ventilation (MV) is a clinical tool providing adequate alveolar ventilation in patients that require respiratory support. Although a life-saving intervention for critically ill patients, prolonged MV results in the rapid development of inspiratory muscle weakness due to both diaphragmatic atrophy and contractile dysfunction; collectively known as "ventilator-induced diaphragm dysfunction" (VIDD). VIDD is a severe clinical problem because diaphragmatic weakness is a risk factor for difficulties in weaning patients from MV. Currently, no standard treatment to prevent VIDD exists. Nonetheless, growing evidence reveals that hydrogen sulfide (H2 S) possesses cytoprotective properties capable of protecting skeletal muscles against several hallmarks of VIDD, including oxidative damage, accelerated proteolysis, and mitochondrial damage. Therefore, we used an established animal model of MV to test the hypothesis that treatment with sodium sulfide (H2 S donor) will defend against VIDD. Our results confirm that sodium sulfide was sufficient to protect the diaphragm against both MV-induced fiber atrophy and contractile dysfunction. H2 S prevents MV-induced damage to diaphragmatic mitochondria as evidenced by protection against mitochondrial uncoupling. Moreover, treatment with sodium sulfide prevented the MV-induced activation of the proteases, calpain, and caspase-3 in the diaphragm. Taken together, these results support the hypothesis that treatment with a H2 S donor protects the diaphragm against VIDD. These outcomes provide the first evidence that H2 S has therapeutic potential to protect against MV-induced diaphragm weakness and to reduce difficulties in weaning patients from the ventilator. Study Highlights WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC? Mechanical ventilation (MV) results in diaphragm atrophy and contractile dysfunction, known as ventilator-induced diaphragm dysfunction (VIDD). VIDD is important because diaphragm weakness is a risk factor for problems in weaning patients from MV. Currently, no accepted treatment exists to protect against VIDD. Growing evidence reveals that hydrogen sulfide (H2 S) donors protect skeletal muscle against ischemia-reperfusion-induced injury. Nonetheless, it is unknown if treatment with a H2 S donor can protect against VIDD. WHAT QUESTION DID THIS STUDY ADDRESS? Can treatment with an H2 S donor protect against VIDD? WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE? This study provides the first evidence that treatment with a H2 S donor protects against VIDD. HOW MIGHT THIS CHANGE CLINICAL PHARMACOLOGY OR TRANSLATIONAL SCIENCE? These new findings provide the basis for further exploration of H2 S donors as a therapy to prevent VIDD and reduce the risk of problems in weaning patients from MV.
Collapse
Affiliation(s)
- Noriko Ichinoseki-Sekine
- Graduate School of Arts and Sciences, The Open University of Japan, Chiba, Japan.,School of Health and Sports Science, Juntendo University, Inzai, Japan
| | - Ashley J Smuder
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Aaron B Morton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - James M Hinkley
- AdventHealth Translational Research Institute, Orlando, Florida, USA
| | - Andres Mor Huertas
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
19
|
Yoshihara T, Deminice R, Hyatt HW, Ozdemir M, Nguyen BL, Powers SK. Angiotensin 1-7 protects against ventilator-induced diaphragm dysfunction. Clin Transl Sci 2021; 14:1512-1523. [PMID: 33742769 PMCID: PMC8301547 DOI: 10.1111/cts.13015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/29/2021] [Accepted: 02/20/2021] [Indexed: 12/24/2022] Open
Abstract
Mechanical ventilation (MV) is a life‐saving instrument used to provide ventilatory support for critically ill patients and patients undergoing surgery. Unfortunately, an unintended consequence of prolonged MV is the development of inspiratory weakness due to both diaphragmatic atrophy and contractile dysfunction; this syndrome is labeled ventilator‐induced diaphragm dysfunction (VIDD). VIDD is clinically important because diaphragmatic weakness is an important contributor to problems in weaning patients from MV. Investigations into the pathogenesis of VIDD reveal that oxidative stress is essential for the rapid development of VIDD as redox disturbances in diaphragm fibers promote accelerated proteolysis. Currently, no standard treatment exists to prevent VIDD and, therefore, developing a strategy to avert VIDD is vital. Guided by evidence indicating that activation of the classical axis of the renin‐angiotensin system (RAS) in diaphragm fibers promotes oxidative stress and VIDD, we hypothesized that activation of the nonclassical RAS signaling pathway via angiotensin 1‐7 (Ang1‐7) will protect against VIDD. Using an established animal model of prolonged MV, our results disclose that infusion of Ang1‐7 protects the diaphragm against MV‐induced contractile dysfunction and fiber atrophy in both fast and slow muscle fibers. Further, Ang1‐7 shielded diaphragm fibers against MV‐induced mitochondrial damage, oxidative stress, and protease activation. Collectively, these results reveal that treatment with Ang1‐7 protects against VIDD, in part, due to diminishing oxidative stress and protease activation. These important findings provide robust evidence that Ang1‐7 has the therapeutic potential to protect against VIDD by preventing MV‐induced contractile dysfunction and atrophy of both slow and fast muscle fibers.
Collapse
Affiliation(s)
- Toshinori Yoshihara
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA.,Graduate School of Health and Sports Science, Juntendo University, Inzai, Japan
| | - Rafael Deminice
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA.,Department of Physical Education, State University of Londrina, Londrina, Brazil
| | - Hayden W Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Mustafa Ozdemir
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Branden L Nguyen
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
20
|
Hyatt HW, Powers SK. Mitochondrial Dysfunction Is a Common Denominator Linking Skeletal Muscle Wasting Due to Disease, Aging, and Prolonged Inactivity. Antioxidants (Basel) 2021; 10:antiox10040588. [PMID: 33920468 PMCID: PMC8070615 DOI: 10.3390/antiox10040588] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 12/29/2022] Open
Abstract
Skeletal muscle is the most abundant tissue in the body and is required for numerous vital functions, including breathing and locomotion. Notably, deterioration of skeletal muscle mass is also highly correlated to mortality in patients suffering from chronic diseases (e.g., cancer). Numerous conditions can promote skeletal muscle wasting, including several chronic diseases, cancer chemotherapy, aging, and prolonged inactivity. Although the mechanisms responsible for this loss of muscle mass is multifactorial, mitochondrial dysfunction is predicted to be a major contributor to muscle wasting in various conditions. This systematic review will highlight the biochemical pathways that have been shown to link mitochondrial dysfunction to skeletal muscle wasting. Importantly, we will discuss the experimental evidence that connects mitochondrial dysfunction to muscle wasting in specific diseases (i.e., cancer and sepsis), aging, cancer chemotherapy, and prolonged muscle inactivity (e.g., limb immobilization). Finally, in hopes of stimulating future research, we conclude with a discussion of important future directions for research in the field of muscle wasting.
Collapse
|
21
|
Acute liver injury following acetaminophen administration does not activate atrophic pathways in the mouse diaphragm. Sci Rep 2021; 11:6302. [PMID: 33737702 PMCID: PMC7973759 DOI: 10.1038/s41598-021-85859-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/04/2021] [Indexed: 11/09/2022] Open
Abstract
N-acetyl-para-amino phenol (APAP, usually named paracetamol), which is commonly used for its analgesic and antipyretic properties may lead to hepatotoxicity and acute liver damage in case of overdoses. Released cytokines and oxidative stress following acute liver damage may affect other organs' function notably the diaphragm, which is particularly sensitive to oxidative stress and circulating cytokines. We addressed this issue in a mouse model of acute liver injury induced by administration of APAP. C57BL/6J mice (each n = 8) were treated with N-acetyl-para-amino phenol (APAP) to induce acute drug caused liver injury and sacrificed 12 or 24 h afterwards. An untreated group served as controls. Key markers of inflammation, proteolysis, autophagy and oxidative stress were measured in diaphragm samples. In APAP treated animals, liver damage was proven by the enhanced serum levels of alanine aminotransferase and aspartate aminotransferase. In the diaphragm, besides a significant increase in IL 6 and lipid peroxidation, no changes were observed in key markers of the proteolytic, and autophagy signaling pathways, other inflammatory markers and fiber dimensions. The first 24 h of acute liver damage did not impair diaphragm atrophic pathways although it slightly enhanced IL-6 and lipid peroxidation. Whether longer exposure might affect the diaphragm needs to be addressed in future experiments.
