1
|
Zhou Y, Zhang X, Baker JS, Davison GW, Yan X. Redox signaling and skeletal muscle adaptation during aerobic exercise. iScience 2024; 27:109643. [PMID: 38650987 PMCID: PMC11033207 DOI: 10.1016/j.isci.2024.109643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Redox regulation is a fundamental physiological phenomenon related to oxygen-dependent metabolism, and skeletal muscle is mainly regarded as a primary site for oxidative phosphorylation. Several studies have revealed the importance of reactive oxygen and nitrogen species (RONS) in the signaling process relating to muscle adaptation during exercise. To date, improving knowledge of redox signaling in modulating exercise adaptation has been the subject of comprehensive work and scientific inquiry. The primary aim of this review is to elucidate the molecular and biochemical pathways aligned to RONS as activators of skeletal muscle adaptation and to further identify the interconnecting mechanisms controlling redox balance. We also discuss the RONS-mediated pathways during the muscle adaptive process, including mitochondrial biogenesis, muscle remodeling, vascular angiogenesis, neuron regeneration, and the role of exogenous antioxidants.
Collapse
Affiliation(s)
- Yingsong Zhou
- Faculty of Sports Science, Ningbo University, Ningbo, China
| | - Xuan Zhang
- School of Wealth Management, Ningbo University of Finance and Economics, Ningbo, China
| | - Julien S. Baker
- Centre for Health and Exercise Science Research, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong
| | - Gareth W. Davison
- Sport and Exercise Sciences Research Institute, Ulster University, Belfast BT15 IED, UK
| | - Xiaojun Yan
- School of Marine Sciences, Ningbo University, Ningbo, China
| |
Collapse
|
2
|
Łoboda A, Dulak J. Nuclear Factor Erythroid 2-Related Factor 2 and Its Targets in Skeletal Muscle Repair and Regeneration. Antioxid Redox Signal 2023; 38:619-642. [PMID: 36597355 DOI: 10.1089/ars.2022.0208] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Significance: Skeletal muscles have a robust regenerative capacity in response to acute and chronic injuries. Muscle repair and redox homeostasis are intimately linked; increased generation of reactive oxygen species leads to cellular dysfunction and contributes to muscle wasting and progression of muscle diseases. In exemplary muscle disease, Duchenne muscular dystrophy (DMD), caused by mutations in the DMD gene that encodes the muscle structural protein dystrophin, the regeneration machinery is severely compromised, while oxidative stress contributes to the progression of the disease. The nuclear factor erythroid 2-related factor 2 (NRF2) and its target genes, including heme oxygenase-1 (HO-1), provide protective mechanisms against oxidative insults. Recent Advances: Relevant advances have been evolving in recent years in understanding the mechanisms by which NRF2 regulates processes that contribute to effective muscle regeneration. To this end, pathways related to muscle satellite cell differentiation, oxidative stress, mitochondrial metabolism, inflammation, fibrosis, and angiogenesis have been studied. The regulatory role of NRF2 in skeletal muscle ferroptosis has been also suggested. Animal studies have shown that NRF2 pathway activation can stop or reverse skeletal muscle pathology, especially when endogenous stress defence mechanisms are imbalanced. Critical Issues: Despite the growing recognition of NRF2 as a factor that regulates various aspects of muscle regeneration, the mechanistic impact on muscle pathology in various models of muscle injury remains imprecise. Future Directions: Further studies are necessary to fully uncover the role of NRF2 in muscle regeneration, both in physiological and pathological conditions, and to investigate the possibilities for development of new therapeutic modalities. Antioxid. Redox Signal. 38, 619-642.
Collapse
Affiliation(s)
- Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| |
Collapse
|
3
|
Bronisz-Budzyńska I, Kozakowska M, Pietraszek-Gremplewicz K, Madej M, Józkowicz A, Łoboda A, Dulak J. NRF2 Regulates Viability, Proliferation, Resistance to Oxidative Stress, and Differentiation of Murine Myoblasts and Muscle Satellite Cells. Cells 2022; 11:cells11203321. [PMID: 36291188 PMCID: PMC9600498 DOI: 10.3390/cells11203321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/27/2022] Open
Abstract
Increased oxidative stress can slow down the regeneration of skeletal muscle and affect the activity of muscle satellite cells (mSCs). Therefore, we evaluated the role of the NRF2 transcription factor (encoded by the Nfe2l2 gene), the main regulator of the antioxidant response, in muscle cell biology. We used (i) an immortalized murine myoblast cell line (C2C12) with stable overexpression of NRF2 and (ii) primary mSCs isolated from wild-type and Nfe2l2 (transcriptionally)-deficient mice (Nfe2l2tKO). NRF2 promoted myoblast proliferation and viability under oxidative stress conditions and decreased the production of reactive oxygen species. Furthermore, NRF2 overexpression inhibited C2C12 cell differentiation by down-regulating the expression of myogenic regulatory factors (MRFs) and muscle-specific microRNAs. We also showed that NRF2 is indispensable for the viability of mSCs since the lack of its transcriptional activity caused high mortality of cells cultured in vitro under normoxic conditions. Concomitantly, Nfe2l2tKO mSCs grown and differentiated under hypoxic conditions were viable and much more differentiated compared to cells isolated from wild-type mice. Taken together, NRF2 significantly influences the properties of myoblasts and muscle satellite cells. This effect might be modulated by the muscle microenvironment.
Collapse
|
4
|
Iacobini C, Vitale M, Pesce C, Pugliese G, Menini S. Diabetic Complications and Oxidative Stress: A 20-Year Voyage Back in Time and Back to the Future. Antioxidants (Basel) 2021; 10:727. [PMID: 34063078 PMCID: PMC8147954 DOI: 10.3390/antiox10050727] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Twenty years have passed since Brownlee and colleagues proposed a single unifying mechanism for diabetic complications, introducing a turning point in this field of research. For the first time, reactive oxygen species (ROS) were identified as the causal link between hyperglycemia and four seemingly independent pathways that are involved in the pathogenesis of diabetes-associated vascular disease. Before and after this milestone in diabetes research, hundreds of articles describe a role for ROS, but the failure of clinical trials to demonstrate antioxidant benefits and some recent experimental studies showing that ROS are dispensable for the pathogenesis of diabetic complications call for time to reflect. This twenty-year journey focuses on the most relevant literature regarding the main sources of ROS generation in diabetes and their role in the pathogenesis of cell dysfunction and diabetic complications. To identify future research directions, this review discusses the evidence in favor and against oxidative stress as an initial event in the cellular biochemical abnormalities induced by hyperglycemia. It also explores possible alternative mechanisms, including carbonyl stress and the Warburg effect, linking glucose and lipid excess, mitochondrial dysfunction, and the activation of alternative pathways of glucose metabolism leading to vascular cell injury and inflammation.
Collapse
Affiliation(s)
- Carla Iacobini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| | - Martina Vitale
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| | - Carlo Pesce
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetic and Maternal Infantile Sciences (DINOGMI), Department of Excellence of MIUR, University of Genoa Medical School, 16132 Genoa, Italy;
| | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| | - Stefano Menini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (S.M.)
| |
Collapse
|
5
|
Zhang B, Wang L, Zhan A, Wang M, Tian L, Guo W, Pan Y. Long-term exposure to a hypomagnetic field attenuates adult hippocampal neurogenesis and cognition. Nat Commun 2021; 12:1174. [PMID: 33608552 PMCID: PMC7896063 DOI: 10.1038/s41467-021-21468-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Adult hippocampal neurogenesis contributes to learning and memory, and is sensitive to a variety of environmental stimuli. Exposure to a hypomagnetic field (HMF) influences the cognitive processes of various animals, from insects to human beings. However, whether HMF exposure affect adult hippocampal neurogenesis and hippocampus-dependent cognitions is still an enigma. Here, we showed that male C57BL/6 J mice exposed to HMF by means of near elimination of the geomagnetic field (GMF) exhibit significant impairments of adult hippocampal neurogenesis and hippocampus-dependent learning, which is strongly correlated with a reduction in the content of reactive oxygen species (ROS). However, these deficits seen in HMF-exposed mice could be rescued either by elevating ROS levels through pharmacological inhibition of ROS removal or by returning them back to GMF. Therefore, our results suggest that GMF plays an important role in adult hippocampal neurogenesis through maintaining appropriate endogenous ROS levels.
Collapse
Affiliation(s)
- Bingfang Zhang
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lei Wang
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Aisheng Zhan
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Min Wang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Lanxiang Tian
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China.
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing, China.
- The Paleomagnetism and Geochronology Laboratory, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China.
| | - Weixiang Guo
- University of Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
| | - Yongxin Pan
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- The Paleomagnetism and Geochronology Laboratory, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
6
|
Daiber A, Andreadou I, Oelze M, Davidson SM, Hausenloy DJ. Discovery of new therapeutic redox targets for cardioprotection against ischemia/reperfusion injury and heart failure. Free Radic Biol Med 2021; 163:325-343. [PMID: 33359685 DOI: 10.1016/j.freeradbiomed.2020.12.026] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023]
Abstract
Global epidemiological studies reported a shift from maternal/infectious communicable diseases to chronic non-communicable diseases and a major part is attributable to atherosclerosis and metabolic disorders. Accordingly, ischemic heart disease was identified as a leading risk factor for global mortality and morbidity with a prevalence of 128 million people. Almost 9 million premature deaths can be attributed to ischemic heart disease and subsequent acute myocardial infarction and heart failure, also representing a substantial socioeconomic burden. As evidenced by typical oxidative stress markers such as lipid peroxidation products or oxidized DNA/RNA bases, the formation of reactive oxygen species by various sources (NADPH oxidases, xanthine oxidase and mitochondrial resperatory chain) plays a central role for the severity of ischemia/reperfusion damage. The underlying mechanisms comprise direct oxidative damage but also adverse redox-regulation of kinase and calcium signaling, inflammation and cardiac remodeling among others. These processes and the role of reactive oxygen species are discussed in the present review. We also present and discuss potential targets for redox-based therapies that are either already established in the clinics (e.g. guanylyl cyclase activators and stimulators) or at least successfully tested in preclinical models of myocardial infarction and heart failure (mitochondria-targeted antioxidants). However, reactive oxygen species have not only detrimental effects but are also involved in essential cellular signaling and may even act protective as seen by ischemic pre- and post-conditioning or eustress - which makes redox therapy quite challenging.
Collapse
Affiliation(s)
- Andreas Daiber
- Department of Cardiology 1, Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Matthias Oelze
- Department of Cardiology 1, Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore; Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan.
| |
Collapse
|
7
|
Zhang B, Tian L. Reactive Oxygen Species: Potential Regulatory Molecules in Response to Hypomagnetic Field Exposure. Bioelectromagnetics 2020; 41:573-580. [PMID: 32997824 DOI: 10.1002/bem.22299] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/27/2020] [Accepted: 09/15/2020] [Indexed: 01/19/2023]
Abstract
Organisms, including humans, could be exposed to hypomagnetic fields (HMFs, intensity <5 μT), e.g. in some artificially shielded magnetic environments and during deep-space flights. Previous studies have demonstrated that HMF exposure could have negative effects on the central nervous system and embryonic development in many animals. However, the underlying mechanisms remain unknown. Studies have revealed that HMFs affect cellular reactive oxygen species (ROS) levels and thereby alter physiological and biological processes in organisms. ROS, the major component of highly active free radicals, which are ubiquitous in biological systems, were hypothesized to be the candidate signaling molecules that regulate diverse physiological processes in response to changes in magnetic fields. Here, we summarize the recent advances in the study of HMF-induced negative effects on the central nervous system and early embryonic development in animals, focusing on cellular ROS and their role in response to HMFs. Furthermore, we discuss the potential mechanism through which HMFs regulate ROS levels in cells. © 2020 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Bingfang Zhang
- Biogeomagnetism Laboratory, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lanxiang Tian
- Biogeomagnetism Laboratory, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
8
|
Göllner M, Ihrig-Biedert I, Petermann V, Saurin S, Oelze M, Kröller-Schön S, Vujacic-Mirski K, Kuntic M, Pautz A, Daiber A, Kleinert H. NOX2ko Mice Show Largely Increased Expression of a Mutated NOX2 mRNA Encoding an Inactive NOX2 Protein. Antioxidants (Basel) 2020; 9:antiox9111043. [PMID: 33114493 PMCID: PMC7692237 DOI: 10.3390/antiox9111043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 11/16/2022] Open
Abstract
Background: The superoxide-generating enzyme nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX2 or gp91phox, the phagocytic isoform) was reported as a major source of oxidative stress in various human diseases. Genetic deletion is widely used to study the impact of NOX2-derived reactive oxygen species (ROS) on disease development and progression in various animal models. Here, we investigate why NOX2 knockout mice show no NOX2 activity but express NOX2 mRNA and protein. Methods and Results: Oxidative burst (NOX2-dependent formation of ROS) was measured by L-012-based chemiluminescence and was largely absent in whole blood of NOX2 knockout mice. Protein expression was still detectable in different tissues of the NOX2 knockout mice, at the expected and a slightly lower molecular weight (determined by Western blot). The NOX2 gene was even largely enhanced at its expressional level in NOX2 knockout mice. RNA sequencing revealed a modified NOX2 mRNA in the knockout mice that is obviously translated to a truncated inactive mutant enzyme. Conclusion: Although the commercial NOX2 knockout mice display no considerable enzymatic NOX2 activity, expression of the NOX2 gene (when using standard primers) and protein (when using antibodies binding to the carboxy-terminal end) can still be detected, which may lead to confusion among investigators.
