51
|
Damal Villivalam S, Ebert SM, Lim HW, Kim J, You D, Jung BC, Palacios HH, Tcheau T, Adams CM, Kang S. A necessary role of DNMT3A in endurance exercise by suppressing ALDH1L1-mediated oxidative stress. EMBO J 2021; 40:e106491. [PMID: 33847380 DOI: 10.15252/embj.2020106491] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 12/25/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
Exercise can alter the skeletal muscle DNA methylome, yet little is known about the role of the DNA methylation machinery in exercise capacity. Here, we show that DNMT3A expression in oxidative red muscle increases greatly following a bout of endurance exercise. Muscle-specific Dnmt3a knockout mice have reduced tolerance to endurance exercise, accompanied by reduction in oxidative capacity and mitochondrial respiration. Moreover, Dnmt3a-deficient muscle overproduces reactive oxygen species (ROS), the major contributors to muscle dysfunction. Mechanistically, we show that DNMT3A suppresses the Aldh1l1 transcription by binding to its promoter region, altering its epigenetic profile. Forced expression of ALDH1L1 elevates NADPH levels, which results in overproduction of ROS by the action of NADPH oxidase complex, ultimately resulting in mitochondrial defects in myotubes. Thus, inhibition of ALDH1L1 pathway can rescue oxidative stress and mitochondrial dysfunction from Dnmt3a deficiency in myotubes. Finally, we show that in vivo knockdown of Aldh1l1 largely rescues exercise intolerance in Dnmt3a-deficient mice. Together, we establish that DNMT3A in skeletal muscle plays a pivotal role in endurance exercise by controlling intracellular oxidative stress.
Collapse
Affiliation(s)
- Sneha Damal Villivalam
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Scott M Ebert
- Departments of Internal Medicine and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA.,Emmyon, Inc., Coralville, IA, USA
| | - Hee Woong Lim
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics & Biomedical Informatics, University of Cincinnati, Cincinnati, OH, USA
| | - Jinse Kim
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Dongjoo You
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Byung Chul Jung
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Hector H Palacios
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Tabitha Tcheau
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Christopher M Adams
- Departments of Internal Medicine and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA.,Emmyon, Inc., Coralville, IA, USA.,Iowa City Department of Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Sona Kang
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| |
Collapse
|
52
|
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: 52] [Impact Index Per Article: 13.0] [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
|
53
|
Beneficial Role of Exercise in the Modulation of mdx Muscle Plastic Remodeling and Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10040558. [PMID: 33916762 PMCID: PMC8066278 DOI: 10.3390/antiox10040558] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/02/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive progressive lethal disorder caused by the lack of dystrophin, which determines myofibers mechanical instability, oxidative stress, inflammation, and susceptibility to contraction-induced injuries. Unfortunately, at present, there is no efficient therapy for DMD. Beyond several promising gene- and stem cells-based strategies under investigation, physical activity may represent a valid noninvasive therapeutic approach to slow down the progression of the pathology. However, ethical issues, the limited number of studies in humans and the lack of consistency of the investigated training interventions generate loss of consensus regarding their efficacy, leaving exercise prescription still questionable. By an accurate analysis of data about the effects of different protocol of exercise on muscles of mdx mice, the most widely-used pre-clinical model for DMD research, we found that low intensity exercise, especially in the form of low speed treadmill running, likely represents the most suitable exercise modality associated to beneficial effects on mdx muscle. This protocol of training reduces muscle oxidative stress, inflammation, and fibrosis process, and enhances muscle functionality, muscle regeneration, and hypertrophy. These conclusions can guide the design of appropriate studies on human, thereby providing new insights to translational therapeutic application of exercise to DMD patients.
Collapse
|
54
|
Bouviere J, Fortunato RS, Dupuy C, Werneck-de-Castro JP, Carvalho DP, Louzada RA. Exercise-Stimulated ROS Sensitive Signaling Pathways in Skeletal Muscle. Antioxidants (Basel) 2021; 10:antiox10040537. [PMID: 33808211 PMCID: PMC8066165 DOI: 10.3390/antiox10040537] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/16/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
Physical exercise represents a major challenge to whole-body homeostasis, provoking acute and adaptative responses at the cellular and systemic levels. Different sources of reactive oxygen species (ROS) have been described in skeletal muscle (e.g., NADPH oxidases, xanthine oxidase, and mitochondria) and are closely related to the physiological changes induced by physical exercise through the modulation of several signaling pathways. Many signaling pathways that are regulated by exercise-induced ROS generation, such as adenosine monophosphate-activated protein kinase (AMPK), mitogen activated protein kinase (MAPK), nuclear respiratory factor2 (NRF2), and PGC-1α are involved in skeletal muscle responses to physical exercise, such as increased glucose uptake, mitochondriogenesis, and hypertrophy, among others. Most of these adaptations are blunted by antioxidants, revealing the crucial role played by ROS during and after physical exercise. When ROS generation is either insufficient or exacerbated, ROS-mediated signaling is disrupted, as well as physical exercise adaptations. Thus, an understanding the limit between "ROS that can promote beneficial effects" and "ROS that can promote harmful effects" is a challenging question in exercise biology. The identification of new mediators that cause reductive stress and thereby disrupt exercise-stimulated ROS signaling is a trending on this topic and are covered in this current review.
Collapse
Affiliation(s)
- Jessica Bouviere
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
| | - Rodrigo S. Fortunato
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
| | - Corinne Dupuy
- Université Paris-Saclay, UMR 9019CNRS, Gustave Roussy, 94800 Villejuif, France;
| | - Joao Pedro Werneck-de-Castro
- Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Denise P. Carvalho
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
| | - Ruy A. Louzada
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
- Université Paris-Saclay, UMR 9019CNRS, Gustave Roussy, 94800 Villejuif, France;
- Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Correspondence:
| |
Collapse
|
55
|
Skeletal muscle redox signaling in rheumatoid arthritis. Clin Sci (Lond) 2021; 134:2835-2850. [PMID: 33146370 PMCID: PMC7642299 DOI: 10.1042/cs20190728] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by synovitis and the presence of serum autoantibodies. In addition, skeletal muscle weakness is a common comorbidity that contributes to inability to work and reduced quality of life. Loss in muscle mass cannot alone account for the muscle weakness induced by RA, but instead intramuscular dysfunction appears as a critical factor underlying the decreased force generating capacity for patients afflicted by arthritis. Oxidative stress and associated oxidative post-translational modifications have been shown to contribute to RA-induced muscle weakness in animal models of arthritis and patients with RA. However, it is still unclear how and which sources of reactive oxygen and nitrogen species (ROS/RNS) that are involved in the oxidative stress that drives the progression toward decreased muscle function in RA. Nevertheless, mitochondria, NADPH oxidases (NOX), nitric oxide synthases (NOS) and phospholipases (PLA) have all been associated with increased ROS/RNS production in RA-induced muscle weakness. In this review, we aim to cover potential ROS sources and underlying mechanisms of oxidative stress and loss of force production in RA. We also addressed the use of antioxidants and exercise as potential tools to counteract oxidative stress and skeletal muscle weakness.
Collapse
|
56
|
Mangner N, Garbade J, Heyne E, van den Berg M, Winzer EB, Hommel J, Sandri M, Jozwiak-Nozdrzykowska J, Meyer AL, Lehmann S, Schmitz C, Malfatti E, Schwarzer M, Ottenheijm CAC, Bowen TS, Linke A, Adams V. Molecular Mechanisms of Diaphragm Myopathy in Humans With Severe Heart Failure. Circ Res 2021; 128:706-719. [PMID: 33535772 DOI: 10.1161/circresaha.120.318060] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Norman Mangner
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Jens Garbade
- Department of Cardiac Surgery (J.G., S.L.), Heart Center Leipzig - University Hospital, Germany
| | - Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Germany (E.H., M.S.)
| | | | - Ephraim B Winzer
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Jennifer Hommel
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Marcus Sandri
- Department of Cardiology (M.S., J.J.-N.), Heart Center Leipzig - University Hospital, Germany
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Germany (E.H., M.S.)
| | | | - Anna L Meyer
- Cardiac Surgery, Heart and Marfan Center, University of Heidelberg, Germany (A.L.M.)
| | - Sven Lehmann
- Department of Cardiac Surgery (J.G., S.L.), Heart Center Leipzig - University Hospital, Germany
| | - Clara Schmitz
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
| | - Edoardo Malfatti
- Neurology, Centre de Référence Maladies Neuromusculaires Nord-Est-Ile-de-France, CHU Raymond-Poincaré, Garches, France (E.M.). U1179 UVSQ-INSERM, Université Versailles-Saint-Quentin-en-Yvelines, France
| | | | - Coen A C Ottenheijm
- Physiology, Amsterdam UMC (location VUmc), the Netherlands (M.v.d.B., C.A.C.O.)
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, United Kingdom (T.S.B.)
| | - Axel Linke
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany (A.L., V.A.)
| | - Volker Adams
- Department of Internal Medicine and Cardiology (N.M., E.B.W., J.H., C.S., A.L. V.A.), Herzzentrum Dresden, Technische Universität Dresden, Germany
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany (A.L., V.A.)
| |
Collapse
|
57
|
Lund M, Andersen KG, Heaton R, Hargreaves IP, Gregersen N, Olsen RKJ. Bezafibrate activation of PPAR drives disturbances in mitochondrial redox bioenergetics and decreases the viability of cells from patients with VLCAD deficiency. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166100. [PMID: 33549744 DOI: 10.1016/j.bbadis.2021.166100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/11/2021] [Accepted: 02/01/2021] [Indexed: 10/22/2022]
Abstract
Very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency is the most common inborn long-chain fatty acid oxidation (FAO) disorder. VLCAD deficiency is characterized by distinct phenotypes. The severe phenotypes are potentially life-threatening and affect the heart or liver, with a comparatively milder phenotype characterized by myopathic symptoms. There is an unmet clinical need for effective treatment options for the myopathic phenotype. The molecular mechanisms driving the gradual decrease in mitochondrial function and associated alterations of muscle fibers are unclear. The peroxisome proliferator-activated receptor (PPAR) pan-agonist bezafibrate is a potent modulator of FAO and multiple other mitochondrial functions and has been proposed as a potential medication for myopathic cases of long-chain FAO disorders. In vitro experiments have demonstrated the ability of bezafibrate to increase VLCAD expression and activity. However, the outcome of small-scale clinical trials has been controversial. We found VLCAD deficient patient fibroblasts to have an increased oxidative stress burden and deranged mitochondrial bioenergetic capacity, compared to controls. Applying heat stress under fasting conditions to bezafibrate pretreated patient cells, caused a marked further increase of mitochondrial superoxide levels. Patient cells failed to maintain levels of the essential thiol peptide antioxidant glutathione and experienced a decrease in cellular viability. Our findings indicate that chronic PPAR activation is a plausible initiator of long-term pathogenesis in VLCAD deficiency. Our findings further implicate disruption of redox homeostasis as a key pathogenic mechanism in VLCAD deficiency and support the notion that a deranged thiol metabolism might be an important pathogenic factor in VLCAD deficiency.
Collapse
Affiliation(s)
- Martin Lund
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Kathrine G Andersen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Robert Heaton
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Iain P Hargreaves
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Niels Gregersen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Rikke K J Olsen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark.
| |
Collapse
|
58
|
Park C, Lee H, Hong S, Molagoda IMN, Jeong JW, Jin CY, Kim GY, Choi SH, Hong SH, Choi YH. Inhibition of Lipopolysaccharide-Induced Inflammatory and Oxidative Responses by Trans-cinnamaldehyde in C2C12 Myoblasts. Int J Med Sci 2021; 18:2480-2492. [PMID: 34104079 PMCID: PMC8176176 DOI: 10.7150/ijms.59169] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
Background: Trans-cinnamaldehyde (tCA), a bioactive component found in Cinnamomum cassia, has been reported to exhibit anti-inflammatory and antioxidant effects, but its efficacy in muscle cells has yet to be found. In this study, we investigated the inhibitory effect of tCA on inflammatory and oxidative stress induced by lipopolysaccharide (LPS) in C2C12 mouse skeletal myoblasts. Methods: To investigate the anti-inflammatory and antioxidant effects of tCA in LPS-treated C2C12 cells, we measured the levels of pro-inflammatory mediator, cytokines, and reactive oxygen species (ROS). To elucidate the mechanism underlying the effect of tCA, the expression of genes involved in the expression of inflammatory and oxidative regulators was also investigated. We further evaluated the anti-inflammatory and antioxidant efficacy of tCA against LPS in the zebrafish model. Results: tCA significantly inhibited the LPS-induced release of pro-inflammatory mediators and cytokines, which was associated with decreased expression of their regulatory genes. tCA also suppressed the expression of Toll-like receptor 4 (TLR4) and myeloid differentiation factor, and attenuated the nuclear translocation of nuclear factor-kappa B (NF-κB) and the binding of LPS to TLR4 on the cell surface in LPS-treated C2C12 cells. Furthermore, tCA abolished LPS-induced generation of ROS and expression levels of ROS producing enzymes, NADPH oxidase 1 (NOX1) and NOX2. However, tCA enhanced the activation of nuclear translocation of nuclear factor-E2-related factor 2 (Nrf2) and the expression of heme oxygenase-1 (HO-1) in LPS-stimulated C2C12 myoblasts. In addition, tCA showed strong protective effects against NO and ROS production in LPS-injected zebrafish larvae. Conclusions: Our findings suggest that tCA exerts its inhibitory ability against LPS-induced inflammatory and antioxidant stress in C2C12 myoblasts by targeting the TLR4/NF-κB, which might be mediated by the NOXs and Nrf2/HO-1 pathways.