Collapse
|
22
|
Sharma S, Chaudhary P, Sandhir R, Bharadwaj A, Gupta RK, Khatri R, Bajaj AC, Baburaj TP, Kumar S, Pal MS, Reddy PK, Kumar B. Heat-induced endoplasmic reticulum stress in soleus and gastrocnemius muscles and differential response to UPR pathway in rats. Cell Stress Chaperones 2021; 26:323-339. [PMID: 33210173 PMCID: PMC7925797 DOI: 10.1007/s12192-020-01178-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to investigate the differential response of oxidative (soleus) and glycolytic (gastrocnemius) muscles to heat-induced endoplasmic reticulum (ER) stress. It was hypothesized that due to compositional and functional differences, both muscles respond differently to acute heat stress. To address this, male Sprague Dawley rats (12/group) were subjected to thermoneutral (25 °C) or heat stress (42 °C) conditions for 1 h. Soleus and gastrocnemius muscles were removed for analysis post-exposure. A significant increase in body temperature and free radical generation was observed in both the muscles following heat exposure. This further caused a significant increase in protein carbonyl content, AOPP, and lipid peroxidation in heat-stressed muscles. These changes were more pronounced in heat-stressed soleus compared to the gastrocnemius muscle. Accumulation of unfolded, denatured proteins results in ER stress, causing activation of unfolded protein response (UPR) pathway. The expressions of UPR transducers were significantly higher in soleus as compared to the gastrocnemius muscle. A significant elevation in resting intracellular calcium ion was also observed in heat-stressed soleus muscle. Overloading of cells with misfolded proteins in soleus muscle activated ER-induced apoptosis as indicated by significant upregulation of C/EBP homologous protein and Caspase12. The study provides a detailed mechanistic representation of the differential response of muscles toward UPR under heat stress. Data suggests that soleus majorly being an oxidative muscle is more prone to heat stress-induced insult indicated by enhanced apoptosis. This study may aid in devising mitigation strategies to improve muscle performance under heat stress.
Collapse
Affiliation(s)
- Shivani Sharma
- Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, Delhi, 110054, India
- Department of Biochemistry, Panjab University, sector 25, Chandigarh, India
| | - Pooja Chaudhary
- Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, Delhi, 110054, India.
| | - Rajat Sandhir
- Department of Biochemistry, Panjab University, sector 25, Chandigarh, India
| | - Abhishek Bharadwaj
- Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, Delhi, 110054, India
| | - Rajinder K Gupta
- Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, Delhi, 110054, India
| | - Rahul Khatri
- Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, Delhi, 110054, India
| | - Amir Chand Bajaj
- Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, Delhi, 110054, India
| | - T P Baburaj
- Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, Delhi, 110054, India
| | - Sachin Kumar
- Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, Delhi, 110054, India
| | - M S Pal
- Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, Delhi, 110054, India
| | - Prasanna K Reddy
- Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, Delhi, 110054, India
| | - Bhuvnesh Kumar
- Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, Delhi, 110054, India
| |
Collapse
|
23
|
Hyatt HW, Powers SK. Disturbances in Calcium Homeostasis Promotes Skeletal Muscle Atrophy: Lessons From Ventilator-Induced Diaphragm Wasting. Front Physiol 2020; 11:615351. [PMID: 33391032 PMCID: PMC7773636 DOI: 10.3389/fphys.2020.615351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
Mechanical ventilation (MV) is often a life-saving intervention for patients in respiratory failure. Unfortunately, a common and undesired consequence of prolonged MV is the development of diaphragmatic atrophy and contractile dysfunction. This MV-induced diaphragmatic weakness is commonly labeled “ventilator-induced diaphragm dysfunction” (VIDD). VIDD is an important clinical problem because diaphragmatic weakness is a major risk factor for the failure to wean patients from MV; this inability to remove patients from ventilator support results in prolonged hospitalization and increased morbidity and mortality. Although several processes contribute to the development of VIDD, it is clear that oxidative stress leading to the rapid activation of proteases is a primary contributor. While all major proteolytic systems likely contribute to VIDD, emerging evidence reveals that activation of the calcium-activated protease calpain plays a required role. This review highlights the signaling pathways leading to VIDD with a focus on the cellular events that promote increased cytosolic calcium levels and the subsequent activation of calpain within diaphragm muscle fibers. In particular, we discuss the emerging evidence that increased mitochondrial production of reactive oxygen species promotes oxidation of the ryanodine receptor/calcium release channel, resulting in calcium release from the sarcoplasmic reticulum, accelerated proteolysis, and VIDD. We conclude with a discussion of important and unanswered questions associated with disturbances in calcium homeostasis in diaphragm muscle fibers during prolonged MV.
Collapse
Affiliation(s)
- Hayden W Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| | - Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
24
|
Hyatt HW, Ozdemir M, Yoshihara T, Nguyen BL, Deminice R, Powers SK. Calpains play an essential role in mechanical ventilation-induced diaphragmatic weakness and mitochondrial dysfunction. Redox Biol 2020; 38:101802. [PMID: 33279868 PMCID: PMC7724197 DOI: 10.1016/j.redox.2020.101802] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Mechanical ventilation (MV) is a life-saving intervention for many critically ill patients. Unfortunately, an unintended consequence of prolonged MV is the rapid development of diaphragmatic atrophy and contractile dysfunction, known as ventilator-induced diaphragm dysfunction (VIDD). Although the mechanism(s) responsible for VIDD are not fully understood, abundant evidence reveals that oxidative stress leading to the activation of the major proteolytic systems (i.e., autophagy, ubiquitin-proteasome, caspase, and calpain) plays a dominant role. Of the proteolytic systems involved in VIDD, calpain has received limited experimental attention due to the longstanding dogma that calpain plays a minor role in inactivity-induced muscle atrophy. Guided by preliminary experiments, we tested the hypothesis that activation of calpains play an essential role in MV-induced oxidative stress and the development of VIDD. This premise was rigorously tested by transgene overexpression of calpastatin, an endogenous inhibitor of calpains. Animals with/without transfection of the calpastatin gene in diaphragm muscle fibers were exposed to 12 h of MV. Results confirmed that overexpression of calpastatin barred MV-induced activation of calpain in diaphragm fibers. Importantly, deterrence of calpain activation protected the diaphragm against MV-induced oxidative stress, fiber atrophy, and contractile dysfunction. Moreover, prevention of calpain activation in the diaphragm forstalled MV-induced mitochondrial dysfunction and prevented MV-induced activation of caspase-3 along with the transcription of muscle specific E3 ligases. Collectively, these results support the hypothesis that calpain activation plays an essential role in the early development of VIDD. Further, these findings provide the first direct evidence that calpain plays an important function in inactivity-induced mitochondrial dysfunction and oxidative stress in skeletal muscle fibers. Inhibiting calpains during mechanical ventilation protects the diaphragm. Calpains play an important role in muscle atrophy and contractile dysfunction. Calpain inhibition during mechanical ventilation prevents mitochondrial dysfunction. Calpain-cleaved molecules may play important signaling roles. Calpain activation cross-talks with other proteolytic systems.
Collapse
Affiliation(s)
- Hayden W Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA.
| | - Mustafa Ozdemir
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Department of Exercise and Sport Sciences, Hacettepe University, Ankara, Turkey
| | - Toshinori Yoshihara
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Department of Exercise Physiology, Juntendo University, Tokyo, Japan
| | - Branden L Nguyen
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Rafael Deminice
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Department of Physical Education, State University of Londrina, Londrina, Brazil
| | - Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
25
|
Hall SE, Ahn B, Smuder AJ, Morton AB, Hinkley JM, Wiggs MP, Sollanek KJ, Hyatt H, Powers SK. Comparative Efficacy of Angiotensin II Type 1 Receptor Blockers Against Ventilator-Induced Diaphragm Dysfunction in Rats. Clin Transl Sci 2020; 14:481-486. [PMID: 33222389 PMCID: PMC7993256 DOI: 10.1111/cts.12916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/29/2020] [Indexed: 12/16/2022] Open
Abstract
Mechanical ventilation (MV) is a life‐saving intervention for many critically ill patients. Unfortunately, prolonged MV results in the rapid development of inspiratory muscle weakness due to diaphragmatic atrophy and contractile dysfunction (termed ventilator‐induced diaphragm dysfunction (VIDD)). Although VIDD is a major risk factor for problems in weaning patients from MV, a standard therapy to prevent VIDD does not exist. However, emerging evidence suggests that pharmacological blockade of angiotensin II type 1 receptors (AT1Rs) protects against VIDD. Nonetheless, the essential characteristics of AT1R blockers (ARBs) required to protect against VIDD remain unclear. To determine the traits of ARBs that are vital for protection against VIDD, we compared the efficacy of two clinically relevant ARBs, irbesartan and olmesartan; these ARBs differ in molecular structure and effects on AT1Rs. Specifically, olmesartan blocks both angiotensin II (AngII) binding and mechanical activation of AT1Rs, whereas irbesartan prevents only AngII binding to AT1Rs. Using a well‐established preclinical model of prolonged MV, we tested the hypothesis that compared with irbesartan, olmesartan provides greater protection against VIDD. Our results reveal that irbesartan does not protect against VIDD whereas olmesartan defends against both MV‐induced diaphragmatic atrophy and contractile dysfunction. These findings support the hypothesis that olmesartan is superior to irbesartan in protecting against VIDD and are consistent with the concept that blockade of mechanical activation of AT1Rs is a required property of ARBs to shield against VIDD. These important findings provide a foundation for future clinical trials to evaluate ARBs as a therapy to protect against VIDD.