Collapse
Affiliation(s)
- Monika Göllner
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
| | - Irmgard Ihrig-Biedert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
| | - Victoria Petermann
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
| | - Sabrina Saurin
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
| | - Matthias Oelze
- Laboratory of Molecular Cardiology, Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.O.); (S.K.-S.); (K.V.-M.); (M.K.)
| | - Swenja Kröller-Schön
- Laboratory of Molecular Cardiology, Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.O.); (S.K.-S.); (K.V.-M.); (M.K.)
| | - Ksenija Vujacic-Mirski
- Laboratory of Molecular Cardiology, Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.O.); (S.K.-S.); (K.V.-M.); (M.K.)
| | - Marin Kuntic
- Laboratory of Molecular Cardiology, Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.O.); (S.K.-S.); (K.V.-M.); (M.K.)
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
| | - Andreas Daiber
- Laboratory of Molecular Cardiology, Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.O.); (S.K.-S.); (K.V.-M.); (M.K.)
- Correspondence: (A.D.); (H.K.); Tel.: +49-(6131)-17-6280 (A.D.); +49-(6131)-17-9150 (H.K.)
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
- Correspondence: (A.D.); (H.K.); Tel.: +49-(6131)-17-6280 (A.D.); +49-(6131)-17-9150 (H.K.)
| |
Collapse
|
9
|
Terzi A, Suter DM. The role of NADPH oxidases in neuronal development. Free Radic Biol Med 2020; 154:33-47. [PMID: 32370993 DOI: 10.1016/j.freeradbiomed.2020.04.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) are critical for maintaining cellular homeostasis and function when produced in physiological ranges. Important sources of cellular ROS include NADPH oxidases (Nox), which are evolutionary conserved multi-subunit transmembrane proteins. Nox-mediated ROS regulate variety of biological processes including hormone synthesis, calcium signaling, cell migration, and immunity. ROS participate in intracellular signaling by introducing post-translational modifications to proteins and thereby altering their functions. The central nervous system (CNS) expresses different Nox isoforms during both development and adulthood. Here, we review the role of Nox-mediated ROS during CNS development. Specifically, we focus on how individual Nox isoforms contribute to signaling in neural stem cell maintenance and neuronal differentiation, as well as neurite outgrowth and guidance. We also discuss how ROS regulates the organization and dynamics of the actin cytoskeleton in the neuronal growth cone. Finally, we review recent evidence that Nox-derived ROS modulate axonal regeneration upon nervous system injury.
Collapse
Affiliation(s)
- Aslihan Terzi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA; Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
10
|
Glutathione and Nitric Oxide: Key Team Players in Use and Disuse of Skeletal Muscle. Nutrients 2019; 11:nu11102318. [PMID: 31575008 PMCID: PMC6836164 DOI: 10.3390/nu11102318] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/11/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Glutathione (GSH) is the main non-enzymatic antioxidant playing an important role in detoxification, signal transduction by modulation of protein thiols redox status and direct scavenging of radicals. The latter function is not only performed against reactive oxygen species (ROS) but GSH also has a fundamental role in buffering nitric oxide (NO), a physiologically-produced molecule having-multifaceted functions. The efficient rate of GSH synthesis and high levels of GSH-dependent enzymes are characteristic features of healthy skeletal muscle where, besides the canonical functions, it is also involved in muscle contraction regulation. Moreover, NO production in skeletal muscle is a direct consequence of contractile activity and influences several metabolic myocyte pathways under both physiological and pathological conditions. In this review, we will consider the homeostasis and intersection of GSH with NO and then we will restrict the discussion on their role in processes related to skeletal muscle function and degeneration.
Collapse
|
11
|
Nouri F, Nematollahi-Mahani SN, Sharifi AM. Preconditioning of Mesenchymal Stem Cells with Non-Toxic Concentration of Hydrogen Peroxide Against Oxidative Stress Induced Cell Death: The Role of Hypoxia-Inducible Factor-1. Adv Pharm Bull 2019; 9:76-83. [PMID: 31011561 PMCID: PMC6468218 DOI: 10.15171/apb.2019.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 11/03/2018] [Accepted: 12/20/2018] [Indexed: 12/30/2022] Open
Abstract
Purpose: To investigate the protective effect of preconditioning with non-toxic dose of hydrogen
peroxide (H2O2) as a possible cell signaling molecule, against cell death induced by toxic
concentration of H2O2 or by serum deprivation in human Wharton’s jelly-derived mesenchymal
stem cells (HWJ-MSCs) and underlying mechanisms.
Methods: HWJ-MSCs were isolated and identified using flow cytometry. After finding non-toxic
concentration of H2O2, cells preconditioning was performed by H2O2 (20 μM) for 12 h and cell
tolerance against serum deprivation or toxic levels of H2O2 was assayed by MTT test. Effect of
preconditioning on mRNA and protein expression of Akt-1, Bcl-2 and Bax were examined using
reverse transcription polymerase chain reaction (RT-PCR) and western blotting respectively. Role
of hypoxia-inducible factor (HIF)-1α was explored in presence HIF-1α inhibitor.
Results: Preconditioning with 20 μM H2O2 for 12 h was non-toxic and decreased cell death
induced by oxidative stress and serum deprivation in MSC cultures. However, the increased
tolerance reversed in the presence of inhibitor of HIF-1α. By regards to RT-PCR and western
blotting data, although expression of Akt-1, Bcl-2 and Bax was not change considerably but
phosphorylated Akt-1 (pAkt-1) was up regulated after treatment with 20 μM H2O2 compared to
control group. Moreover after exposure to 100 μM H2O2, western blotting analysis showed that
cell pretreatment with 20 μM H2O2, decremented Bax/Bcl2 ratio and up-regulated HIF-1α and
pAkt-1 compared to the control group.
Conclusion: Increased tolerance of H2O2-pretreated cells led to the suggestion that transplantation
of H2O2 preconditioned MSCs may improve therapeutic potential of stem cells in cell therapy
procedures.
Collapse
Affiliation(s)
- Fatemeh Nouri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Ali Mohammad Sharifi
- Razi Drug Research Center, Department of Pharmacology, School of Medicine, Iran University of Medical Science, Tehran, Iran
| |
Collapse
|
12
|
Cervelli M, Leonetti A, Duranti G, Sabatini S, Ceci R, Mariottini P. Skeletal Muscle Pathophysiology: The Emerging Role of Spermine Oxidase and Spermidine. Med Sci (Basel) 2018; 6:medsci6010014. [PMID: 29443878 PMCID: PMC5872171 DOI: 10.3390/medsci6010014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/07/2018] [Accepted: 02/09/2018] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle comprises approximately 40% of the total body mass. Preserving muscle health and function is essential for the entire body in order to counteract chronic diseases such as type II diabetes, cardiovascular diseases, and cancer. Prolonged physical inactivity, particularly among the elderly, causes muscle atrophy, a pathological state with adverse outcomes such as poor quality of life, physical disability, and high mortality. In murine skeletal muscle C2C12 cells, increased expression of the spermine oxidase (SMOX) enzyme has been found during cell differentiation. Notably, SMOX overexpression increases muscle fiber size, while SMOX reduction was enough to induce muscle atrophy in multiple murine models. Of note, the SMOX reaction product spermidine appears to be involved in skeletal muscle atrophy/hypertrophy. It is effective in reactivating autophagy, ameliorating the myopathic defects of collagen VI-null mice. Moreover, spermidine treatment, if combined with exercise, can affect D-gal-induced aging-related skeletal muscle atrophy. This review hypothesizes a role for SMOX during skeletal muscle differentiation and outlines its role and that of spermidine in muscle atrophy. The identification of new molecular pathways involved in the maintenance of skeletal muscle health could be beneficial in developing novel therapeutic lead compounds to treat muscle atrophy.
Collapse
Affiliation(s)
- Manuela Cervelli
- Department of Science, Università degli Studi di Roma "Roma Tre", 00146 Rome, Italy.
| | - Alessia Leonetti
- Department of Science, Università degli Studi di Roma "Roma Tre", 00146 Rome, Italy.
| | - Guglielmo Duranti
- Department of of Movement Human and Health Sciences, Unit of Biology, Genetics and Biochemistry, Università degli Studi di Roma "Foro Italico", Piazza Lauro De Bosis 15, 00135 Rome, Italy.
| | - Stefania Sabatini
- Department of of Movement Human and Health Sciences, Unit of Biology, Genetics and Biochemistry, Università degli Studi di Roma "Foro Italico", Piazza Lauro De Bosis 15, 00135 Rome, Italy.
| | - Roberta Ceci
- Department of of Movement Human and Health Sciences, Unit of Biology, Genetics and Biochemistry, Università degli Studi di Roma "Foro Italico", Piazza Lauro De Bosis 15, 00135 Rome, Italy.
| | - Paolo Mariottini
- Department of Science, Università degli Studi di Roma "Roma Tre", 00146 Rome, Italy.
| |
Collapse
|
13
|
Sakellariou GK, McDonagh B. Redox Homeostasis in Age-Related Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:281-306. [PMID: 30390257 DOI: 10.1007/978-981-13-1435-3_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Muscle atrophy and weakness, characterized by loss of lean muscle mass and function, has a significant effect on the independence and quality of life of older people. The cellular mechanisms that drive the age-related decline in neuromuscular integrity and function are multifactorial. Quiescent and contracting skeletal muscle can endogenously generate reactive oxygen and nitrogen species (RONS) from various cellular sites. Excessive RONS can potentially cause oxidative damage and disruption of cellular signaling pathways contributing to the initiation and progression of age-related muscle atrophy. Altered redox homeostasis and modulation of intracellular signal transduction processes have been proposed as an underlying mechanism of sarcopenia. This chapter summarizes the current evidence that has associated disrupted redox homeostasis and muscle atrophy as a result of skeletal muscle inactivity and aging.