Collapse
Affiliation(s)
- Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong-Eui University, Busan 47340, Republic of Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Suhyun Hong
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Ilandarage Menu Neelaka Molagoda
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Jin-Woo Jeong
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Republic of Korea
| | - Cheng-Yun Jin
- School of Pharmaceutical Sciences, Zhengzhou University, Henan 450001, China
| | - Gi-Young Kim
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Sung Hyun Choi
- Department of System Management, Korea Lift College, Geochang 50141, Republic of Korea
| | - Sang Hoon Hong
- Department of Internal Medicine, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| |
Collapse
|
59
|
ABCG1 Attenuates Oxidative Stress Induced by H 2O 2 through the Inhibition of NADPH Oxidase and the Upregulation of Nrf2-Mediated Antioxidant Defense in Endothelial Cells. ACTA ACUST UNITED AC 2020; 2020:2095645. [PMID: 33344146 PMCID: PMC7732382 DOI: 10.1155/2020/2095645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/23/2020] [Indexed: 11/18/2022]
Abstract
Summary. Oxidative stress is an important factor that is related to endothelial dysfunction. ATP-binding cassette transporter G1 (ABCG1), a regulator of intracellular cholesterol efflux, has been found to prevent endothelial activation in vessel walls. To explore the role of ABCG1 in oxidative stress production in endothelial cells, HUAECs were exposed to H2O2 and transfected with the specific ABCG1 siRNA or ABCG1 overexpression plasmid. The results showed that overexpression of ABCG1 by ABCG1 plasmid or liver X receptor (LXR) agonist T0901317 treatment inhibited ROS production and MDA content induced by H2O2 in HUAECs. Furthermore, ABCG1 upregulation blunted the activity of prooxidant NADPH oxidase and the expression of Nox4, one of the NADPH oxidase subunits. Moreover, the increased migration of Nrf2 from the cytoplasm to the nucleus and antioxidant HO-1 expression were detected in HUAECs with upregulation of ABCG1. Conversely, ABCG1 downregulation by ABCG1 siRNA increased NADPH oxidase activity and Nox4 expression and abrogated the increase at Nrf2 nuclear protein levels. In addition, intracellular cholesterol load interfered with the balance between NADPH oxidase activity and HO-1 expression. It was suggested that ABCG1 attenuated oxidative stress induced by H2O2 in endothelial cells, which might be involved in the balance between decreased NADPH oxidase activity and increased Nrf2/OH-1 antioxidant defense signaling via its regulation for intracellular cholesterol accumulation.
Collapse
|
60
|
ACE2, angiotensin 1-7 and skeletal muscle: review in the era of COVID-19. Clin Sci (Lond) 2020; 134:3047-3062. [PMID: 33231620 PMCID: PMC7687025 DOI: 10.1042/cs20200486] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Angiotensin converting enzyme-2 (ACE2) is a multifunctional transmembrane protein recently recognised as the entry receptor of the virus causing COVID-19. In the renin–angiotensin system (RAS), ACE2 cleaves angiotensin II (Ang II) into angiotensin 1-7 (Ang 1-7), which is considered to exert cellular responses to counteract the activation of the RAS primarily through a receptor, Mas, in multiple organs including skeletal muscle. Previous studies have provided abundant evidence suggesting that Ang 1-7 modulates multiple signalling pathways leading to protection from pathological muscle remodelling and muscle insulin resistance. In contrast, there is relatively little evidence to support the protective role of ACE2 in skeletal muscle. The potential contribution of endogenous ACE2 to the regulation of Ang 1-7-mediated protection of these muscle pathologies is discussed in this review. Recent studies have suggested that ACE2 protects against ageing-associated muscle wasting (sarcopenia) through its function to modulate molecules outside of the RAS. Thus, the potential association of sarcopenia with ACE2 and the associated molecules outside of RAS is also presented herein. Further, we introduce the transcriptional regulation of muscle ACE2 by drugs or exercise, and briefly discuss the potential role of ACE2 in the development of COVID-19.
Collapse
|
61
|
Wei X, Franke J, Ost M, Wardelmann K, Börno S, Timmermann B, Meierhofer D, Kleinridders A, Klaus S, Stricker S. Cell autonomous requirement of neurofibromin (Nf1) for postnatal muscle hypertrophic growth and metabolic homeostasis. J Cachexia Sarcopenia Muscle 2020; 11:1758-1778. [PMID: 33078583 PMCID: PMC7749575 DOI: 10.1002/jcsm.12632] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/09/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is a multi-organ disease caused by mutations in neurofibromin 1 (NF1). Amongst other features, NF1 patients frequently show reduced muscle mass and strength, impairing patients' mobility and increasing the risk of fall. The role of Nf1 in muscle and the cause for the NF1-associated myopathy are mostly unknown. METHODS To dissect the function of Nf1 in muscle, we created muscle-specific knockout mouse models for NF1, inactivating Nf1 in the prenatal myogenic lineage either under the Lbx1 promoter or under the Myf5 promoter. Mice were analysed during prenatal and postnatal myogenesis and muscle growth. RESULTS Nf1Lbx1 and Nf1Myf5 animals showed only mild defects in prenatal myogenesis. Nf1Lbx1 animals were perinatally lethal, while Nf1Myf5 animals survived only up to approximately 25 weeks. A comprehensive phenotypic characterization of Nf1Myf5 animals showed decreased postnatal growth, reduced muscle size, and fast fibre atrophy. Proteome and transcriptome analyses of muscle tissue indicated decreased protein synthesis and increased proteasomal degradation, and decreased glycolytic and increased oxidative activity in muscle tissue. High-resolution respirometry confirmed enhanced oxidative metabolism in Nf1Myf5 muscles, which was concomitant to a fibre type shift from type 2B to type 2A and type 1. Moreover, Nf1Myf5 muscles showed hallmarks of decreased activation of mTORC1 and increased expression of atrogenes. Remarkably, loss of Nf1 promoted a robust activation of AMPK with a gene expression profile indicative of increased fatty acid catabolism. Additionally, we observed a strong induction of genes encoding catabolic cytokines in muscle Nf1Myf5 animals, in line with a drastic reduction of white, but not brown adipose tissue. CONCLUSIONS Our results demonstrate a cell autonomous role for Nf1 in myogenic cells during postnatal muscle growth required for metabolic and proteostatic homeostasis. Furthermore, Nf1 deficiency in muscle drives cross-tissue communication and mobilization of lipid reserves.
Collapse
Affiliation(s)
- Xiaoyan Wei
- Musculoskeletal Development and Regeneration Group, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Development and Disease Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Julia Franke
- Musculoskeletal Development and Regeneration Group, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Development and Disease Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Mario Ost
- Department of Physiology of Energy Metabolism, German Institute for Human Nutrition, Nuthetal, Germany.,Department of Neuropathology, University Hospital Leipzig, Leipzig, Germany
| | - Kristina Wardelmann
- Junior Research Group Central Regulation of Metabolism, German Institute for Human Nutrition, Nuthetal, Germany.,Institute of Nutritional Science, Department of Molecular and Experimental Nutritional Medicine, University of Potsdam, Potsdam, Germany
| | - Stefan Börno
- Sequencing Core Unit, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Bernd Timmermann
- Sequencing Core Unit, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - David Meierhofer
- Mass Spectrometry Core Unit, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Andre Kleinridders
- Junior Research Group Central Regulation of Metabolism, German Institute for Human Nutrition, Nuthetal, Germany.,Institute of Nutritional Science, Department of Molecular and Experimental Nutritional Medicine, University of Potsdam, Potsdam, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Susanne Klaus
- Department of Physiology of Energy Metabolism, German Institute for Human Nutrition, Nuthetal, Germany.,Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Sigmar Stricker
- Musculoskeletal Development and Regeneration Group, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Development and Disease Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
62
|
The Universal Soldier: Enzymatic and Non-Enzymatic Antioxidant Functions of Serum Albumin. Antioxidants (Basel) 2020; 9:antiox9100966. [PMID: 33050223 PMCID: PMC7601824 DOI: 10.3390/antiox9100966] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
As a carrier of many biologically active compounds, blood is exposed to oxidants to a greater extent than the intracellular environment. Serum albumin plays a key role in antioxidant defence under both normal and oxidative stress conditions. This review evaluates data published in the literature and from our own research on the mechanisms of the enzymatic and non-enzymatic activities of albumin that determine its participation in redox modulation of plasma and intercellular fluid. For the first time, the results of numerous clinical, biochemical, spectroscopic and computational experiments devoted to the study of allosteric modulation of the functional properties of the protein associated with its participation in antioxidant defence are analysed. It has been concluded that it is fundamentally possible to regulate the antioxidant properties of albumin with various ligands, and the binding and/or enzymatic features of the protein by changing its redox status. The perspectives for using the antioxidant properties of albumin in practice are discussed.
Collapse
|
63
|
Antioxidants in Sport Sarcopenia. Nutrients 2020; 12:nu12092869. [PMID: 32961753 PMCID: PMC7551250 DOI: 10.3390/nu12092869] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/12/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022] Open
Abstract
The decline of skeletal muscle mass and strength that leads to sarcopenia is a pathology that might represent an emergency healthcare issue in future years. Decreased muscle mass is also a condition that mainly affects master athletes involved in endurance physical activities. Skeletal muscles respond to exercise by reshaping the biochemical, morphological, and physiological state of myofibrils. Adaptive responses involve the activation of intracellular signaling pathways and genetic reprogramming, causing alterations in contractile properties, metabolic status, and muscle mass. One of the mechanisms leading to sarcopenia is an increase in reactive oxygen and nitrogen species levels and a reduction in enzymatic antioxidant protection. The present review shows the recent experimental models of sarcopenia that explore molecular mechanisms. Furthermore, the clinical aspect of sport sarcopenia will be highlighted, and new strategies based on nutritional supplements, which may contribute to reducing indices of oxidative stress by reinforcing natural endogenous protection, will be suggested.
Collapse
|
64
|
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
|
65
|
Powers SK, Deminice R, Ozdemir M, Yoshihara T, Bomkamp MP, Hyatt H. Exercise-induced oxidative stress: Friend or foe? JOURNAL OF SPORT AND HEALTH SCIENCE 2020; 9:415-425. [PMID: 32380253 PMCID: PMC7498668 DOI: 10.1016/j.jshs.2020.04.001] [Citation(s) in RCA: 321] [Impact Index Per Article: 64.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/21/2020] [Accepted: 02/18/2020] [Indexed: 05/02/2023]
Abstract
The first report demonstrating that prolonged endurance exercise promotes oxidative stress in humans was published more than 4 decades ago. Since this discovery, many ensuing investigations have corroborated the fact that muscular exercise increases the production of reactive oxygen species (ROS) and results in oxidative stress in numerous tissues including blood and skeletal muscles. Although several tissues may contribute to exercise-induced ROS production, it is predicted that muscular contractions stimulate ROS production in active muscle fibers and that skeletal muscle is a primary source of ROS production during exercise. This contraction-induced ROS generation is associated with (1) oxidant damage in several tissues (e.g., increased protein oxidation and lipid peroxidation), (2) accelerated muscle fatigue, and (3) activation of biochemical signaling pathways that contribute to exercise-induced adaptation in the contracting muscle fibers. While our understanding of exercise and oxidative stress has advanced rapidly during the last decades, questions remain about whether exercise-induced increases in ROS production are beneficial or harmful to health. This review addresses this issue by discussing the site(s) of oxidant production during exercise and detailing the health consequences of exercise-induced ROS production.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32608, USA
| | - Rafael Deminice
- Department of Physical Education, State University of Londrina, Londrina, 10011, Brazil
| | - Mustafa Ozdemir
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32608, USA; Department of Exercise and Sport Sciences, Hacettepe University, Ankara, 06800, Turkey.
| | - Toshinori Yoshihara
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32608, USA; Department of Exercise Physiology, Juntendo University, Tokyo, 270-1695, Japan
| | - Matthew P Bomkamp
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32608, USA
| | - Hayden Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32608, USA
| |
Collapse
|
66
|
Mason SA, Trewin AJ, Parker L, Wadley GD. Antioxidant supplements and endurance exercise: Current evidence and mechanistic insights. Redox Biol 2020; 35:101471. [PMID: 32127289 PMCID: PMC7284926 DOI: 10.1016/j.redox.2020.101471] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/11/2020] [Accepted: 02/17/2020] [Indexed: 01/07/2023] Open
Abstract
Antioxidant supplements are commonly consumed by endurance athletes to minimize exercise-induced oxidative stress, with the intention of enhancing recovery and improving performance. There are numerous commercially available nutritional supplements that are targeted to athletes and health enthusiasts that allegedly possess antioxidant properties. However, most of these compounds are poorly investigated with respect to their in vivo redox activity and efficacy in humans. Therefore, this review will firstly provide a background to endurance exercise-related redox signalling and the subsequent adaptations in skeletal muscle and vascular function. The review will then discuss commonly available compounds with purported antioxidant effects for use by athletes. N-acetyl cysteine may be of benefit over the days prior to an endurance event; while chronic intake of combined 1000 mg vitamin C + vitamin E is not recommended during periods of heavy training associated with adaptations in skeletal muscle. Melatonin, vitamin E and α-lipoic acid appear effective at decreasing markers of exercise-induced oxidative stress. However, evidence on their effects on endurance performance are either lacking or not supportive. Catechins, anthocyanins, coenzyme Q10 and vitamin C may improve vascular function, however, evidence is either limited to specific sub-populations and/or does not translate to improved performance. Finally, additional research should clarify the potential benefits of curcumin in improving muscle recovery post intensive exercise; and the potential hampering effects of astaxanthin, selenium and vitamin A on skeletal muscle adaptations to endurance training. Overall, we highlight the lack of supportive evidence for most antioxidant compounds to recommend to athletes.
