101
|
Pereira EJ, Smolko CM, Janes KA. Computational Models of Reactive Oxygen Species as Metabolic Byproducts and Signal-Transduction Modulators. Front Pharmacol 2016; 7:457. [PMID: 27965578 PMCID: PMC5126069 DOI: 10.3389/fphar.2016.00457] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/14/2016] [Indexed: 12/30/2022] Open
Abstract
Reactive oxygen species (ROS) are widely involved in intracellular signaling and human pathologies, but their precise roles have been difficult to enumerate and integrate holistically. The context- and dose-dependent intracellular effects of ROS can lead to contradictory experimental results and confounded interpretations. For example, lower levels of ROS promote cell signaling and proliferation, whereas abundant ROS cause overwhelming damage to biomolecules and cellular apoptosis or senescence. These complexities raise the question of whether the many facets of ROS biology can be joined under a common mechanistic framework using computational modeling. Here, we take inventory of some current models for ROS production or ROS regulation of signaling pathways. Several models captured non-intuitive observations or made predictions that were later verified by experiment. There remains a need for systems-level analyses that jointly incorporate ROS production, handling, and modulation of multiple signal-transduction cascades.
Collapse
Affiliation(s)
- Elizabeth J Pereira
- Department of Biomedical Engineering, University of Virginia, Charlottesville VA, USA
| | - Christian M Smolko
- Department of Biomedical Engineering, University of Virginia, Charlottesville VA, USA
| | - Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville VA, USA
| |
Collapse
|
102
|
Haque ZK, Wang DZ. How cardiomyocytes sense pathophysiological stresses for cardiac remodeling. Cell Mol Life Sci 2016; 74:983-1000. [PMID: 27714411 DOI: 10.1007/s00018-016-2373-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/01/2016] [Accepted: 09/19/2016] [Indexed: 12/14/2022]
Abstract
In the past decades, the cardiovascular community has laid out the fundamental signaling cascades that become awry in the cardiomyocyte during the process of pathologic cardiac remodeling. These pathways are initiated at the cell membrane and work their way to the nucleus to mediate gene expression. Complexity is multiplied as the cardiomyocyte is subjected to cross talk with other cells as well as a barrage of extracellular stimuli and mechanical stresses. In this review, we summarize the signaling cascades that play key roles in cardiac function and then we proceed to describe emerging concepts of how the cardiomyocyte senses the mechanical and environmental stimuli to transition to the deleterious genetic program that defines pathologic cardiac remodeling. As a highlighting example of these processes, we illustrate the transition from a compensated hypertrophied myocardium to a decompensated failing myocardium, which is clinically manifested as decompensated heart failure.
Collapse
Affiliation(s)
- Zaffar K Haque
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 1260 John F. Enders Research Bldg, 320 Longwood Ave, Boston, MA, 02115, USA.
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 1260 John F. Enders Research Bldg, 320 Longwood Ave, Boston, MA, 02115, USA
| |
Collapse
|
103
|
Kurz FT, Kembro JM, Flesia AG, Armoundas AA, Cortassa S, Aon MA, Lloyd D. Network dynamics: quantitative analysis of complex behavior in metabolism, organelles, and cells, from experiments to models and back. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2016; 9. [PMID: 27599643 DOI: 10.1002/wsbm.1352] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/20/2016] [Accepted: 06/23/2016] [Indexed: 12/15/2022]
Abstract
Advancing from two core traits of biological systems: multilevel network organization and nonlinearity, we review a host of novel and readily available techniques to explore and analyze their complex dynamic behavior within the framework of experimental-computational synergy. In the context of concrete biological examples, analytical methods such as wavelet, power spectra, and metabolomics-fluxomics analyses, are presented, discussed, and their strengths and limitations highlighted. Further shown is how time series from stationary and nonstationary biological variables and signals, such as membrane potential, high-throughput metabolomics, O2 and CO2 levels, bird locomotion, at the molecular, (sub)cellular, tissue, and whole organ and animal levels, can reveal important information on the properties of the underlying biological networks. Systems biology-inspired computational methods start to pave the way for addressing the integrated functional dynamics of metabolic, organelle and organ networks. As our capacity to unravel the control and regulatory properties of these networks and their dynamics under normal or pathological conditions broadens, so is our ability to address endogenous rhythms and clocks to improve health-span in human aging, and to manage complex metabolic disorders, neurodegeneration, and cancer. WIREs Syst Biol Med 2017, 9:e1352. doi: 10.1002/wsbm.1352 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Felix T Kurz
- Massachusetts General Hospital, Cardiovascular Research Center, Harvard Medical School, Charlestown, MA, USA.,Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jackelyn M Kembro
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT-CONICET), and Instituto de Ciencia y Tecnología de los Alimentos, Cátedra de Química Biológica, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ana G Flesia
- Centro de Investigaciones y Estudios de Matemática (CIEM-CONICET), and Facultad de Matemática, Astronomía y Física FAMAF, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Antonis A Armoundas
- Massachusetts General Hospital, Cardiovascular Research Center, Harvard Medical School, Charlestown, MA, USA
| | - Sonia Cortassa
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Miguel A Aon
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David Lloyd
- Cardiff University School of Biosciences, Cardiff, UK
| |
Collapse
|
104
|
Yadetie F, Bjørneklett S, Garberg HK, Oveland E, Berven F, Goksøyr A, Karlsen OA. Quantitative analyses of the hepatic proteome of methylmercury-exposed Atlantic cod (Gadus morhua) suggest oxidative stress-mediated effects on cellular energy metabolism. BMC Genomics 2016; 17:554. [PMID: 27496535 PMCID: PMC4974784 DOI: 10.1186/s12864-016-2864-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 06/30/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Methylmecury (MeHg) is a widely distributed environmental pollutant with considerable risk to both human health and wildlife. To gain better insight into the underlying mechanisms of MeHg-mediated toxicity, we have used label-free quantitative mass spectrometry to analyze the liver proteome of Atlantic cod (Gadus morhua) exposed in vivo to MeHg (0, 0.5, 2 mg/kg body weight) for 2 weeks. RESULTS Out of a toltal of 1143 proteins quantified, 125 proteins were differentially regulated between MeHg-treated samples and controls. Using various bioinformatics tools, we performed gene ontology, pathway and network enrichment analysis, which indicated that proteins and pathways mainly related to energy metabolism, antioxidant defense, cytoskeleton remodeling, and protein synthesis were regulated in the hepatic proteome after MeHg exposure. Comparison with previous gene expression data strengthened these results, and further supported that MeHg predominantly affects many energy metabolism pathways, presumably through its strong induction of oxidative stress. Some enzymes known to have functionally important oxidation-sensitive cysteine residues in other animals are among the differentially regulated proteins, suggesting their modulations by MeHg-induced oxidative stress. Integrated analysis of the proteomics dataset combined with previous gene expression dataset showed a more pronounced effect of MeHg on amino acid, glucose and fatty acid metabolic pathways, and suggested possible interactions of the cellular energy metabolism and antioxidant defense pathways. CONCLUSIONS MeHg disrupts mainly redox homeostasis and energy generating metabolic pathways in cod liver. The energy pathways appear to be modulated through MeHg-induced oxidative stress, possibly mediated by oxidation sensitive enzymes.
Collapse
Affiliation(s)
- Fekadu Yadetie
- Department of Biology, University of Bergen, PO Box 7803, N-5020, Bergen, Norway
| | - Silje Bjørneklett
- Department of Biology, University of Bergen, PO Box 7803, N-5020, Bergen, Norway
| | - Hilde Kristin Garberg
- Department of Biomedicine, Proteomics Unit (PROBE) at the University of Bergen, Bergen, Norway
| | - Eystein Oveland
- Department of Biomedicine, Proteomics Unit (PROBE) at the University of Bergen, Bergen, Norway
| | - Frode Berven
- Department of Biomedicine, Proteomics Unit (PROBE) at the University of Bergen, Bergen, Norway
| | - Anders Goksøyr
- Department of Biology, University of Bergen, PO Box 7803, N-5020, Bergen, Norway
| | - Odd André Karlsen
- Department of Biology, University of Bergen, PO Box 7803, N-5020, Bergen, Norway.
| |
Collapse
|
105
|
Ma J, Banerjee P, Whelan SA, Liu T, Wei AC, Genaro Ramirez-Correa, McComb ME, Costello CE, O’Rourke B, Murphy A, Hart GW. Comparative Proteomics Reveals Dysregulated Mitochondrial O-GlcNAcylation in Diabetic Hearts. J Proteome Res 2016; 15:2254-64. [PMID: 27213235 PMCID: PMC7814404 DOI: 10.1021/acs.jproteome.6b00250] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
O-linked β-N-acetylglucosamine (O-GlcNAc), a post-translational modification on serine and threonine residues of many proteins, plays crucial regulatory roles in diverse biological events. As a nutrient sensor, O-GlcNAc modification (O-GlcNAcylation) on nuclear and cytoplasmic proteins underlies the pathology of diabetic complications including cardiomyopathy. However, mitochondrial O-GlcNAcylation, especially in response to chronic hyperglycemia in diabetes, has been poorly explored. We performed a comparative O-GlcNAc profiling of mitochondria from control and streptozotocin (STZ)-induced diabetic rat hearts by using an improved β-elimination/Michael addition with isotopic DTT reagents (BEMAD) followed by tandem mass spectrometric analysis. In total, 86 mitochondrial proteins, involved in diverse pathways, were O-GlcNAcylated. Among them, many proteins have site-specific alterations in O-GlcNAcylation in response to diabetes, which suggests that protein O-GlcNAcylation is a novel layer of regulation mediating adaptive changes in mitochondrial metabolism during the progression of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Junfeng Ma
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Partha Banerjee
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Stephen A. Whelan
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, United States
| | - Ting Liu
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - An-Chi Wei
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Genaro Ramirez-Correa
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Mark E. McComb
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, United States
| | - Catherine E. Costello
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, United States
| | - Brian O’Rourke
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Anne Murphy
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Gerald W. Hart
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
106
|
Munro D, Banh S, Sotiri E, Tamanna N, Treberg JR. The thioredoxin and glutathione-dependent H2O2 consumption pathways in muscle mitochondria: Involvement in H2O2 metabolism and consequence to H2O2 efflux assays. Free Radic Biol Med 2016; 96:334-46. [PMID: 27101737 DOI: 10.1016/j.freeradbiomed.2016.04.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 04/07/2016] [Accepted: 04/15/2016] [Indexed: 11/25/2022]
Abstract
The most common methods of measuring mitochondrial hydrogen peroxide production are based on the extramitochondrial oxidation of a fluorescent probe such as amplex ultra red (AUR) by horseradish peroxidase (HRP). These traditional HRP-based assays only detect H2O2 that has escaped the matrix, raising the potential for substantial underestimation of production if H2O2 is consumed by matrix antioxidant pathways. To measure this underestimation, we characterized matrix consumers of H2O2 in rat skeletal muscle mitochondria, and developed specific means to inhibit these consumers. Mitochondria removed exogenously added H2O2 (2.5µM) at rates of 4.7 and 5.0nmol min(-1) mg protein(-1) when respiring on glutamate+malate and succinate+rotenone, respectively. In the absence of respiratory substrate, or after disrupting membranes by cycles of freeze-thaw, rates of H2O2 consumption were negligible. We concluded that matrix consumers are respiration-dependent (requiring respiratory substrates), suggesting the involvement of either the thioredoxin (Trx) and/or glutathione (GSH)-dependent enzymatic pathways. The Trx-reductase inhibitor auranofin (2µM), and a pre-treatment of mitochondria with 35µM of 1-chloro-2,4-dintrobenzene (CDNB) to deplete GSH specifically compromise these two consumption pathways. These inhibition approaches presented no undesirable "off-target" effects during extensive preliminary tests. These inhibition approaches independently and additively decreased the rate of consumption of H2O2 exogenously added to the medium (2.5µM). During traditional HRP-based H2O2 efflux assays, these inhibition approaches independently and additively increased apparent efflux rates. When used in combination (double inhibition), these inhibition approaches allowed accumulation of (endogenously produced) H2O2 in the medium at a comparable rate whether it was measured with an end point assay where 2.5µM H2O2 is initially added to the medium or with traditional HRP-based efflux assays. This finding confirms that a high degree of inhibition of all matrix consumers is attained with the double inhibition. Importantly, this double inhibition of the matrix consumers allowed revealing that a large part of the H2O2 produced in muscle mitochondria is consumed before escaping the matrix during traditional HRP-based efflux assays. The degree of this underestimation was substrate dependent, reaching >80% with malate, which complicates comparisons of substrates for their capacity to generate H2O2 in normal conditions i.e. when matrix consumers are active. Our results also urge caution in interpreting changes in H2O2 efflux in response to a treatment; when HRP-based assays are used, large changes in apparent H2O2 efflux may come from altered capacity of the matrix consumers.