Collapse
Affiliation(s)
| | - Brian McDonagh
- Discipline of Physiology, School of Medicine, NUI Galway, Galway, Ireland
| |
Collapse
|
14
|
Sakellariou GK, Lightfoot AP, Earl KE, Stofanko M, McDonagh B. Redox homeostasis and age-related deficits in neuromuscular integrity and function. J Cachexia Sarcopenia Muscle 2017; 8:881-906. [PMID: 28744984 PMCID: PMC5700439 DOI: 10.1002/jcsm.12223] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 04/06/2017] [Accepted: 05/22/2017] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle is a major site of metabolic activity and is the most abundant tissue in the human body. Age-related muscle atrophy (sarcopenia) and weakness, characterized by progressive loss of lean muscle mass and function, is a major contributor to morbidity and has a profound effect on the quality of life of older people. With a continuously growing older population (estimated 2 billion of people aged >60 by 2050), demand for medical and social care due to functional deficits, associated with neuromuscular ageing, will inevitably increase. Despite the importance of this 'epidemic' problem, the primary biochemical and molecular mechanisms underlying age-related deficits in neuromuscular integrity and function have not been fully determined. Skeletal muscle generates reactive oxygen and nitrogen species (RONS) from a variety of subcellular sources, and age-associated oxidative damage has been suggested to be a major factor contributing to the initiation and progression of muscle atrophy inherent with ageing. RONS can modulate a variety of intracellular signal transduction processes, and disruption of these events over time due to altered redox control has been proposed as an underlying mechanism of ageing. The role of oxidants in ageing has been extensively examined in different model organisms that have undergone genetic manipulations with inconsistent findings. Transgenic and knockout rodent studies have provided insight into the function of RONS regulatory systems in neuromuscular ageing. This review summarizes almost 30 years of research in the field of redox homeostasis and muscle ageing, providing a detailed discussion of the experimental approaches that have been undertaken in murine models to examine the role of redox regulation in age-related muscle atrophy and weakness.
Collapse
Affiliation(s)
| | - Adam P. Lightfoot
- School of Healthcare ScienceManchester Metropolitan UniversityManchesterM1 5GDUK
| | - Kate E. Earl
- MRC‐Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing, Department of Musculoskeletal Biology, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolL7 8TXUK
| | - Martin Stofanko
- Microvisk Technologies LtdThe Quorum7600 Oxford Business ParkOxfordOX4 2JZUK
| | - Brian McDonagh
- MRC‐Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing, Department of Musculoskeletal Biology, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolL7 8TXUK
- Department of Physiology, School of MedicineNational University of IrelandGalwayIreland
| |
Collapse
|
15
|
Le Moal E, Pialoux V, Juban G, Groussard C, Zouhal H, Chazaud B, Mounier R. Redox Control of Skeletal Muscle Regeneration. Antioxid Redox Signal 2017; 27:276-310. [PMID: 28027662 PMCID: PMC5685069 DOI: 10.1089/ars.2016.6782] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 12/24/2016] [Accepted: 12/27/2016] [Indexed: 12/12/2022]
Abstract
Skeletal muscle shows high plasticity in response to external demand. Moreover, adult skeletal muscle is capable of complete regeneration after injury, due to the properties of muscle stem cells (MuSCs), the satellite cells, which follow a tightly regulated myogenic program to generate both new myofibers and new MuSCs for further needs. Although reactive oxygen species (ROS) and reactive nitrogen species (RNS) have long been associated with skeletal muscle physiology, their implication in the cell and molecular processes at work during muscle regeneration is more recent. This review focuses on redox regulation during skeletal muscle regeneration. An overview of the basics of ROS/RNS and antioxidant chemistry and biology occurring in skeletal muscle is first provided. Then, the comprehensive knowledge on redox regulation of MuSCs and their surrounding cell partners (macrophages, endothelial cells) during skeletal muscle regeneration is presented in normal muscle and in specific physiological (exercise-induced muscle damage, aging) and pathological (muscular dystrophies) contexts. Recent advances in the comprehension of these processes has led to the development of therapeutic assays using antioxidant supplementation, which result in inconsistent efficiency, underlying the need for new tools that are aimed at precisely deciphering and targeting ROS networks. This review should provide an overall insight of the redox regulation of skeletal muscle regeneration while highlighting the limits of the use of nonspecific antioxidants to improve muscle function. Antioxid. Redox Signal. 27, 276-310.
Collapse
Affiliation(s)
- Emmeran Le Moal
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
- Movement, Sport and Health Sciences Laboratory, M2S, EA1274, University of Rennes 2, Bruz, France
| | - Vincent Pialoux
- Laboratoire Interuniversitaire de Biologie de la Motricité, EA7424, Université Claude Bernard Lyon 1, Univ Lyon, Villeurbanne, France
- Institut Universitaire de France, Paris, France
| | - Gaëtan Juban
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
| | - Carole Groussard
- Movement, Sport and Health Sciences Laboratory, M2S, EA1274, University of Rennes 2, Bruz, France
| | - Hassane Zouhal
- Movement, Sport and Health Sciences Laboratory, M2S, EA1274, University of Rennes 2, Bruz, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
| |
Collapse
|
16
|
Daiber A, Di Lisa F, Oelze M, Kröller‐Schön S, Steven S, Schulz E, Münzel T. Crosstalk of mitochondria with NADPH oxidase via reactive oxygen and nitrogen species signalling and its role for vascular function. Br J Pharmacol 2017; 174:1670-1689. [PMID: 26660451 PMCID: PMC5446573 DOI: 10.1111/bph.13403] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/22/2015] [Accepted: 11/30/2015] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases are associated with and/or caused by oxidative stress. This concept has been proven by using the approach of genetic deletion of reactive species producing (pro-oxidant) enzymes as well as by the overexpression of reactive species detoxifying (antioxidant) enzymes leading to a marked reduction of reactive oxygen and nitrogen species (RONS) and in parallel to an amelioration of the severity of diseases. Likewise, the development and progression of cardiovascular diseases is aggravated by overexpression of RONS producing enzymes as well as deletion of antioxidant RONS detoxifying enzymes. Thus, the consequences of the interaction (redox crosstalk) of superoxide/hydrogen peroxide produced by mitochondria with other ROS producing enzymes such as NADPH oxidases (Nox) are of outstanding importance and will be discussed including the consequences for endothelial nitric oxide synthase (eNOS) uncoupling as well as the redox regulation of the vascular function/tone in general (soluble guanylyl cyclase, endothelin-1, prostanoid synthesis). Pathways and potential mechanisms leading to this crosstalk will be analysed in detail and highlighted by selected examples from the current literature including hypoxia, angiotensin II-induced hypertension, nitrate tolerance, aging and others. The general concept of redox-based activation of RONS sources via "kindling radicals" and enzyme-specific "redox switches" will be discussed providing evidence that mitochondria represent key players and amplifiers of the burden of oxidative stress. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Andreas Daiber
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Fabio Di Lisa
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Matthias Oelze
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Swenja Kröller‐Schön
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Sebastian Steven
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
- Center of Thrombosis and HemostasisMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Eberhard Schulz
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Thomas Münzel
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| |
Collapse
|
17
|
Di Meo S, Iossa S, Venditti P. Skeletal muscle insulin resistance: role of mitochondria and other ROS sources. J Endocrinol 2017; 233:R15-R42. [PMID: 28232636 DOI: 10.1530/joe-16-0598] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
Abstract
At present, obesity is one of the most important public health problems in the world because it causes several diseases and reduces life expectancy. Although it is well known that insulin resistance plays a pivotal role in the development of type 2 diabetes mellitus (the more frequent disease in obese people) the link between obesity and insulin resistance is yet a matter of debate. One of the most deleterious effects of obesity is the deposition of lipids in non-adipose tissues when the capacity of adipose tissue is overwhelmed. During the last decade, reduced mitochondrial function has been considered as an important contributor to 'toxic' lipid metabolite accumulation and consequent insulin resistance. More recent reports suggest that mitochondrial dysfunction is not an early event in the development of insulin resistance, but rather a complication of the hyperlipidemia-induced reactive oxygen species (ROS) production in skeletal muscle, which might promote mitochondrial alterations, lipid accumulation and inhibition of insulin action. Here, we review the literature dealing with the mitochondria-centered mechanisms proposed to explain the onset of obesity-linked IR in skeletal muscle. We conclude that the different pathways leading to insulin resistance may act synergistically because ROS production by mitochondria and other sources can result in mitochondrial dysfunction, which in turn can further increase ROS production leading to the establishment of a harmful positive feedback loop.
Collapse
Affiliation(s)
- Sergio Di Meo
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Susanna Iossa
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Paola Venditti
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| |
Collapse
|
18
|
Henríquez-Olguín C, Díaz-Vegas A, Utreras-Mendoza Y, Campos C, Arias-Calderón M, Llanos P, Contreras-Ferrat A, Espinosa A, Altamirano F, Jaimovich E, Valladares DM. NOX2 Inhibition Impairs Early Muscle Gene Expression Induced by a Single Exercise Bout. Front Physiol 2016; 7:282. [PMID: 27471471 PMCID: PMC4944119 DOI: 10.3389/fphys.2016.00282] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/22/2016] [Indexed: 01/07/2023] Open
Abstract
Reactive oxygen species (ROS) participate as signaling molecules in response to exercise in skeletal muscle. However, the source of ROS and the molecular mechanisms involved in these phenomena are still not completely understood. The aim of this work was to study the role of skeletal muscle NADPH oxidase isoform 2 (NOX2) in the molecular response to physical exercise in skeletal muscle. BALB/c mice, pre-treated with a NOX2 inhibitor, apocynin, (3 mg/kg) or vehicle for 3 days, were swim-exercised for 60 min. Phospho–p47phox levels were significantly upregulated by exercise in flexor digitorum brevis (FDB). Moreover, exercise significantly increased NOX2 complex assembly (p47phox–gp91phox interaction) demonstrated by both proximity ligation assay and co-immunoprecipitation. Exercise-induced NOX2 activation was completely inhibited by apocynin treatment. As expected, exercise increased the mRNA levels of manganese superoxide dismutase (MnSOD), glutathione peroxidase (GPx), citrate synthase (CS), mitochondrial transcription factor A (tfam) and interleukin-6 (IL-I6) in FDB muscles. Moreover, the apocynin treatment was associated to a reduced activation of p38 MAP kinase, ERK 1/2, and NF-κB signaling pathways after a single bout of exercise. Additionally, the increase in plasma IL-6 elicited by exercise was decreased in apocynin-treated mice compared with the exercised vehicle-group (p < 0.001). These results were corroborated using gp91-dstat in an in vitro exercise model. In conclusion, NOX2 inhibition by both apocynin and gp91dstat, alters the intracellular signaling to exercise and electrical stimuli in skeletal muscle, suggesting that NOX2 plays a critical role in molecular response to an acute exercise.