Collapse
Affiliation(s)
- Stephanie E Hall
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, USA
| | - Bumsoo Ahn
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Ashley J Smuder
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | | | - J Matthew Hinkley
- Advent Health Translational Research Institute, Orlando, Florida, USA
| | | | | | - Hayden Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
26
|
Almeida MR, Horta JGÁ, de Matos NA, de Souza ABF, Castro TDF, Cândido LDS, Andrade MC, Cangussú SD, Costa GDP, Talvani A, Bezerra FS. The effects of different ventilatory modes in female adult rats submitted to mechanical ventilation. Respir Physiol Neurobiol 2020; 284:103583. [PMID: 33202295 DOI: 10.1016/j.resp.2020.103583] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 10/30/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022]
Abstract
This study aimed to analyze the effects of volume-controlled ventilation (VCV) and pressure-controlled ventilation (PCV) modes in female Wistar rats. 18 Wistar female adult rats were divided into three groups: control (CG), pressure-controlled ventilation (PCVG), and volume-controlled ventilation (VCVG). PCVG and VCVG were submitted to MV for one hour with a tidal volume (TV) of 8 mL/Kg, respiratory rate of 80 breaths/min, and positive end-expiratory pressure of 0 cmH2O. At the end of the experiment, all animals were euthanized. The neutrophils and lymphocytes influx to lung were higher in VCVG and PCVG compared to CG. The activities of superoxide dismutase, catalase and myeloperoxidase were higher in PCVG compared to CG. There was an increase in lipid peroxidation and protein oxidation in PCVG compared to CG. The levels of CCL3 and CCL5 were higher in PCVG compared to CG. In conclusions, the PCV mode promoted structural changes in the lung parenchyma, redox imbalance and inflammation in healthy adult female rats submitted to MV.
Collapse
Affiliation(s)
- Matheus Rocha Almeida
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Brazil
| | - Jacques Gabriel Álvares Horta
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Brazil; Department of Clinical Medicine/Pediatrics, School of Medicine, Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - Natália Alves de Matos
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Brazil
| | - Ana Beatriz Farias de Souza
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Brazil
| | - Thalles de Freitas Castro
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Brazil
| | - Leandro da Silva Cândido
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Brazil
| | - Mônica Campos Andrade
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Brazil
| | - Sílvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Brazil
| | - Guilherme de Paula Costa
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Brazil
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Brazil.
| |
Collapse
|
27
|
Powers SK, Ozdemir M, Hyatt H. Redox Control of Proteolysis During Inactivity-Induced Skeletal Muscle Atrophy. Antioxid Redox Signal 2020; 33:559-569. [PMID: 31941357 PMCID: PMC7454189 DOI: 10.1089/ars.2019.8000] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Significance: Skeletal muscles play essential roles in key body functions including breathing, locomotion, and glucose homeostasis; therefore, maintaining healthy skeletal muscles is important. Prolonged periods of muscle inactivity (e.g., bed rest, mechanical ventilation, or limb immobilization) result in skeletal muscle atrophy and weakness. Recent Advances: Disuse skeletal muscle atrophy occurs due to both accelerated proteolysis and decreased protein synthesis with proteolysis playing a leading role in some types of inactivity-induced atrophy. Although all major proteolytic systems are involved in inactivity-induced proteolysis in skeletal muscles, growing evidence indicates that both calpain and autophagy play an important role. Regulation of proteolysis in skeletal muscle is under complex control, but it is established that activation of both calpain and autophagy is directly linked to oxidative stress. Critical Issues: In this review, we highlight the experimental evidence that supports a cause and effect link between reactive oxygen species (ROS) and activation of both calpain and autophagy in skeletal muscle fibers during prolonged inactivity. We also review the sources of oxidant production in muscle fibers during inactivity-induced atrophy, and provide a detailed discussion on how ROS activates both calpain and autophagy during disuse muscle wasting. Future Directions: Future studies are required to delineate the specific mechanisms by which ROS activates both calpain and autophagy in skeletal muscles during prolonged periods of contractile inactivity. This knowledge is essential to develop the most effective strategies to protect against disuse muscle atrophy. Antioxid. Redox Signal. 33, 559-569.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Mustafa Ozdemir
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Hayden Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
28
|
Effect of Long-Term Polytrauma on Ventilator-Induced Diaphragmatic Dysfunction in a Piglet Model. Shock 2020; 52:443-448. [PMID: 30300316 DOI: 10.1097/shk.0000000000001272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Mechanical ventilation is known to activate oxidative stress and proteolytic pathways in the diaphragm. Trauma by inducing inflammation and activating proteolytic pathways may potentiate the effects of mechanical ventilation on the diaphragm. In a blunt chest trauma with concomitant injuries we tested the hypothesis that trauma via inflammation further activates the proteolytic pathways and worsens atrophy in the diaphragm. MATERIAL AND METHODS Piglets were separated into two groups and underwent 72 h of mechanical ventilation. One group received a polytrauma (PT) by unilateral femur fracture, blunt chest trauma with lung contusion, laparotomy with standardized liver incision, and a predefined hemorrhagic shock. The second mechanically ventilated group (MV) did not receive any trauma. A non-ventilated group (Con) served as control.Diaphragmatic fiber dimensions, Western Blot analyses of proteolytic pathways, and lipid peroxidation and messenger ribonucleic acid (mRNA) levels of cytokines and nuclear factor kappa b subunit p65 were measured. RESULTS Active Caspase-3 was significantly increased in MV (P = 0.019), and in PT (P = 0.02) compared with Con. Nuclear factor kappa b subunit p65, was upregulated in PT (P = 0.010) compared with Con. IL-6 mRNA increased significantly in PT compared with Con (P = 0.0024) but did not differ between Con and MV. CONCLUSION Trauma and mechanical ventilation induced proteolysis and atrophy in the diaphragm, but only polytrauma induced an inflammatory response in the diaphragm. The additional traumatic inflammatory stimulus did not increase the levels of the prementioned variables. These data underline that inflammation is not a major contributor to ventilator-induced diaphragmatic dysfunction. TRIAL REGISTRY NUMBER AZ 84-02.04.2014.A265 (Landesamt für Natur-, Umwelt- und Verbraucherschutz, LANUV NRW, Germany).
Collapse
|
29
|
Molecular changes in transcription and metabolic pathways underlying muscle atrophy in the CuZnSOD null mouse model of sarcopenia. GeroScience 2020; 42:1101-1118. [PMID: 32394347 DOI: 10.1007/s11357-020-00189-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/02/2020] [Indexed: 12/19/2022] Open
Abstract
Mice lacking the superoxide anion scavenger CuZn superoxide dismutase (Sod1-/- mice) develop a number of age-related phenotypes, including an early progression of muscle atrophy and weakness (sarcopenia) associated with loss of innervation. The purpose of this study was to delineate the early development of sarcopenia in the Sod1-/- mice and to measure changes in the muscle transcriptome, proteome, and eicosanoid profile at the stage when sarcopenia is markedly induced in this model (7-9 months of age). We found a strong correlation between muscle atrophy and mitochondrial state 1 hydroperoxide production, which was 40% higher in isolated mitochondria from Sod1-/- mouse gastrocnemius muscle by 2 months of age. The primary pathways showing altered gene expression in Sod1-/- mice identified by RNA-seq transcriptomic analysis are protein ubiquitination, synaptic long-term potentiation, calcium signaling, phospholipase C signaling, AMPK, and TWEAK signaling. Targeted proteomics shows elevated expression of mitochondrial proteins, fatty acid metabolism enzymes, tricarboxylic acid (TCA) cycle enzymes, and antioxidants, while enzymes involved in carbohydrate metabolism are downregulated in Sod1-/- mice. LC-MS analysis of lipids in gastrocnemius muscle detected 78 eicosanoids, of which 31 are significantly elevated in muscle from Sod1-/- mice. These data suggest that mitochondrial hydroperoxide generation is elevated prior to muscle atrophy and may be a potential driving factor of changes in the transcriptome, proteome, and eicosanoid profile of the Sod1-/- mice. Together, these analyses revealed important molecular events that occur during muscle atrophy, which will pave the way for future studies using new approaches to treat sarcopenia.