Collapse
Affiliation(s)
- Shaun A Mason
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Adam J Trewin
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Lewan Parker
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Glenn D Wadley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia.
| |
Collapse
|
67
|
Grotle AK, Macefield VG, Farquhar WB, O'Leary DS, Stone AJ. Recent advances in exercise pressor reflex function in health and disease. Auton Neurosci 2020; 228:102698. [PMID: 32861944 DOI: 10.1016/j.autneu.2020.102698] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 01/11/2023]
Abstract
Autonomic alterations at the onset of exercise are critical to redistribute cardiac output towards the contracting muscles while preventing a fall in arterial pressure due to excessive vasodilation within the contracting muscles. Neural mechanisms responsible for these adjustments include central command, the exercise pressor reflex, and arterial and cardiopulmonary baroreflexes. The exercise pressor reflex evokes reflex increases in sympathetic activity to the heart and systemic vessels and decreases in parasympathetic activity to the heart, which increases blood pressure (BP), heart rate, and total peripheral resistance through vasoconstriction of systemic vessels. In this review, we discuss recent advancements in our understanding of exercise pressor reflex function in health and disease. Specifically, we discuss emerging evidence suggesting that sympathetic vasoconstrictor drive to the contracting and non-contracting skeletal muscle is differentially controlled by central command and the metaboreflex in healthy conditions. Further, we discuss evidence from animal and human studies showing that cardiovascular diseases, including hypertension, diabetes, and heart failure, lead to an altered exercise pressor reflex function. We also provide an update on the mechanisms thought to underlie this altered exercise pressor reflex function in each of these diseases. Although these mechanisms are complex, multifactorial, and dependent on the etiology of the disease, there is a clear consensus that several mechanisms are involved. Ultimately, approaches targeting these mechanisms are clinically significant as they provide alternative therapeutic strategies to prevent adverse cardiovascular events while also reducing symptoms of exercise intolerance.
Collapse
Affiliation(s)
- Ann-Katrin Grotle
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, United States of America
| | | | - William B Farquhar
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, United States of America
| | - Donal S O'Leary
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Audrey J Stone
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, United States of America.
| |
Collapse
|
68
|
Yu T, Dohl J, Wang L, Chen Y, Gasier HG, Deuster PA. Curcumin Ameliorates Heat-Induced Injury through NADPH Oxidase-Dependent Redox Signaling and Mitochondrial Preservation in C2C12 Myoblasts and Mouse Skeletal Muscle. J Nutr 2020; 150:2257-2267. [PMID: 32692359 PMCID: PMC7919340 DOI: 10.1093/jn/nxaa201] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/13/2020] [Accepted: 06/22/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and the mitochondrial electron transport chain are the primary sources of reactive oxygen species (ROS). Previous studies have shown that severe heat exposure damages mitochondria and causes excessive mitochondrial ROS production that contributes to the pathogenesis of heat-related illnesses. OBJECTIVES We tested whether the antioxidant curcumin could protect against heat-induced mitochondrial dysfunction and skeletal muscle injury, and characterized the possible mechanism. METHODS Mouse C2C12 myoblasts and rat flexor digitorum brevis (FDB) myofibers were treated with 5 μM curcumin; adult male C57BL/6J mice received daily curcumin (15, 50, or 100 mg/kg body weight) by gavage for 10 consecutive days. We compared ROS levels and mitochondrial morphology and function between treatment and nontreatment groups under unheated or heat conditions, and investigated the upstream mechanism and the downstream effect of curcumin-regulated ROS production. RESULTS In C2C12 myoblasts, curcumin prevented heat-induced mitochondrial fragmentation, ROS overproduction, and apoptosis (all P < 0.05). Curcumin treatment for 2 and 4 h at 37°C induced increases in ROS levels by 42% and 59% (dihydroethidium-derived fluorescence), accompanied by increases in NADPH oxidase protein expression by 24% and 32%, respectively (all P < 0.01). In curcumin-treated cells, chemical inhibition and genetic knockdown of NADPH oxidase restored ROS to levels similar to those of controls, indicating NADPH oxidase mediates curcumin-stimulated ROS production. Moreover, curcumin induced ROS-dependent shifting of the mitochondrial fission-fusion balance toward fusion, and increases in mitochondrial mass by 143% and membrane potential by 30% (both P < 0.01). In rat FDB myofibers and mouse gastrocnemius muscles, curcumin preserved mitochondrial morphology and function during heat stress, and prevented heat-induced mitochondrial ROS overproduction and tissue injury (all P < 0.05). CONCLUSIONS Curcumin regulates ROS hormesis favoring mitochondrial fusion/elongation, biogenesis, and improved function in rodent skeletal muscle. Curcumin may be an effective therapeutic target for heat-related illness and other mitochondrial diseases.
Collapse
Affiliation(s)
| | - Jacob Dohl
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA,Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Li Wang
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yifan Chen
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Heath G Gasier
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA,Department of Anesthesiology, Center for Hyperbaric Medicine & Environmental Physiology, Duke University School of Medicine, Durham, NC, USA
| | - Patricia A Deuster
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| |
Collapse
|
69
|
Sokolov SS, Severin FF. Manipulating Cellular Energetics to Slow Aging of Tissues and Organs. BIOCHEMISTRY (MOSCOW) 2020; 85:651-659. [PMID: 32586228 DOI: 10.1134/s0006297920060024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Up to now numerous studies in the field of gerontology have been published. Nevertheless, a well-known food restriction remains the most reliable and efficient way of lifespan extension. Physical activity is also a well-documented anti-aging intervention being especially efficient in slowing down the age-associated decline of skeletal muscle mass. In this review we focus on the molecular mechanisms of the effect of physical exercise on muscle tissues. We also discuss the possibilities of pharmacological extension of this effect to the rest of the tissues. During the exercise, the level of ATP decreases triggering activation of AMP-dependent protein kinase (AMPK). This kinase stimulates antioxidant potential of the cells and their mitochondrial respiratory capacity. The exercise also induces mild oxidative stress, which, in turn, mediates the stimulation via hormetic response. Furthermore, during the exercise cells generate activators of mammalian target of rapamycin (mTOR). The intracellular ATP level increases during the rest periods between exercises thus promoting mTOR activation. Therefore, regular exercise intermittently activates anti-oxidant defenses and mitochondrial biogenesis (via AMPK and the hormetic response) of the muscle tissue, as well as its proliferative potential (via mTOR), which, in turn, impedes the age-dependent muscle atrophy. Thus, the intermittent treatment with activators of (i) AMPK combined with the inducers of hormetic response and of (ii) mTOR might partly mimic the effects of physical exercise. Importantly, pharmacological activation of AMPK takes place in the absence of ATP level decrease. The use of uncouplers of respiration and oxidative phosphorylation at the phase of AMPK activation could also prevent negative consequences of the cellular hyper-energization. It is believed that the decline of both antioxidant and proliferative potentials of the cells causes the age-dependent decline of multiple tissues, rather than only the muscular one. We argue that the approach above is applicable for the majority of tissues in an organism.
Collapse
Affiliation(s)
- S S Sokolov
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, 119991, Russia
| | - F F Severin
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, 119991, Russia.
| |
Collapse
|
70
|
Coccurello R, Volonté C. P2X7 Receptor in the Management of Energy Homeostasis: Implications for Obesity, Dyslipidemia, and Insulin Resistance. Front Endocrinol (Lausanne) 2020; 11:199. [PMID: 32528404 PMCID: PMC7247848 DOI: 10.3389/fendo.2020.00199] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/19/2020] [Indexed: 12/15/2022] Open
Abstract
Whole-body energy metabolism entails the highly regulated balance between food intake, nutrient breakdown, energy generation (ATP), and energy storage for the preservation of vital functions and body mass. Purinergic signaling has attracted increasing attention in the regulatory mechanisms not only for the reverse processes of white adipose tissue lipogenesis and lipolysis, but also for brown adipocyte-dependent thermogenesis and leptin production. This regulatory role has remarkable implications in the handling of body's energy expenditure and energy reservoir. Hence, selected purinergic receptors can play a relevant function in lipid metabolism, endocrine activity, glucose uptake, ATP-dependent increased expression of uncoupling protein 1, and browning of adipose tissue. Indeed, purinergic P2 receptors regulate adipogenesis and lipid metabolism and are involved in adipogenic differentiation. In particular, the ionotropic ATP-activated P2X7 subtype is involved in fat distribution, as well as in the modulation of inflammatory pathways in white adipose tissue. Within this context, very recent evidence has established a direct function of P2X7 in energy metabolism. Specifically, either genetic deletion (P2X7 knockout mice) or subchronic pharmacological inhibition of the receptor produces a decrease of whole-body energy expenditure and, concurrently, an increase of carbohydrate oxidation. As further evidence, lipid accumulation, increased fat mass distribution, and weight gain are reported in P2X7-depleted mice. Conversely, the stimulation of P2X7 enhances energy expenditure. Altogether, this knowledge supports the role of P2X7 signaling in the fight against obesity and insulin resistance, as well as in the promotion of adaptive thermogenesis.
Collapse
Affiliation(s)
- Roberto Coccurello
- Institute for Complex System (ISC), National Research Council (CNR), Rome, Italy
- Preclinical Neuroscience, European Center for Brain Research (CERC)/IRCCS Santa Lucia Foundation, Rome, Italy
| | - Cinzia Volonté
- Preclinical Neuroscience, European Center for Brain Research (CERC)/IRCCS Santa Lucia Foundation, Rome, Italy
- Institute for Systems Analysis and Computer Science, National Research Council (CNR), Rome, Italy
| |
Collapse
|
71
|
Eccardt AM, Pelzel RJ, Mattathil L, Moon YA, Mannino MH, Janowiak BE, Fisher JS. A peroxidase mimetic protects skeletal muscle cells from peroxide challenge and stimulates insulin signaling. Am J Physiol Cell Physiol 2020; 318:C1214-C1225. [PMID: 32348172 DOI: 10.1152/ajpcell.00167.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Reactive oxygen species such as hydrogen peroxide have been implicated in causing metabolic dysfunction such as insulin resistance. Heme groups, either by themselves or when incorporated into proteins, have been shown to scavenge peroxide and demonstrate protective effects in various cell types. Thus, we hypothesized that a metalloporphyrin similar in structure to heme, Fe(III)tetrakis(4-benzoic acid)porphyrin (FeTBAP), would be a peroxidase mimetic that could defend cells against oxidative stress. After demonstrating that FeTBAP has peroxidase activity with reduced nicotinamide adenine dinucleotide phosphate (NADPH) and NADH as reducing substrates, we determined that FeTBAP partially rescued C2C12 myotubes from peroxide-induced insulin resistance as measured by phosphorylation of AKT (S473) and insulin receptor substrate 1 (IRS-1, Y612). Furthermore, we found that FeTBAP stimulates insulin signaling in myotubes and mouse soleus skeletal muscle to about the same level as insulin for phosphorylation of AKT, IRS-1, and glycogen synthase kinase 3β (S9). We found that FeTBAP lowers intracellular peroxide levels and protects against carbonyl formation in myotubes exposed to peroxide. Additionally, we found that FeTBAP stimulates glucose transport in myotubes and skeletal muscle to about the same level as insulin. We conclude that a peroxidase mimetic can blunt peroxide-induced insulin resistance and also stimulate insulin signaling and glucose transport, suggesting a possible role of peroxidase activity in regulation of insulin signaling.