Collapse
Affiliation(s)
- Carlos Henríquez-Olguín
- Facultad de Medicina, Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Universidad de ChileSantiago, Chile; Laboratory of Exercise Sciences, Clínica MEDSSantiago, Chile
| | - Alexis Díaz-Vegas
- Facultad de Medicina, Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Universidad de Chile Santiago, Chile
| | - Yildy Utreras-Mendoza
- Facultad de Medicina, Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Universidad de Chile Santiago, Chile
| | - Cristian Campos
- Facultad de Medicina, Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Universidad de Chile Santiago, Chile
| | - Manuel Arias-Calderón
- Facultad de Medicina, Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Universidad de Chile Santiago, Chile
| | - Paola Llanos
- Facultad de Odontología, Institute for Research in Dental Sciences, Universidad de Chile Santiago, Chile
| | - Ariel Contreras-Ferrat
- Facultad de Medicina, School of Medical Technology, Universidad de Chile Santiago, Chile
| | - Alejandra Espinosa
- Facultad de Medicina, School of Medical Technology, Universidad de Chile Santiago, Chile
| | - Francisco Altamirano
- Facultad de Medicina, Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Universidad de Chile Santiago, Chile
| | - Enrique Jaimovich
- Facultad de Medicina, Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Universidad de Chile Santiago, Chile
| | - Denisse M Valladares
- Facultad de Medicina, Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Universidad de Chile Santiago, Chile
| |
Collapse
|
19
|
Hussain SNA, Cornachione AS, Guichon C, Al Khunaizi A, de Souza Leite F, Petrof BJ, Mofarrahi M, Moroz N, de Varennes B, Goldberg P, Rassier DE. Prolonged controlled mechanical ventilation in humans triggers myofibrillar contractile dysfunction and myofilament protein loss in the diaphragm. Thorax 2016; 71:436-45. [DOI: 10.1136/thoraxjnl-2015-207559] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 01/06/2016] [Indexed: 12/16/2022]
|
20
|
Nguyen A, Duquette N, Mamarbachi M, Thorin E. Epigenetic Regulatory Effect of Exercise on Glutathione Peroxidase 1 Expression in the Skeletal Muscle of Severely Dyslipidemic Mice. PLoS One 2016; 11:e0151526. [PMID: 27010651 PMCID: PMC4806847 DOI: 10.1371/journal.pone.0151526] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 02/28/2016] [Indexed: 11/19/2022] Open
Abstract
Exercise is an effective approach for primary and secondary prevention of cardiovascular diseases (CVD) and loss of muscular mass and function. Its benefits are widely documented but incompletely characterized. It has been reported that exercise can induce changes in the expression of antioxidant enzymes including Sod2, Trx1, Prdx3 and Gpx1 and limits the rise in oxidative stress commonly associated with CVD. These enzymes can be subjected to epigenetic regulation, such as DNA methylation, in response to environmental cues. The aim of our study was to determine whether in the early stages of atherogenesis, in young severely dyslipidemic mice lacking LDL receptors and overexpressing human ApoB100 (LDLR-/-; hApoB+/+), exercise regulates differentially the expression of antioxidant enzymes by DNA methylation in the skeletal muscles that consume high levels of oxygen and thus generate high levels of reactive oxygen species. Expression of Sod2, Txr1, Prdx3 and Gpx1 was altered by 3 months of exercise and/or severe dyslipidemia in 6-mo dyslipidemic mice. Of these genes, only Gpx1 exhibited changes in DNA methylation associated with dyslipidemia and exercise: we observed both increased DNA methylation with dyslipidemia and a transient decrease in DNA methylation with exercise. These epigenetic alterations are found in the second exon of the Gpx1 gene and occur alongside with inverse changes in mRNA expression. Inhibition of expression by methylation of this specific locus was confirmed in vitro. In conclusion, Gpx1 expression in the mouse skeletal muscle can be altered by both exercise and dyslipidemia through changes in DNA methylation, leading to a fine regulation of free radical metabolism.
Collapse
Affiliation(s)
- Albert Nguyen
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Natacha Duquette
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Maya Mamarbachi
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Eric Thorin
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
21
|
Allen DG, Whitehead NP, Froehner SC. Absence of Dystrophin Disrupts Skeletal Muscle Signaling: Roles of Ca2+, Reactive Oxygen Species, and Nitric Oxide in the Development of Muscular Dystrophy. Physiol Rev 2016; 96:253-305. [PMID: 26676145 DOI: 10.1152/physrev.00007.2015] [Citation(s) in RCA: 309] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dystrophin is a long rod-shaped protein that connects the subsarcolemmal cytoskeleton to a complex of proteins in the surface membrane (dystrophin protein complex, DPC), with further connections via laminin to other extracellular matrix proteins. Initially considered a structural complex that protected the sarcolemma from mechanical damage, the DPC is now known to serve as a scaffold for numerous signaling proteins. Absence or reduced expression of dystrophin or many of the DPC components cause the muscular dystrophies, a group of inherited diseases in which repeated bouts of muscle damage lead to atrophy and fibrosis, and eventually muscle degeneration. The normal function of dystrophin is poorly defined. In its absence a complex series of changes occur with multiple muscle proteins showing reduced or increased expression or being modified in various ways. In this review, we will consider the various proteins whose expression and function is changed in muscular dystrophies, focusing on Ca(2+)-permeable channels, nitric oxide synthase, NADPH oxidase, and caveolins. Excessive Ca(2+) entry, increased membrane permeability, disordered caveolar function, and increased levels of reactive oxygen species are early changes in the disease, and the hypotheses for these phenomena will be critically considered. The aim of the review is to define the early damage pathways in muscular dystrophy which might be appropriate targets for therapy designed to minimize the muscle degeneration and slow the progression of the disease.
Collapse
Affiliation(s)
- David G Allen
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Nicholas P Whitehead
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Stanley C Froehner
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| |
Collapse
|
22
|
Abrigo J, Rivera JC, Simon F, Cabrera D, Cabello-Verrugio C. Transforming growth factor type beta (TGF-β) requires reactive oxygen species to induce skeletal muscle atrophy. Cell Signal 2016; 28:366-376. [PMID: 26825874 DOI: 10.1016/j.cellsig.2016.01.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 12/28/2015] [Accepted: 01/24/2016] [Indexed: 12/13/2022]
Abstract
Transforming growth factor beta 1 (TGF-β1) is a classical modulator of skeletal muscle and regulates several processes, such as myogenesis, regeneration, and muscle function in skeletal muscle diseases. Skeletal muscle atrophy, characterised by the loss of muscle strength and mass, is one of the pathological conditions regulated by TGF-β. Atrophy also results in increased myosin heavy chain (MHC) degradation and the expression of two muscle-specific E3 ubiquitin ligases, atrogin-1 and MuRF-1. Reactive oxygen species (ROS) are modulators of muscle wasting, and NAD(P)H oxidase (NOX) is one of the main sources of ROS. While it was recently found that TGF-β1 induces atrophy in skeletal muscle, the underlying mechanism is not fully understood. In this study, the role of NOX-derived ROS in skeletal muscle atrophy induced by TGF-β was assessed. TGF-β1 induced an atrophic effect in C2C12 myotubes, as evidenced by decreased myotube diameter and MHC levels, together with increased MuRF-1 levels. Concomitantly, TGF-β increased NOX-induced ROS contents. Interestingly, NOX inhibition through apocynin and the antioxidant treatment with N-acetyl cysteine (NAC) decreased increased ROS levels in myotubes. Additionally, both apocynin and NAC completely prevented the decreased MHC, decreased myotube diameter, and increased MuRF-1 induced by TGF-β. Injection of TGF-β1 into the tibialis anterior muscle induced atrophy, as observed by decreased fibre diameter and MHC levels, together with increased MuRF-1 levels. Likewise, TGF-β increased the ROS contents in the smaller fibres of skeletal muscle. Additionally, the administration of NAC to mice prevented all atrophic effects and the increase in ROS induced by TGF-β in the tibialis anterior. This is the first study to report that TGF-β has an atrophic effect dependent on NOX-induced ROS in skeletal muscle.
Collapse
Affiliation(s)
- Johanna Abrigo
- Laboratory of Biology and Molecular Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Juan Carlos Rivera
- Laboratory of Biology and Molecular Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Laboratory of Integrative Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile
| | - Daniel Cabrera
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Ciencias Químicas y Biológicas, Facultad de Salud, Universidad Bernardo O Higgins, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Biology and Molecular Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
| |
Collapse
|
23
|
Schmidt HHHW, Stocker R, Vollbracht C, Paulsen G, Riley D, Daiber A, Cuadrado A. Antioxidants in Translational Medicine. Antioxid Redox Signal 2015; 23:1130-43. [PMID: 26154592 PMCID: PMC4657516 DOI: 10.1089/ars.2015.6393] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE It is generally accepted that reactive oxygen species (ROS) scavenging molecules or antioxidants exert health-promoting effects and thus their consumption as food additives and nutraceuticals has been greatly encouraged. Antioxidants may be beneficial in situations of subclinical deficiency and increased demand or acutely upon high-dose infusion. However, to date, there is little clinical evidence for the long-term benefit of most antioxidants. Alarmingly, recent evidence points even to health risks, in particular for supplements of lipophilic antioxidants. RECENT ADVANCES The biological impact of ROS depends not only on their quantities but also on their chemical nature, (sub)cellular and tissue location, and the rates of their formation and degradation. Moreover, ROS serve important physiological functions; thus, inappropriate removal of ROS may cause paradoxical reductive stress and thereby induce or promote disease. CRITICAL ISSUES Any recommendation on antioxidants must be based on solid clinical evidence and patient-relevant outcomes rather than surrogate parameters. FUTURE DIRECTIONS Such evidence-based use may include site-directed application, time-limited high dosing, (functional) pharmacological repair of oxidized biomolecules, and triggers of endogenous antioxidant response systems. Ideally, these approaches need guidance by patient stratification through predictive biomarkers and possibly imaging modalities.
Collapse
Affiliation(s)
- Harald H H W Schmidt
- 1 Department of Pharmacology, CARIM, FHML, MIAS, Maastricht University , Maastricht, The Netherlands
| | - Roland Stocker
- 2 Victor Chang Cardiac Research Institute , Sydney, Australia .,3 University of New South Wales , Sydney, Australia
| | - Claudia Vollbracht
- 4 Hochschule Fresenius, University of Applied Sciences , Idstein, Germany
| | | | - Dennis Riley
- 6 Galera Therapeutics Inc., Malvern, Pennsylvania
| | - Andreas Daiber
- 7 Labor für Molekulare Kardiologie, II. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes Gutenberg-Universität , Mainz, Germany
| | - Antonio Cuadrado
- 8 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , ISCIII, Madrid, Spain .,9 Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC , Madrid, Spain .,10 Instituto de Investigación Sanitaria La Paz (IdiPaz) , Madrid, Spain .,11 Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid , Madrid, Spain
| |
Collapse
|
24
|
Diebold BA, Smith SM, Li Y, Lambeth JD. NOX2 As a Target for Drug Development: Indications, Possible Complications, and Progress. Antioxid Redox Signal 2015; 23:375-405. [PMID: 24512192 PMCID: PMC4545678 DOI: 10.1089/ars.2014.5862] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/08/2014] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE NOX2 is important for host defense, and yet is implicated in a large number of diseases in which inflammation plays a role in pathogenesis. These include acute and chronic lung inflammatory diseases, stroke, traumatic brain injury, and neurodegenerative diseases, including Alzheimer's and Parkinson's Diseases. RECENT ADVANCES Recent drug development programs have targeted several NOX isoforms that are implicated in a variety of diseases. The focus has been primarily on NOX4 and NOX1 rather than on NOX2, due, in part, to concerns about possible immunosuppressive side effects. Nevertheless, NOX2 clearly contributes to the pathogenesis of many inflammatory diseases, and its inhibition is predicted to provide a novel therapeutic approach. CRITICAL ISSUES Possible side effects that might arise from targeting NOX2 are discussed, including the possibility that such inhibition will contribute to increased infections and/or autoimmune disorders. The state of the field with regard to existing NOX2 inhibitors and targeted development of novel inhibitors is also summarized. FUTURE DIRECTIONS NOX2 inhibitors show particular promise for the treatment of inflammatory diseases, both acute and chronic. Theoretical side effects include pro-inflammatory and autoimmune complications and should be considered in any therapeutic program, but in our opinion, available data do not indicate that they are sufficiently likely to eliminate NOX2 as a drug target, particularly when weighed against the seriousness of many NOX2-related indications. Model studies demonstrating efficacy with minimal side effects are needed to encourage future development of NOX2 inhibitors as therapeutic agents.
Collapse
Affiliation(s)
- Becky A. Diebold
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Susan M.E. Smith
- Department of Biology and Physics, Kennesaw State University, Kennesaw, Georgia
| | - Yang Li
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - J. David Lambeth
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
25
|
Guo S, Chen X. The human Nox4: gene, structure, physiological function and pathological significance. J Drug Target 2015; 23:888-96. [PMID: 25950600 DOI: 10.3109/1061186x.2015.1036276] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Increased generation of reactive oxygen species (ROS) has been implicated in the pathogenesis of a variety of diseases such as cardiovascular diseases and cancer. NADPH oxidase (Nox), a multicomponent enzyme, has been identified as one of the key sources of ROS. Nox4, one of the seven members of Nox family (Nox1, Nox2, Nox3, Nox4, Nox5, Duox1 and Duox2), has been extensively investigated in recent years. Its unique structures result in the constitutive generation of hydrogen peroxide (H2O2) as the main product. As a key oxygen sensor, Nox4-derived H2O2 plays diverse roles in cell proliferation, migration and death. Increased expression of Nox4 in cancer has been observed, which participates in metastasis, angiogenesis and apoptosis. Expression of Nox4 in endothelial cells actively mediated endothelial activation, dysfunction and injury, which contributes to the development of atherosclerosis, hypertension, cardiac hypertrophy and among others. This article explores the experimental studies related to the gene, structure, physiological function and pathological significance of Nox4. As Nox4 might serve as a potential target for the therapy of cardiovascular diseases and cancer, the Nox4 inhibitor is also discussed in this article.