Collapse
|
30
|
Petushkova AI, Zamyatnin AA. Redox-Mediated Post-Translational Modifications of Proteolytic Enzymes and Their Role in Protease Functioning. Biomolecules 2020; 10:biom10040650. [PMID: 32340246 PMCID: PMC7226053 DOI: 10.3390/biom10040650] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 12/13/2022] Open
Abstract
Proteolytic enzymes play a crucial role in metabolic processes, providing the cell with amino acids through the hydrolysis of multiple endogenous and exogenous proteins. In addition to this function, proteases are involved in numerous protein cascades to maintain cellular and extracellular homeostasis. The redox regulation of proteolysis provides a flexible dose-dependent mechanism for proteolytic activity control. The excessive reactive oxygen species (ROS) and reactive nitrogen species (RNS) in living organisms indicate pathological conditions, so redox-sensitive proteases can swiftly induce pro-survival responses or regulated cell death (RCD). At the same time, severe protein oxidation can lead to the dysregulation of proteolysis, which induces either protein aggregation or superfluous protein hydrolysis. Therefore, oxidative stress contributes to the onset of age-related dysfunction. In the present review, we consider the post-translational modifications (PTMs) of proteolytic enzymes and their impact on homeostasis.
Collapse
Affiliation(s)
- Anastasiia I. Petushkova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Correspondence:
| |
Collapse
|
31
|
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
|
32
|
Redox modulation of muscle mass and function. Redox Biol 2020; 35:101531. [PMID: 32371010 PMCID: PMC7284907 DOI: 10.1016/j.redox.2020.101531] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/28/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022] Open
Abstract
Muscle mass and strength are very important for exercise performance. Training-induced musculoskeletal injuries usually require periods of complete immobilization to prevent any muscle contraction of the affected muscle groups. Disuse muscle wasting will likely affect every sport practitioner in his or her lifetime. Even short periods of disuse results in significant declines in muscle size, fiber cross sectional area, and strength. To understand the molecular signaling pathways involved in disuse muscle atrophy is of the utmost importance to develop more effective countermeasures in sport science research. We have divided our review in four different sections. In the first one we discuss the molecular mechanisms involved in muscle atrophy including the main protein synthesis and protein breakdown signaling pathways. In the second section of the review we deal with the main cellular, animal, and human atrophy models. The sources of reactive oxygen species in disuse muscle atrophy and the mechanism through which they regulate protein synthesis and proteolysis are reviewed in the third section of this review. The last section is devoted to the potential interventions to prevent muscle disuse atrophy with especial consideration to studies on which the levels of endogenous antioxidants enzymes or dietary antioxidants have been tested.
Collapse
|
33
|
Luo Z, Han S, Sun W, Wang Y, Liu S, Yang L, Pang B, Jin J, Chen H, Cao Z, Ma Y. Maintenance of spontaneous breathing at an intensity of 60%-80% may effectively prevent mechanical ventilation-induced diaphragmatic dysfunction. PLoS One 2020; 15:e0229944. [PMID: 32131083 PMCID: PMC7056322 DOI: 10.1371/journal.pone.0229944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 02/18/2020] [Indexed: 11/18/2022] Open
Abstract
Controlled mechanical ventilation (CMV) can cause diaphragmatic motionlessness to induce diaphragmatic dysfunction. Partial maintenance of spontaneous breathing (SB) can reduce ventilation-induced diaphragmatic dysfunction (VIDD). However, to what extent SB is maintained in CMV can attenuate or even prevent VIDD has been rarely reported. The current study aimed to investigate the relationship between SB intensity and VIDD and to identify what intensity of SB maintained in CMV can effectively avoid VIDD. Adult rats were randomly divided according to different SB intensities: SB (0% pressure controlled ventilation (PCV)), high-intensity SB (20% PCV), medium-intensity SB (40% PCV), medium-low intensity SB (60% PCV), low-intensity SB (80% PCV), and PCV (100% PCV). The animals underwent 24-h controlled mechanical ventilation (CMV). The transdiaphragmatic pressure (Pdi), the maximal Pdi (Pdi max) when phrenic nerves were stimulated, Pdi/Pdi max, and the diaphragmatic tonus under different frequencies of electric stimulations were determined. Calpain and caspase-3 were detected using ELISA and the cross-section areas (CSAs) of different types of muscle fibers were measured. The Pdi showed a significant decrease from 20% PCV and the Pdi max showed a significant decrease from 40% PCV (P<0.05). In vivo and vitro diaphragmatic tonus exhibited a significant decrease from 40% PCV and 20% PCV, respectively (P<0.05). From 20% PCV, the CSAs of types I, IIa, and IIb/x muscle fibers showed significant differences, which reached the lowest levels at 100% PCV. SB intensity is negatively associated with the development of VIDD. Maintenance of SB at an intensity of 60%-80% may effectively prevent the occurrence of VIDD.
Collapse
Affiliation(s)
- Zujin Luo
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Silu Han
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Wei Sun
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Sijie Liu
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Liu Yang
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Baosen Pang
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiawei Jin
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Hong Chen
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhixin Cao
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- * E-mail: (ZC); (YM)
| | - Yingmin Ma
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- * E-mail: (ZC); (YM)
| |
Collapse
|
34
|
Qaisar R, Karim A, Elmoselhi AB. Muscle unloading: A comparison between spaceflight and ground-based models. Acta Physiol (Oxf) 2020; 228:e13431. [PMID: 31840423 DOI: 10.1111/apha.13431] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 12/09/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022]
Abstract
Prolonged unloading of skeletal muscle, a common outcome of events such as spaceflight, bed rest and hindlimb unloading, can result in extensive metabolic, structural and functional changes in muscle fibres. With advancement in investigations of cellular and molecular mechanisms, understanding of disuse muscle atrophy has significantly increased. However, substantial gaps exist in our understanding of the processes dictating muscle plasticity during unloading, which prevent us from developing effective interventions to combat muscle loss. This review aims to update the status of knowledge and underlying mechanisms leading to cellular and molecular changes in skeletal muscle during unloading. We have also discussed advances in the understanding of contractile dysfunction during spaceflights and in ground-based models of muscle unloading. Additionally, we have elaborated on potential therapeutic interventions that show promising results in boosting muscle mass and strength during mechanical unloading. Finally, we have identified key gaps in our knowledge as well as possible research direction for the future.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Department of Basic Medical Sciences College of Medicine University of Sharjah Sharjah UAE
| | - Asima Karim
- Department of Basic Medical Sciences College of Medicine University of Sharjah Sharjah UAE
| | - Adel B. Elmoselhi
- Department of Basic Medical Sciences College of Medicine University of Sharjah Sharjah UAE
- Department of Physiology Michigan State University East Lansing MI USA
| |
Collapse
|
35
|
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: 20] [Impact Index Per Article: 4.0] [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
|
36
|
Abstract
WHAT WE ALREADY KNOW ABOUT THIS TOPIC Diaphragm dysfunction and atrophy develop during controlled mechanical ventilation. Although oxidative stress injures muscle during controlled mechanical ventilation, it is unclear whether it causes autophagy or fiber atrophy. WHAT THIS ARTICLE TELLS US THAT IS NEW Pretreatment of rats undergoing 24 h of mechanical ventilation with N-acetylcysteine prevents decreases in diaphragm contractility, inhibits the autophagy and proteasome pathways, but has no influence on the development of diaphragm fiber atrophy. BACKGROUND Diaphragm dysfunction and atrophy develop during prolonged controlled mechanical ventilation. Fiber atrophy has been attributed to activation of the proteasome and autophagy proteolytic pathways. Oxidative stress activates the proteasome during controlled mechanical ventilation, but it is unclear whether it also activates autophagy. This study investigated whether pretreatment with the antioxidant N-acetylcysteine affects controlled mechanical ventilation-induced diaphragm contractile dysfunction, fiber atrophy, and proteasomal and autophagic pathway activation. The study also explored whether proteolytic pathway activity during controlled mechanical ventilation is mediated by microRNAs that negatively regulate ubiquitin E3 ligases and autophagy-related genes. METHODS Three groups of adult male rats were studied (n = 10 per group). The animals in the first group were anesthetized and allowed to spontaneously breathe. Animals in the second group were pretreated with saline before undergoing controlled mechanical ventilation for 24 h. The animals in the third group were pretreated with N-acetylcysteine (150 mg/kg) before undergoing controlled mechanical ventilation for 24 h. Diaphragm contractility and activation of the proteasome and autophagy pathways were measured. Expressions of microRNAs that negatively regulate ubiquitin E3 ligases and autophagy-related genes were measured with quantitative polymerase chain reaction. RESULTS Controlled mechanical ventilation decreased diaphragm twitch force from 428 ± 104 g/cm (mean ± SD) to 313 ± 50 g/cm and tetanic force from 2,491 ± 411 g/cm to 1,618 ± 177 g/cm. Controlled mechanical ventilation also decreased diaphragm fiber size, increased expression of several autophagy genes, and augmented Atrogin-1, MuRF1, and Nedd4 expressions by 36-, 41-, and 8-fold, respectively. Controlled mechanical ventilation decreased the expressions of six microRNAs (miR-20a, miR-106b, miR-376, miR-101a, miR-204, and miR-93) that regulate autophagy genes. Pretreatment with N-acetylcysteine prevented diaphragm contractile dysfunction, attenuated protein ubiquitination, and downregulated E3 ligase and autophagy gene expression. It also reversed controlled mechanical ventilation-induced microRNA expression decreases. N-Acetylcysteine pretreatment had no affect on fiber atrophy. CONCLUSIONS Prolonged controlled mechanical ventilation activates the proteasome and autophagy pathways in the diaphragm through oxidative stress. Pathway activation is accomplished, in part, through inhibition of microRNAs that negatively regulate autophagy-related genes.