Collapse
Affiliation(s)
- Amanda M Eccardt
- Department of Biology, Saint Louis University, St. Louis, Missouri
| | - Ross J Pelzel
- Department of Biology, Saint Louis University, St. Louis, Missouri
| | - Lyn Mattathil
- Department of Biology, Saint Louis University, St. Louis, Missouri
| | - Yerin A Moon
- Department of Biology, Saint Louis University, St. Louis, Missouri
| | - Mark H Mannino
- Department of Biology, Saint Louis University, St. Louis, Missouri
| | | | | |
Collapse
|
72
|
Osório Alves J, Matta Pereira L, Cabral Coutinho do Rêgo Monteiro I, Pontes dos Santos LH, Soares Marreiros Ferraz A, Carneiro Loureiro AC, Calado Lima C, Leal-Cardoso JH, Pires Carvalho D, Soares Fortunato R, Marilande Ceccatto V. Strenuous Acute Exercise Induces Slow and Fast Twitch-Dependent NADPH Oxidase Expression in Rat Skeletal Muscle. Antioxidants (Basel) 2020; 9:antiox9010057. [PMID: 31936265 PMCID: PMC7022445 DOI: 10.3390/antiox9010057] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/13/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
The enzymatic complex Nicotinamide Adenine Dinucleotide Phosphate (NADPH) oxidase (NOx) may be the principal source of reactive oxygen species (ROS). The NOX2 and NOX4 isoforms are tissue-dependent and are differentially expressed in slow-twitch fibers (type I fibers) and fast-twitch fibers (type II fibers) of skeletal muscle, making them different markers of ROS metabolism induced by physical exercise. The aim of this study was to investigate NOx signaling, as a non-adaptive and non-cumulative response, in the predominant fiber types of rat skeletal muscles 24 h after one strenuous treadmill exercise session. The levels of mRNA, reduced glycogen, thiol content, NOx, superoxide dismutase, catalase, glutathione peroxidase activity, and PPARGC1α and SLC2A4 gene expression were measured in the white gastrocnemius (WG) portion, the red gastrocnemius (RG) portion, and the soleus muscle (SOL). NOx activity showed higher values in the SOL muscle compared to the RG and WG portions. The same was true of the NOX2 and NOX4 mRNA levels, antioxidant enzymatic activities, glycogen content. Twenty-four hours after the strenuous exercise session, NOx expression increased in slow-twitch oxidative fibers. The acute strenuous exercise condition showed an attenuation of oxidative stress and an upregulation of antioxidant activity through PPARGC1α gene activity, antioxidant defense adaptations, and differential gene expression according to the predominant fiber type. The most prominent location of detoxification (indicated by NOX4 activation) in the slow-twitch oxidative SOL muscle was the mitochondria, while the fast-twitch oxidative RG portion showed a more cytosolic location. Glycolytic metabolism in the WG portion suggested possible NOX2/NOX4 non-regulation, indicating other possible ROS regulation pathways.
Collapse
Affiliation(s)
- Juliana Osório Alves
- Laboratório de Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza 60.714-903, Brazil; (J.O.A.); (L.H.P.d.S.); (A.C.C.L.)
| | - Leonardo Matta Pereira
- Laboratório de Fisiologia e Sinalização redox, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.M.P.); (I.C.C.d.R.M.); (R.S.F.)
| | - Igor Cabral Coutinho do Rêgo Monteiro
- Laboratório de Fisiologia e Sinalização redox, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.M.P.); (I.C.C.d.R.M.); (R.S.F.)
| | - Luiz Henrique Pontes dos Santos
- Laboratório de Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza 60.714-903, Brazil; (J.O.A.); (L.H.P.d.S.); (A.C.C.L.)
| | | | - Adriano Cesar Carneiro Loureiro
- Laboratório de Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza 60.714-903, Brazil; (J.O.A.); (L.H.P.d.S.); (A.C.C.L.)
| | - Crystianne Calado Lima
- Laboratório de Eletrofisiologia Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza 60.714-903, Brazil; (C.C.L.); (J.H.L.-C.)
| | - José Henrique Leal-Cardoso
- Laboratório de Eletrofisiologia Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza 60.714-903, Brazil; (C.C.L.); (J.H.L.-C.)
| | - Denise Pires Carvalho
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Rodrigo Soares Fortunato
- Laboratório de Fisiologia e Sinalização redox, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.M.P.); (I.C.C.d.R.M.); (R.S.F.)
| | - Vânia Marilande Ceccatto
- Laboratório de Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza 60.714-903, Brazil; (J.O.A.); (L.H.P.d.S.); (A.C.C.L.)
- Correspondence:
| |
Collapse
|
73
|
Qi J, Luo X, Ma Z, Zhang B, Li S, Duan X, Yang B, Zhang J. Swimming Exercise Protects against Insulin Resistance via Regulating Oxidative Stress through Nox4 and AKT Signaling in High-Fat Diet-Fed Mice. J Diabetes Res 2020; 2020:2521590. [PMID: 32051831 PMCID: PMC6995488 DOI: 10.1155/2020/2521590] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 12/26/2019] [Indexed: 12/31/2022] Open
Abstract
Nonpharmaceutical therapies such as exercise training and diet intervention are widely used for the treatment of insulin resistance (IR). Although the skeletal muscle is the major peripheral tissue of glucose metabolism under insulin stimulation, the mechanism underlying muscle IR is poorly understood. Using a high-fat diet-induced IR mouse model, we here show that NADPH oxidase 4 (Nox4) upregulation mediates the production of reactive oxygen species (ROS) that causes metabolic syndrome featuring IR. The Nox4 expression level was markedly elevated in IR mice, and Nox4 overexpression was sufficient to trigger IR. Conversely, downregulation of Nox4 expression through exercise training prevented diet-induced IR by reducing the production of ROS and enhancing the AKT signaling pathway. Thus, this study indicates that exercise might improve IR through a reduction of Nox4-induced ROS in the skeletal muscle and enhancement of AKT signal transduction.
Collapse
Affiliation(s)
- Jie Qi
- College of Physical Education, Shanghai Normal University, Shanghai 200234, China
| | - Xue Luo
- Medical College of Yangzhou Polytechnic College, Jiangsu 225009, China
| | - Zhichao Ma
- The School of Physical Education, Wuhan Business University, Hubei 430056, China
| | - Bo Zhang
- College of Physical Education, Shanghai Normal University, Shanghai 200234, China
| | - Shuyan Li
- College of Physical Education, Yangzhou University, Jiangsu 225009, China
| | - Xuyang Duan
- College of Physical Education, Shanghai Normal University, Shanghai 200234, China
| | - Bo Yang
- College of Public Health and Management, Wenzhou Medical University, Zhejiang 325000, China
| | - Jun Zhang
- College of Physical Education, Shanghai Normal University, Shanghai 200234, China
| |
Collapse
|
74
|
Henríquez-Olguín C, Boronat S, Cabello-Verrugio C, Jaimovich E, Hidalgo E, Jensen TE. The Emerging Roles of Nicotinamide Adenine Dinucleotide Phosphate Oxidase 2 in Skeletal Muscle Redox Signaling and Metabolism. Antioxid Redox Signal 2019; 31:1371-1410. [PMID: 31588777 PMCID: PMC6859696 DOI: 10.1089/ars.2018.7678] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Skeletal muscle is a crucial tissue to whole-body locomotion and metabolic health. Reactive oxygen species (ROS) have emerged as intracellular messengers participating in both physiological and pathological adaptations in skeletal muscle. A complex interplay between ROS-producing enzymes and antioxidant networks exists in different subcellular compartments of mature skeletal muscle. Recent evidence suggests that nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are a major source of contraction- and insulin-stimulated oxidants production, but they may paradoxically also contribute to muscle insulin resistance and atrophy. Recent Advances: Pharmacological and molecular biological tools, including redox-sensitive probes and transgenic mouse models, have generated novel insights into compartmentalized redox signaling and suggested that NOX2 contributes to redox control of skeletal muscle metabolism. Critical Issues: Major outstanding questions in skeletal muscle include where NOX2 activation occurs under different conditions in health and disease, how NOX2 activation is regulated, how superoxide/hydrogen peroxide generated by NOX2 reaches the cytosol, what the signaling mediators are downstream of NOX2, and the role of NOX2 for different physiological and pathophysiological processes. Future Directions: Future research should utilize and expand the current redox-signaling toolbox to clarify the NOX2-dependent mechanisms in skeletal muscle and determine whether the proposed functions of NOX2 in cells and animal models are conserved into humans.
Collapse
Affiliation(s)
- Carlos Henríquez-Olguín
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark.,Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Susanna Boronat
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Elena Hidalgo
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
75
|
Grotle AK, Stone AJ. Exaggerated exercise pressor reflex in type 2 diabetes: Potential role of oxidative stress. Auton Neurosci 2019; 222:102591. [PMID: 31669797 PMCID: PMC6858935 DOI: 10.1016/j.autneu.2019.102591] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes mellitus (T2DM) leads to exaggerated cardiovascular responses to exercise, in part due to an exaggerated exercise pressor reflex. Accumulating data suggest excessive oxidative stress contributes to an exaggerated exercise pressor reflex in cardiovascular-related diseases. Excessive oxidative stress is also a primary underlying mechanism for the development and progression of T2DM. However, whether oxidative stress plays a role in mediating the exaggerated exercise pressor reflex in T2DM is not known. Therefore, this review explores the potential role of oxidative stress leading to increased activation of the afferent arm of the exercise pressor reflex. Several lines of evidence support direct and indirect effects of oxidative stress on the exercise pressor reflex. For example, intramuscular ROS may directly and indirectly (by attenuating contracting muscle blood flow) increase group III and IV afferent activity. Oxidative stress is a primary underlying mechanism for the development of neuropathic pain, which in turn is associated with increased group III and IV afferent activity. These are the same type of afferents that evoke muscle pain and the exercise pressor reflex. Furthermore, oxidative stress-induced release of inflammatory mediators may modulate afferent activity. Collectively, these alterations may result in a positive feedback loop that further amplifies the exercise pressor reflex. An exaggerated reflex increases the risk of adverse cardiovascular events. Thus, identifying the contribution of oxidative stress could provide a potential therapeutic target to reduce this risk in T2DM.
Collapse
Affiliation(s)
- Ann-Katrin Grotle
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX 78712, United States of America
| | - Audrey J Stone
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX 78712, United States of America.
| |
Collapse
|
76
|
Torma F, Gombos Z, Jokai M, Takeda M, Mimura T, Radak Z. High intensity interval training and molecular adaptive response of skeletal muscle. SPORTS MEDICINE AND HEALTH SCIENCE 2019; 1:24-32. [PMID: 35782463 PMCID: PMC9219277 DOI: 10.1016/j.smhs.2019.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Increased cardiovascular fitness, V˙O2max, is associated with enhanced endurance capacity and a decreased rate of mortality. High intensity interval training (HIIT) is one of the best methods to increase V˙O2max and endurance capacity for top athletes and for the general public as well. Because of the high intensity of this type of training, the adaptive response is not restricted to Type I fibers, as found for moderate intensity exercise of long duration. Even with a short exercise duration, HIIT can induce activation of AMPK, PGC-1α, SIRT1 and ROS pathway as well as by the modulation of Ca2+ homeostasis, leading to enhanced mitochondrial biogenesis, and angiogenesis. The present review summarizes the current knowledge of the adaptive response of HIIT.
Collapse
Affiliation(s)
- Ferenc Torma
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Zoltan Gombos
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Matyas Jokai
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Masaki Takeda
- Faculty of Health and Sports Science, Doshisha University, Kyotanabe, Japan
| | - Tatsuya Mimura
- Faculty of Sport and Health Sciences, Osaka Sangyo University, Osaka, Japan
| | - Zsolt Radak
- Research Center of Molecular Exercise Science, University of Physical Education, Budapest, Hungary
- Corresponding author. Alkotas u. 44, Budapest, H-1123, Hungary.
| |
Collapse
|
77
|
Increased Circulating Levels of Interleukin-6 Affect the Redox Balance in Skeletal Muscle. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3018584. [PMID: 31827671 PMCID: PMC6881749 DOI: 10.1155/2019/3018584] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/01/2019] [Accepted: 09/26/2019] [Indexed: 12/25/2022]
Abstract
The extent of oxidative stress and chronic inflammation are closely related events which coexist in a muscle environment under pathologic conditions. It has been generally accepted that the inflammatory cells, as well as myofibers, are sources of reactive species which are, in turn, able to amplify the activation of proinflammatory pathways. However, the precise mechanism underlining the physiopathologic interplay between ROS generation and inflammatory response has to be fully clarified. Thus, the identification of key molecular players in the interconnected pathogenic network between the two processes might help to design more specific therapeutic approaches for degenerative diseases. Here, we investigated whether elevated circulating levels of the proinflammatory cytokine Interleukin-6 (IL-6) are sufficient to perturb the physiologic redox balance in skeletal muscle, independently of tissue damage and inflammatory response. We observed that the overexpression of circulating IL-6 enhances the generation and accumulation of free radicals in the diaphragm muscle of adult NSE/IL-6 mice, by deregulating redox-associated molecular circuits and impinging the nuclear factor erythroid 2-related factor 2- (Nrf2-) mediated antioxidant response. Our findings are coherent with a model in which uncontrolled levels of IL-6 in the bloodstream can influence the local redox homeostasis, inducing the establishment of prooxidative conditions in skeletal muscle tissue.