Collapse
Affiliation(s)
- Shuhui Guo
- a State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao , China
| | - Xiuping Chen
- a State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao , China
| |
Collapse
|
26
|
Souto Padron de Figueiredo A, Salmon AB, Bruno F, Jimenez F, Martinez HG, Halade GV, Ahuja SS, Clark RA, DeFronzo RA, Abboud HE, El Jamali A. Nox2 mediates skeletal muscle insulin resistance induced by a high fat diet. J Biol Chem 2015; 290:13427-39. [PMID: 25825489 DOI: 10.1074/jbc.m114.626077] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Indexed: 01/16/2023] Open
Abstract
Inflammation and oxidative stress through the production of reactive oxygen species (ROS) are consistently associated with metabolic syndrome/type 2 diabetes. Although the role of Nox2, a major ROS-generating enzyme, is well described in host defense and inflammation, little is known about its potential role in insulin resistance in skeletal muscle. Insulin resistance induced by a high fat diet was mitigated in Nox2-null mice compared with wild-type mice after 3 or 9 months on the diet. High fat feeding increased Nox2 expression, superoxide production, and impaired insulin signaling in skeletal muscle tissue of wild-type mice but not in Nox2-null mice. Exposure of C2C12 cultured myotubes to either high glucose concentration, palmitate, or H2O2 decreases insulin-induced Akt phosphorylation and glucose uptake. Pretreatment with catalase abrogated these effects, indicating a key role for H2O2 in mediating insulin resistance. Down-regulation of Nox2 in C2C12 cells by shRNA prevented insulin resistance induced by high glucose or palmitate but not H2O2. These data indicate that increased production of ROS in insulin resistance induced by high glucose in skeletal muscle cells is a consequence of Nox2 activation. This is the first report to show that Nox2 is a key mediator of insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
| | - Adam B Salmon
- The Sam and Ann Barshop Institute for Longevity and Aging Studies 15355 Lambda Drive, San Antonio, Texas 78245, and Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78229-3900
| | - Francesca Bruno
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900
| | - Fabio Jimenez
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900
| | - Herman G Martinez
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900
| | - Ganesh V Halade
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900
| | - Seema S Ahuja
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900, Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78229-3900
| | - Robert A Clark
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900, Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78229-3900
| | - Ralph A DeFronzo
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900, Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78229-3900
| | - Hanna E Abboud
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900, Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78229-3900
| | - Amina El Jamali
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900,
| |
Collapse
|
27
|
Zhou N, Lee WR, Abasht B. Messenger RNA sequencing and pathway analysis provide novel insights into the biological basis of chickens' feed efficiency. BMC Genomics 2015; 16:195. [PMID: 25886891 PMCID: PMC4414306 DOI: 10.1186/s12864-015-1364-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 02/20/2015] [Indexed: 11/21/2022] Open
Abstract
Background Advanced selection technologies have been developed and continually optimized to improve traits of agricultural importance; however, these methods have been primarily applied without knowledge of underlying biological changes that may be induced by selection. This study aims to characterize the biological basis of differences between chickens with low and high feed efficiency (FE) with a long-term goal of improving the ability to select for FE. Results High-throughput RNA sequencing was performed on 23 breast muscle samples from commercial broiler chickens with extremely high (n = 10) and low (n = 13) FE. An average of 34 million paired-end reads (75 bp) were produced for each sample, 80% of which were properly mapped to the chicken reference genome (Ensembl Galgal4). Differential expression analysis identified 1,059 genes (FDR < 0.05) that significantly divergently expressed in breast muscle between the high- and low-FE chickens. Gene function analysis revealed that genes involved in muscle remodeling, inflammatory response and free radical scavenging were mostly up-regulated in the high-FE birds. Additionally, growth hormone and IGFs/PI3K/Akt signaling pathways were enriched in differentially expressed genes, which might contribute to the high breast muscle yield in high-FE birds and partly explain the FE advantage of high-FE chickens. Conclusions This study provides novel insights into transcriptional differences in breast muscle between high- and low-FE broiler chickens. Our results show that feed efficiency is associated with breast muscle growth in these birds; furthermore, some physiological changes, e.g., inflammatory response and oxidative stress, may occur in the breast muscle of the high-FE chickens, which may be of concern for continued selection for both of these traits together in modern broiler chickens. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1364-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nan Zhou
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, 19716, USA.
| | | | - Behnam Abasht
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
28
|
Kamli MR, Kim J, Pokharel S, Jan AT, Lee EJ, Choi I. Expressional studies of the aldehyde oxidase (AOX1) gene during myogenic differentiation in C2C12 cells. Biochem Biophys Res Commun 2014; 450:1291-6. [DOI: 10.1016/j.bbrc.2014.06.126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 06/25/2014] [Indexed: 02/05/2023]
|
29
|
Van Thienen R, D'Hulst G, Deldicque L, Hespel P. Biochemical artifacts in experiments involving repeated biopsies in the same muscle. Physiol Rep 2014; 2:e00286. [PMID: 24819751 PMCID: PMC4098731 DOI: 10.14814/phy2.286] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Needle biopsies are being extensively used in clinical trials addressing muscular adaptation to exercise and diet. Still, the potential artifacts due to biopsy sampling are often overlooked. Healthy volunteers (n = 9) underwent two biopsies through a single skin incision in a pretest. Two days later (posttest) another biopsy was taken 3 cm proximally and 3 cm distally to the pretest incision. Muscle oxygenation status (tissue oxygenation index [TOI]) was measured by near‐infrared spectroscopy. Biopsy samples were analyzed for 40 key markers (mRNA and protein contents) of myocellular O2 sensing, inflammation, cell proliferation, mitochondrial biogenesis, protein synthesis and breakdown, oxidative stress, and energy metabolism. In the pretest, all measurements were identical between proximal and distal biopsies. However, compared with the pretest, TOI in the posttest was reduced in the proximal (−10%, P < 0.05), but not in the distal area. Conversely, most inflammatory markers were upregulated at the distal (100–500%, P < 0.05), but not at the proximal site. Overall, 29 of the 40 markers measured, equally distributed over all pathways studied, were either up‐ or downregulated by 50–500% (P < 0.05). In addition, 19 markers yielded conflicting results between the proximal and distal measurements (P < 0.05). This study clearly documents that prior muscle biopsies can cause major disturbances in myocellular signaling pathways in needle biopsies specimens sampled 48 h later. In addition, different biopsy sites within identical experimental conditions yielded conflicting results. This study clearly demonstrates that skeletal muscle biopsying per se, at least by causing local tissue inflammation and/or topical deoxygenation, can substantially alter biochemical events happening in needle biopsy specimens sampled at a later day in the same muscle belly. It is crucial to take into account these potential artifacts whenever investigating the cellular mechanisms implicated in adaptation to exercise, recovery, or hypoxia.
Collapse
Affiliation(s)
- Ruud Van Thienen
- Exercise Physiology Research Group - Department of Kinesiology, KU Leuven, Tervuursevest 101, Leuven, B-3001, Belgium
| | | | | | | |
Collapse
|
30
|
Guo Y, Gosker HR, Schols AMWJ, Kapchinsky S, Bourbeau J, Sandri M, Jagoe RT, Debigaré R, Maltais F, Taivassalo T, Hussain SNA. Autophagy in locomotor muscles of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2014; 188:1313-20. [PMID: 24228729 DOI: 10.1164/rccm.201304-0732oc] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Locomotor muscle atrophy develops in patients with chronic obstructive pulmonary disease (COPD) partly because of increased protein degradation by the ubiquitin-proteasome system. It is not known if autophagy also contributes to protein degradation. OBJECTIVES To investigate whether autophagy is enhanced in locomotor muscles of stable patients with COPD, to quantify autophagy-related gene expression in these muscles, and to identify mechanisms of autophagy induction. METHODS Muscle biopsies were obtained from two cohorts of control subjects and patients with COPD and the numbers of autophagosomes in the vastus lateralis and tibialis anterior muscles, the levels of LC3B protein lipidation, and the expression of autophagy-related genes were measured in the vastus lateralis muscle. To investigate potential pathways that might induce the activation of autophagy, measures were taken of protein kinase B (AKT), mTORC1, and AMPK pathway activation, transcription factor regulation, proteasome activation, and oxidative stress. MEASUREMENTS AND MAIN RESULTS Autophagy is enhanced in the locomotor muscles of patients with COPD as shown by significantly higher numbers of autophagosomes in affected muscles as compared with control subjects. Autophagosome number inversely correlates with FEV1. In the vastus lateralis, LC3B protein lipidation is increased by COPD and the expression of autophagy-related gene expressions is up-regulated. LC3B lipidation inversely correlates with thigh cross-sectional area, FEV1, and FEV1/FVC ratio. Enhanced autophagy is associated with activation of the AMPK pathway and FOXO transcription factors, inhibition of the mTORC1 and AKT pathways, and the development of oxidative stress. CONCLUSIONS Autophagy is significantly enhanced in locomotor muscles of stable patients with COPD. The degree of autophagy correlates with severity of muscle atrophy and lung function impairment.
Collapse
|
31
|
Schulz E, Wenzel P, Münzel T, Daiber A. Mitochondrial redox signaling: Interaction of mitochondrial reactive oxygen species with other sources of oxidative stress. Antioxid Redox Signal 2014; 20:308-24. [PMID: 22657349 PMCID: PMC3887453 DOI: 10.1089/ars.2012.4609] [Citation(s) in RCA: 189] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Oxidative stress is a well established hallmark of cardiovascular disease and there is strong evidence for a causal role of reactive oxygen and nitrogen species (RONS) therein. RECENT ADVANCES Improvement of cardiovascular complications by genetic deletion of RONS producing enzymes and overexpression of RONS degrading enzymes proved the involvement of these species in cardiovascular disease at a molecular level. Vice versa, overexpression of RONS producing enzymes as well as deletion of antioxidant enzymes was demonstrated to aggravate cardiovascular complications. CRITICAL ISSUES With the present overview we present and discuss different pathways how mitochondrial RONS interact (crosstalk) with other sources of oxidative stress, namely NADPH oxidases, xanthine oxidase and an uncoupled nitric oxide synthase. The potential mechanisms of how this crosstalk proceeds are discussed in detail. Several examples from the literature are summarized (including hypoxia, angiotensin II mediated vascular dysfunction, cellular starvation, nitrate tolerance, aging, hyperglycemia, β-amyloid stress and others) and the underlying mechanisms are put together to a more general concept of redox-based activation of different sources of RONS via enzyme-specific "redox switches". Mitochondria play a key role in this concept providing redox triggers for oxidative damage in the cardiovascular system but also act as amplifiers to increase the burden of oxidative stress. FUTURE DIRECTIONS Based on these considerations, the characterization of the role of mitochondrial RONS formation in cardiac disease as well as inflammatory processes but also the role of mitochondria as potential therapeutic targets in these pathophysiological states should be addressed in more detail in the future.