Collapse
|
37
|
Souza PRMD, da Palma RK, Vieira RP, Santos FD, Monteiro-De-Moraes WMA, Medeiros A, Koike MK, Arantes-Costa FM, De Angelis K, Irigoyen MC, Consolim Colombo FM. Early activation of ubiquitin-proteasome system at the diaphragm tissue occurs independently of left ventricular dysfunction in SHR rats. Exp Biol Med (Maywood) 2020; 245:245-253. [PMID: 31986909 DOI: 10.1177/1535370219897883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Hypertensive status induces modifications in the respiratory profile. Previous studies have indicated that hypertensive rats show increased respiratory-sympathetic coupling compared to normotensive rats. However, these effects and especially the mechanisms underlying such effects are not well known. Thus, we evaluated the influence of high blood pressure and autonomic dysfunction on a ventilatory pattern associated with lung injury and on the ubiquitin-proteasome system of the diaphragm muscle. Autonomic cardiovascular modulation (systolic BP variance and low-frequency band and pulse interval variance) and arterial blood gases patterns (pH, pO2, HCO3, SpO2), can be changed by hypertension, as well exacerbated chemoreflex pressor response. We observed that the diaphragm muscle of SHR showed increase in type I cross-sectional fiber (16%) and reduction in type II cross-sectional fiber area (41%), increased activity of the ubiquitin-proteasome system and lipid peroxidation, with no differences between groups in the analysis of ubiquitinated proteins and misfolded proteins. Our results showed that hypertension induced functional compensatory/adverse alterations associated with diaphragm fiber type changes and protein degradation as well as changed autonomic control of circulation. In conclusion, we believe there is an adaptation in ventilatory pattern in regarding to prevent the development of fatigue and muscle weakness and improve ventilatory endurance. Impact statement It was well known that hypertension can be driven by increased sympathetic activity and has been documented as a central link between autonomic dysfunction and alterations in the respiratory pattern. Our study demonstrated the impact of hypertension in ventilatory mechanics and their relationship with diaphragm muscle protein degradation. These findings may assist us in future alternative treatments to prevent diaphragm fatigue and weakness in hypertensive patients.
Collapse
Affiliation(s)
- Pamella Ramona Moraes de Souza
- Hypertension Unit, Heart Institute (InCor), School of Medicine, University of São Paulo (FMUSP), Avenida Dr Eneas de Carvalho Aguiar 44, São Paulo/SP, Brazil 05403-000.,Department of Post-graduation in Medicine, Nove de Julho University (UNINOVE), Rua Vergueiro 235/249, São Paulo/SP, Brazil 01504-001
| | - Renata Kelly da Palma
- Department of Post-graduation in Medicine, Nove de Julho University (UNINOVE), Rua Vergueiro 235/249, São Paulo/SP, Brazil 01504-001.,Institute for Bioengineering of Catalonia, Biomimetic systems for cell engineering. Barcelona, Spain (IBEC). C. Baldiri Reixac, 15-21 Barcelona. 08028
| | - Rodolfo Paula Vieira
- Department of Post-graduation in Medicine, Nove de Julho University (UNINOVE), Rua Vergueiro 235/249, São Paulo/SP, Brazil 01504-001.,Institute for Bioengineering of Catalonia, Biomimetic systems for cell engineering. Barcelona, Spain (IBEC). C. Baldiri Reixac, 15-21 Barcelona. 08028
| | - Fernando Dos Santos
- Hypertension Unit, Heart Institute (InCor), School of Medicine, University of São Paulo (FMUSP), Avenida Dr Eneas de Carvalho Aguiar 44, São Paulo/SP, Brazil 05403-000
| | - Wilson Max Almeida Monteiro-De-Moraes
- Hypertension Unit, Heart Institute (InCor), School of Medicine, University of São Paulo (FMUSP), Avenida Dr Eneas de Carvalho Aguiar 44, São Paulo/SP, Brazil 05403-000
| | | | - Marcia Kiyomi Koike
- Department of Medicine, Center of Development of Medical Education, CEDEM, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Fernanda Magalhães Arantes-Costa
- Department of Medicine, Center of Development of Medical Education, CEDEM, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Kátia De Angelis
- Federal University of Sao Paulo.Rua Pedro de Toledo, 1800. São Paulo/SP, Brazil 04021-001
| | - Maria Claudia Irigoyen
- Hypertension Unit, Heart Institute (InCor), School of Medicine, University of São Paulo (FMUSP), Avenida Dr Eneas de Carvalho Aguiar 44, São Paulo/SP, Brazil 05403-000
| | - Fernanda Marciano Consolim Colombo
- Hypertension Unit, Heart Institute (InCor), School of Medicine, University of São Paulo (FMUSP), Avenida Dr Eneas de Carvalho Aguiar 44, São Paulo/SP, Brazil 05403-000.,Department of Post-graduation in Medicine, Nove de Julho University (UNINOVE), Rua Vergueiro 235/249, São Paulo/SP, Brazil 01504-001
| |
Collapse
|
38
|
Molina Peña ME, Sánchez CM, Rodríguez-Triviño CY. Physiopathological mechanisms of diaphragmatic dysfunction associated with mechanical ventilation. ACTA ACUST UNITED AC 2020; 67:195-203. [PMID: 31982168 DOI: 10.1016/j.redar.2019.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/06/2019] [Accepted: 12/16/2019] [Indexed: 02/07/2023]
Abstract
Ventilator-induced diaphragm dysfunction (VIDD) is the loss of diaphragmatic muscle strength'related to of mechanical ventilation, noticed during the first day or 48hours after initiating controlled mechanical ventilation. This alteration has been related to disruption on the insulin growth factor/phosphoinositol 3-kinase/kinase B protein pathway (IGF/PI3K/AKT), in addition to an overexpression of FOXO, expression of NF-kB signaling, increase function of muscular ubiquitin ligase and activation of caspasa-3. VIDD has a negative impact on quality of life, duration of mechanical ventilation, and hospitalization stance and cost. More studies are necessary to understated the process and impact of VIDD. This is a narrative review of non-systematic literature, aiming to explain the molecular pathways involved in VIDD.