Collapse
Affiliation(s)
- Daniel Munro
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada; Centre on Aging, University of Manitoba, Winnipeg, MB, Canada.
| | - Sheena Banh
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Emianka Sotiri
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Nahid Tamanna
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jason R Treberg
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada; Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada; Centre on Aging, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
107
|
Margaritelis NV. Antioxidants as therapeutics in the intensive care unit: Have we ticked the redox boxes? Pharmacol Res 2016; 111:126-132. [PMID: 27270047 DOI: 10.1016/j.phrs.2016.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 05/29/2016] [Accepted: 06/03/2016] [Indexed: 02/07/2023]
Abstract
Critically ill patients are under oxidative stress and antioxidant administration reasonably emerged as a promising approach to combat the aberrant redox homeostasis in this patient cohort. However, the results of the antioxidant treatments in the intensive care unit are conflicting and inconclusive. The main objective of the present review is to highlight some inherent, yet widely overlooked redox-related issues about the equivocal effectiveness of antioxidants in the intensive care unit, beyond methodological considerations. In particular, the discrepancy in the literature partially stems from: (1) the largely unspecified role of reactive species in disease onset and progression, (2) our fragmentary understanding on the interplay between inflammation and oxidative stress, (3) the complex spatiotemporal specificity of in vivo redox biology, (4) the pleiotropic effects of antioxidants and (5) the divergent effects of antioxidants according to the temporal administration pattern. In addition, two novel and sophisticated practices with promising pre-clinical results are presented: (1) the selective neutralization of reactive species in key organelles after they are formed (i.e., in mitochondria) and (2) the targeted complete inhibition of dominant reactive species sources (i.e., NADPH oxidases). Finally, the reductive potential of NADPH as a key pharmacological target for redox therapies is rationalized. In light of the above, the recontextualization of knowledge from basic redox biology to translational medicine seems imperative to perform more realistic in vivo studies in the fast-growing field of critical care pharmacology.
Collapse
Affiliation(s)
- Nikos V Margaritelis
- Intensive Care Unit, 424 General Military Hospital of Thessaloniki, Thessaloniki, Greece; Department of Physical Education and Sport Science at Serres, Aristotle University of Thessaloniki, Serres, Greece.
| |
Collapse
|
108
|
Dey S, Sidor A, O'Rourke B. Compartment-specific Control of Reactive Oxygen Species Scavenging by Antioxidant Pathway Enzymes. J Biol Chem 2016; 291:11185-97. [PMID: 27048652 PMCID: PMC4900267 DOI: 10.1074/jbc.m116.726968] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/01/2016] [Indexed: 11/06/2022] Open
Abstract
Oxidative stress arises from an imbalance in the production and scavenging rates of reactive oxygen species (ROS) and is a key factor in the pathophysiology of cardiovascular disease and aging. The presence of parallel pathways and multiple intracellular compartments, each having its own ROS sources and antioxidant enzymes, complicates the determination of the most important regulatory nodes of the redox network. Here we quantified ROS dynamics within specific intracellular compartments in the cytosol and mitochondria and determined which scavenging enzymes exert the most control over antioxidant fluxes in H9c2 cardiac myoblasts. We used novel targeted viral gene transfer vectors expressing redox-sensitive GFP fused to sensor domains to measure H2O2 or oxidized glutathione. Using genetic manipulation in heart-derived H9c2 cells, we explored the contribution of specific antioxidant enzymes to ROS scavenging and glutathione redox potential within each intracellular compartment. Our findings reveal that antioxidant flux is strongly dependent on mitochondrial substrate catabolism, with availability of NADPH as a major rate-controlling step. Moreover, ROS scavenging by mitochondria significantly contributes to cytoplasmic ROS handling. The findings provide fundamental information about the control of ROS scavenging by the redox network and suggest novel interventions for circumventing oxidative stress in cardiac cells.
Collapse
Affiliation(s)
- Swati Dey
- From the Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, Maryland 21205
| | - Agnieszka Sidor
- From the Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, Maryland 21205
| | - Brian O'Rourke
- From the Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, Maryland 21205
| |
Collapse
|
109
|
Dwivedi G, Gran MA, Bagchi P, Kemp ML. Dynamic Redox Regulation of IL-4 Signaling. PLoS Comput Biol 2015; 11:e1004582. [PMID: 26562652 PMCID: PMC4642971 DOI: 10.1371/journal.pcbi.1004582] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/29/2015] [Indexed: 12/22/2022] Open
Abstract
Quantifying the magnitude and dynamics of protein oxidation during cell signaling is technically challenging. Computational modeling provides tractable, quantitative methods to test hypotheses of redox mechanisms that may be simultaneously operative during signal transduction. The interleukin-4 (IL-4) pathway, which has previously been reported to induce reactive oxygen species and oxidation of PTP1B, may be controlled by several other putative mechanisms of redox regulation; widespread proteomic thiol oxidation observed via 2D redox differential gel electrophoresis upon IL-4 treatment suggests more than one redox-sensitive protein implicated in this pathway. Through computational modeling and a model selection strategy that relied on characteristic STAT6 phosphorylation dynamics of IL-4 signaling, we identified reversible protein tyrosine phosphatase (PTP) oxidation as the primary redox regulatory mechanism in the pathway. A systems-level model of IL-4 signaling was developed that integrates synchronous pan-PTP oxidation with ROS-independent mechanisms. The model quantitatively predicts the dynamics of IL-4 signaling over a broad range of new redox conditions, offers novel hypotheses about regulation of JAK/STAT signaling, and provides a framework for interrogating putative mechanisms involving receptor-initiated oxidation. Incomplete reduction of oxygen during respiration results in the formation of highly reactive molecules known as reactive oxygen species (ROS) that react indiscriminately with cellular components and adversely affect cellular function. For a long time ROS were thought solely to be undesirable byproducts of respiration. Indeed, high levels of ROS are associated with a number of diseases. Despite these facts, antioxidants, agents that neutralize ROS, have not shown any clinical benefits when used as oral supplements. This paradox is partially explained by discoveries over the last two decades demonstrating that ROS are not always detrimental and may be essential for controlling physiological processes like cell signaling. However, the mechanisms by which ROS react with biomolecules are not well understood. In this work we have combined biological experiments with novel computational methods to identify the most important mechanisms of ROS-mediated regulation in the IL-4 signaling pathway of the immune system. We have also developed a detailed computer model of the IL-4 pathway and its regulation by ROS dependent and independent methods. Our work enhances the understanding of principles underlying regulation of cell signaling by ROS and has potential implications in advancing therapeutic methods targeting ROS and their adverse effects.
Collapse
Affiliation(s)
- Gaurav Dwivedi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
| | - Margaret A. Gran
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
| | - Pritha Bagchi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
| | - Melissa L. Kemp
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
110
|
Bartz RR, Suliman HB, Piantadosi CA. Redox mechanisms of cardiomyocyte mitochondrial protection. Front Physiol 2015; 6:291. [PMID: 26578967 PMCID: PMC4620408 DOI: 10.3389/fphys.2015.00291] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 10/02/2015] [Indexed: 12/30/2022] Open
Abstract
Oxidative and nitrosative stress are primary contributors to the loss of myocardial tissue in insults ranging from ischemia/reperfusion injury from coronary artery disease and heart transplantation to sepsis-induced myocardial dysfunction and drug-induced myocardial damage. This cell damage caused by oxidative and nitrosative stress leads to mitochondrial protein, DNA, and lipid modifications, which inhibits energy production and contractile function, potentially leading to cell necrosis and/or apoptosis. However, cardiomyocytes have evolved an elegant set of redox-sensitive mechanisms that respond to and contain oxidative and nitrosative damage. These responses include the rapid induction of antioxidant enzymes, mitochondrial DNA repair mechanisms, selective mitochondrial autophagy (mitophagy), and mitochondrial biogenesis. Coordinated cytoplasmic to nuclear cell-signaling and mitochondrial transcriptional responses to the presence of elevated cytoplasmic oxidant production, e.g., H2O2, allows nuclear translocation of the Nfe2l2 transcription factor and up-regulation of downstream cytoprotective genes such as heme oxygenase-1 which generates physiologic signals, such as CO that up-regulates Nfe212 gene transcription. Simultaneously, a number of other DNA binding transcription factors are expressed and/or activated under redox control, such as Nuclear Respiratory Factor-1 (NRF-1), and lead to the induction of genes involved in both intracellular and mitochondria-specific repair mechanisms. The same insults, particularly those related to vascular stress and inflammation also produce elevated levels of nitric oxide, which also has mitochondrial protein thiol-protective functions and induces mitochondrial biogenesis through cyclic GMP-dependent and perhaps other pathways. This brief review provides an overview of these pathways and interconnected cardiac repair mechanisms.