Collapse
Affiliation(s)
- Eberhard Schulz
- 1 2nd Medical Clinic, Molecular Cardiology, Medical Center of the Johannes Gutenberg University , Mainz, Germany
| | | | | | | |
Collapse
|
32
|
Clanton TL, Hogan MC, Gladden LB. Regulation of cellular gas exchange, oxygen sensing, and metabolic control. Compr Physiol 2013; 3:1135-90. [PMID: 23897683 DOI: 10.1002/cphy.c120030] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cells must continuously monitor and couple their metabolic requirements for ATP utilization with their ability to take up O2 for mitochondrial respiration. When O2 uptake and delivery move out of homeostasis, cells have elaborate and diverse sensing and response systems to compensate. In this review, we explore the biophysics of O2 and gas diffusion in the cell, how intracellular O2 is regulated, how intracellular O2 levels are sensed and how sensing systems impact mitochondrial respiration and shifts in metabolic pathways. Particular attention is paid to how O2 affects the redox state of the cell, as well as the NO, H2S, and CO concentrations. We also explore how these agents can affect various aspects of gas exchange and activate acute signaling pathways that promote survival. Two kinds of challenges to gas exchange are also discussed in detail: when insufficient O2 is available for respiration (hypoxia) and when metabolic requirements test the limits of gas exchange (exercising skeletal muscle). This review also focuses on responses to acute hypoxia in the context of the original "unifying theory of hypoxia tolerance" as expressed by Hochachka and colleagues. It includes discourse on the regulation of mitochondrial electron transport, metabolic suppression, shifts in metabolic pathways, and recruitment of cell survival pathways preventing collapse of membrane potential and nuclear apoptosis. Regarding exercise, the issues discussed relate to the O2 sensitivity of metabolic rate, O2 kinetics in exercise, and influences of available O2 on glycolysis and lactate production.
Collapse
Affiliation(s)
- T L Clanton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA.
| | | | | |
Collapse
|
33
|
Sullivan-Gunn MJ, Lewandowski PA. Elevated hydrogen peroxide and decreased catalase and glutathione peroxidase protection are associated with aging sarcopenia. BMC Geriatr 2013; 13:104. [PMID: 24093947 PMCID: PMC3853025 DOI: 10.1186/1471-2318-13-104] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 09/30/2013] [Indexed: 12/22/2022] Open
Abstract
Background Sarcopenia is the progressive loss of skeletal muscle that contributes to the decline in physical function during aging. A higher level of oxidative stress has been implicated in aging sarcopenia. The current study aims to determine if the higher level of oxidative stress is a result of increased superoxide (O2‾) production by the NADPH oxidase (NOX) enzyme or decrease in endogenous antioxidant enzyme protection. Methods Female Balb/c mice were assigned to 4 age groups; 6, 12, 18 and 24 months. Body weight and animal survival rates were recorded over the course of the study. Skeletal muscle tissues were collected and used to measure NOX subunit mRNA, O2‾ levels and antioxidant enzymes. Results Key subunit components of NOX expression were elevated in skeletal muscle at 18 months, when sarcopenia was first evident. Increased superoxide dismutase 1 (SOD1) activity suggests an increase in O2‾ dismutation and this was further supported by elevated levels of hydrogen peroxide (H2O2) and decline in catalase and glutathione peroxidase (GPx) antioxidant protection in skeletal muscle at this time. NOX expression was also higher in skeletal muscle at 24 months, however this was coupled with elevated levels of O2‾ and a decline in SOD1 activity, compared to 6 and 12 months but was not associated with further loss of muscle mass. Conclusions While the source of ROS in sarcopenic muscle remains unknown, this study provides evidence that the NOX enzyme could be involved in ROS production by regulating superoxide in ageing muscles. This study also suggests that H2O2 is the key ROS in the onset of sarcopenia and that the decline in antioxidant protection by catalase and GPx is indicative of antioxidant dysfunction and may therefore be a major contributing factor in the development or onset of sarcopenia. Furthermore, the changes in ROS and antioxidant activity after sarcopenia was first evident gives some evidence for a compensatory mechanism, in response to insult, in order to maintain muscle integrity.
Collapse
|
34
|
Sakellariou GK, Jackson MJ, Vasilaki A. Redefining the major contributors to superoxide production in contracting skeletal muscle. The role of NAD(P)H oxidases. Free Radic Res 2013; 48:12-29. [PMID: 23915064 DOI: 10.3109/10715762.2013.830718] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The production of reactive oxygen and nitrogen species (RONS) by skeletal muscle is important as it (i) underlies oxidative damage in many degenerative muscle pathologies and (ii) plays multiple regulatory roles by fulfilling important cellular functions. Superoxide and nitric oxide (NO) are the primary radical species produced by skeletal muscle and studies in the early 1980s demonstrated that their generation is augmented during contractile activity. Over the past 30 years considerable research has been undertaken to identify the major sites that contribute to the increased rate of RONS generation in response to contractions. It is widely accepted that NO is regulated by the nitric oxide synthases, however the sites that modulate changes in superoxide during exercise remain unclear. Despite the initial indications that the mitochondrial electron transport chain was the predominant source of superoxide during activity, with the development of analytical methods a number of alternative potential sites have been identified including the NAD(P)H oxidases, xanthine oxidase, cyclooxygenases, and lipoxygenases linked to the activity of the phospholipase A2 enzymes. In the present review we outline the subcellular sites that modulate intracellular changes in superoxide in skeletal muscle and based on the available experimental evidence in the literature we conclude that the NAD(P)H oxidases are likely to be the major superoxide generating sources in contracting skeletal muscle.
Collapse
Affiliation(s)
- G K Sakellariou
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing, Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool , Liverpool , UK
| | | | | |
Collapse
|
35
|
Espinosa A, Campos C, Díaz-Vegas A, Galgani JE, Juretic N, Osorio-Fuentealba C, Bucarey JL, Tapia G, Valenzuela R, Contreras-Ferrat A, Llanos P, Jaimovich E. Insulin-dependent H2O2 production is higher in muscle fibers of mice fed with a high-fat diet. Int J Mol Sci 2013; 14:15740-54. [PMID: 23899788 PMCID: PMC3759883 DOI: 10.3390/ijms140815740] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 07/20/2013] [Accepted: 07/24/2013] [Indexed: 12/13/2022] Open
Abstract
Insulin resistance is defined as a reduced ability of insulin to stimulate glucose utilization. C57BL/6 mice fed with a high-fat diet (HFD) are a model of insulin resistance. In skeletal muscle, hydrogen peroxide (H2O2) produced by NADPH oxidase 2 (NOX2) is involved in signaling pathways triggered by insulin. We evaluated oxidative status in skeletal muscle fibers from insulin-resistant and control mice by determining H2O2 generation (HyPer probe), reduced-to-oxidized glutathione ratio and NOX2 expression. After eight weeks of HFD, insulin-dependent glucose uptake was impaired in skeletal muscle fibers when compared with control muscle fibers. Insulin-resistant mice showed increased insulin-stimulated H2O2 release and decreased reduced-to-oxidized glutathione ratio (GSH/GSSG). In addition, p47phox and gp91phox (NOX2 subunits) mRNA levels were also high (~3-fold in HFD mice compared to controls), while protein levels were 6.8- and 1.6-fold higher, respectively. Using apocynin (NOX2 inhibitor) during the HFD feeding period, the oxidative intracellular environment was diminished and skeletal muscle insulin-dependent glucose uptake restored. Our results indicate that insulin-resistant mice have increased H2O2 release upon insulin stimulation when compared with control animals, which appears to be mediated by an increase in NOX2 expression.
Collapse
Affiliation(s)
- Alejandra Espinosa
- School of Medical Technology, Faculty of Medicine, University of Chile, Santiago 8380455, Chile; E-Mails: (C.C.); (A.D.-V.)
- Center for Molecular Studies of the Cell, Santiago 8380453, Chile; E-Mails: (C.O.-F.); (A.C.-F.); (P.L.); (E.J.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +56-02-297-866-64; Fax: +56-02-297-866-82
| | - Cristian Campos
- School of Medical Technology, Faculty of Medicine, University of Chile, Santiago 8380455, Chile; E-Mails: (C.C.); (A.D.-V.)
| | - Alexis Díaz-Vegas
- School of Medical Technology, Faculty of Medicine, University of Chile, Santiago 8380455, Chile; E-Mails: (C.C.); (A.D.-V.)
- Center for Molecular Studies of the Cell, Santiago 8380453, Chile; E-Mails: (C.O.-F.); (A.C.-F.); (P.L.); (E.J.)
| | - José E. Galgani
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; E-Mail:
| | - Nevenka Juretic
- Faculty of Medicine, Institute of Biomedical Sciences, Santiago 8380453, Chile; E-Mails: (N.J.); (G.T.); (R.V.)
| | - César Osorio-Fuentealba
- Center for Molecular Studies of the Cell, Santiago 8380453, Chile; E-Mails: (C.O.-F.); (A.C.-F.); (P.L.); (E.J.)
| | - José L. Bucarey
- School of Medicine, University of Valparaíso, Valparaíso 2341369, Chile; E-Mail:
| | - Gladys Tapia
- Faculty of Medicine, Institute of Biomedical Sciences, Santiago 8380453, Chile; E-Mails: (N.J.); (G.T.); (R.V.)
| | - Rodrigo Valenzuela
- Faculty of Medicine, Institute of Biomedical Sciences, Santiago 8380453, Chile; E-Mails: (N.J.); (G.T.); (R.V.)
| | - Ariel Contreras-Ferrat
- Center for Molecular Studies of the Cell, Santiago 8380453, Chile; E-Mails: (C.O.-F.); (A.C.-F.); (P.L.); (E.J.)
| | - Paola Llanos
- Center for Molecular Studies of the Cell, Santiago 8380453, Chile; E-Mails: (C.O.-F.); (A.C.-F.); (P.L.); (E.J.)
| | - Enrique Jaimovich
- Center for Molecular Studies of the Cell, Santiago 8380453, Chile; E-Mails: (C.O.-F.); (A.C.-F.); (P.L.); (E.J.)
- Faculty of Medicine, Institute of Biomedical Sciences, Santiago 8380453, Chile; E-Mails: (N.J.); (G.T.); (R.V.)
| |
Collapse
|
36
|
Rajakumar D, Alexander M, Oommen A. Oxidative stress, NF-κB and the ubiquitin proteasomal pathway in the pathology of calpainopathy. Neurochem Res 2013; 38:2009-18. [PMID: 23846623 DOI: 10.1007/s11064-013-1107-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 05/19/2013] [Accepted: 06/29/2013] [Indexed: 01/11/2023]
Abstract
The neuromuscular disorder, calpainopathy (LGMD 2A), is a major muscular dystrophy classified under limb girdle muscular dystrophies. Genetic mutations of the enzyme calpain 3 cause LGMD 2A. Calpainopathy is phenotypically observed as progressive muscle wasting and weakness. Pathomechanisms of muscle wasting of calpainopathy remain poorly understood. Oxidative stress, NF-κB and the ubiquitin proteasomal pathway underlie the pathology of several muscle wasting conditions but their role in calpainopathic dystrophy is not known. Oxidative and nitrosative stress, the source of reactive oxygen species, NF-κB signaling and protein ubiquitinylation were studied in 15 calpainopathic and 8 healthy control human muscle biopsies. Oxidative stress and NF-κB/IKK β signaling were increased in calpainopathic muscle and may contribute to increased protein ubiquitinylation and muscle protein loss. Preventing oxidative stress or inhibition of NF-κB signaling could be considered for treatment of LGMD 2A.