Collapse
|
78
|
Scalabrin M, Pollock N, Staunton CA, Brooks SV, McArdle A, Jackson MJ, Vasilaki A. Redox responses in skeletal muscle following denervation. Redox Biol 2019; 26:101294. [PMID: 31450104 PMCID: PMC6831873 DOI: 10.1016/j.redox.2019.101294] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/01/2019] [Accepted: 08/07/2019] [Indexed: 10/30/2022] Open
Abstract
Previous studies have shown a significant increase in the mitochondrial generation of hydrogen peroxide (H2O2) and other peroxides in recently denervated muscle fibers. The mechanisms for generation of these peroxides and how the muscle responds to these peroxides are not fully established. The aim of this work was to determine the effect of denervation on the muscle content of proteins that may contribute to mitochondrial peroxide release and the muscle responses to this generation. Denervation of the tibialis anterior (TA) and extensor digitorum longus (EDL) muscles in mice was achieved by surgical removal of a small section of the peroneal nerve prior to its entry into the muscle. An increase in mitochondrial peroxide generation has been observed from 7 days and sustained up to 21 days following denervation in the TA muscle fibers. This increased peroxide generation was reduced by incubation of skinned fibers with inhibitors of monoamine oxidases, NADPH oxidases or phospholipase A2 enzymes and the muscle content of these enzymes together with peroxiredoxin 6 were increased following denervation. Denervated muscle also showed significant adaptations in the content of several enzymes involved in the protection of cells against oxidative damage. Morphological analyses indicated a progressive significant loss of muscle mass in the TA muscle from 7 days up to 21 days following denervation due to fiber atrophy but without fiber loss. These results support the possibility that, at least initially, the increase in peroxide production may stimulate adaptations in an attempt to protect the muscle fibers, but that these processes are insufficient and the increased peroxide generation over the longer term may activate degenerative and atrophic processes in the denervated muscle fibers.
Collapse
Affiliation(s)
- Mattia Scalabrin
- MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, UK
| | - Natalie Pollock
- MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, UK
| | - Caroline A Staunton
- MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, UK
| | - Susan V Brooks
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Anne McArdle
- MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, UK
| | - Malcolm J Jackson
- MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, UK
| | - Aphrodite Vasilaki
- MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, UK.
| |
Collapse
|
79
|
Damiano S, Muscariello E, La Rosa G, Di Maro M, Mondola P, Santillo M. Dual Role of Reactive Oxygen Species in Muscle Function: Can Antioxidant Dietary Supplements Counteract Age-Related Sarcopenia? Int J Mol Sci 2019; 20:ijms20153815. [PMID: 31387214 PMCID: PMC6696113 DOI: 10.3390/ijms20153815] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Sarcopenia is characterized by the progressive loss of skeletal muscle mass and strength. In older people, malnutrition and physical inactivity are often associated with sarcopenia, and, therefore, dietary interventions and exercise must be considered to prevent, delay, or treat it. Among the pathophysiological mechanisms leading to sarcopenia, a key role is played by an increase in reactive oxygen and nitrogen species (ROS/RNS) levels and a decrease in enzymatic antioxidant protection leading to oxidative stress. Many studies have evaluated, in addition to the effects of exercise, the effects of antioxidant dietary supplements in limiting age-related muscle mass and performance, but the data which have been reported are conflicting. In skeletal muscle, ROS/RNS have a dual function: at low levels they increase muscle force and adaptation to exercise, while at high levels they lead to a decline of muscle performance. Controversial results obtained with antioxidant supplementation in older persons could in part reflect the lack of univocal effects of ROS on muscle mass and function. The purpose of this review is to examine the molecular mechanisms underlying the dual effects of ROS in skeletal muscle function and the analysis of literature data on dietary antioxidant supplementation associated with exercise in normal and sarcopenic subjects.
Collapse
Affiliation(s)
- Simona Damiano
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II", Via S. Pansini, 5, 80131 Naples, Italy
| | - Espedita Muscariello
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II", Via S. Pansini, 5, 80131 Naples, Italy
| | - Giuliana La Rosa
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II", Via S. Pansini, 5, 80131 Naples, Italy
| | - Martina Di Maro
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II", Via S. Pansini, 5, 80131 Naples, Italy
| | - Paolo Mondola
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II", Via S. Pansini, 5, 80131 Naples, Italy
| | - Mariarosaria Santillo
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II", Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
80
|
Meza CA, La Favor JD, Kim DH, Hickner RC. Endothelial Dysfunction: Is There a Hyperglycemia-Induced Imbalance of NOX and NOS? Int J Mol Sci 2019; 20:ijms20153775. [PMID: 31382355 PMCID: PMC6696313 DOI: 10.3390/ijms20153775] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
NADPH oxidases (NOX) are enzyme complexes that have received much attention as key molecules in the development of vascular dysfunction. NOX have the primary function of generating reactive oxygen species (ROS), and are considered the main source of ROS production in endothelial cells. The endothelium is a thin monolayer that lines the inner surface of blood vessels, acting as a secretory organ to maintain homeostasis of blood flow. The enzymatic production of nitric oxide (NO) by endothelial NO synthase (eNOS) is critical in mediating endothelial function, and oxidative stress can cause dysregulation of eNOS and endothelial dysfunction. Insulin is a stimulus for increases in blood flow and endothelium-dependent vasodilation. However, cardiovascular disease and type 2 diabetes are characterized by poor control of the endothelial cell redox environment, with a shift toward overproduction of ROS by NOX. Studies in models of type 2 diabetes demonstrate that aberrant NOX activation contributes to uncoupling of eNOS and endothelial dysfunction. It is well-established that endothelial dysfunction precedes the onset of cardiovascular disease, therefore NOX are important molecular links between type 2 diabetes and vascular complications. The aim of the current review is to describe the normal, healthy physiological mechanisms involved in endothelial function, and highlight the central role of NOX in mediating endothelial dysfunction when glucose homeostasis is impaired.
Collapse
Affiliation(s)
- Cesar A Meza
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Justin D La Favor
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Do-Houn Kim
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Robert C Hickner
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA.
- Institute of Sports Sciences and Medicine, College of Human Sciences, Florida State University, Tallahassee, FL 32306, USA.
- Department of Biokinetics, Exercise and Leisure Sciences, School of Health Sciences, University of KwaZulu-Natal, Westville 4041, South Africa.
| |
Collapse
|
81
|
Yeo YH, Lai YC. Redox Regulation of Metabolic Syndrome: Recent Developments in Skeletal Muscle Insulin Resistance and Non-alcoholic Fatty Liver Disease (NAFLD). CURRENT OPINION IN PHYSIOLOGY 2019; 9:79-86. [PMID: 32818162 DOI: 10.1016/j.cophys.2019.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Several new discoveries over the past decade have shown that metabolic syndrome, a cluster of metabolic disorders, including increased visceral obesity, hyperglycemia, hypertension, dyslipidemia and low HDL-cholesterol, is commonly associated with skeletal muscle insulin resistance. More recently, non-alcoholic fatty liver disease (NAFLD) was recognized as an additional condition that is strongly associated with features of metabolic syndrome. While the pathogenesis of skeletal muscle insulin resistance and fatty liver is multifactorial, the role of dysregulated redox signaling has been clearly demonstrated in the regulation of skeletal muscle insulin resistance and NAFLD. In this review, we aim to provide recent updates on redox regulation with respect to (a) pro-oxidant enzymes (e.g. NAPDH oxidase and xanthine oxidase); (b) mitochondrial dysfunction; (c) endoplasmic reticulum (ER) stress; (d) iron metabolism derangements; and (e) gut-skeletal muscle or gut-liver connection in the development of skeletal muscle insulin resistance and NAFLD. Furthermore, we discuss promising new therapeutic strategies targeting redox regulation currently under investigation for the treatment of skeletal muscle insulin resistance and NAFLD.
Collapse
Affiliation(s)
- Yee-Hui Yeo
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, Palo Alto, California, USA
| | - Yen-Chun Lai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine; Indianapolis, IN, USA
| |
Collapse
|
82
|
Touyz RM, Anagnostopoulou A, Rios F, Montezano AC, Camargo LL. NOX5: Molecular biology and pathophysiology. Exp Physiol 2019; 104:605-616. [PMID: 30801870 PMCID: PMC6519284 DOI: 10.1113/ep086204] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
NEW FINDINGS What is the topic of this review? This review provides a comprehensive overview of Nox5 from basic biology to human disease and highlights unique features of this Nox isoform What advances does it highlight? Major advances in Nox5 biology relate to crystallization of the molecule and new insights into the pathophysiological role of Nox5. Recent discoveries have unravelled the crystal structure of Nox5, the first Nox isoform to be crystalized. This provides new opportunities to develop drugs or small molecules targeted to Nox5 in an isoform-specific manner, possibly for therapeutic use. Moreover genome wide association studies (GWAS) identified Nox5 as a new blood pressure-associated gene and studies in mice expressing human Nox5 in a cell-specific manner have provided new information about the (patho) physiological role of Nox5 in the cardiovascular system and kidneys. Nox5 seems to be important in the regulation of vascular contraction and kidney function. In cardiovascular disease and diabetic nephropathy, Nox5 activity is increased and this is associated with increased production of reactive oxygen species and oxidative stress implicated in tissue damage. ABSTRACT Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Nox), comprise seven family members (Nox1-Nox5 and dual oxidase 1 and 2) and are major producers of reactive oxygen species in mammalian cells. Reactive oxygen species are crucially involved in cell signalling and function. All Noxs share structural homology comprising six transmembrane domains with two haem-binding regions and an NADPH-binding region on the intracellular C-terminus, whereas their regulatory systems, mechanisms of activation and tissue distribution differ. This explains the diverse function of Noxs. Of the Noxs, NOX5 is unique in that rodents lack the gene, it is regulated by Ca2+ , it does not require NADPH oxidase subunits for its activation, and it is not glycosylated. NOX5 localizes in the perinuclear and endoplasmic reticulum regions of cells and traffics to the cell membrane upon activation. It is tightly regulated through numerous post-translational modifications and is activated by vasoactive agents, growth factors and pro-inflammatory cytokines. The exact pathophysiological significance of NOX5 remains unclear, but it seems to be important in the physiological regulation of sperm motility, vascular contraction and lymphocyte differentiation, and NOX5 hyperactivation has been implicated in cardiovascular disease, kidney injury and cancer. The field of NOX5 biology is still in its infancy, but with new insights into its biochemistry and cellular regulation, discovery of the NOX5 crystal structure and genome-wide association studies implicating NOX5 in disease, the time is now ripe to advance NOX5 research. This review provides a comprehensive overview of our current understanding of NOX5, from basic biology to human disease, and highlights the unique characteristics of this enigmatic Nox isoform.
Collapse
Affiliation(s)
- Rhian M. Touyz
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Aikaterini Anagnostopoulou
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Francisco Rios
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Augusto C. Montezano
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Livia L. Camargo
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| |
Collapse
|
83
|
Atractylenolide III Attenuates Muscle Wasting in Chronic Kidney Disease via the Oxidative Stress-Mediated PI3K/AKT/mTOR Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1875471. [PMID: 31178951 PMCID: PMC6501186 DOI: 10.1155/2019/1875471] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/11/2019] [Accepted: 03/13/2019] [Indexed: 01/08/2023]
Abstract
Oxidative stress contributes to muscle wasting in advanced chronic kidney disease (CKD) patients. Atractylenolide III (ATL-III), the major active constituent of Atractylodes rhizome, has been previously reported to function as an antioxidant. This study is aimed at investigating whether ATL-III has protective effects against CKD-induced muscle wasting by alleviating oxidative stress. The results showed that the levels of serum creatinine (SCr), blood urea nitrogen (BUN), and urinary protein significantly decreased in the ATL-III treatment group compared with the 5/6 nephrectomy (5/6 Nx) model group but were higher than those in the sham operation group. Skeletal muscle weight was increased, while inflammation was alleviated in the ATL-III administration group compared with the 5/6 Nx model group. ATL-III-treated rats also showed reduced dilation of the mitochondria, increased CAT, GSH-Px, and SOD activity, and decreased levels of MDA both in skeletal muscles and serum compared with 5/6 Nx model rats, suggesting that ATL-III alleviated mitochondrial damage and increased the activity of antioxidant enzymes, thus reducing the production of ROS. Furthermore, accumulated autophagosomes (APs) and autolysosomes (ALs) were reduced in the gastrocnemius (Gastroc) muscles of ATL-III-treated rats under transmission electron microscopy (TEM) together with the downregulation of LC3-II and upregulation of p62 according to Western blotting. This evidence indicated that ATL-III improved skeletal muscle atrophy and alleviated oxidative stress and autophagy in CKD rats. Furthermore, ATL-III could also increase the protein levels of p-PI3K, p-AKT, and p-mTOR in skeletal muscles in CKD rats. To further reveal the relevant mechanism, the oxidative stress-mediated PI3K/AKT/mTOR pathway was assessed, which showed that a reduced expression of p-PI3K, p-AKT, and p-mTOR in C2C12 myoblast atrophy induced by TNF-α could be upregulated by ATL-III; however, after the overexpression of Nox2 to increase ROS production, the attenuated effect was reversed. Our findings indicated that ATL-III is a potentially protective drug against muscle wasting via activation of the oxidative stress-mediated PI3K/AKT/mTOR pathway.