Collapse
Affiliation(s)
- Raquel R Bartz
- Department of Anesthesiology, Duke University School of Medicine Durham, NC, USA ; Department of Medicine, Duke University School of Medicine Durham, NC, USA
| | - Hagir B Suliman
- Department of Anesthesiology, Duke University School of Medicine Durham, NC, USA ; Department of Pathology, Duke University School of Medicine Durham, NC, USA
| | - Claude A Piantadosi
- Department of Anesthesiology, Duke University School of Medicine Durham, NC, USA ; Department of Medicine, Duke University School of Medicine Durham, NC, USA ; Department of Pathology, Duke University School of Medicine Durham, NC, USA ; Durham Veterans Affairs Hospital Durham, NC, USA
| |
Collapse
|
111
|
Granger DN, Kvietys PR. Reperfusion injury and reactive oxygen species: The evolution of a concept. Redox Biol 2015; 6:524-551. [PMID: 26484802 PMCID: PMC4625011 DOI: 10.1016/j.redox.2015.08.020] [Citation(s) in RCA: 1032] [Impact Index Per Article: 103.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 12/11/2022] Open
Abstract
Reperfusion injury, the paradoxical tissue response that is manifested by blood flow-deprived and oxygen-starved organs following the restoration of blood flow and tissue oxygenation, has been a focus of basic and clinical research for over 4-decades. While a variety of molecular mechanisms have been proposed to explain this phenomenon, excess production of reactive oxygen species (ROS) continues to receive much attention as a critical factor in the genesis of reperfusion injury. As a consequence, considerable effort has been devoted to identifying the dominant cellular and enzymatic sources of excess ROS production following ischemia-reperfusion (I/R). Of the potential ROS sources described to date, xanthine oxidase, NADPH oxidase (Nox), mitochondria, and uncoupled nitric oxide synthase have gained a status as the most likely contributors to reperfusion-induced oxidative stress and represent priority targets for therapeutic intervention against reperfusion-induced organ dysfunction and tissue damage. Although all four enzymatic sources are present in most tissues and are likely to play some role in reperfusion injury, priority and emphasis has been given to specific ROS sources that are enriched in certain tissues, such as xanthine oxidase in the gastrointestinal tract and mitochondria in the metabolically active heart and brain. The possibility that multiple ROS sources contribute to reperfusion injury in most tissues is supported by evidence demonstrating that redox-signaling enables ROS produced by one enzymatic source (e.g., Nox) to activate and enhance ROS production by a second source (e.g., mitochondria). This review provides a synopsis of the evidence implicating ROS in reperfusion injury, the clinical implications of this phenomenon, and summarizes current understanding of the four most frequently invoked enzymatic sources of ROS production in post-ischemic tissue. Reperfusion injury is implicated in a variety of human diseases and disorders. Evidence implicating ROS in reperfusion injury continues to grow. Several enzymes are candidate sources of ROS in post-ischemic tissue. Inter-enzymatic ROS-dependent signaling enhances the oxidative stress caused by I/R. .
Collapse
Affiliation(s)
- D Neil Granger
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, United States.
| | - Peter R Kvietys
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
112
|
Ma J, Liu T, Wei AC, Banerjee P, O'Rourke B, Hart GW. O-GlcNAcomic Profiling Identifies Widespread O-Linked β-N-Acetylglucosamine Modification (O-GlcNAcylation) in Oxidative Phosphorylation System Regulating Cardiac Mitochondrial Function. J Biol Chem 2015; 290:29141-53. [PMID: 26446791 DOI: 10.1074/jbc.m115.691741] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Indexed: 11/06/2022] Open
Abstract
Dynamic cycling of O-linked β-N-acetylglucosamine (O-GlcNAc) on nucleocytoplasmic proteins serves as a nutrient sensor to regulate numerous biological processes. However, mitochondrial protein O-GlcNAcylation and its effects on function are largely unexplored. In this study, we performed a comparative analysis of the proteome and O-GlcNAcome of cardiac mitochondria from rats acutely (12 h) treated without or with thiamet-G (TMG), a potent and specific inhibitor of O-GlcNAcase. We then determined the functional consequences in mitochondria isolated from the two groups. O-GlcNAcomic profiling finds that over 88 mitochondrial proteins are O-GlcNAcylated, with the oxidative phosphorylation system as a major target. Moreover, in comparison with controls, cardiac mitochondria from TMG-treated rats did not exhibit altered protein abundance but showed overall elevated O-GlcNAcylation of many proteins. However, O-GlcNAc was unexpectedly down-regulated at certain sites of specific proteins. Concomitantly, TMG treatment resulted in significantly increased mitochondrial oxygen consumption rates, ATP production rates, and enhanced threshold for permeability transition pore opening by Ca(2+). Our data reveal widespread and dynamic mitochondrial protein O-GlcNAcylation, serving as a regulator to their function.
Collapse
Affiliation(s)
- Junfeng Ma
- From the Department of Biological Chemistry and
| | - Ting Liu
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - An-Chi Wei
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | | | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | | |
Collapse
|
113
|
Abstract
AbstractThe bovine mammary gland is a dynamic and complex organ composed of various cell types that work together for the purpose of milk synthesis and secretion. A layer of endothelial cells establishes the blood–milk barrier, which exists to facilitate the exchange of solutes and macromolecules necessary for optimal milk production. During bacterial challenge, however, endothelial cells divert some of their lactation function to protect the underlying tissue from damage by initiating inflammation. At the onset of inflammation, endothelial cells tightly regulate the movement of plasma components and leukocytes into affected tissue. Unfortunately, endothelial dysfunction as a result of exacerbated or sustained inflammation can negatively affect both barrier integrity and the health of surrounding extravascular tissue. The objective of this review is to highlight the role of endothelial cells in supporting milk production and regulating optimal inflammatory responses. The consequences of endothelial dysfunction and sustained inflammation on milk synthesis and secretion are discussed. Given the important role of endothelial cells in orchestrating the inflammatory response, a better understanding of endothelial function during mastitis may support development of targeted therapies to protect bovine mammary tissue and mammary endothelium.
Collapse
|
114
|
Abstract
SIGNIFICANCE Mitochondria are structurally and biochemically diverse, even within a single type of cell. Protein complexes localized to the inner mitochondrial membrane synthesize ATP by coupling electron transport and oxidative phosphorylation. The organelles produce reactive oxygen species (ROS) from mitochondrial oxygen and ROS can, in turn, alter the function and expression of proteins used for aerobic respiration by post-translational and transcriptional regulation. RECENT ADVANCES New interest is emerging not only into the roles of mitochondria in disease development and progression but also as a target for environmental toxicants. CRITICAL ISSUES Dysregulation of respiration has been linked to cell death and is a major contributor to acute neuronal trauma, peripheral diseases, as well as chronic neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. FUTURE DIRECTIONS Here, we discuss the mechanisms underlying the sensitivity of the mitochondrial respiratory complexes to redox modulation, as well as examine the effects of environmental contaminants that have well-characterized mitochondrial toxicity. The contaminants discussed in this review are some of the most prevalent and potent environmental contaminants that have been linked to neurological dysfunction, altered cellular respiration, and oxidation.
Collapse
Affiliation(s)
- Samuel W Caito
- Department of Molecular Pharmacology, Albert Einstein College of Medicine , Bronx, New York
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine , Bronx, New York
| |
Collapse
|
115
|
Belmonte F, Das S, Sysa-Shah P, Sivakumaran V, Stanley B, Guo X, Paolocci N, Aon MA, Nagane M, Kuppusamy P, Steenbergen C, Gabrielson K. ErbB2 overexpression upregulates antioxidant enzymes, reduces basal levels of reactive oxygen species, and protects against doxorubicin cardiotoxicity. Am J Physiol Heart Circ Physiol 2015; 309:H1271-80. [PMID: 26254336 DOI: 10.1152/ajpheart.00517.2014] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 07/31/2015] [Indexed: 11/22/2022]
Abstract
Levels of the HER2/ErbB2 protein in the heart are upregulated in some women during breast cancer therapy, and these women are at high risk for developing heart dysfunction after sequential treatment with anti-ErbB2/trastuzumab or doxorubicin. Doxorubicin is known to increase oxidative stress in the heart, and thus we considered the possibility that ErbB2 protein influences the status of cardiac antioxidant defenses in cardiomyocytes. In this study, we measured reactive oxygen species (ROS) in cardiac mitochondria and whole hearts from mice with cardiac-specific overexpression of ErbB2 (ErbB2(tg)) and found that, compared with control mice, high levels of ErbB2 in myocardium result in lower levels of ROS in mitochondria (P = 0.0075) and whole hearts (P = 0.0381). Neonatal cardiomyocytes isolated from ErbB2(tg) hearts have lower ROS levels and less cellular death (P < 0.0001) following doxorubicin treatment. Analyzing antioxidant enzyme levels and activities, we found that ErbB2(tg) hearts have increased levels of glutathione peroxidase 1 (GPx1) protein (P < 0.0001) and GPx activity (P = 0.0031) in addition to increased levels of two known GPx activators, c-Abl (P = 0.0284) and Arg (P < 0.0001). Interestingly, although mitochondrial ROS emission is reduced in the ErbB2(tg) hearts, oxygen consumption rates and complex I activity are similar to control littermates. Compared with these in vivo studies, H9c2 cells transfected with ErbB2 showed less cellular toxicity and produced less ROS (P < 0.0001) after doxorubicin treatment but upregulated GR activity (P = 0.0237) instead of GPx. Our study shows that ErbB2-dependent signaling contributes to antioxidant defenses and suggests a novel mechanism by which anticancer therapies involving ErbB2 antagonists can harm myocardial structure and function.