Collapse
Affiliation(s)
- Dhanarajan Rajakumar
- Neurochemistry Laboratory, Department of Neurological Sciences, Christian Medical College, Vellore, Tamil Nadu, India,
| | | | | |
Collapse
|
37
|
Michailidis Y, Karagounis LG, Terzis G, Jamurtas AZ, Spengos K, Tsoukas D, Chatzinikolaou A, Mandalidis D, Stefanetti RJ, Papassotiriou I, Athanasopoulos S, Hawley JA, Russell AP, Fatouros IG. Thiol-based antioxidant supplementation alters human skeletal muscle signaling and attenuates its inflammatory response and recovery after intense eccentric exercise. Am J Clin Nutr 2013; 98:233-45. [PMID: 23719546 DOI: 10.3945/ajcn.112.049163] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The major thiol-disulfide couple of reduced glutathione (GSH) and oxidized glutathione is a key regulator of major transcriptional pathways regulating aseptic inflammation and recovery of skeletal muscle after aseptic injury. Antioxidant supplementation may hamper exercise-induced cellular adaptations. OBJECTIVE The objective was to examine how thiol-based antioxidant supplementation affects skeletal muscle's performance and redox-sensitive signaling during the inflammatory and repair phases associated with exercise-induced microtrauma. DESIGN In a double-blind, crossover design, 10 men received placebo or N-acetylcysteine (NAC; 20 mg · kg(-1) · d(-1)) after muscle-damaging exercise (300 eccentric contractions). In each trial, muscle performance was measured at baseline, after exercise, 2 h after exercise, and daily for 8 consecutive days. Muscle biopsy samples from vastus lateralis and blood samples were collected before exercise and 2 h, 2 d, and 8 d after exercise. RESULTS NAC attenuated the elevation of inflammatory markers of muscle damage (creatine kinase activity, C-reactive protein, proinflammatory cytokines), nuclear factor κB phosphorylation, and the decrease in strength during the first 2 d of recovery. NAC also blunted the increase in phosphorylation of protein kinase B, mammalian target of rapamycin, p70 ribosomal S6 kinase, ribosomal protein S6, and mitogen activated protein kinase p38 at 2 and 8 d after exercise. NAC also abolished the increase in myogenic determination factor and reduced tumor necrosis factor-α 8 d after exercise. Performance was completely recovered only in the placebo group. CONCLUSION Although thiol-based antioxidant supplementation enhances GSH availability in skeletal muscle, it disrupts the skeletal muscle inflammatory response and repair capability, potentially because of a blunted activation of redox-sensitive signaling pathways. This trial was registered at clinicaltrials.gov as NCT01778309.
Collapse
Affiliation(s)
- Yannis Michailidis
- Democritus University of Thrace, Department of Physical Education and Sport Sciences, Komotini, Greece
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Acharya S, Peters AM, Norton AS, Murdoch GK, Hill RA. Change in Nox4 expression is accompanied by changes in myogenic marker expression in differentiating C2C12 myoblasts. Pflugers Arch 2013; 465:1181-96. [PMID: 23503725 DOI: 10.1007/s00424-013-1241-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 01/10/2013] [Accepted: 02/04/2013] [Indexed: 01/10/2023]
Abstract
Myoblast differentiation is mediated by a cascade of changes in gene expression including transcription factors such as myogenin. Subsequent to myoblast differentiation, there is an increase in expression of the transmembrane protein NADPH oxidase (Nox). Nox is one of the primary factors for the generation of reactive oxygen species (ROS) in myogenic (C2C12) cells. Recently, ROS have been shown to be important regulators of several intracellular signaling pathways, and the full extent of their regulatory roles is yet to be discovered. In the present study, qRT PCR analysis demonstrated that Nox4 isoform is primarily expressed in differentiating C2C12 cells and contributes to the generation of ROS in C2C12 myoblast during differentiation. Over-expression and silencing of Nox4 expression during myoblast differentiation was accompanied by a reduction in intracellular ROS concentrations and an alteration in the expression patterns of Myf5, Pax7, MyoD1, and myogenin. This modulation was found to be associated with ERK1/2 phosphorylation. In both over-expression and reduced expression of Nox4, we found significant reductions in ERK1/2 phosphorylation. This indicates that cellular differentiation may be affected by Nox4-mediated endogenous ROS generation. These data suggest a new opportunity to study the temporal expression of Nox4 in the generation of ROS accompanying changes in myogenic differentiation.
Collapse
Affiliation(s)
- S Acharya
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID 83844, USA
| | | | | | | | | |
Collapse
|
39
|
Sakellariou GK, Vasilaki A, Palomero J, Kayani A, Zibrik L, McArdle A, Jackson MJ. Studies of mitochondrial and nonmitochondrial sources implicate nicotinamide adenine dinucleotide phosphate oxidase(s) in the increased skeletal muscle superoxide generation that occurs during contractile activity. Antioxid Redox Signal 2013; 18:603-21. [PMID: 23050834 PMCID: PMC3549212 DOI: 10.1089/ars.2012.4623] [Citation(s) in RCA: 197] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS The sources of cytosolic superoxide in skeletal muscle have not been defined. This study examined the subcellular sites that contribute to cytosolic superoxide in mature single muscle fibers at rest and during contractile activity. RESULTS Isolated fibers from mouse flexor digitorum brevis loaded with superoxide and nitric-oxide-sensitive fluorescent probes, specific pathway inhibitors and immunolocalization techniques were used to identify subcellular sites contributing to cytosolic superoxide. Treatment with the electron transport chain complex III inhibitor, antimycin A, but not the complex I inhibitor, rotenone, caused increased cytosolic superoxide through release from the mitochondrial intermembrane space via voltage-dependent anion or Bax channels, but inhibition of these channels did not affect contraction-induced increases in cytosolic superoxide. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors decreased cytosolic superoxide at rest and following contractions. Protein and mRNA expression of NADPH oxidase subunits was demonstrated in single fibers. NOX2, NOX4, and p22(phox) subunits localized to the sarcolemma and transverse tubules; NOX4 was additionally expressed in mitochondria. Regulatory p40(phox) and p67(phox) proteins were found in the cytoplasm of resting fibers, but following contractions, p40(phox) appeared to translocate to the sarcolemma. INNOVATION Superoxide and other reactive oxygen species generated by skeletal muscle are important regulators of muscle force production and adaptations to contractions. This study has defined the relative contribution of mitochondrial and cytosolic sources of superoxide within the cytosol of single muscle fibers at rest and during contractions. CONCLUSION Muscle mitochondria do not modulate cytosolic superoxide in skeletal muscle but NADPH oxidase is a major contributor both at rest and during contractions.
Collapse
Affiliation(s)
- Giorgos Konstantinos Sakellariou
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
40
|
Sciancalepore M, Luin E, Parato G, Ren E, Giniatullin R, Fabbretti E, Lorenzon P. Reactive oxygen species contribute to the promotion of the ATP-mediated proliferation of mouse skeletal myoblasts. Free Radic Biol Med 2012; 53:1392-8. [PMID: 22917975 DOI: 10.1016/j.freeradbiomed.2012.08.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 07/09/2012] [Accepted: 08/02/2012] [Indexed: 11/29/2022]
Abstract
Reactive oxygen species (ROS) and extracellular adenosine 5'-triphosphate (ATP) participate in autocrine and paracrine regulation in skeletal muscle. However, the link between these two signaling systems is not well established. Here, we studied cell proliferation as a possible consequence of the trophic effect of ATP in cultured skeletal mouse myoblasts and we tested the possibility that low concentrations of ROS represent the intermediate signaling molecule mediating this effect. Exposure to 10 μM ATP increased proliferation of mouse myoblasts by ~20%. ATP also induced intracellular Ca(2+) oscillations, which were independent of extracellular Ca(2+). Both effects of ATP were prevented by suramin, a broad-spectrum purinergic P2 receptor antagonist. In contrast, the adenosine receptor blocker CGS-15943 did not modify the ATP-mediated effects. Consistent with this, adenosine per se did not change myoblast growth, indicating the direct action of ATP via P2 receptor activation. The proliferative effect of ATP was prevented after depletion of hydrogen peroxide (H(2)O(2)) by the peroxidase enzyme catalase. Low-micromolar concentrations of exogenous H(2)O(2) mimicked the stimulatory effect of ATP on myoblast growth. DCF imaging revealed ATP-induced catalase and DPI-sensitive ROS production in myoblasts. In conclusion, our results indicate that extracellular ATP controls mouse myoblast proliferation via induction of ROS generation.
Collapse
Affiliation(s)
- Marina Sciancalepore
- Department of Life Sciences and Centre for Neuroscience (BRAIN), University of Trieste, I-34127 Trieste, Italy.
| | | | | | | | | | | | | |
Collapse
|
41
|
Choi J. Oxidative stress, endogenous antioxidants, alcohol, and hepatitis C: pathogenic interactions and therapeutic considerations. Free Radic Biol Med 2012; 52:1135-50. [PMID: 22306508 DOI: 10.1016/j.freeradbiomed.2012.01.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 01/04/2012] [Accepted: 01/12/2012] [Indexed: 12/16/2022]
Abstract
Hepatitis C virus (HCV) is a blood-borne pathogen that was identified as an etiologic agent of non-A, non-B hepatitis in 1989. HCV is estimated to have infected at least 170 million people worldwide. The majority of patients infected with HCV do not clear the virus and become chronically infected, and chronic HCV infection increases the risk for hepatic steatosis, cirrhosis, and hepatocellular carcinoma. HCV induces oxidative/nitrosative stress from multiple sources, including inducible nitric oxide synthase, the mitochondrial electron transport chain, hepatocyte NAD(P)H oxidases, and inflammation, while decreasing glutathione. The cumulative oxidative burden is likely to promote both hepatic and extrahepatic conditions precipitated by HCV through a combination of local and more distal effects of reactive species, and clinical, animal, and in vitro studies strongly point to a role of oxidative/nitrosative stress in HCV-induced pathogenesis. Oxidative stress and hepatopathogenesis induced by HCV are exacerbated by even low doses of alcohol. Alcohol and reactive species may have other effects on hepatitis C patients such as modulation of the host immune system, viral replication, and positive selection of HCV sequence variants that contribute to antiviral resistance. This review summarizes the current understanding of redox interactions of HCV, outlining key experimental findings, directions for future research, and potential applications to therapy.
Collapse
Affiliation(s)
- Jinah Choi
- Department of Molecular Cell Biology, School of Natural Sciences, University of California at Merced, Merced, CA 95343, USA.
| |
Collapse
|
42
|
Kennedy KAM, Sandiford SDE, Skerjanc IS, Li SSC. Reactive oxygen species and the neuronal fate. Cell Mol Life Sci 2012; 69:215-21. [PMID: 21947442 PMCID: PMC11114775 DOI: 10.1007/s00018-011-0807-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 07/29/2011] [Accepted: 08/18/2011] [Indexed: 10/17/2022]
Abstract
Aberrant or elevated levels of reactive oxygen species (ROS) can mediate deleterious cellular effects, including neuronal toxicity and degeneration observed in the etiology of a number of pathological conditions, including Alzheimer's and Parkinson's diseases. Nevertheless, ROS can be generated in a controlled manner and can regulate redox sensitive transcription factors such as NFκB, AP-1 and NFAT. Moreover, ROS can modulate the redox state of tyrosine phosphorylated proteins, thereby having an impact on many transcriptional networks and signaling cascades important for neurogenesis. A large body of literature links the controlled generation of ROS at low-to-moderate levels with the stimulation of differentiation in certain developmental programs such as neurogenesis. In this regard, ROS are involved in governing the acquisition of the neural fate-from neural induction to the elaboration of axons. Here, we summarize and discuss the growing body of literature that describe a role for ROS signaling in neuronal development.
Collapse
Affiliation(s)
- Karen A. M. Kennedy
- Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, ON N6A 5C1 Canada
| | - Shelley D. E. Sandiford
- Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, ON N6A 5C1 Canada
| | - Ilona S. Skerjanc
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5 Canada
| | - Shawn S.-C. Li
- Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, ON N6A 5C1 Canada
| |
Collapse
|
43
|
Jiang F, Zhang Y, Dusting GJ. NADPH oxidase-mediated redox signaling: roles in cellular stress response, stress tolerance, and tissue repair. Pharmacol Rev 2011; 63:218-42. [PMID: 21228261 DOI: 10.1124/pr.110.002980] [Citation(s) in RCA: 444] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
NADPH oxidase (Nox) has a dedicated function of generating reactive oxygen species (ROS). Accumulating evidence suggests that Nox has an important role in signal transduction in cellular stress responses. We have reviewed the current evidence showing that the Nox system can be activated by a collection of chemical, physical, and biological cellular stresses. In many circumstances, Nox activation fits to the cellular stress response paradigm, in that (1) the response can be initiated by various forms of cellular stresses; (2) Nox-derived ROS may activate mitogen-activated protein kinases (extracellular signal-regulated kinase, p38) and c-Jun NH(2)-terminal kinase, which are the core of the cell stress-response signaling network; and (3) Nox is involved in the development of stress cross-tolerance. Activation of the cell survival pathway by Nox may promote cell adaptation to stresses, whereas Nox may also convey signals toward apoptosis in irreversibly injured cells. At later stage after injury, Nox is involved in tissue repair by modulating cell proliferation, angiogenesis, and fibrosis. We suggest that Nox may have an integral role in cell stress responses and the subsequent tissue repair process. Understanding Nox-mediated redox signaling mechanisms may be of prominent significance at the crossroads of directing cellular responses to stress, aiming at either enhancing the stress resistance (in such situations as preventing ischemia-reperfusion injuries and accelerating wound healing) or sensitizing the stress-induced cytotoxicity for proliferative diseases such as cancer. Therefore, an optimal outcome of interventions on Nox will only be achieved when this is dealt with in a timely and disease-and stage-specific manner.