Collapse
|
84
|
NADPH Oxidase Isoforms Are Involved in Glucocorticoid-Induced Preosteoblast Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9192413. [PMID: 31049140 PMCID: PMC6458927 DOI: 10.1155/2019/9192413] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 12/17/2018] [Indexed: 01/01/2023]
Abstract
Oxidative stress induced by long-term glucocorticoid (GC) use weakens the repair capacity of bone tissue. Nicotinamide adenine dinucleotide phosphate, reduced form (NADPH) oxidase (NOX) is a superoxide-generating enzyme that plays an important role in regulating bone metabolism. To clarify the role of nonphagocytic NOX isoforms in osteoblast reactive oxygen species (ROS) generation and apoptosis, dexamethasone was used to establish a high-dose GC environment in vitro. A dose-dependent increase in intracellular ROS generation was demonstrated, which was accompanied by increased osteoblastic MC3T3-E1 cell apoptosis. Addition of the ROS inhibitor NAC (N-acetyl-L-cysteine) or NOX inhibitor DPI (diphenyleneiodonium) reversed this effect, indicating that NOX-derived ROS can induce osteoblast apoptosis under high-dose dexamethasone stimulation. NOX1, NOX2, and NOX4 are NOX homologs recently identified in bone tissue. To clarify the NOX isoforms that play a role in osteoblast ROS generation, Nox1, Nox2, and Nox4 mRNA expression and NOX2 and NOX4 protein expression were analyzed. Nox1 and Nox4 mRNA expression was elevated in a dose-dependent manner after culture in 100 nM, 250 nM, 500 nM, or 1000 nM dexamethasone, and the increased expression of NOX1 mRNA was more significant compared with NOX4 mRNA. Small interfering RNAs (siRNAs) were used to confirm the role of NOX1 and NOX4 in ROS generation. To clarify the signaling pathway in ROS-induced osteoblast apoptosis, mitogen-activated protein kinase (MAPK) signaling molecules were analyzed. Phosphorylated ASK1 and p38 levels were significantly higher in the 1000 nM dexamethasone group, which NAC or DPI markedly attenuated. However, the total mRNA and protein levels of ASK1 and p38 between the dexamethasone group and control were not significantly different. This is related to ROS regulating the posttranslational modification of ASK1 and p38 in MC3T3-E1 cell apoptosis. Altogether, NOX1- and NOX4-derived ROS plays a pivotal role in high-dose dexamethasone-induced preosteoblast apoptosis by increasing phosphorylated ASK1 and p38 and may be an important mechanism in steroid-induced avascular necrosis of the femoral head (SANFH).
Collapse
|
85
|
Venditti P, Reed TT, Victor VM, Di Meo S. Insulin resistance and diabetes in hyperthyroidism: a possible role for oxygen and nitrogen reactive species. Free Radic Res 2019; 53:248-268. [PMID: 30843740 DOI: 10.1080/10715762.2019.1590567] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In addition to insulin, glycemic control involves thyroid hormones. However, an excess of thyroid hormone can disturb the blood glucose equilibrium, leading to alterations of carbohydrate metabolism and, eventually, diabetes. Indeed, experimental and clinical hyperthyroidism is often accompanied by abnormal glucose tolerance. A common characteristic of hyperthyroidism and type 2 diabetes is the altered mitochondrial efficiency caused by the enhanced production of reactive oxygen and nitrogen species. It is known that an excess of thyroid hormone leads to increased oxidant production and mitochondrial oxidative damage. It can be hypothesised that these species represent the link between hyperthyroidism and development of insulin resistance and diabetes, even though direct evidence of this relationship is lacking. In this review, we examine the literature concerning the effects of insulin and thyroid hormones on glucose metabolism and discuss alterations of glucose metabolism in hyperthyroid conditions and the cellular and molecular mechanisms that may underline them.
Collapse
Affiliation(s)
- Paola Venditti
- a Dipartimento di Biologia , Università di Napoli Federico II , Napoli , Italy
| | - Tanea T Reed
- b Department of Chemistry , Eastern Kentucky University , Richmond , KY , USA
| | - Victor M Victor
- c Service of Endocrinology, Dr. Peset University Hospital, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO) , Valencia , Spain.,d Department of Physiology , University of Valencia , Valencia , Spain
| | - Sergio Di Meo
- a Dipartimento di Biologia , Università di Napoli Federico II , Napoli , Italy
| |
Collapse
|
86
|
Lassiter K, Kong BC, Piekarski-Welsher A, Dridi S, Bottje WG. Gene Expression Essential for Myostatin Signaling and Skeletal Muscle Development Is Associated With Divergent Feed Efficiency in Pedigree Male Broilers. Front Physiol 2019; 10:126. [PMID: 30873041 PMCID: PMC6401619 DOI: 10.3389/fphys.2019.00126] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/31/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Feed efficiency (FE, gain to feed) is an important genetic trait as 70% of the cost of raising animals is due to feed costs. The objective of this study was to determine mRNA expression of genes involved in muscle development and hypertrophy, and the insulin receptor-signaling pathway in breast muscle associated with the phenotypic expression of FE. Methods: Breast muscle samples were obtained from Pedigree Male (PedM) broilers (8 to 10 week old) that had been individually phenotyped for FE between 6 and 7 week of age. The high FE group gained more weight but consumed the same amount of feed compared to the low FE group. Total RNA was extracted from breast muscle (n = 6 per group) and mRNA expression of target genes was determined by real-time quantitative PCR. Results: Targeted gene expression analysis in breast muscle of the high FE phenotype revealed that muscle development may be fostered in the high FE PedM phenotype by down-regulation several components of the myostatin signaling pathway genes combined with upregulation of genes that enhance muscle formation and growth. There was also evidence of genetic architecture that would foster muscle protein synthesis in the high FE phenotype. A clear indication of differences in insulin signaling between high and low FE phenotypes was not apparent in this study. Conclusion: These findings indicate that a gene expression architecture is present in breast muscle of PedM broilers exhibiting high FE that would support enhanced muscle development-differentiation as well as protein synthesis compared to PedM broilers exhibiting low FE.
Collapse
Affiliation(s)
- Kentu Lassiter
- Department of Poultry Science, Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Byungwhi Caleb Kong
- Department of Poultry Science, Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | | | - Sami Dridi
- Department of Poultry Science, Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Walter Gay Bottje
- Department of Poultry Science, Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
87
|
Cobley JN, Sakellariou GK, Husi H, McDonagh B. Proteomic strategies to unravel age-related redox signalling defects in skeletal muscle. Free Radic Biol Med 2019; 132:24-32. [PMID: 30219702 DOI: 10.1016/j.freeradbiomed.2018.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/22/2018] [Accepted: 09/12/2018] [Indexed: 01/06/2023]
Abstract
Increased oxidative damage and disrupted redox signalling are consistently associated with age-related loss of skeletal muscle mass and function. Redox signalling can directly regulate biogenesis and degradation pathways and indirectly via activation of key transcription factors. Contracting skeletal muscle fibres endogenously generate free radicals (e.g. superoxide) and non-radical derivatives (e.g. hydrogen peroxide). Exercise induced redox signalling can promote beneficial adaptive responses that are disrupted by age-related redox changes. Identifying and quantifying the redox signalling pathways responsible for successful adaptation to exercise makes skeletal muscle an attractive physiological model for redox proteomic approaches. Site specific identification of the redox modification and quantification of site occupancy in the context of protein abundance remains a crucial concept for redox proteomics approaches. Notwithstanding, the technical limitations associated with skeletal muscle for proteomic analysis, we discuss current approaches for the identification and quantification of transient and stable redox modifications that have been employed to date in ageing research. We also discuss recent developments in proteomic approaches in skeletal muscle and potential implications and opportunities for investigating disrupted redox signalling in skeletal muscle ageing.
Collapse
Affiliation(s)
- James N Cobley
- Free Radical Laboratory, Departments of Diabetes and Cardiovascular Sciences, Centre for Health Sciences, University of the Highlands and Islands, Inverness IV2 3JH, UK
| | | | - Holger Husi
- Free Radical Laboratory, Departments of Diabetes and Cardiovascular Sciences, Centre for Health Sciences, University of the Highlands and Islands, Inverness IV2 3JH, UK
| | - Brian McDonagh
- Discipline of Physiology, School of Medicine, NUI Galway, Ireland.
| |
Collapse
|
88
|
Mortensen SP, Winding KM, Iepsen UW, Munch GW, Marcussen N, Hellsten Y, Pedersen BK, Baum O. The effect of two exercise modalities on skeletal muscle capillary ultrastructure in individuals with type 2 diabetes. Scand J Med Sci Sports 2019; 29:360-368. [PMID: 30480353 DOI: 10.1111/sms.13348] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/23/2018] [Indexed: 12/21/2022]
Abstract
Type 2 diabetes is associated with microvascular dysfunction, but little is known about how capillary ultrastructure is affected by exercise training. To investigate the effect of two types of exercise training on skeletal muscle capillary ultrastructure and capillarization in individuals with type 2 diabetes, 21 individuals with type 2 diabetes were allocated (randomized controlled trial) to 11 weeks of aerobic exercise training consisting of either moderate-intensity endurance training (END; n = 10) or low-volume high-intensity interval training (HIIT; n = 11). Skeletal muscle biopsies (m vastus lateralis) were obtained before and after the training intervention. At baseline, there was no difference in capillarization, capillary structure, and exercise hyperemia between the two groups. After the training intervention, capillary-to-fiber ratio increased by 8% ± 3% in the END group (P < 0.05) and was unchanged in the HIIT group with no difference between groups. Endothelium thickness increased (P < 0.05), basement membrane thickness decreased (P < 0.05), and the capillary lumen tended (P = 0.07) to increase in the END group, whereas these structural indicators were unchanged after HIIT. In contrast, skeletal muscle endothelial nitric oxide synthase (eNOS) increased after HIIT (P < 0.05), but not END, whereas there was no change in vascular endothelial growth factor (VEGF), superoxide dismutase (SOD)-2, or NADPH oxidase after both training protocols. In contrast to END training, HIIT did not alter capillarization or capillary structure in individuals with type 2 diabetes. In conclusion, HIIT appears to be a less effective strategy to treat capillary rarefaction and reduce basement thickening in type 2 diabetes.
Collapse
Affiliation(s)
- Stefan Peter Mortensen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Kamilla Munch Winding
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,The Danish Diabetes Academy, Odense, Denmark
| | - Ulrik Winning Iepsen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Gregers Winding Munch
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Niels Marcussen
- Institute of Pathology, Odense University Hospital, Odense, Denmark
| | - Ylva Hellsten
- Department of Nutrition, Exercise and Sport, University of Copenhagen, Copenhagen, Denmark
| | - Bente Klarlund Pedersen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Oliver Baum
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
89
|
Wong HS, Benoit B, Brand MD. Mitochondrial and cytosolic sources of hydrogen peroxide in resting C2C12 myoblasts. Free Radic Biol Med 2019; 130:140-150. [PMID: 30389498 DOI: 10.1016/j.freeradbiomed.2018.10.448] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 10/28/2022]
Abstract
The relative contributions of different mitochondrial and cytosolic sources of superoxide and hydrogen peroxide in cells are not well established because of a lack of suitable quantitative assays. To address this problem using resting C2C12 myoblasts we measured the effects of specific inhibitors that do not affect other pathways on the rate of appearance of hydrogen peroxide in the extracellular medium. We used inhibitors of NADPH oxidases (NOXs), suppressors of site IQ electron leak (S1QELs) at mitochondrial Complex I, and suppressors of site IIIQo electron leak (S3QELs) at mitochondrial Complex III. Around 40% of net cellular hydrogen peroxide release was from NOXs and approximately 45% was from the two mitochondrial sites; 30% from site IIIQo and 15% from site IQ. As expected, decreasing cytosolic antioxidant capacity by lowering glutathione levels increased the absolute rates from all sites without changing their proportions, whereas decreasing antioxidant defenses in the mitochondrial matrix increased only the absolute and relative contributions of the two mitochondrial sites. These results show directly that mitochondria are a major contributor to cytosolic hydrogen peroxide in resting C2C12 myoblasts, and provide the first direct evidence of superoxide/hydrogen peroxide production from site IQ in unstressed cells.