Collapse
Affiliation(s)
- Frances Belmonte
- Program in Molecular and Translational Toxicology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Samarjit Das
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Polina Sysa-Shah
- Department of Molecular and Comparative Pathobiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Vidhya Sivakumaran
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, Maryland; and
| | - Brian Stanley
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, Maryland; and
| | - Xin Guo
- Department of Molecular and Comparative Pathobiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, Maryland; and
| | - Miguel A Aon
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, Maryland; and
| | - Masaki Nagane
- Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Periannan Kuppusamy
- Department of Radiology, EPR Center for the Study of Viable Systems, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Charles Steenbergen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kathleen Gabrielson
- Program in Molecular and Translational Toxicology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Molecular and Comparative Pathobiology, Johns Hopkins Medical Institutions, Baltimore, Maryland;
| |
Collapse
|
116
|
Yoshioka J. Thioredoxin Reductase 2 (Txnrd2) Regulates Mitochondrial Integrity in the Progression of Age-Related Heart Failure. J Am Heart Assoc 2015. [PMID: 26199229 PMCID: PMC4608096 DOI: 10.1161/jaha.115.002278] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Jun Yoshioka
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA (J.Y.)
| |
Collapse
|
117
|
Sharaf MS, van den Heuvel MR, Stevens D, Kamunde C. Zinc and calcium modulate mitochondrial redox state and morphofunctional integrity. Free Radic Biol Med 2015; 84:142-153. [PMID: 25841782 DOI: 10.1016/j.freeradbiomed.2015.03.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 03/03/2015] [Accepted: 03/08/2015] [Indexed: 01/08/2023]
Abstract
Zinc and calcium have highly interwoven functions that are essential for cellular homeostasis. Here we first present a novel real-time flow cytometric technique to measure mitochondrial redox state and show it is modulated by zinc and calcium, individually and combined. We then assess the interactions of zinc and calcium on mitochondrial H2O2 production, membrane potential (ΔΨm), morphological status, oxidative phosphorylation (OXPHOS), complex I activity, and structural integrity. Whereas zinc at low doses and both cations at high doses individually and combined promoted H2O2 production, the two cations individually did not alter mitochondrial redox state. However, when combined at low and high doses the two cations synergistically suppressed and promoted, respectively, mitochondrial shift to a more oxidized state. Surprisingly, the antioxidants vitamin E and N-acetylcysteine showed pro-oxidant activity at low doses, whereas at high antioxidant doses NAC inhibited OXPHOS and dyscoupled mitochondria. Individually, zinc was more potent than calcium in inhibiting OXPHOS, whereas calcium more potently dissipated the ΔΨm and altered mitochondrial volume and ultrastructure. The two cations synergistically inhibited OXPHOS but antagonistically dissipated ΔΨm and altered mitochondrial volume and morphology. Overall, our study highlights the importance of zinc and calcium in mitochondrial redox regulation and functional integrity. Importantly, we uncovered previously unrecognized bidirectional interactions of zinc and calcium that reveal distinctive foci for modulating mitochondrial function in normal and disease states because they are potentially protective or damaging depending on conditions.
Collapse
Affiliation(s)
- Mahmoud S Sharaf
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada; Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Michael R van den Heuvel
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada; Canadian Rivers Institute, Department of Biology, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada
| | - Don Stevens
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada.
| |
Collapse
|
118
|
Impaired mitochondrial energy supply coupled to increased H2O2 emission under energy/redox stress leads to myocardial dysfunction during Type I diabetes. Clin Sci (Lond) 2015; 129:561-74. [PMID: 26186741 DOI: 10.1042/cs20150204] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/29/2015] [Indexed: 12/23/2022]
Abstract
In Type I diabetic (T1DM) patients, both peaks of hyperglycaemia and increased sympathetic tone probably contribute to impair systolic and diastolic function. However, how these stressors eventually alter cardiac function during T1DM is not fully understood. In the present study, we hypothesized that impaired mitochondrial energy supply and excess reactive oxygen species (ROS) emission is centrally involved in T1DM cardiac dysfunction due to metabolic/redox stress and aimed to determine the mitochondrial sites implicated in these alterations. To this end, we used isolated myocytes and mitochondria from Sham and streptozotocin (STZ)-induced T1DM guinea pigs (GPs), untreated or treated with insulin. Relative to controls, T1DM myocytes exhibited higher oxidative stress when challenged with high glucose (HG) combined with β-adrenergic stimulation [via isoprenaline (isoproterenol) (ISO)], leading to contraction/relaxation deficits. T1DM mitochondria had decreased respiration with complex II and IV substrates and markedly lower ADP phosphorylation rates and higher H2O2 emission when challenged with oxidants to mimic the more oxidized redox milieu present in HG + ISO-treated cardiomyocytes. Since in T1DM hearts insulin-sensitivity is preserved and a glucose-to-fatty acid (FA) shift occurs, we next tested whether insulin therapy or acute palmitate (Palm) infusion prevents HG + ISO-induced cardiac dysfunction. We found that insulin rescued proper cardiac redox balance, but not mitochondrial respiration or contractile performance. Conversely, Palm restored redox balance and preserved myocyte function. Thus, stressors such as peaks of HG and adrenergic hyperactivity impair mitochondrial respiration, hampering energy supply while exacerbating ROS emission. Our study suggests that an ideal therapeutic measure to treat metabolically/redox-challenged T1DM hearts should concomitantly correct energetic and redox abnormalities to fully maintain cardiac function.
Collapse
|
119
|
Abstract
SIGNIFICANCE Cardiac function is energetically demanding, reliant on efficient well-coupled mitochondria to generate adenosine triphosphate and fulfill the cardiac demand. Predictably then, mitochondrial dysfunction is associated with cardiac pathologies, often related to metabolic disease, most commonly diabetes. Diabetic cardiomyopathy (DCM), characterized by decreased left ventricular function, arises independently of coronary artery disease and atherosclerosis. Dysregulation of Ca(2+) handling, metabolic changes, and oxidative stress are observed in DCM, abnormalities reflected in alterations in mitochondrial energetics. Cardiac tissue from DCM patients also presents with altered mitochondrial morphology, suggesting a possible role of mitochondrial dynamics in its pathological progression. RECENT ADVANCES Abnormal mitochondrial morphology is associated with pathologies across diverse tissues, suggesting that this highly regulated process is essential for proper cell maintenance and physiological homeostasis. Highly structured cardiac myofibers were hypothesized to limit alterations in mitochondrial morphology; however, recent work has identified morphological changes in cardiac tissue, specifically in DCM. CRITICAL ISSUES Mitochondrial dysfunction has been reported independently from observations of altered mitochondrial morphology in DCM. The temporal relationship and causative nature between functional and morphological changes of mitochondria in the establishment/progression of DCM is unclear. FUTURE DIRECTIONS Altered mitochondrial energetics and morphology are not only causal for but also consequential to reactive oxygen species production, hence exacerbating oxidative damage through reciprocal amplification, which is integral to the progression of DCM. Therefore, targeting mitochondria for DCM will require better mechanistic characterization of morphological distortion and bioenergetic dysfunction.
Collapse
Affiliation(s)
- Chad A Galloway
- 1Department of Anesthesiology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Yisang Yoon
- 2Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
120
|
Protective effects of dietary avocado oil on impaired electron transport chain function and exacerbated oxidative stress in liver mitochondria from diabetic rats. J Bioenerg Biomembr 2015; 47:337-53. [PMID: 26060181 DOI: 10.1007/s10863-015-9614-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 05/29/2015] [Indexed: 02/07/2023]
Abstract
Electron transport chain (ETC) dysfunction, excessive ROS generation and lipid peroxidation are hallmarks of mitochondrial injury in the diabetic liver, with these alterations also playing a role in the development of non-alcoholic fatty liver disease (NAFLD). Enhanced mitochondrial sensitivity to lipid peroxidation during diabetes has been also associated to augmented content of C22:6 in membrane phospholipids. Thus, we aimed to test whether avocado oil, a rich source of C18:1 and antioxidants, attenuates the deleterious effects of diabetes on oxidative status of liver mitochondria by decreasing unsaturation of acyl chains of membrane lipids and/or by improving ETC functionality and decreasing ROS generation. Streptozocin-induced diabetes elicited a noticeable increase in the content of C22:6, leading to augmented mitochondrial peroxidizability index and higher levels of lipid peroxidation. Mitochondrial respiration and complex I activity were impaired in diabetic rats with a concomitant increase in ROS generation using a complex I substrate. This was associated to a more oxidized state of glutathione, All these alterations were prevented by avocado oil except by the changes in mitochondrial fatty acid composition. Avocado oil did not prevented hyperglycemia and polyphagia although did normalized hyperlipidemia. Neither diabetes nor avocado oil induced steatosis. These results suggest that avocado oil improves mitochondrial ETC function by attenuating the deleterious effects of oxidative stress in the liver of diabetic rats independently of a hypoglycemic effect or by modifying the fatty acid composition of mitochondrial membranes. These findings might have also significant implications in the progression of NAFLD in experimental models of steatosis.
Collapse
|
121
|
Aon MA, Tocchetti CG, Bhatt N, Paolocci N, Cortassa S. Protective mechanisms of mitochondria and heart function in diabetes. Antioxid Redox Signal 2015; 22:1563-86. [PMID: 25674814 PMCID: PMC4449630 DOI: 10.1089/ars.2014.6123] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE The heart depends on continuous mitochondrial ATP supply and maintained redox balance to properly develop force, particularly under increased workload. During diabetes, however, myocardial energetic-redox balance is perturbed, contributing to the systolic and diastolic dysfunction known as diabetic cardiomyopathy (DC). CRITICAL ISSUES How these energetic and redox alterations intertwine to influence the DC progression is still poorly understood. Excessive bioavailability of both glucose and fatty acids (FAs) play a central role, leading, among other effects, to mitochondrial dysfunction. However, where and how this nutrient excess affects mitochondrial and cytoplasmic energetic/redox crossroads remains to be defined in greater detail. RECENT ADVANCES We review how high glucose alters cellular redox balance and affects mitochondrial DNA. Next, we address how lipid excess, either stored in lipid droplets or utilized by mitochondria, affects performance in diabetic hearts by influencing cardiac energetic and redox assets. Finally, we examine how the reciprocal energetic/redox influence between mitochondrial and cytoplasmic compartments shapes myocardial mechanical activity during the course of DC, focusing especially on the glutathione and thioredoxin systems. FUTURE DIRECTIONS Protecting mitochondria from losing their ability to generate energy, and to control their own reactive oxygen species emission is essential to prevent the onset and/or to slow down DC progression. We highlight mechanisms enforced by the diabetic heart to counteract glucose/FAs surplus-induced damage, such as lipid storage, enhanced mitochondria-lipid droplet interaction, and upregulation of key antioxidant enzymes. Learning more on the nature and location of mechanisms sheltering mitochondrial functions would certainly help in further optimizing therapies for human DC.
Collapse
Affiliation(s)
- Miguel A Aon
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Carlo G Tocchetti
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Niraj Bhatt
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sonia Cortassa
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
122
|
Markevich NI, Hoek JB. Computational modeling analysis of mitochondrial superoxide production under varying substrate conditions and upon inhibition of different segments of the electron transport chain. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1847:656-79. [PMID: 25868872 PMCID: PMC4426091 DOI: 10.1016/j.bbabio.2015.04.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 03/24/2015] [Accepted: 04/06/2015] [Indexed: 12/13/2022]
Abstract
A computational mechanistic model of superoxide (O2•-) formation in the mitochondrial electron transport chain (ETC) was developed to facilitate the quantitative analysis of factors controlling mitochondrial O2•- production and assist in the interpretation of experimental studies. The model takes into account all individual electron transfer reactions in Complexes I and III. The model accounts for multiple, often seemingly contradictory observations on the effects of ΔΨ and ΔpH, and for the effects of multiple substrate and inhibitor conditions, including differential effects of Complex III inhibitors antimycin A, myxothiazol and stigmatellin. Simulation results confirm that, in addition to O2•- formation in Complex III and at the flavin site of Complex I, the quinone binding site of Complex I is an additional superoxide generating site that accounts for experimental observations on O2•- production during reverse electron transfer. However, our simulation results predict that, when cytochrome c oxidase is inhibited during oxidation of succinate, ROS production at this site is eliminated and almost all superoxide in Complex I is generated by reduced FMN, even when the redox pressure for reverse electron transfer from succinate is strong. In addition, the model indicates that conflicting literature data on the kinetics of electron transfer in Complex III involving the iron-sulfur protein-cytochrome bL complex can be resolved in favor of a dissociation of the protein only after electron transfer to cytochrome bH. The model predictions can be helpful in understanding factors driving mitochondrial superoxide formation in intact cells and tissues.