Collapse
Affiliation(s)
- M E Molina Peña
- Semillero de Fisiología Pr ctica aplicada, Grupo Navarra Medicina, Departamento de Ciencias Fisiológicas, Facultad de Ciencias de la Salud, Fundación Universitaria Navarra-UNINAVARRA, Neiva, Huila, Colombia.
| | - C M Sánchez
- Semillero de Fisiología Pr ctica aplicada, Grupo Navarra Medicina, Departamento de Ciencias Fisiológicas, Facultad de Ciencias de la Salud, Fundación Universitaria Navarra-UNINAVARRA, Neiva, Huila, Colombia
| | - C Y Rodríguez-Triviño
- Grupo Navarra Medicina, Departamento de Ciencias Fisiológicas, Facultad de Ciencias de la Salud, Fundación Universitaria Navarra-UNINAVARRA, Neiva, Huila, Colombia; Grupo Cuidar, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| |
Collapse
|
39
|
Dexmedetomidine Impairs Diaphragm Function and Increases Oxidative Stress but Does Not Aggravate Diaphragmatic Atrophy in Mechanically Ventilated Rats. Anesthesiology 2019; 128:784-795. [PMID: 29346133 DOI: 10.1097/aln.0000000000002081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Anesthetics in ventilated patients are critical as any cofactor hampering diaphragmatic function may have a negative impact on the weaning progress and therefore on patients' mortality. Dexmedetomidine may display antioxidant and antiproteolytic properties, but it also reduced glucose uptake by the muscle, which may impair diaphragm force production. This study tested the hypothesis that dexmedetomidine could inhibit ventilator-induced diaphragmatic dysfunction. METHODS Twenty-four rats were separated into three groups (n = 8/group). Two groups were mechanically ventilated during either dexmedetomidine or pentobarbital exposure for 24 h, referred to as interventional groups. A third group of directly euthanized rats served as control. Force generation, fiber dimensions, proteolysis markers, protein oxidation and lipid peroxidation, calcium homeostasis markers, and glucose transporter-4 (Glut-4) translocation were measured in the diaphragm. RESULTS Diaphragm force, corrected for cross-sectional area, was significantly decreased in both interventional groups compared to controls and was significantly lower with dexmedetomidine compared to pentobarbital (e.g., 100 Hz: -18%, P < 0.0001). In contrast to pentobarbital, dexmedetomidine did not lead to diaphragmatic atrophy, but it induced more protein oxidation (200% vs. 73% in pentobarbital, P = 0.0015), induced less upregulation of muscle atrophy F-box (149% vs. 374% in pentobarbital, P < 0.001) and impaired Glut-4 translocation (-73%, P < 0.0005). It activated autophagy, the calcium-dependent proteases, and caused lipid peroxidation similarly to pentobarbital. CONCLUSIONS Twenty-four hours of mechanical ventilation during dexmedetomidine sedation led to a worsening of ventilation-induced diaphragm dysfunction, possibly through impaired Glut-4 translocation. Although dexmedetomidine prevented diaphragmatic fiber atrophy, it did not inhibit oxidative stress and activation of the proteolytic pathways.
Collapse
|
40
|
Peñuelas O, Keough E, López-Rodríguez L, Carriedo D, Gonçalves G, Barreiro E, Lorente JÁ. Ventilator-induced diaphragm dysfunction: translational mechanisms lead to therapeutical alternatives in the critically ill. Intensive Care Med Exp 2019; 7:48. [PMID: 31346802 PMCID: PMC6658639 DOI: 10.1186/s40635-019-0259-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 02/08/2023] Open
Abstract
Mechanical ventilation [MV] is a life-saving technique delivered to critically ill patients incapable of adequately ventilating and/or oxygenating due to respiratory or other disease processes. This necessarily invasive support however could potentially result in important iatrogenic complications. Even brief periods of MV may result in diaphragm weakness [i.e., ventilator-induced diaphragm dysfunction [VIDD]], which may be associated with difficulty weaning from the ventilator as well as mortality. This suggests that VIDD could potentially have a major impact on clinical practice through worse clinical outcomes and healthcare resource use. Recent translational investigations have identified that VIDD is mainly characterized by alterations resulting in a major decline of diaphragmatic contractile force together with atrophy of diaphragm muscle fibers. However, the signaling mechanisms responsible for VIDD have not been fully established. In this paper, we summarize the current understanding of the pathophysiological pathways underlying VIDD and highlight the diagnostic approach, as well as novel and experimental therapeutic options.
Collapse
Affiliation(s)
- Oscar Peñuelas
- Intensive Care Unit, Hospital Universitario de Getafe, Carretera de Toledo, km 12.5, 28905, Getafe, Madrid, Spain.
- Centro de Investigación en Red de Enfermedades Respiratorias [CIBERES], Instituto de Salud Carlos III [ISCIII], Madrid, Spain.
| | - Elena Keough
- Intensive Care Unit, Hospital Universitario de Getafe, Carretera de Toledo, km 12.5, 28905, Getafe, Madrid, Spain
| | - Lucía López-Rodríguez
- Intensive Care Unit, Hospital Universitario de Getafe, Carretera de Toledo, km 12.5, 28905, Getafe, Madrid, Spain
| | - Demetrio Carriedo
- Intensive Care Unit, Hospital Universitario de Getafe, Carretera de Toledo, km 12.5, 28905, Getafe, Madrid, Spain
| | - Gesly Gonçalves
- Intensive Care Unit, Hospital Universitario de Getafe, Carretera de Toledo, km 12.5, 28905, Getafe, Madrid, Spain
| | - Esther Barreiro
- Centro de Investigación en Red de Enfermedades Respiratorias [CIBERES], Instituto de Salud Carlos III [ISCIII], Madrid, Spain
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department [CEXS], Barcelona, Spain
- Universitat Pompeu Fabra [UPF], Barcelona Biomedical Research Park [PRBB], Barcelona, Spain
| | - José Ángel Lorente
- Intensive Care Unit, Hospital Universitario de Getafe, Carretera de Toledo, km 12.5, 28905, Getafe, Madrid, Spain
- Centro de Investigación en Red de Enfermedades Respiratorias [CIBERES], Instituto de Salud Carlos III [ISCIII], Madrid, Spain
- Universidad Europea, Madrid, Spain
| |
Collapse
|
41
|
Interactions between Cytosolic Phospholipase A2 Activation and Mitochondrial Reactive Oxygen Species Production in the Development of Ventilator-Induced Diaphragm Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2561929. [PMID: 31178955 PMCID: PMC6501131 DOI: 10.1155/2019/2561929] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/13/2019] [Indexed: 12/29/2022]
Abstract
Cytosolic phospholipase A2 (cPLA2) has been reported to be critical for infection-induced mitochondrial reactive oxygen species (ROS) production and diaphragm dysfunction (DD). In the present study, we aim to investigate whether cPLA2 was involved in ventilator-induced diaphragm dysfunction (VIDD). Our results showed that mechanical ventilation (MV) induced cPLA2 activation in the diaphragm with excessive mitochondrial ROS generation and muscle weakness. Specific inhibition of cPLA2 with CDIBA resulted in decreased mitochondrial ROS levels and improved diaphragm forces. In addition, mitochondria-targeted antioxidant MitoTEMPO attenuated ventilator-induced mitochondrial oxidative stress and downregulated cPLA2 activation in vivo. Both CDIBA and MitoTEMPO were able to attenuate protein degradation, muscle atrophy, and weakness following prolonged MV. Furthermore, laser Doppler imaging showed that MV decreased diaphragm tissue perfusion and induced subsequent hypoxia. An in vitro study also demonstrated a positive association between cPLA2 activation and mitochondrial ROS generation in C2C12 cells cultured under hypoxic condition. Collectively, our study showed that cPLA2 activation positively interacts with mitochondrial ROS generation in the development of VIDD, and ventilator-induced diaphragm hypoxia serves as a possible contributor to this positive feedback loop.
Collapse
|
42
|
Tang H, Shrager JB. The Signaling Network Resulting in Ventilator-induced Diaphragm Dysfunction. Am J Respir Cell Mol Biol 2019; 59:417-427. [PMID: 29768017 DOI: 10.1165/rcmb.2018-0022tr] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Mechanical ventilation (MV) is a life-saving measure for those incapable of adequately ventilating or oxygenating without assistance. Unfortunately, even brief periods of MV result in diaphragm weakness (i.e., ventilator-induced diaphragm dysfunction [VIDD]) that may render it difficult to wean the ventilator. Prolonged MV is associated with cascading complications and is a strong risk factor for death. Thus, prevention of VIDD may have a dramatic impact on mortality rates. Here, we summarize the current understanding of the pathogenic events underlying VIDD. Numerous alterations have been proven important in both human and animal MV diaphragm. These include protein degradation via the ubiquitin proteasome system, autophagy, apoptosis, and calpain activity-all causing diaphragm muscle fiber atrophy, altered energy supply via compromised oxidative phosphorylation and upregulation of glycolysis, and also mitochondrial dysfunction and oxidative stress. Mitochondrial oxidative stress in fact appears to be a central factor in each of these events. Recent studies by our group and others indicate that mitochondrial function is modulated by several signaling molecules, including Smad3, signal transducer and activator of transcription 3, and FoxO. MV rapidly activates Smad3 and signal transducer and activator of transcription 3, which upregulate mitochondrial oxidative stress. Additional roles may be played by angiotensin II and leaky ryanodine receptors causing elevated calcium levels. We present, here, a hypothetical scaffold for understanding the molecular pathogenesis of VIDD, which links together these elements. These pathways harbor several drug targets that could soon move toward testing in clinical trials. We hope that this review will shape a short list of the most promising candidates.