Collapse
Affiliation(s)
- Hoi-Shan Wong
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945, USA.
| | - Bérengère Benoit
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945, USA.
| | - Martin D Brand
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945, USA.
| |
Collapse
|
90
|
Carriker CR, Rombach P, Stevens BM, Vaughan RA, Gibson AL. Acute dietary nitrate supplementation does not attenuate oxidative stress or the hemodynamic response during submaximal exercise in hypobaric hypoxia. Appl Physiol Nutr Metab 2018; 43:1268-1274. [DOI: 10.1139/apnm-2017-0813] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The purpose of this study was to investigate changes in oxidative stress, arterial oxygen saturation (SaO2), blood pressure (BP), and heart rate (HR) during exercise in hypobaric hypoxia following acute dietary nitrate supplementation. Nine well-trained (maximal oxygen consumption, 60.8 ± 7.8 mL·kg−1·min−1) males (age, 29 ± 7 years) visited the laboratory on 3 occasions, each separated by 1 week. Visit 1 included a maximal aerobic cycling test and five 5-min increasing-intensity exercise bouts in a normobaric environment (1600 m). A single dose of either a nitrate-depleted placebo (PL) or a nitrate-rich beverage (NR; 12.8 mmol nitrate) was consumed 2.5 h prior to exercise during visits 2 and 3 (3500 m) in a double-blind, placebo-controlled, crossover study consisting of a 5-min cycling warm-up and 4 bouts, each 5 min in duration, separated by 4-min periods of passive rest. Exercise wattages were determined during visit 1 and corresponded to 25%, 40%, 50%, 60%, and 70% of normobaric maximal oxygen consumption. Catalase and 8-isoprostane were measured before and after exercise (immediately before and 1 h postexercise, respectively). NR increased plasma nitrite (1.53 ± 0.83 μmol·L−1) compared with PL (0.88 ± 0.56 μmol·L−1) (p < 0.05). In both conditions, postexercise (3500 m) 8-isoprostane (PL, 23.49 ± 3.38 to 60.90 ± 14.95 pg·mL−1; NR, 23.23 ± 4.12 to 52.11 ± 19.76 pg·mL−1) and catalase (PL, 63.89 ± 25.69 to 128.15 ± 41.80 mmol·min−1·mL−1; NR, 78.89 ± 30.95 to 109.96 ± 35.05 mmol·min−1·mL−1) were elevated compared with baseline resting values (p < 0.05). However, both 8-isoprostane and catalase were similar in the 2 groups (PL and NR) (p = 0.217 and p = 0.080, respectively). We concluded that an acute, pre-exercise dose of dietary nitrate yielded no beneficial changes in oxidative stress, SaO2, BP, or HR in healthy, aerobically fit men exercising at 3500 m.
Collapse
Affiliation(s)
- Colin R. Carriker
- Department of Exercise Science, High Point University, High Point, NC 27268, USA
- Department of Health, Exercise and Sports Sciences, University of New Mexico, Albuquerque, NM 87131, USA
| | - Paige Rombach
- Department of Exercise Science, High Point University, High Point, NC 27268, USA
| | - Brooke M. Stevens
- Department of Exercise Science, High Point University, High Point, NC 27268, USA
| | - Roger A. Vaughan
- Department of Exercise Science, High Point University, High Point, NC 27268, USA
| | - Ann L. Gibson
- Department of Health, Exercise and Sports Sciences, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
91
|
Sfyri PP, Yuldasheva NY, Tzimou A, Giallourou N, Crispi V, Aburima A, Beltran-Alvarez P, Patel K, Mougios V, Swann JR, Kearney MT, Matsakas A. Attenuation of oxidative stress-induced lesions in skeletal muscle in a mouse model of obesity-independent hyperlipidaemia and atherosclerosis through the inhibition of Nox2 activity. Free Radic Biol Med 2018; 129:504-519. [PMID: 30342191 DOI: 10.1016/j.freeradbiomed.2018.10.422] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 09/12/2018] [Accepted: 10/09/2018] [Indexed: 02/08/2023]
Abstract
Obesity leading to hyperlipidaemia and atherosclerosis is recognised to induce morphological and metabolic changes in many tissues. However, hyperlipidaemia can occur in the absence of obesity. The impact of the latter scenario on skeletal muscle and liver is not understood sufficiently. In this regard, we used the Apolipoprotein E-deficient (ApoE-/-) mouse model, an established model of hyperlipidaemia and atherosclerosis, that does not become obese when subjected to a high-fat diet, to determine the impact of Western-type diet (WD) and ApoE deficiency on skeletal muscle morphological, metabolic and biochemical properties. To establish the potential of therapeutic targets, we further examined the impact of Nox2 pharmacological inhibition on skeletal muscle redox biology. We found ectopic lipid accumulation in skeletal muscle and the liver, and altered skeletal muscle morphology and intramuscular triacylglycerol fatty acid composition. WD and ApoE deficiency had a detrimental impact in muscle metabolome, followed by perturbed gene expression for fatty acid uptake and oxidation. Importantly, there was enhanced oxidative stress in the skeletal muscle and development of liver steatosis, inflammation and oxidative protein modifications. Pharmacological inhibition of Nox2 decreased reactive oxygen species production and protein oxidative modifications in the muscle of ApoE-/- mice subjected to a Western-type diet. This study provides key evidence to better understand the pathophysiology of skeletal muscle in the context of hyperlipidaemia and atherosclerosis and identifies Nox2 as a potential target for attenuating oxidative stress in skeletal muscle in a mouse model of obesity-independent hyperlipidaemia.
Collapse
Affiliation(s)
- Pagona Panagiota Sfyri
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, United Kingdom
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom
| | - Anastasia Tzimou
- Laboratory of Evaluation of Human Biological Performance, School of Physical Education and Sports Science at Thessaloniki, Aristotle University of Thessaloniki, Greece
| | - Natasa Giallourou
- Department of Surgery and Cancer, Division of Computational and Systems Medicine, Imperial College London, United Kingdom
| | - Vassili Crispi
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, United Kingdom
| | - Ahmed Aburima
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, United Kingdom
| | | | - Ketan Patel
- School of Biological Sciences, University of Reading, United Kingdom
| | - Vassilis Mougios
- Laboratory of Evaluation of Human Biological Performance, School of Physical Education and Sports Science at Thessaloniki, Aristotle University of Thessaloniki, Greece
| | - Jonathan R Swann
- Department of Surgery and Cancer, Division of Computational and Systems Medicine, Imperial College London, United Kingdom
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, United Kingdom.
| |
Collapse
|
92
|
Sárközy M, Kovács ZZA, Kovács MG, Gáspár R, Szűcs G, Dux L. Mechanisms and Modulation of Oxidative/Nitrative Stress in Type 4 Cardio-Renal Syndrome and Renal Sarcopenia. Front Physiol 2018; 9:1648. [PMID: 30534079 PMCID: PMC6275322 DOI: 10.3389/fphys.2018.01648] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/31/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic kidney disease (CKD) is a public health problem and a recognized risk factor for cardiovascular diseases (CVD). CKD could amplify the progression of chronic heart failure leading to the development of type 4 cardio-renal syndrome (T4CRS). The severity and persistence of heart failure are strongly associated with mortality risk in T4CRS. CKD is also a catabolic state leading to renal sarcopenia which is characterized by the loss of skeletal muscle strength and physical function. Renal sarcopenia also promotes the development of CVD and increases the mortality in CKD patients. In turn, heart failure developed in T4CRS could result in chronic muscle hypoperfusion and metabolic disturbances leading to or aggravating the renal sarcopenia. The interplay of multiple factors (e.g., comorbidities, over-activated renin-angiotensin-aldosterone system [RAAS], sympathetic nervous system [SNS], oxidative/nitrative stress, inflammation, etc.) may result in the progression of T4CRS and renal sarcopenia. Among these factors, oxidative/nitrative stress plays a crucial role in the complex pathomechanism and interrelationship between T4CRS and renal sarcopenia. In the heart and skeletal muscle, mitochondria, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, uncoupled nitric oxide synthase (NOS) and xanthine oxidase are major ROS sources producing superoxide anion (O2·−) and/or hydrogen peroxide (H2O2). O2·− reacts with nitric oxide (NO) forming peroxynitrite (ONOO−) which is a highly reactive nitrogen species (RNS). High levels of ROS/RNS cause lipid peroxidation, DNA damage, interacts with both DNA repair enzymes and transcription factors, leads to the oxidation/nitration of key proteins involved in contractility, calcium handling, metabolism, antioxidant defense mechanisms, etc. It also activates the inflammatory response, stress signals inducing cardiac hypertrophy, fibrosis, or cell death via different mechanisms (e.g., apoptosis, necrosis) and dysregulates autophagy. Therefore, the thorough understanding of the mechanisms which lead to perturbations in oxidative/nitrative metabolism and its relationship with pro-inflammatory, hypertrophic, fibrotic, cell death and other pathways would help to develop strategies to counteract systemic and tissue oxidative/nitrative stress in T4CRS and renal sarcopenia. In this review, we also focus on the effects of some well-known and novel pharmaceuticals, nutraceuticals, and physical exercise on cardiac and skeletal muscle oxidative/nitrative stress in T4CRS and renal sarcopenia.
Collapse
Affiliation(s)
- Márta Sárközy
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Zsuzsanna Z A Kovács
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Mónika G Kovács
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Renáta Gáspár
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Gergő Szűcs
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - László Dux
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
93
|
Reyes DRA, Gomes MJ, Rosa CM, Pagan LU, Zanati SG, Damatto RL, Rodrigues EA, Carvalho RF, Fernandes AAH, Martinez PF, Lima ARR, Cezar MDM, Carvalho LEFM, Okoshi K, Okoshi MP. Exercise during transition from compensated left ventricular hypertrophy to heart failure in aortic stenosis rats. J Cell Mol Med 2018; 23:1235-1245. [PMID: 30456799 PMCID: PMC6349163 DOI: 10.1111/jcmm.14025] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/20/2018] [Accepted: 10/22/2018] [Indexed: 12/18/2022] Open
Abstract
We evaluated the influence of aerobic exercise on cardiac remodelling during the transition from compensated left ventricular (LV) hypertrophy to clinical heart failure in aortic stenosis (AS) rats. Eighteen weeks after AS induction, rats were assigned into sedentary (AS) and exercised (AS-Ex) groups. Results were compared to Sham rats. Exercise was performed on treadmill for 8 weeks. Exercise improved functional capacity. Echocardiogram showed no differences between AS-Ex and AS groups. After exercise, fractional shortening and ejection fraction were lower in AS-Ex than Sham. Myocyte diameter and interstitial collagen fraction were higher in AS and AS-Ex than Sham; however, myocyte diameter was higher in AS-Ex than AS. Myocardial oxidative stress, evaluated by lipid hydroperoxide concentration, was higher in AS than Sham and was normalized by exercise. Gene expression of the NADPH oxidase subunits NOX2 and NOX4, which participate in ROS generation, did not differ between groups. Activity of the antioxidant enzyme superoxide dismutase was lower in AS and AS-Ex than Sham and glutathione peroxidase was lower in AS-Ex than Sham. Total and reduced myocardial glutathione, which is involved in cellular defence against oxidative stress, was lower in AS than Sham and total glutathione was higher in AS-Ex than AS. The MAPK JNK was higher in AS-Ex than Sham and AS groups. Phosphorylated P38 was lower in AS-Ex than AS. Despite improving functional capacity, aerobic exercise does not change LV function in AS rats. Exercise restores myocardial glutathione, reduces oxidative stress, impairs JNK signalling and further induces myocyte hypertrophy.
Collapse
Affiliation(s)
- David R A Reyes
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Mariana J Gomes
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Camila M Rosa
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Luana U Pagan
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Silmeia G Zanati
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Ricardo L Damatto
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Eder A Rodrigues
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Robson F Carvalho
- Institute of Biosciences of Botucatu, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Ana A H Fernandes
- Institute of Biosciences of Botucatu, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Paula F Martinez
- School of Physical Therapy, Federal University of Mato Grosso do Sul, Campo Grande, Brazil
| | - Aline R R Lima
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Marcelo D M Cezar
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Luiz E F M Carvalho
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Katashi Okoshi
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Marina P Okoshi
- Botucatu Medical School, Internal Medicine Department, Sao Paulo State University, UNESP, Botucatu, Brazil
| |
Collapse
|
94
|
Hyatt H, Deminice R, Yoshihara T, Powers SK. Mitochondrial dysfunction induces muscle atrophy during prolonged inactivity: A review of the causes and effects. Arch Biochem Biophys 2018; 662:49-60. [PMID: 30452895 DOI: 10.1016/j.abb.2018.11.005] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/05/2018] [Indexed: 02/08/2023]
Abstract
Prolonged skeletal muscle inactivity (e.g. limb immobilization, bed rest, mechanical ventilation, spinal cord injury, etc.) results in muscle atrophy that manifests into a decreased quality of life and in select patient populations, a higher risk of morbidity and mortality. Thus, understanding the processes that contribute to muscle atrophy during prolonged periods of muscle disuse is an important area of research. In this regard, mitochondrial dysfunction has been directly linked to the muscle wasting that occurs during extended periods of skeletal muscle inactivity. While the concept that mitochondrial dysfunction contributes to disuse muscle atrophy has been contemplated for nearly 50 years, the mechanisms connecting mitochondrial signaling events to skeletal muscle atrophy remained largely unexplained until recently. Indeed, emerging evidence reveals that mitochondrial dysfunction and the associated mitochondrial signaling events are a requirement for several forms of inactivity-induced skeletal muscle atrophy. Specifically, inactivity-induced alterations in skeletal muscle mitochondria phenotype and increased ROS emission, impaired Ca2+ handling, and release of mitochondria-specific proteolytic activators are established occurrences that promote fiber atrophy during prolonged periods of muscle inactivity. This review highlights the evidence that directly connects mitochondrial dysfunction and aberrant mitochondrial signaling with skeletal muscle atrophy and discusses the mechanisms linking these interconnected phenomena.