Collapse
Affiliation(s)
- Nikolai I Markevich
- MitoCare Center for Mitochondrial Research, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 14290, Russia.
| | - Jan B Hoek
- MitoCare Center for Mitochondrial Research, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
123
|
Kariž S, Mankoč S, Petrovič D. Association of thioredoxin reductase 2 (TXNRD2) gene polymorphisms with myocardial infarction in Slovene patients with type 2 diabetes mellitus. Diabetes Res Clin Pract 2015; 108:323-8. [PMID: 25703281 DOI: 10.1016/j.diabres.2015.01.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 12/28/2014] [Accepted: 01/30/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVES The aim of our study was to investigate the relationship between genetic polymorphisms in the mitochondrial thioredoxin reductase 2 (TrxR2) and myocardial infarction (MI) in subjects with type 2 diabetes mellitus (T2DM) of Slovenian origin. METHODS The study population consisted of 972 Caucasian subjects with T2DM of more than 10 years' duration: 161 patients with MI and 811 patients with no history of coronary artery disease. Polymorphisms in thioredoxin reductase 2 (TXNRD2) gene, rs1548357, rs4485648, and rs5748469, were studied. RESULTS Individuals carrying CC+CT genotypes of rs1548357 TXNRD2 polymorphism had lower prevalence of MI compared with TT genotype group (41.6% vs 52.8%, OR=0.589, 95% CI=0.368-0.942, P=0.027). CONCLUSIONS The TXNRD2 rs 1548357 polymorphism might be a genetic risk factor for MI in subjects with T2DM of Slovenian origin.
Collapse
Affiliation(s)
- Stojan Kariž
- Internal Department, General Hospital Izola, Polje 35, 6310 Izola, Slovenia
| | - Sara Mankoč
- Institute of Histology and Embryology, Faculty of Medicine, University Ljubljana, Korytkova 2, 1105 Ljubljana, Slovenia
| | - Daniel Petrovič
- Institute of Histology and Embryology, Faculty of Medicine, University Ljubljana, Korytkova 2, 1105 Ljubljana, Slovenia.
| |
Collapse
|
124
|
Ristić AJ, Savić D, Sokić D, Bogdanović Pristov J, Nestorov J, Baščarević V, Raičević S, Savić S, Spasojević I. Hippocampal antioxidative system in mesial temporal lobe epilepsy. Epilepsia 2015; 56:789-99. [DOI: 10.1111/epi.12981] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2015] [Indexed: 01/01/2023]
Affiliation(s)
- Aleksandar J. Ristić
- Center for Epilepsy and Sleep Disorders; Neurology Clinic; Clinical Center of Serbia; Belgrade Serbia
| | - Danijela Savić
- Department of Neurobiology; Institute for Biological Research “Siniša Stanković”; University of Belgrade; Belgrade Serbia
| | - Dragoslav Sokić
- Center for Epilepsy and Sleep Disorders; Neurology Clinic; Clinical Center of Serbia; Belgrade Serbia
| | - Jelena Bogdanović Pristov
- Life Sciences Department; Institute for Multidisciplinary Research; University of Belgrade; Belgrade Serbia
| | - Jelena Nestorov
- Department of Biochemistry; Institute for Biological Research “Siniša Stanković”; University of Belgrade; Belgrade Serbia
| | | | - Savo Raičević
- Institute for Neurosurgery; Clinical Center of Serbia; Belgrade Serbia
| | - Slobodan Savić
- Institute of Forensic Medicine; Medical School; University of Belgrade; Belgrade Serbia
| | - Ivan Spasojević
- Life Sciences Department; Institute for Multidisciplinary Research; University of Belgrade; Belgrade Serbia
| |
Collapse
|
125
|
Otero-Losada M, Cao G, González J, Muller A, Ottaviano G, Lillig C, Capani F, Ambrosio G, Milei J. Functional and morphological changes in endocrine pancreas following cola drink consumption in rats. PLoS One 2015; 10:e0118700. [PMID: 25790473 PMCID: PMC4366068 DOI: 10.1371/journal.pone.0118700] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 01/22/2015] [Indexed: 12/26/2022] Open
Abstract
Aim We report the effects of long-term cola beverage drinking on glucose homeostasis, endocrine pancreas function and morphology in rats. Methods Wistar rats drank: water (group W), regular cola beverage (group C, sucrose sweetened) or “light” cola beverage (group L, artificially sweetened). After 6 months, 50% of the animals in each group were euthanized and the remaining animals consumed water for the next 6 months when euthanasia was performed. Biochemical assays, insulinemia determination, estimation of insulin resistance (HOMA-IR), morphometry and immunohistochemistry evaluations were performed in pancreas. Results Hyperglycemia (16%, p<0.05), CoQ10 (coenzyme-Q10) decrease (−52%,p<0.01), strong hypertriglyceridemia (2.8-fold, p<0.01), hyperinsulinemia (2.4 fold, p<0.005) and HOMA-IR increase (2.7 fold, p<0.01) were observed in C. Group C showed a decrease in number of α cells (−42%, p<0.01) and β cells (−58%, p<0.001) and a moderate increase in α cells’ size after wash-out (+14%, p<0.001). Group L showed reduction in β cells’ size (−9%, p<0.001) and only after wash-out (L12) a 19% increase in size (p<0.0001) with 35% decrease in number of α cells (p<0.01). Groups C and L showed increase in α/β-cell ratio which was irreversible only in C (α/β = +38% in C6,+30% in C12, p<0.001vs.W6). Regular cola induced a striking increase in the cytoplasmic expression of Trx1 (Thioredoxin-1) (2.25-fold in C6 vs. W6; 2.7-fold in C12 vs. W12, p<0.0001) and Prx2 (Peroxiredoxin-2) (3-fold in C6 vs. W6; 2-fold in C12 vs. W12, p<0.0001). Light cola induced increase in Trx1 (3-fold) and Prx2 (2-fold) after wash-out (p<0.0001, L12 vs. W12). Conclusion Glucotoxicity may contribute to the loss of β cell function with depletion of insulin content. Oxidative stress, suggested by increased expression of thioredoxins and low circulating levels of CoQ10, may follow sustained hyperglycemia. A likely similar panorama may result from the effects of artificially sweetened cola though via other downstream routes.
Collapse
Affiliation(s)
- Matilde Otero-Losada
- Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, ININCA.UBA.CONICET, Buenos Aires, Argentina
- * E-mail:
| | - Gabriel Cao
- Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, ININCA.UBA.CONICET, Buenos Aires, Argentina
| | - Julián González
- Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, ININCA.UBA.CONICET, Buenos Aires, Argentina
| | - Angélica Muller
- Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, ININCA.UBA.CONICET, Buenos Aires, Argentina
| | - Graciela Ottaviano
- Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, ININCA.UBA.CONICET, Buenos Aires, Argentina
| | - Christopher Lillig
- Institut für Biochemie und Molekularbiologie, Universitätsmedizin Greifswald KdöR, Ernst Moritz Arndt Universität, Greifswald, Germany
| | - Francisco Capani
- Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, ININCA.UBA.CONICET, Buenos Aires, Argentina
| | - Giuseppe Ambrosio
- Università di Perugia, Cardiologia e Fisiopatologia Cardiovascolare, Perugia, Italy
| | - José Milei
- Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, ININCA.UBA.CONICET, Buenos Aires, Argentina
| |
Collapse
|
126
|
Wiggs MP. Can endurance exercise preconditioning prevention disuse muscle atrophy? Front Physiol 2015; 6:63. [PMID: 25814955 PMCID: PMC4356230 DOI: 10.3389/fphys.2015.00063] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/17/2015] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence suggests that exercise training can provide a level of protection against disuse muscle atrophy. Endurance exercise training imposes oxidative, metabolic, and heat stress on skeletal muscle which activates a variety of cellular signaling pathways that ultimately leads to the increased expression of proteins that have been demonstrated to protect muscle from inactivity -induced atrophy. This review will highlight the effect of exercise-induced oxidative stress on endogenous enzymatic antioxidant capacity (i.e., superoxide dismutase, glutathione peroxidase, and catalase), the role of oxidative and metabolic stress on PGC1-α, and finally highlight the effect heat stress and HSP70 induction. Finally, this review will discuss the supporting scientific evidence that these proteins can attenuate muscle atrophy through exercise preconditioning.
Collapse
Affiliation(s)
- Michael P Wiggs
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida Gainesville, FL, USA
| |
Collapse
|
127
|
Seidlmayer LK, Juettner VV, Kettlewell S, Pavlov EV, Blatter LA, Dedkova EN. Distinct mPTP activation mechanisms in ischaemia-reperfusion: contributions of Ca2+, ROS, pH, and inorganic polyphosphate. Cardiovasc Res 2015; 106:237-48. [PMID: 25742913 DOI: 10.1093/cvr/cvv097] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 01/30/2015] [Indexed: 12/16/2022] Open
Abstract
AIMS The mitochondrial permeability transition pore (mPTP) plays a central role for tissue damage and cell death during ischaemia-reperfusion (I/R). We investigated the contribution of mitochondrial inorganic polyphosphate (polyP), a potent activator of Ca(2+)-induced mPTP opening, towards mPTP activation and cardiac cell death in I/R. METHODS AND RESULTS A significant increase in mitochondrial free calcium concentration ([Ca(2+)]m), reactive oxygen species (ROS) generation, mitochondrial membrane potential depolarization (ΔΨm), and mPTP activity, but no cell death, was observed after 20 min of ischaemia. The [Ca(2+)]m increase during ischaemia was partially prevented by the mitochondrial Ca(2+) uniporter (MCU) inhibitor Ru360 and completely abolished by the combination of Ru360 and the ryanodine receptor type 1 blocker dantrolene, suggesting two complimentary Ca(2+) uptake mechanisms. In the absence of Ru360 and dantrolene, mPTP closing by polyP depletion or CSA decreased mitochondrial Ca(2+) uptake, suggesting that during ischaemia Ca(2+) can enter mitochondria through mPTP. During reperfusion, a burst of endogenous polyP production coincided with a decrease in [Ca(2+)]m, a decline in superoxide generation, and an acceleration of hydrogen peroxide (H2O2) production. An increase in H2O2 correlated with restoration of mitochondrial pHm and an increase in cell death. mPTP opening and cell death on reperfusion were prevented by antioxidants Trolox and MnTBAP [Mn (III) tetrakis (4-benzoic acid) porphyrin chloride]. Enzymatic polyP depletion did not affect mPTP opening during reperfusion, but increased ROS generation and cell death, suggesting that polyP plays a protective role in cellular stress response. CONCLUSIONS Transient Ca(2+)/polyP-mediated mPTP opening during ischaemia may serve to protect cells against cytosolic Ca(2+) overload, whereas ROS/pH-mediated sustained mPTP opening on reperfusion induces cell death.