Collapse
Affiliation(s)
- Huibin Tang
- Stanford University School of Medicine, Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford, California; and Veterans Affairs Palo Alto Healthcare System, Palo Alto, California
| | - Joseph B Shrager
- Stanford University School of Medicine, Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford, California; and Veterans Affairs Palo Alto Healthcare System, Palo Alto, California
| |
Collapse
|
43
|
Gorgey AS, Witt O, O’Brien L, Cardozo C, Chen Q, Lesnefsky EJ, Graham ZA. Mitochondrial health and muscle plasticity after spinal cord injury. Eur J Appl Physiol 2018; 119:315-331. [DOI: 10.1007/s00421-018-4039-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 11/22/2018] [Indexed: 01/15/2023]
|
44
|
Endurance exercise protects skeletal muscle against both doxorubicin-induced and inactivity-induced muscle wasting. Pflugers Arch 2018; 471:441-453. [PMID: 30426248 DOI: 10.1007/s00424-018-2227-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/09/2018] [Accepted: 10/18/2018] [Indexed: 12/20/2022]
Abstract
Repeated bouts of endurance exercise promotes numerous biochemical adaptations in skeletal muscle fibers resulting in a muscle phenotype that is protected against a variety of homeostatic challenges; these exercise-induced changes in muscle phenotype are often referred to as "exercise preconditioning." Importantly, exercise preconditioning provides protection against several threats to skeletal muscle health including cancer chemotherapy (e.g., doxorubicin) and prolonged muscle inactivity. This review summarizes our current understanding of the mechanisms responsible for exercise-induced protection of skeletal muscle fibers against both doxorubicin-induced muscle wasting and a unique form of inactivity-induced muscle atrophy (i.e., ventilator-induced diaphragm atrophy). Specifically, the first section of this article will highlight the potential mechanisms responsible for exercise-induced protection of skeletal muscle fibers against doxorubicin-induced fiber atrophy. The second segment will discuss the biochemical changes that are responsible for endurance exercise-mediated protection of diaphragm muscle against ventilator-induced diaphragm wasting. In each section, we highlight gaps in our knowledge in hopes of stimulating future research in this evolving field of investigation.
Collapse
|
45
|
Powers SK, Morton AB, Hyatt H, Hinkley MJ. The Renin-Angiotensin System and Skeletal Muscle. Exerc Sport Sci Rev 2018; 46:205-214. [PMID: 30001274 DOI: 10.1249/jes.0000000000000158] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The renin-angiotensin system (RAS) plays a key role in the control of blood pressure and fluid homeostasis. Emerging evidence also reveals that hyperactivity of the RAS contributes to skeletal muscle wasting. This review discusses the key role that the RAS plays in skeletal muscle wasting due to congestive heart failure, chronic kidney disease, and ventilator-induced diaphragmatic wasting.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
| | | | | | | |
Collapse
|
46
|
Kaur N, Gupta P, Saini V, Sherawat S, Gupta S, Dua A, Kumar V, Injeti E, Mittal A. Cinnamaldehyde regulates H 2 O 2 -induced skeletal muscle atrophy by ameliorating the proteolytic and antioxidant defense systems. J Cell Physiol 2018; 234:6194-6208. [PMID: 30317570 DOI: 10.1002/jcp.27348] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 08/17/2018] [Indexed: 12/20/2022]
Abstract
Skeletal muscle atrophy/wasting is associated with impaired protein metabolism in diverse physiological and pathophysiological conditions. Elevated levels of reactive oxygen species (ROS), disturbed redox status, and weakened antioxidant defense system are the major contributing factors toward atrophy. Regulation of protein metabolism by controlling ROS levels and its associated catabolic pathways may help in treating atrophy and related clinical conditions. Although cinnamaldehyde (CNA) enjoys the established status of antioxidant and its role in ROS management is reported, impact of CNA on skeletal muscle atrophy and related pathways is still unexplored. In the current study, the impact of CNA on C2C12 myotubes and the possible protection of cultured cells from H 2 O 2 -induced atrophy is examined. Myotubes were treated with H 2 O 2 in the presence and absence of CNA and the changes in the antioxidative, proteolytic systems, and mitochondrial functions were scored. Morphological analysis showed significant protective effects of CNA on length, diameter, and nuclei fusion index of myotubes. The evaluation of biochemical markers of atrophy; creatine kinase, lactate dehydrogenase, succinate dehydrogenase along with the study of muscle-specific structural protein (i.e., myosin heavy chain-fast [MHCf] type) showed significant protection of proteins by CNA. CNA pretreatment not only checked the activation of proteolytic systems (ubiquitin-proteasome E3-ligases [MuRF1/Atrogin1]), autophagy [Beclin1/LC3B], cathepsin L, calpain, caspase), but also prevented any alteration in the activities of antioxidative defense enzymes (catalase, glutathione- S-transferase, glutathione-peroxidase, superoxide dismutase, glutathione reductase). The results suggest that CNA protects myotubes from H 2 O 2 -induced atrophy by inhibiting/resisting the amendments in proteolytic systems and maintains cellular redox-balance.
Collapse
Affiliation(s)
- Nirmaljeet Kaur
- Skeletal Muscle Lab, University College, Kurukshetra University, Kurukshetra, India
| | - Prachi Gupta
- Skeletal Muscle Lab, University College, Kurukshetra University, Kurukshetra, India
| | - Vikram Saini
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep Sherawat
- Skeletal Muscle Lab, University College, Kurukshetra University, Kurukshetra, India
| | - Sanjeev Gupta
- Skeletal Muscle Lab, University College, Kurukshetra University, Kurukshetra, India
| | - Anita Dua
- Skeletal Muscle Lab, University College, Kurukshetra University, Kurukshetra, India
| | - Vinod Kumar
- Department of Chemistry, M.M. University, Ambala, India
| | - Elisha Injeti
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, Ohio
| | - Ashwani Mittal
- Skeletal Muscle Lab, University College, Kurukshetra University, Kurukshetra, India
| |
Collapse
|
47
|
Song Y, Dahl M, Leavitt W, Alvord J, Bradford CY, Albertine KH, Pillow JJ. Vitamin A Protects the Preterm Lamb Diaphragm Against Adverse Effects of Mechanical Ventilation. Front Physiol 2018; 9:1119. [PMID: 30150942 PMCID: PMC6099107 DOI: 10.3389/fphys.2018.01119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/25/2018] [Indexed: 11/13/2022] Open
Abstract
Background: Preterm infants are deficient in vitamin A, which is essential for growth and development of the diaphragm. Preterm infants often require mechanical ventilation (MV) for respiratory distress. In adults, MV is associated with the development of ventilation-induced diaphragm dysfunction and difficulty weaning from the ventilator. We assessed the impact of MV on the preterm diaphragm and the protective effect of vitamin A during MV. Methods: Preterm lambs delivered operatively at ∼131 days gestation (full gestation: 150 days) received respiratory support by synchronized intermittent mandatory ventilation for 3 days. Lambs in the treated group received daily (24 h) enteral doses of 2500 IU/kg/day vitamin A combined with 250 IU/kg/day retinoic acid (VARA) during MV, while MV control lambs received saline. Unventilated fetal reference lambs were euthanized at birth, without being allowed to breathe. The fetal diaphragm was collected to quantify mRNA levels of myosin heavy chain (MHC) isoforms, atrophy genes, antioxidant genes, and pro-inflammatory genes; to determine ubiquitin proteasome pathway activity; to measure the abundance of protein carbonyl, and to investigate metabolic signaling. Results: Postnatal MV significantly decreased expression level of the neonatal MHC gene but increased expression level of MHC IIx mRNA level (p < 0.05). Proteasome activity increased after 3 days MV, accompanied by increased MuRF1 mRNA level and accumulated protein carbonyl abundance. VARA supplementation decreased proteasome activity and FOXO1 signaling, down-regulated MuRF1 expression, and reduced reactive oxidant production. Conclusion: These findings suggest that 3 days of MV results in abnormal myofibrillar composition, activation of the proteolytic pathway, and oxidative injury of diaphragms in mechanically ventilated preterm lambs. Daily enteral VARA protects the preterm diaphragm from these adverse effects.