Collapse
Affiliation(s)
- Hayden Hyatt
- 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, University of Estadual of Londrina, Londrina, Brazil
| | - Toshinori Yoshihara
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Department of Exercise Physiology, Juntendo University, Tokyo, Japan
| | - Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
95
|
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
|
96
|
Laitano O, Sheikh LH, Mattingly AJ, Murray KO, Ferreira LF, Clanton TL. Osmolality Selectively Offsets the Impact of Hyperthermia on Mouse Skeletal Muscle in vitro. Front Physiol 2018; 9:1496. [PMID: 30429796 PMCID: PMC6220237 DOI: 10.3389/fphys.2018.01496] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/03/2018] [Indexed: 11/24/2022] Open
Abstract
Hyperthermia and dehydration can occur during exercise in hot environments. Nevertheless, whether elevations in extracellular osmolality contributes to the increased skeletal muscle tension, sarcolemmal injury, and oxidative stress reported in warm climates remains unknown. We simulated osmotic and heat stress, in vitro, in mouse limb muscles with different fiber compositions. Extensor digitorum longus (EDL) and soleus (SOL) were dissected from 36 male C57BL6J and mounted at optimal length in tissue baths containing oxygenated buffer. Muscles were stimulated with non-fatiguing twitches for 30 min. Four experimental conditions were tested: isotonic-normothermia (285 mOsm•kg-1 and 35°C), hypertonic-normothermia (300 mOsm•kg-1 and 35°C), isotonic-hyperthermia (285 mOsm•kg-1 and 41°C), and hypertonic-hyperthermia (300 mOsm•kg-1 and 41°C). Passive tension was recorded continuously. The integrity of the sarcolemma was determined using a cell-impermeable fluorescent dye and immunoblots were used for detection of protein carbonyls. In EDL muscles, isotonic and hypertonic-hyperthermia increased resting tension (P < 0.001). Whereas isotonic-hyperthermia increased sarcolemmal injury in EDL (P < 0.001), this effect was absent in hypertonic-hyperthermia. Similarly, isotonic-hyperthermia elevated protein carbonyls (P = 0.018), a response not observed with hypertonic-hyperthermia. In SOL muscles, isotonic-hyperthermia also increases resting tension (P < 0.001); however, these effects were eliminated in hypertonic-hyperthermia. Unlike EDL, there were no effects of hyperthermia and/or hyperosmolality on sarcolemmal injury or protein carbonyls. Osmolality selectively modifies skeletal muscle response to hyperthermia in this model. Fast-glycolytic muscle appears particularly vulnerable to isotonic-hyperthermia, resulting in elevated muscle tension, sarcolemmal injury and protein oxidation; whereas slow-oxidative muscle exhibits increased tension but no injury or protein oxidation under the conditions and duration tested.
Collapse
Affiliation(s)
- Orlando Laitano
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
- Colegiado de Educação Física, Federal University of Vale do São Francisco, Petrolina, Brazil
| | - Laila H. Sheikh
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| | - Alex J. Mattingly
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| | - Kevin O. Murray
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| | - Leonardo F. Ferreira
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| | - Thomas L. Clanton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
97
|
TRPC channels in exercise-mimetic therapy. Pflugers Arch 2018; 471:507-517. [PMID: 30298191 PMCID: PMC6515694 DOI: 10.1007/s00424-018-2211-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/18/2018] [Accepted: 09/25/2018] [Indexed: 11/21/2022]
Abstract
Physical exercise yields beneficial effects on all types of muscle cells, which are essential for the maintenance of cardiovascular homeostasis and good blood circulation. Daily moderate exercise increases systemic antioxidative capacity, which can lead to the prevention of the onset and progression of oxidative stress-related diseases. Therefore, exercise is now widely accepted as one of the best therapeutic strategies for the treatment of ischemic (hypoxic) diseases. Canonical transient receptor potential (TRPC) proteins are non-selective cation channels activated by mechanical stress and/or stimulation of phospholipase C-coupled surface receptors. TRPC channels, especially diacylglycerol-activated TRPC channels (TRPC3 and TRPC6; TRPC3/6), play a key role in the development of cardiovascular remodeling. We have recently found that physical interaction between TRPC3 and NADPH oxidase (Nox) 2 under hypoxic stress promotes Nox2-dependent reactive oxygen species (ROS) production and mediates rodent cardiac plasticity, and inhibition of the TRPC3-Nox2 protein complex results in enhancement of myocardial compliance and flexibility similar to that observed in exercise-treated hearts. In this review, we describe current understanding of the roles of TRPC channels in striated muscle (patho)physiology and propose that targeting TRPC-based protein complexes could be a new strategy to imitate exercise therapy.
Collapse
|
98
|
Abstract
SIGNIFICANCE Hydrogen peroxide (H2O2) is a key signaling molecule involved in the regulation of both physiological and pathological cellular processes. Genetically encoded HyPer probes are currently among the most effective approaches for monitoring H2O2 dynamics in various biological systems because they can be easily targeted to specific cells and organelles. Since its development in 2006, HyPer has proved to be a robust and powerful tool in redox biology research. Recent Advances: HyPer probes were used in a variety of models to study the role of H2O2 in various redox processes. HyPer has been increasingly used in the past few years for in vivo studies, which has already led to many important discoveries, for example, that H2O2 plays a key role in the regulation of signaling cascades involved in development and aging, inflammation, regeneration, photosynthetic signaling, and other biological processes. CRITICAL ISSUES In this review, we focus on the main achievements in the field of redox biology that have been obtained from in vivo experiments using HyPer probes. FUTURE DIRECTIONS Further in vivo studies of the role of H2O2 largely depend on the development of more suitable versions of HyPer for in vivo models: those having brighter fluorescence and a more stable signal in response to physiological changes in pH. Antioxid. Redox Signal. 29, 569-584.
Collapse
Affiliation(s)
- Dmitry S Bilan
- 1 Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry , Moscow, Russia .,2 Pirogov Russian National Research Medical University , Moscow, Russia
| | - Vsevolod V Belousov
- 1 Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry , Moscow, Russia .,2 Pirogov Russian National Research Medical University , Moscow, Russia .,3 Institute for Cardiovascular Physiology, Georg August University Göttingen , Göttingen, Germany
| |
Collapse
|
99
|
Heydemann A. Skeletal Muscle Metabolism in Duchenne and Becker Muscular Dystrophy-Implications for Therapies. Nutrients 2018; 10:nu10060796. [PMID: 29925809 PMCID: PMC6024668 DOI: 10.3390/nu10060796] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/14/2018] [Accepted: 06/16/2018] [Indexed: 02/06/2023] Open
Abstract
The interactions between nutrition and metabolism and skeletal muscle have long been known. Muscle is the major metabolic organ—it consumes more calories than other organs—and therefore, there is a clear need to discuss these interactions and provide some direction for future research areas regarding muscle pathologies. In addition, new experiments and manuscripts continually reveal additional highly intricate, reciprocal interactions between metabolism and muscle. These reciprocal interactions include exercise, age, sex, diet, and pathologies including atrophy, hypoxia, obesity, diabetes, and muscle myopathies. Central to this review are the metabolic changes that occur in the skeletal muscle cells of muscular dystrophy patients and mouse models. Many of these metabolic changes are pathogenic (inappropriate body mass changes, mitochondrial dysfunction, reduced adenosine triphosphate (ATP) levels, and increased Ca2+) and others are compensatory (increased phosphorylated AMP activated protein kinase (pAMPK), increased slow fiber numbers, and increased utrophin). Therefore, reversing or enhancing these changes with therapies will aid the patients. The multiple therapeutic targets to reverse or enhance the metabolic pathways will be discussed. Among the therapeutic targets are increasing pAMPK, utrophin, mitochondrial number and slow fiber characteristics, and inhibiting reactive oxygen species. Because new data reveals many additional intricate levels of interactions, new questions are rapidly arising. How does muscular dystrophy alter metabolism, and are the changes compensatory or pathogenic? How does metabolism affect muscular dystrophy? Of course, the most profound question is whether clinicians can therapeutically target nutrition and metabolism for muscular dystrophy patient benefit? Obtaining the answers to these questions will greatly aid patients with muscular dystrophy.
Collapse
Affiliation(s)
- Ahlke Heydemann
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA.
- Center for Cardiovascular Research, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
100
|
Barreiro E, Puig-Vilanova E, Salazar-Degracia A, Pascual-Guardia S, Casadevall C, Gea J. The phosphodiesterase-4 inhibitor roflumilast reverts proteolysis in skeletal muscle cells of patients with COPD cachexia. J Appl Physiol (1985) 2018; 125:287-303. [PMID: 29648516 DOI: 10.1152/japplphysiol.00798.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Peripheral muscle weakness and mass loss are characteristic features in severe chronic obstructive pulmonary disease (COPD). We hypothesized that the phosphodiesterase (PDE)-4 inhibitor roflumilast-induced cAMP may ameliorate proteolysis and metabolism in skeletal muscles of COPD patients with severe muscle wasting. In myogenic precursor cells (isolated from muscle biopsies and cultured up to obtain differentiated myotubes) from 10 severe COPD patients and 10 healthy controls, which were treated with 1 μM roflumilast N-oxide (RNO) for three time cohorts (1, 6, and 24 h), genes of antioxidant defense and oxidative stress marker, myogenesis and muscle metabolism, proteolysis (tyrosine release assay) and ubiquitin-proteasome system markers, autophagy, and myosin isoforms were analyzed using RT-PCR and immunoblotting. In COPD patients at 6 h RNO treatment, myotube tyrosine release, total protein ubiquitination, and tripartite motif-containing protein 32 levels were significantly lower than healthy controls, whereas at 24 h RNO treatment, myotube myosin heavy chain ( MyHC) -I and MyHC-IIx expression levels were upregulated in both patients and controls. In the 6-h RNO cohort, in patients and controls, myotube expression of nuclear factor (erythroid-derived 2)-like 2 ( NRF2) and its downstream antioxidants sirtuin-1, FGF-inducible 14, and insulin-like growth factor-1 was upregulated, whereas that of myocyte-specific enhancer factor 2C, myogenic differentiation, myogenin, myostatin, atrogin-1, and muscle RING-finger protein-1 was downregulated. In myotubes of severe COPD patients with cachexia, roflumilast-induced cAMP signaling exerts beneficial effects by targeting muscle protein breakdown (tyrosine release), along with reduced expression of proteolytic markers of the ubiquitin-proteasome system and that of myostatin. In both patients and controls, roflumilast also favored antioxidant defense through upregulation of the NRF2 pathway and that of the histone deacetylase sirtuin-1, whereas it improved the expression of slow- and fast-twitch myosin isoforms. These findings show that muscle dysfunction and wasting may be targeted by roflumilast-induced cAMP signaling in COPD. These results have potential therapeutic implications, as this PDE-4 inhibitor is currently available for the treatment of systemic inflammation and exacerbations in patients with severe COPD. NEW & NOTEWORTHY In myotubes of cachectic chronic obstructive pulmonary disease (COPD) patients, cAMP signaling exerted beneficial effects by targeting muscle proteolysis and reducing gene expression of proteolytic markers of the ubiquitin-proteasome system and that of myostatin. In myotubes of patients and controls, roflumilast also favored antioxidant defense through upregulation of the nuclear factor (erythroid-derived 2)-like 2 pathway, of sirtuin-1, and of gene expression of slow- and fast-twitch isoforms. These findings have potential clinical implications for the treatment of muscle wasting in patients with COPD and cachexia.
Collapse
Affiliation(s)
- Esther Barreiro
- Pulmonology Department, Muscle and Respiratory System Research Unit, Hospital del Mar Medical Research Institute, Parc de Salut Mar, and Health and Experimental Sciences Department, Universitat Pompeu Fabra , Barcelona , Spain.,Centro de Investigación en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III , Madrid , Spain
| | - Ester Puig-Vilanova
- Pulmonology Department, Muscle and Respiratory System Research Unit, Hospital del Mar Medical Research Institute, Parc de Salut Mar, and Health and Experimental Sciences Department, Universitat Pompeu Fabra , Barcelona , Spain
| | - Anna Salazar-Degracia
- Pulmonology Department, Muscle and Respiratory System Research Unit, Hospital del Mar Medical Research Institute, Parc de Salut Mar, and Health and Experimental Sciences Department, Universitat Pompeu Fabra , Barcelona , Spain
| | - Sergi Pascual-Guardia
- Pulmonology Department, Muscle and Respiratory System Research Unit, Hospital del Mar Medical Research Institute, Parc de Salut Mar, and Health and Experimental Sciences Department, Universitat Pompeu Fabra , Barcelona , Spain
| | - Carme Casadevall
- Pulmonology Department, Muscle and Respiratory System Research Unit, Hospital del Mar Medical Research Institute, Parc de Salut Mar, and Health and Experimental Sciences Department, Universitat Pompeu Fabra , Barcelona , Spain.,Centro de Investigación en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III , Madrid , Spain
| | - Joaquim Gea
- Pulmonology Department, Muscle and Respiratory System Research Unit, Hospital del Mar Medical Research Institute, Parc de Salut Mar, and Health and Experimental Sciences Department, Universitat Pompeu Fabra , Barcelona , Spain.,Centro de Investigación en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III , Madrid , Spain
| |
Collapse
|