Collapse
Affiliation(s)
- Lea K Seidlmayer
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, 1750 W. Harrison St, Chicago, IL 60612, USA
| | - Vanessa V Juettner
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, 1750 W. Harrison St, Chicago, IL 60612, USA
| | - Sarah Kettlewell
- Institute of Cardiovascular and Medical Sciences, College of Veterinary Medical and Life Sciences, University of Glasgow, Glasgow, UK
| | - Evgeny V Pavlov
- Dalhousie University, Halifax, NS, Canada New York University, NY, USA
| | - Lothar A Blatter
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, 1750 W. Harrison St, Chicago, IL 60612, USA
| | - Elena N Dedkova
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, 1750 W. Harrison St, Chicago, IL 60612, USA
| |
Collapse
|
128
|
Bhatt NM, Aon MA, Tocchetti CG, Shen X, Dey S, Ramirez-Correa G, O'Rourke B, Gao WD, Cortassa S. Restoring redox balance enhances contractility in heart trabeculae from type 2 diabetic rats exposed to high glucose. Am J Physiol Heart Circ Physiol 2014; 308:H291-302. [PMID: 25485897 DOI: 10.1152/ajpheart.00378.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hearts from type 2 diabetic (T2DM) subjects are chronically subjected to hyperglycemia and hyperlipidemia, both thought to contribute to oxidizing conditions and contractile dysfunction. How redox alterations and contractility interrelate, ultimately diminishing T2DM heart function, remains poorly understood. Herein we tested whether the fatty acid palmitate (Palm), in addition to its energetic contribution, rescues function by improving redox [glutathione (GSH), NAD(P)H, less oxidative stress] in T2DM rat heart trabeculae subjected to high glucose. Using cardiac trabeculae from Zucker Diabetic Fatty (ZDF) rats, we assessed the impact of low glucose (EG) and high glucose (HG), in absence or presence of Palm or insulin, on force development, energetics, and redox responses. We found that in EG ZDF and lean trabeculae displayed similar contractile work, yield of contractile work (Ycw), representing the ratio of force time integral over rate of O2 consumption. Conversely, HG had a negative impact on Ycw, whereas Palm, but not insulin, completely prevented contractile loss. This effect was associated with higher GSH, less oxidative stress, and augmented matrix GSH/thioredoxin (Trx) in ZDF mitochondria. Restoration of myocardial redox with GSH ethyl ester also rescued ZDF contractile function in HG, independently from Palm. These results support the idea that maintained redox balance, via increased GSH and Trx antioxidant activities to resist oxidative stress, is an essential protective response of the diabetic heart to keep contractile function.
Collapse
Affiliation(s)
- Niraj M Bhatt
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Miguel A Aon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Carlo G Tocchetti
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Xiaoxu Shen
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Swati Dey
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Genaro Ramirez-Correa
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sonia Cortassa
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| |
Collapse
|
129
|
Vajapey R, Rini D, Walston J, Abadir P. The impact of age-related dysregulation of the angiotensin system on mitochondrial redox balance. Front Physiol 2014; 5:439. [PMID: 25505418 PMCID: PMC4241834 DOI: 10.3389/fphys.2014.00439] [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] [Received: 08/25/2014] [Accepted: 10/27/2014] [Indexed: 12/13/2022] Open
Abstract
Aging is associated with the accumulation of various deleterious changes in cells. According to the free radical and mitochondrial theory of aging, mitochondria initiate most of the deleterious changes in aging and govern life span. The failure of mitochondrial reduction-oxidation (redox) homeostasis and the formation of excessive free radicals are tightly linked to dysregulation in the Renin Angiotensin System (RAS). A main rate-controlling step in RAS is renin, an enzyme that hydrolyzes angiotensinogen to generate angiotensin I. Angiotensin I is further converted to Angiotensin II (Ang II) by angiotensin-converting enzyme (ACE). Ang II binds with equal affinity to two main angiotensin receptors—type 1 (AT1R) and type 2 (AT2R). The binding of Ang II to AT1R activates NADPH oxidase, which leads to increased generation of cytoplasmic reactive oxygen species (ROS). This Ang II-AT1R–NADPH-ROS signal triggers the opening of mitochondrial KATP channels and mitochondrial ROS production in a positive feedback loop. Furthermore, RAS has been implicated in the decrease of many of ROS scavenging enzymes, thereby leading to detrimental levels of free radicals in the cell. AT2R is less understood, but evidence supports an anti-oxidative and mitochondria-protective function for AT2R. The overlap between age related changes in RAS and mitochondria, and the consequences of this overlap on age-related diseases are quite complex. RAS dysregulation has been implicated in many pathological conditions due to its contribution to mitochondrial dysfunction. Decreased age-related, renal and cardiac mitochondrial dysfunction was seen in patients treated with angiotensin receptor blockers. The aim of this review is to: (a) report the most recent information elucidating the role of RAS in mitochondrial redox hemostasis and (b) discuss the effect of age-related activation of RAS on generation of free radicals.
Collapse
Affiliation(s)
- Ramya Vajapey
- School of Medicine, Northeast Ohio Medical University Rootstown, OH, USA
| | - David Rini
- Division of Cellular and Molecular Medicine, Art as Applied to Medicine, Johns Hopkins University Baltimore, MD, USA
| | - Jeremy Walston
- Division of Geriatrics Medicine and Gerontology, Department of Medicine, Johns Hopkins University Baltimore, MD, USA
| | - Peter Abadir
- Division of Geriatrics Medicine and Gerontology, Department of Medicine, Johns Hopkins University Baltimore, MD, USA
| |
Collapse
|
130
|
Ghosh M, Sangwan N, Sangwan AK. Variations in free radical scavenging activities and antioxidant responses in salivary glands of Hyalomma anatolicum anatolicum and Hyalomma dromedarii (Acari: Ixodidae) ticks. Vet World 2014. [DOI: 10.14202/vetworld.2014.876-881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
131
|
Birkedal R, Laasmaa M, Vendelin M. The location of energetic compartments affects energetic communication in cardiomyocytes. Front Physiol 2014; 5:376. [PMID: 25324784 PMCID: PMC4178378 DOI: 10.3389/fphys.2014.00376] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 09/10/2014] [Indexed: 01/08/2023] Open
Abstract
The heart relies on accurate regulation of mitochondrial energy supply to match energy demand. The main regulators are Ca2+ and feedback of ADP and Pi. Regulation via feedback has intrigued for decades. First, the heart exhibits a remarkable metabolic stability. Second, diffusion of ADP and other molecules is restricted specifically in heart and red muscle, where a fast feedback is needed the most. To explain the regulation by feedback, compartmentalization must be taken into account. Experiments and theoretical approaches suggest that cardiomyocyte energetic compartmentalization is elaborate with barriers obstructing diffusion in the cytosol and at the level of the mitochondrial outer membrane (MOM). A recent study suggests the barriers are organized in a lattice with dimensions in agreement with those of intracellular structures. Here, we discuss the possible location of these barriers. The more plausible scenario includes a barrier at the level of MOM. Much research has focused on how the permeability of MOM itself is regulated, and the importance of the creatine kinase system to facilitate energetic communication. We hypothesize that at least part of the diffusion restriction at the MOM level is not by MOM itself, but due to the close physical association between the sarcoplasmic reticulum (SR) and mitochondria. This will explain why animals with a disabled creatine kinase system exhibit rather mild phenotype modifications. Mitochondria are hubs of energetics, but also ROS production and signaling. The close association between SR and mitochondria may form a diffusion barrier to ADP added outside a permeabilized cardiomyocyte. But in vivo, it is the structural basis for the mitochondrial-SR coupling that is crucial for the regulation of mitochondrial Ca2+-transients to regulate energetics, and for avoiding Ca2+-overload and irreversible opening of the mitochondrial permeability transition pore.
Collapse
Affiliation(s)
- Rikke Birkedal
- Laboratory of Systems Biology, Institute of Cybernetics, Tallinn University of Technology Tallinn, Estonia
| | - Martin Laasmaa
- Laboratory of Systems Biology, Institute of Cybernetics, Tallinn University of Technology Tallinn, Estonia
| | - Marko Vendelin
- Laboratory of Systems Biology, Institute of Cybernetics, Tallinn University of Technology Tallinn, Estonia
| |
Collapse
|
132
|
Zorov DB, Juhaszova M, Sollott SJ. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev 2014; 94:909-50. [PMID: 24987008 DOI: 10.1152/physrev.00026.2013] [Citation(s) in RCA: 3715] [Impact Index Per Article: 337.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Byproducts of normal mitochondrial metabolism and homeostasis include the buildup of potentially damaging levels of reactive oxygen species (ROS), Ca(2+), etc., which must be normalized. Evidence suggests that brief mitochondrial permeability transition pore (mPTP) openings play an important physiological role maintaining healthy mitochondria homeostasis. Adaptive and maladaptive responses to redox stress may involve mitochondrial channels such as mPTP and inner membrane anion channel (IMAC). Their activation causes intra- and intermitochondrial redox-environment changes leading to ROS release. This regenerative cycle of mitochondrial ROS formation and release was named ROS-induced ROS release (RIRR). Brief, reversible mPTP opening-associated ROS release apparently constitutes an adaptive housekeeping function by the timely release from mitochondria of accumulated potentially toxic levels of ROS (and Ca(2+)). At higher ROS levels, longer mPTP openings may release a ROS burst leading to destruction of mitochondria, and if propagated from mitochondrion to mitochondrion, of the cell itself. The destructive function of RIRR may serve a physiological role by removal of unwanted cells or damaged mitochondria, or cause the pathological elimination of vital and essential mitochondria and cells. The adaptive release of sufficient ROS into the vicinity of mitochondria may also activate local pools of redox-sensitive enzymes involved in protective signaling pathways that limit ischemic damage to mitochondria and cells in that area. Maladaptive mPTP- or IMAC-related RIRR may also be playing a role in aging. Because the mechanism of mitochondrial RIRR highlights the central role of mitochondria-formed ROS, we discuss all of the known ROS-producing sites (shown in vitro) and their relevance to the mitochondrial ROS production in vivo.