Collapse
Affiliation(s)
- Yong Song
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia.,Centre for Neonatal Research and Education, Division of Paediatrics and Child Health, Medical School, The University of Western Australia, Crawley, WA, Australia.,School of Public Health, Curtin University, Bentley, WA, Australia.,Centre for Genetic Origins of Health and Disease, The University of Western Australia, Curtin University, Crawley, WA, Australia
| | - MarJanna Dahl
- Department of Pediatrics, University of Utah, Salt Lake City, UT, United States
| | - Wendy Leavitt
- Department of Pediatrics, University of Utah, Salt Lake City, UT, United States
| | - Jeremy Alvord
- Department of Pediatrics, University of Utah, Salt Lake City, UT, United States
| | - Calan Y Bradford
- Department of Pediatrics, University of Utah, Salt Lake City, UT, United States
| | - Kurt H Albertine
- Department of Pediatrics, University of Utah, Salt Lake City, UT, United States
| | - J Jane Pillow
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia.,Centre for Neonatal Research and Education, Division of Paediatrics and Child Health, Medical School, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
48
|
Lee J, Mun S, Park A, Kim D, Heun Cha B, Kang HG. Bicalutamide enhances fodrin-mediated apoptosis through calpain in LNCaP. Exp Biol Med (Maywood) 2018; 243:843-851. [PMID: 29860890 DOI: 10.1177/1535370218779780] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Prostate cancer is the most common cancer in men, and before it progresses and metastasizes, the anticancer drug bicalutamide is often administered to patients. Many cases of androgen-dependent prostate cancer develop resistance during treatment with bicalutamide. Therefore, the effect of bicalutamide on androgen-dependent LNCaP prostate cancer cells is of clinical interest. The aim of this study was to demonstrate the effects of the anticancer drug bicalutamide on LNCaP prostate cancer cells by using a proteomics approach. Based on the results, 314 proteins were differentially expressed between the LNCaP and LNCaP treated with bicalutamide. The apoptosis pathway associated with differentially expressed proteins was shown in the Kyoto Encyclopedia of Gene and Genome pathway mapper. The Kyoto Encyclopedia of Gene and Genome pathway mapper results revealed that the fodrin-mediated apoptosis pathway is associated with the actions of bicalutamide and Western blotting was performed to validate these results. Impact statement We studied bicalutamide's anticancer action by using proteomics. The effect of bicalutamide on androgen-exposed LNCaP cells was also studied. KEGG identified >1.8-fold differentially expressed proteins between test group cells. KEGG mapper showed fodrin-mediated apoptosis involvement in bicalutamide's action. The anticancer effects of bicalutamide, which was further confirmed using Western blotting. Therefore, this drug is a potential candidate for understanding bicalutamide's effect on LNCaP and fodrin can be used as a biomarker monitoring status in metastatic carcinoma.
Collapse
Affiliation(s)
- Jiyeong Lee
- 1 Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam 13135, Korea
| | - Sora Mun
- 2 Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea
| | - Arum Park
- 2 Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea
| | - Doojin Kim
- 1 Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam 13135, Korea
| | - Byung Heun Cha
- 1 Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam 13135, Korea
| | - Hee-Gyoo Kang
- 1 Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam 13135, Korea.,2 Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea
| |
Collapse
|
49
|
Wang D, Wei L, Yang Y, Liu H. Dietary supplementation with ketoacids protects against CKD-induced oxidative damage and mitochondrial dysfunction in skeletal muscle of 5/6 nephrectomised rats. Skelet Muscle 2018; 8:18. [PMID: 29855350 PMCID: PMC5984473 DOI: 10.1186/s13395-018-0164-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 05/16/2018] [Indexed: 02/07/2023] Open
Abstract
Background A low-protein diet supplemented with ketoacids (LPD + KA) maintains the nutritional status of patients with chronic kidney disease (CKD). Oxidative damage and mitochondrial dysfunction associated with the upregulation of p66SHC and FoxO3a have been shown to contribute to muscle atrophy. This study aimed to determine whether LPD + KA improves muscle atrophy and attenuates the oxidative stress and mitochondrial damage observed in CKD rats. Methods 5/6 nephrectomy rats were randomly divided into three groups and fed with either 22% protein (normal-protein diet; NPD), 6% protein (low-protein diets; LPD) or 5% protein plus 1% ketoacids (LPD + KA) for 24 weeks. Sham-operated rats with NPD intake were used as the control. Results KA supplementation improved muscle atrophy and function in CKD + LPD rats. It also reduced the upregulation of genes related to the ubiquitin-proteasome system and 26S proteasome activity, as well as protein and mitochondrial oxidative damage in the muscles of CKD + LPD rats. Moreover, KA supplementation prevented the drastic decrease in activities of mitochondrial electron transport chain complexes, mitochondrial respiration, and content in the muscles of CKD + LPD rats. Furthermore, KA supplementation reversed the elevation in p66Shc and FoxO3a expression in the muscles of CKD + LPD rats. Conclusions Our results showed that KA supplementation to be beneficial to muscle atrophy in CKD + LPD, which might be associated with improvement of oxidative damage and mitochondrial dysfunction through suppression of p66Shc and FoxO3a.
Collapse
Affiliation(s)
- Dongtao Wang
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, Guangdong, China. .,Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine, Shenzhen, 518033, Guangdong, China. .,Department of Nephrology, Ruikang Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, 530011, Guangxi, China.
| | - Lianbo Wei
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, Guangdong, China
| | - Yajun Yang
- Department of Pharmacology, Guangdong Key Laboratory for R&D of Natural Drug, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Huan Liu
- Department of Nephrology, Ruikang Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, 530011, Guangxi, China
| |
Collapse
|
50
|
Bolus DJ, Shanmugam G, Narasimhan M, Rajasekaran NS. Recurrent heat shock impairs the proliferation and differentiation of C2C12 myoblasts. Cell Stress Chaperones 2018; 23:399-410. [PMID: 29063376 PMCID: PMC5904084 DOI: 10.1007/s12192-017-0851-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 10/05/2017] [Accepted: 10/07/2017] [Indexed: 12/21/2022] Open
Abstract
Heat-related illness and injury are becoming a growing safety concern for the farmers, construction workers, miners, firefighters, manufacturing workers, and other outdoor workforces who are exposed to heat stress in their routine lives. A primary response by a cell to an acute heat shock (HS) exposure is the induction of heat-shock proteins (HSPs), which chaperone and facilitate cellular protein folding and remodeling processes. While acute HS is well studied, the effect of repeated bouts of hyperthermia and the sustained production of HSPs in the myoblast-myotube model system of C2C12 cells are poorly characterized. In C2C12 myoblasts, we found that robust HS (43 °C, dose/time) significantly decreased the proliferation by 50% as early as on day 1 and maintained at the same level on days 2 and 3 of HS. This was accompanied by an accumulation of cells at G2 phase with reduced cell number in G1 phase indicating cell cycle arrest. FACS analysis indicates that there was no apparent change in apoptosis (markers) and cell death upon repeated HS. Immunoblot analysis and qPCR demonstrated a significant increase in the baseline expression of HSP25, 70, and 90 (among others) in cells after a single HS (43 °C) for 60 min as a typical HS response. Importantly, the repeated HS for 60 min each on days 2 and 3 maintained the elevated levels of HSPs compared to the control cells. Further, the continuous HS exposure resulted in significant inhibition of the differentiation of C2C12 myocytes to myotubes and only 1/10th of the cells underwent differentiation in HS relative to control. This was associated with significantly higher levels of HSPs and reduced expression of myogenin and Myh2 (P < 0.05), the genes involved in the differentiation process. Finally, the cell migration (scratch) assay indicated that the wound closure was significantly delayed in HS cells relative to the control cells. Overall, these results suggest that a repeated HS may perturb the active process of proliferation, motility, and differentiation processes in an in vitro murine myoblast-myotube model.
Collapse
Affiliation(s)
- Daniel J Bolus
- Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, 35294-2180, USA
| | - Gobinath Shanmugam
- Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, 35294-2180, USA
| | - Madhusudhanan Narasimhan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| | - Namakkal S Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, 35294-2180, USA.
- Division of Cardiovascular Medicine, Department of Medicine, The University of Utah School of Medicine, Salt Lake City, UT, 84132, USA.
- Center for Free Radical Biology, The University of Alabama at Birmingham, Birmingham, AL, 35294-2180, USA.
| |
Collapse
|