Collapse
Affiliation(s)
- Fan Jiang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan, Shandong 250012, China.
| | | | | |
Collapse
|
44
|
Whitehead NP, Yeung EW, Froehner SC, Allen DG. Skeletal muscle NADPH oxidase is increased and triggers stretch-induced damage in the mdx mouse. PLoS One 2010; 5:e15354. [PMID: 21187957 PMCID: PMC3004864 DOI: 10.1371/journal.pone.0015354] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 11/11/2010] [Indexed: 11/19/2022] Open
Abstract
Recent studies have shown that oxidative stress contributes to the pathogenesis of muscle damage in dystrophic (mdx) mice. In this study we have investigated the role of NADPH oxidase as a source of the oxidative stress in these mice. The NADPH oxidase subunits gp91phox, p67phox and rac 1 were increased 2–3 fold in tibilais anterior muscles from mdx mice compared to wild type. Importantly, this increase occurred in 19 day old mice, before the onset of muscle necrosis and inflammation, suggesting that NADPH oxidase is an important source of oxidative stress in mdx muscle. In muscles from 9 week old mdx mice, gp91phox and p67phox were increased 3–4 fold and NADPH oxidase superoxide production was 2 times greater than wild type. In single fibers from mdx muscle NADPH oxidase subunits were all located on or near the sarcolemma, except for p67phox,which was expressed in the cytosol. Pharmacological inhibition of NADPH oxidase significantly reduced the intracellular Ca2+ rise following stretched contractions in mdx single fibers, and also attenuated the loss of muscle force. These results suggest that NADPH oxidase is a major source of reactive oxygen species in dystrophic muscle and its enhanced activity has a stimulatory effect on stretch-induced Ca2+ entry, a key mechanism for muscle damage and functional impairment.
Collapse
Affiliation(s)
- Nicholas P Whitehead
- Discipline of Physiology, Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia.
| | | | | | | |
Collapse
|
45
|
Hussain SNA, Mofarrahi M, Sigala I, Kim HC, Vassilakopoulos T, Maltais F, Bellenis I, Chaturvedi R, Gottfried SB, Metrakos P, Danialou G, Matecki S, Jaber S, Petrof BJ, Goldberg P. Mechanical ventilation-induced diaphragm disuse in humans triggers autophagy. Am J Respir Crit Care Med 2010; 182:1377-86. [PMID: 20639440 DOI: 10.1164/rccm.201002-0234oc] [Citation(s) in RCA: 174] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Controlled mechanical ventilation (CMV) results in atrophy of the human diaphragm. The autophagy-lysosome pathway (ALP) contributes to skeletal muscle proteolysis, but its contribution to diaphragmatic protein degradation in mechanically ventilated patients is unknown. OBJECTIVES To evaluate the autophagy pathway responses to CMV in the diaphragm and limb muscles of humans and to identify the roles of FOXO transcription factors in these responses. METHODS Muscle biopsies were obtained from nine control subjects and nine brain-dead organ donors. Subjects were mechanically ventilated for 2 to 4 hours and 15 to 276 hours, respectively. Activation of the ubiquitin-proteasome system was detected by measuring mRNA expressions of Atrogin-1, MURF1, and protein expressions of UBC2, UBC4, and the α subunits of the 20S proteasome (MCP231). Activation of the ALP was detected by electron microscopy and by measuring the expressions of several autophagy-related genes. Total carbonyl content and HNE-protein adduct formation were measured to assess oxidative stress. Total AKT, phosphorylated and total FOXO1, and FOXO3A protein levels were also measured. MEASUREMENTS AND MAIN RESULTS Prolonged CMV triggered activation of the ALP as measured by the appearance of autophagosomes in the diaphragm and increased expressions of autophagy-related genes, as compared with controls. Induction of autophagy was associated with increased protein oxidation and enhanced expression of the FOXO1 gene, but not the FOXO3A gene. CMV also triggered the inhibition of both AKT expression and FOXO1 phosphorylation. CONCLUSIONS We propose that prolonged CMV causes diaphragm disuse, which, in turn, leads to activation of the ALP through oxidative stress and the induction of the FOXO1 transcription factor.
Collapse
|
46
|
Gordillo G, Fang H, Park H, Roy S. Nox-4-dependent nuclear H2O2 drives DNA oxidation resulting in 8-OHdG as urinary biomarker and hemangioendothelioma formation. Antioxid Redox Signal 2010; 12:933-43. [PMID: 19817625 PMCID: PMC2935344 DOI: 10.1089/ars.2009.2917] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hemangioendotheliomas are classified as endothelial cell tumors, which are the most common soft tissue tumors in infants. In a murine model of hemangioendothelioma, we previously showed that MCP-1 is required for its development and that the expression of MCP-1 in EOMA cells is redox sensitive. Here, we sought to identify the source of oxidants that drive hemangioendothelioma formation. Seven known isoforms exist of the catalytic subunit gp91. Only the nox-4 isoform of gp91 was present in EOMA cells, in contrast with non-tumor-forming murine endothelial cells that contained multiple forms of nox. Nox-4 knockdown markedly attenuated MCP-1 expression and hemangioendothelioma formation. We report that in EOMA cells, nox-4 is localized such that it delivers H2O2 to the nuclear compartment. Such delivery of H2O2 causes oxidative modification of DNA, which can be detected in the urine of tumor-bearing mice as 8-hydroxy-2-deoxyguanosine. Iron chelation by in vivo administration of deferoxamine improved tumor outcomes. The current state of information connects nox-4 to MCP-1 to form a major axis of control that regulates the fate of hemangioendothelioma development in vivo.
Collapse
Affiliation(s)
- Gayle Gordillo
- Division of Plastic Surgery, Department of Surgery, Davis Heart Lung Research Institute, The Ohio State University, Columbus, Ohio 43212, USA.
| | | | | | | |
Collapse
|
47
|
Kim HC, Mofarrahi M, Vassilakopoulos T, Maltais F, Sigala I, Debigare R, Bellenis I, Hussain SNA. Expression and Functional Significance of Nicotinamide N-methyl Transferase in Skeletal Muscles of Patients with Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2010; 181:797-805. [DOI: 10.1164/rccm.200906-0936oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
48
|
Abstract
The free radical theory of aging proposes that ROS (reactive oxygen species) are major driving forces of aging, and are also critically involved in cellular senescence. Besides the mitochondrial respiratory chain, alternative sources of ROS have been described that might contribute to cellular senescence. Noxs (NADPH oxidases) are well-known sources of superoxide, which contribute to the antimicrobial capabilities of macrophages, a process involving the prototypical member of the family referred to as Nox2. However, in recent years non-phagocytic homologues of Nox2 have been identified that are involved in processes other than the host defence. Superoxide anions produced by these enzymes are believed to play a major role in signalling by MAPKs (mitogen-activated protein kinases) and stress-activated kinases, but could also contribute to cellular senescence, which is known to involve oxygen radicals. In HUVECs (human umbilical vein endothelial cells), Nox4 is predominantly expressed, but its role in replicative senescence of HUVECs remains to be elucidated. Using shRNA (small-hairpin RNA)-mediated knockdown of Nox4, implicating lentiviral vectors, we addressed the question of whether lifelong depletion of Nox4 in HUVECs would influence the senescent phenotype. We found a significant extension of the replicative lifespan of HUVECs upon knockdown of Nox4. Surprisingly, mean telomere length was significantly reduced in Nox4-depleted cells. Nox4 depletion had no discernable influence on the activity of MAPKs and stress-activated kinases, but reduced the degree of oxidative DNA damage. These results suggest that Nox4 activity increases oxidative damage in HUVECs, leading to loss of replicative potential, which is at least partly independent of telomere attrition.
Collapse
|
49
|
Santos CXC, Tanaka LY, Wosniak J, Laurindo FRM. Mechanisms and implications of reactive oxygen species generation during the unfolded protein response: roles of endoplasmic reticulum oxidoreductases, mitochondrial electron transport, and NADPH oxidase. Antioxid Redox Signal 2009; 11:2409-27. [PMID: 19388824 DOI: 10.1089/ars.2009.2625] [Citation(s) in RCA: 416] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cellular mechanisms governing redox homeostasis likely involve their integration with other stresses. Endoplasmic reticulum (ER) stress triggers complex adaptive or proapoptotic signaling defined as the unfolded protein response (UPR), involved in several pathophysiological processes. Since protein folding is highly redox-dependent, convergence between ER stress and oxidative stress has attracted interest. Evidence suggests that ROS production and oxidative stress are not only coincidental to ER stress, but are integral UPR components, being triggered by distinct types of ER stressors and contributing to support proapoptotic, as well as proadaptive UPR signaling. Thus, ROS generation can be upstream or downstream UPR targets and may display a UPR-specific plus a nonspecific component. Enzymatic mechanisms of ROS generation during UPR include: (a) Multiple thiol-disulfide exchanges involving ER oxidoreductases including flavooxidase Ero1 and protein disulfide isomerase (PDI); (b) Mitochondrial electron transport; (c) Nox4 NADPH oxidase complex, particularly Nox4. Understanding the roles of such mechanisms and how they interconnect with the UPR requires more investigation. Integration among such ROS sources may depend on Ca(2+) levels, ROS themselves, and PDI, which associates with NADPH oxidase and regulates its function. Oxidative stress may frequently integrate with a background of ER stress/UPR in several diseases; here we discuss a focus in the vascular system.
Collapse
Affiliation(s)
- Célio X C Santos
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, CEP 05403-000, São Paulo, Brazil
| | | | | | | |
Collapse
|
50
|
Ushio-Fukai M, Urao N. Novel role of NADPH oxidase in angiogenesis and stem/progenitor cell function. Antioxid Redox Signal 2009; 11:2517-33. [PMID: 19309262 PMCID: PMC2821135 DOI: 10.1089/ars.2009.2582] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neovascularization is involved in normal development and wound repair as well as ischemic heart disease and peripheral artery disease. Both angiogenesis and vasculogenesis [de novo new vessel formation through mobilization of stem/progenitor cells from bone marrow (BM) and their homing to the ischemic sites] contribute to the formation of new blood vessels after tissue ischemia. Angiogenesis is dependent on cell proliferation, migration, and capillary tube formation in endothelial cells (ECs). Stem/progenitor cells have been used for cell-based therapy to promote revascularization after peripheral or myocardial ischemia. Excess amounts of reactive oxygen species (ROS) are involved in senescence and apoptosis of ECs and stem/progenitor cells, causing defective neovascularization. ROS at low levels function as signaling molecules to mediate cell proliferation, migration, differentiation, and gene expression. NADPH oxidase is one of the major sources of ROS in ECs and stem/progenitor cells, and is activated by various growth factors, cytokines, hypoxia, and ischemia. ROS derived from NADPH oxidase play an important role in redox signaling linked to angiogenesis ECs, as well as stem/progenitor cell mobilization, homing, and differentiation, thereby promoting neovascularization. Understanding these mechanisms may provide insight into NADPH oxidase and its mediators as potential therapeutic targets for ischemic heart and limb disease.
Collapse
Affiliation(s)
- Masuko Ushio-Fukai
- Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois at Chicago, Chicago, Illinois 60612, USA.
| | | |
Collapse
|