Collapse
Affiliation(s)
- Dmitry B Zorov
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; and Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Magdalena Juhaszova
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; and Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Steven J Sollott
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; and Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
133
|
Benfeitas R, Selvaggio G, Antunes F, Coelho PMBM, Salvador A. Hydrogen peroxide metabolism and sensing in human erythrocytes: a validated kinetic model and reappraisal of the role of peroxiredoxin II. Free Radic Biol Med 2014; 74:35-49. [PMID: 24952139 DOI: 10.1016/j.freeradbiomed.2014.06.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/26/2014] [Accepted: 06/10/2014] [Indexed: 01/09/2023]
Abstract
Hydrogen peroxide (H2O2) metabolism in human erythrocytes has been thoroughly investigated, but unclear points persist. By integrating the available data into a mathematical model that accurately represents the current understanding and comparing computational predictions to observations we sought to (a) identify inconsistencies in present knowledge, (b) propose resolutions, and (c) examine their functional implications. The systematic confrontation of computational predictions with experimental observations of the responses of intact erythrocytes highlighted the following important discrepancy. The high rate constant (10(7)-10(8) M(-1) s(-1)) for H2O2 reduction determined for purified peroxiredoxin II (Prx2) and the high abundance of this protein indicate that under physiological conditions it consumes practically all the H2O2. However, this is inconsistent with extensive evidence that Prx2's contribution to H2O2 elimination is comparable to that of catalase. Models modified such that Prx2's effective peroxidase activity is just 10(5) M(-1) s(-1) agree near quantitatively with extensive experimental observations. This low effective activity is probably due to a strong but readily reversible inhibition of Prx2's peroxidatic activity in intact cells, implying that the main role of Prx2 in human erythrocytes is not to eliminate peroxide substrates. Simulations of the responses to physiological H2O2 stimuli highlight that a design combining abundant Prx2 with a low effective peroxidase activity spares NADPH while improving potential signaling properties of the Prx2/thioredoxin/thioredoxin reductase system.
Collapse
Affiliation(s)
- Rui Benfeitas
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Gianluca Selvaggio
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Fernando Antunes
- Departamento de Química e Bioquímica and Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Pedro M B M Coelho
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Armindo Salvador
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Coimbra Chemistry Center, University of Coimbra, 3004-535 Coimbra, Portugal.
| |
Collapse
|
134
|
O-Uchi J, Ryu SY, Jhun BS, Hurst S, Sheu SS. Mitochondrial ion channels/transporters as sensors and regulators of cellular redox signaling. Antioxid Redox Signal 2014; 21:987-1006. [PMID: 24180309 PMCID: PMC4116125 DOI: 10.1089/ars.2013.5681] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Mitochondrial ion channels/transporters and the electron transport chain (ETC) serve as key sensors and regulators for cellular redox signaling, the production of reactive oxygen species (ROS) and nitrogen species (RNS) in mitochondria, and balancing cell survival and death. Although the functional and pharmacological characteristics of mitochondrial ion transport mechanisms have been extensively studied for several decades, the majority of the molecular identities that are responsible for these channels/transporters have remained a mystery until very recently. RECENT ADVANCES Recent breakthrough studies uncovered the molecular identities of the diverse array of major mitochondrial ion channels/transporters, including the mitochondrial Ca2+ uniporter pore, mitochondrial permeability transition pore, and mitochondrial ATP-sensitive K+ channel. This new information enables us to form detailed molecular and functional characterizations of mitochondrial ion channels/transporters and their roles in mitochondrial redox signaling. CRITICAL ISSUES Redox-mediated post-translational modifications of mitochondrial ion channels/transporters and ETC serve as key mechanisms for the spatiotemporal control of mitochondrial ROS/RNS generation. FUTURE DIRECTIONS Identification of detailed molecular mechanisms for redox-mediated regulation of mitochondrial ion channels will enable us to find novel therapeutic targets for many diseases that are associated with cellular redox signaling and mitochondrial ion channels/transporters.
Collapse
Affiliation(s)
- Jin O-Uchi
- 1 Department of Medicine, Center for Translational Medicine, Jefferson Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | | | | | | | | |
Collapse
|
135
|
Wang S, Wang X, Luo F, Tang X, Li K, Hu X, Bai J. Panaxatriol saponin ameliorated liver injury by acetaminophen via restoring thioredoxin-1 and pro-caspase-12. Liver Int 2014; 34:1068-73. [PMID: 24119161 DOI: 10.1111/liv.12329] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 08/31/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Acetaminophen (APAP) is widely used as an antipyretic agent which is safe at therapeutic doses. However, overdose of APAP induces fatal and non-fatal hepatic necroses. The chemical reactive metabolites of APAP initiate toxicity and inflammatory response within the liver and lead to acute liver failure. However, the mechanism underlying APAP-induced liver injury is unknown. Thioredoxin-1 (TRX-1) is an important redox regulator, which plays roles in resisting oxidative stress, regulating inflammation and inhibiting apoptosis. Panaxatriol saponin (PTS) is one of the biologically active fractions of Panax notoginseng which is a traditional Chinese medicine. The aim of this study was to investigate the mechanism on PTS protecting liver from APAP hepatotoxicity. METHODS Mice were divided into three groups, control group, APAP group and APAP combined with PTS group. Alanine aminotransferase (ALT) and tumour necrosis factor-alpha (TNF-α) were detected by ELISA. TRX-1 and pro-caspase-12 were examined by Western blotting. RESULTS Our results showed PTS inhibited the levels of ALT and TNF-α by APAP. Pretreatment with PTS ameliorated liver injury induced by APAP. The decrease in TRX-1 expression was restored by PTS, as well as decreased pro-caspase-12 expression was inhibited by PTS. These data suggest that PTS has roles in suppressing the hepatotoxicity by APAP. CONCLUSION Panaxatriol saponin ameliorated liver injury by APAP through restoring the expression TRX-1 and inhibiting pro-caspase-12 decrease.
Collapse
Affiliation(s)
- Shengdong Wang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, China; Department of Science and Education Section, the seventh People's Hospital of Hangzhou, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
136
|
Antioxidant defences of Spironucleus vortens: Glutathione is the major non-protein thiol. Mol Biochem Parasitol 2014; 196:45-52. [DOI: 10.1016/j.molbiopara.2014.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/21/2014] [Accepted: 07/28/2014] [Indexed: 11/17/2022]
|
137
|
Aon MA, Bhatt N, Cortassa SC. Mitochondrial and cellular mechanisms for managing lipid excess. Front Physiol 2014; 5:282. [PMID: 25132820 PMCID: PMC4116787 DOI: 10.3389/fphys.2014.00282] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 07/10/2014] [Indexed: 12/16/2022] Open
Abstract
Current scientific debates center on the impact of lipids and mitochondrial function on diverse aspects of human health, nutrition and disease, among them the association of lipotoxicity with the onset of insulin resistance in skeletal muscle, and with heart dysfunction in obesity and diabetes. Mitochondria play a fundamental role in aging and in prevalent acute or chronic diseases. Lipids are main mitochondrial fuels however these molecules can also behave as uncouplers and inhibitors of oxidative phosphorylation. Knowledge about the functional composition of these contradictory effects and their impact on mitochondrial-cellular energetics/redox status is incomplete. Cells store fatty acids (FAs) as triacylglycerol and package them into cytoplasmic lipid droplets (LDs). New emerging data shows the LD as a highly dynamic storage pool of FAs that can be used for energy reserve. Lipid excess packaging into LDs can be seen as an adaptive response to fulfilling energy supply without hindering mitochondrial or cellular redox status and keeping low concentration of lipotoxic intermediates. Herein we review the mechanisms of action and utilization of lipids by mitochondria reported in liver, heart and skeletal muscle under relevant physiological situations, e.g., exercise. We report on perilipins, a family of proteins that associate with LDs in response to loading of cells with lipids. Evidence showing that in addition to physical contact, mitochondria and LDs exhibit metabolic interactions is presented and discussed. A hypothetical model of channeled lipid utilization by mitochondria is proposed. Direct delivery and channeled processing of lipids in mitochondria could represent a reliable and efficient way to maintain reactive oxygen species (ROS) within levels compatible with signaling while ensuring robust and reliable energy supply.
Collapse
Affiliation(s)
- Miguel A Aon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Niraj Bhatt
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Sonia C Cortassa
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine Baltimore, MD, USA
| |
Collapse
|
138
|
Kembro JM, Cortassa S, Aon MA. Complex oscillatory redox dynamics with signaling potential at the edge between normal and pathological mitochondrial function. Front Physiol 2014; 5:257. [PMID: 25071602 PMCID: PMC4085651 DOI: 10.3389/fphys.2014.00257] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 06/17/2014] [Indexed: 12/31/2022] Open
Abstract
The time-keeping properties bestowed by oscillatory behavior on functional rhythms represent an evolutionarily conserved trait in living systems. Mitochondrial networks function as timekeepers maximizing energetic output while tuning reactive oxygen species (ROS) within physiological levels compatible with signaling. In this work, we explore the potential for timekeeping functions dependent on mitochondrial dynamics with the validated two-compartment mitochondrial energetic-redox (ME-R) computational model, that takes into account (a) four main redox couples [NADH, NADPH, GSH, Trx(SH)2], (b) scavenging systems (glutathione, thioredoxin, SOD, catalase) distributed in matrix and extra-matrix compartments, and (c) transport of ROS species between them. Herein, we describe that the ME-R model can exhibit highly complex oscillatory dynamics in energetic/redox variables and ROS species, consisting of at least five frequencies with modulated amplitudes and period according to power spectral analysis. By stability analysis we describe that the extent of steady state—as against complex oscillatory behavior—was dependent upon the abundance of Mn and Cu, Zn SODs, and their interplay with ROS production in the respiratory chain. Large parametric regions corresponding to oscillatory dynamics of increasingly complex waveforms were obtained at low Cu, Zn SOD concentration as a function of Mn SOD. This oscillatory domain was greatly reduced at higher levels of Cu, Zn SOD. Interestingly, the realm of complex oscillations was located at the edge between normal and pathological mitochondrial energetic behavior, and was characterized by oxidative stress. We conclude that complex oscillatory dynamics could represent a frequency- and amplitude-modulated H2O2 signaling mechanism that arises under intense oxidative stress. By modulating SOD, cells could have evolved an adaptive compromise between relative constancy and the flexibility required under stressful redox/energetic conditions.
Collapse
Affiliation(s)
- Jackelyn M Kembro
- Facultad de Ciencias Exactas, Físicas y Naturales, Instituto de Investigaciones Biológicas y Tecnológicas (Consejo Nacional de Investigaciones Científicas y Técnicas-UNC) and Instituto de Ciencia y Tecnología de los Alimentos, Universidad Nacional de Córdoba Córdoba, Argentina
| | - Sonia Cortassa
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Miguel A Aon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine Baltimore, MD, USA
| |
Collapse
|
139
|
Duivenvoorde LPM, van Schothorst EM, Derous D, van der Stelt I, Masania J, Rabbani N, Thornalley PJ, Keijer J. Oxygen restriction as challenge test reveals early high-fat-diet-induced changes in glucose and lipid metabolism. Pflugers Arch 2014; 467:1179-93. [PMID: 24974902 DOI: 10.1007/s00424-014-1553-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 05/06/2014] [Accepted: 06/09/2014] [Indexed: 01/17/2023]
Abstract
Challenge tests stress homeostasis and may reveal deviations in health that remain masked under unchallenged conditions. Ideally, challenge tests are non-invasive and applicable in an early phase of an animal experiment. Oxygen restriction (OxR; based on ambient, mild normobaric hypoxia) is a non-invasive challenge test that measures the flexibility to adapt metabolism. Metabolic inflexibility is one of the hallmarks of the metabolic syndrome. To test whether OxR can be used to reveal early diet-induced health effects, we exposed mice to a low-fat (LF) or high-fat (HF) diet for only 5 days. The response to OxR was assessed by calorimetric measurements, followed by analysis of gene expression in liver and epididymal white adipose tissue (eWAT) and serum markers for e.g. protein glycation and oxidation. Although HF feeding increased body weight, HF and LF mice did not differ in indirect calorimetric values under normoxic conditions and in a fasting state. Exposure to OxR; however, increased oxygen consumption and lipid oxidation in HF mice versus LF mice. Furthermore, OxR induced gluconeogenesis and an antioxidant response in the liver of HF mice, whereas it induced de novo lipogenesis and an antioxidant response in eWAT of LF mice, indicating that HF and LF mice differed in their adaptation to OxR. OxR also increased serum markers of protein glycation and oxidation in HF mice, whereas these changes were absent in LF mice. Cumulatively, OxR is a promising new method to test food products on potential beneficial effects for human health.
Collapse
Affiliation(s)
- Loes P M Duivenvoorde
- Human and Animal Physiology, Wageningen University, De Elst 1, 6708 WD, Wageningen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
140
|
Grattagliano I, Calamita G, Cocco T, Wang DQH, Portincasa P. Pathogenic role of oxidative and nitrosative stress in primary biliary cirrhosis. World J Gastroenterol 2014; 20:5746-5759. [PMID: 24914336 PMCID: PMC4024785 DOI: 10.3748/wjg.v20.i19.5746] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 01/18/2014] [Accepted: 03/07/2014] [Indexed: 02/06/2023] Open
Abstract
Primary biliary cirrhosis is a multifactor autoimmune disease characterized by hepatic and systemic manifestations, with immune system dysregulation and abnormalities in the hepatic metabolism of bile salts, lipids, and nutrients, as well as destruction of membrane lipids and mitochondrial dysfunction. Both oxidative and nitrosative stress are associated with ongoing manifestations of the disease. In particular, abnormalities in nitric oxide metabolism and thiol oxidation already occur at early stages, thus leading to the hypothesis that these biochemical events play a pathogenic role in primary biliary cirrhosis. Moreover, the association of these metabolic abnormalities with the progression of the disease may indicate some biochemical parameters as early diagnostic markers of disease evolution, and may open up the potential for pharmacological intervention to inhibit intra- and extra-cellular stress events for resuming hepatocellular functions. The following paragraphs summarize the current knowledge by outlining molecular mechanisms of the disease related to these stress events.
Collapse
|
141
|
The Glutathione System: A New Drug Target in Neuroimmune Disorders. Mol Neurobiol 2014; 50:1059-84. [DOI: 10.1007/s12035-014-8705-x] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 03/31/2014] [Indexed: 01/17/2023]
|
142
|
Mitochondrial reactive oxygen species production and elimination. J Mol Cell Cardiol 2014; 73:26-33. [PMID: 24657720 DOI: 10.1016/j.yjmcc.2014.03.011] [Citation(s) in RCA: 234] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 02/24/2014] [Accepted: 03/14/2014] [Indexed: 12/31/2022]
Abstract
Reactive oxygen species (ROS) play an important role in cardiovascular diseases, and one important source for ROS are mitochondria. Emission of ROS from mitochondria is the net result of ROS production at the electron transport chain (ETC) and their elimination by antioxidative enzymes. Both of these processes are highly dependent on the mitochondrial redox state, which is dynamically altered under different physiological and pathological conditions. The concept of "redox-optimized ROS balance" integrates these aspects and implies that oxidative stress occurs when the optimal equilibrium of an intermediate redox state is disturbed towards either strong oxidation or reduction. Furthermore, mitochondria integrate ROS signals from other cellular sources, presumably through a process termed "ROS-induced ROS release" that involves mitochondrial ion channels. Here, we attempt to integrate these recent advances in our understanding of the control of mitochondrial ROS emission and develop a concept of how in heart failure, defects in ion handling can lead to mitochondrial oxidative stress. This article is part of a Special Issue entitled "Redox Signalling in the Cardiovascular System".
Collapse
|
143
|
Pannala VR, Bazil JN, Camara AKS, Dash RK. A mechanistic mathematical model for the catalytic action of glutathione peroxidase. Free Radic Res 2014; 48:487-502. [PMID: 24456207 DOI: 10.3109/10715762.2014.886775] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Glutathione peroxidase (GPx) is a well-known seleno-enzyme that protects cells from oxidative stress (e.g., lipid peroxidation and oxidation of other cellular proteins and macromolecules), by catalyzing the reduction of harmful peroxides (e.g., hydrogen peroxide: H₂O₂) with reduced glutathione (GSH). However, the catalytic mechanism of GPx kinetics is not well characterized in terms of a mathematical model. We developed here a mechanistic mathematical model of GPx kinetics by considering a unified catalytic scheme and estimated the unknown model parameters based on different experimental data from the literature on the kinetics of the enzyme. The model predictions are consistent with the consensus that GPx operates via a ping-pong mechanism. The unified catalytic scheme proposed here for GPx kinetics clarifies various anomalies, such as what are the individual steps in the catalytic scheme by estimating their associated rate constant values and a plausible rationale for the contradicting experimental results. The developed model presents a unique opportunity to understand the effects of pH and product GSSG on the GPx activity under both physiological and pathophysiological conditions. Although model parameters related to the product GSSG were not identifiable due to lack of product-inhibition data, the preliminary model simulations with the assumed range of parameters show that the inhibition by the product GSSG is negligible, consistent with what is known in the literature. In addition, the model is able to simulate the bi-modal behavior of the GPx activity with respect to pH with the pH-range for maximal GPx activity decreasing significantly as the GSH levels decrease and H₂O₂ levels increase (characteristics of oxidative stress). The model provides a key component for an integrated model of H₂O₂ balance under normal and oxidative stress conditions.
Collapse
Affiliation(s)
- V R Pannala
- Biotechnology and Bioengineering Center and Department of Physiology , Milwaukee , US
| | | | | | | |
Collapse
|
144
|
Dröse S, Brandt U, Wittig I. Mitochondrial respiratory chain complexes as sources and targets of thiol-based redox-regulation. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:1344-54. [PMID: 24561273 DOI: 10.1016/j.bbapap.2014.02.006] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 02/05/2014] [Accepted: 02/08/2014] [Indexed: 02/06/2023]
Abstract
The respiratory chain of the inner mitochondrial membrane is a unique assembly of protein complexes that transfers the electrons of reducing equivalents extracted from foodstuff to molecular oxygen to generate a proton-motive force as the primary energy source for cellular ATP-synthesis. Recent evidence indicates that redox reactions are also involved in regulating mitochondrial function via redox-modification of specific cysteine-thiol groups in subunits of respiratory chain complexes. Vice versa the generation of reactive oxygen species (ROS) by respiratory chain complexes may have an impact on the mitochondrial redox balance through reversible and irreversible thiol-modification of specific target proteins involved in redox signaling, but also pathophysiological processes. Recent evidence indicates that thiol-based redox regulation of the respiratory chain activity and especially S-nitrosylation of complex I could be a strategy to prevent elevated ROS production, oxidative damage and tissue necrosis during ischemia-reperfusion injury. This review focuses on the thiol-based redox processes involving the respiratory chain as a source as well as a target, including a general overview on mitochondria as highly compartmentalized redox organelles and on methods to investigate the redox state of mitochondrial proteins. This article is part of a Special Issue entitled: Thiol-Based Redox Processes.
Collapse
Affiliation(s)
- Stefan Dröse
- Clinic of Anesthesiology, Intensive-Care Medicine and Pain Therapy, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Ulrich Brandt
- Radboud University Medical Centre, Nijmegen Centre for Mitochondrial Disorders, Geert Grooteplein-Zuid 10, 6525 GA Nijmegen, The Netherlands; Cluster of Excellence "Macromolecular Complexes", Goethe-University, Frankfurt am Main, Germany.
| | - Ilka Wittig
- Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Johann Wolfgang Goethe University, 60590 Frankfurt am Main, Germany; Cluster of Excellence "Macromolecular Complexes", Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
145
|
Analysis of the kinetics and bistability of ubiquinol:cytochrome c oxidoreductase. Biophys J 2014; 105:343-55. [PMID: 23870256 DOI: 10.1016/j.bpj.2013.05.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 03/28/2013] [Accepted: 05/13/2013] [Indexed: 11/21/2022] Open
Abstract
Ubiquinol:cytochrome c oxidoreductase, bc1 complex, is the enzyme in the respiratory chain of mitochondria responsible for the transfer reducing potential from ubiquinol to cytochrome c coupled to the movement of charge against the electrostatic potential across the mitochondrial inner membrane. The complex is also implicated in the generation of reactive oxygen species under certain conditions and is thus a contributor to cellular oxidative stress. Here, a biophysically detailed, thermodynamically consistent model of the bc1 complex for mammalian mitochondria is developed. The model incorporates the major redox centers near the Qo- and Qi-site of the enzyme, includes the pH-dependent redox reactions, accounts for the effect of the proton-motive force of the reaction rate, and simulates superoxide production at the Qo-site. The model consists of six distinct states characterized by the mobile electron distribution in the enzyme. Within each state, substates that correspond to various electron localizations exist in a rapid equilibrium distribution. The steady-state equation for the six-state system is parameterized using five independent data sets and validated in comparison to additional experimental data. Model analysis suggests that the pH-dependence on turnover is primarily due to the pKa values of cytochrome bH and Rieske iron sulfur protein. A previously proposed kinetic scheme at the Qi-site where ubiquinone binds to only the reduced enzyme and ubiquinol binds to only the oxidized enzyme is shown to be thermodynamically infeasible. Moreover, the model is able to reproduce the bistability phenomenon where at a given overall flux through the enzyme, different rates of superoxide production are attained when the enzyme is differentially reduced.
Collapse
|
146
|
Complex Systems Biology of Networks: The Riddle and the Challenge. SYSTEMS BIOLOGY OF METABOLIC AND SIGNALING NETWORKS 2014. [DOI: 10.1007/978-3-642-38505-6_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
147
|
Gauthier LD, Greenstein JL, O'Rourke B, Winslow RL. An integrated mitochondrial ROS production and scavenging model: implications for heart failure. Biophys J 2013; 105:2832-42. [PMID: 24359755 PMCID: PMC3882515 DOI: 10.1016/j.bpj.2013.11.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 08/30/2013] [Accepted: 11/05/2013] [Indexed: 12/27/2022] Open
Abstract
It has been observed experimentally that cells from failing hearts exhibit elevated levels of reactive oxygen species (ROS) upon increases in energetic workload. One proposed mechanism for this behavior is mitochondrial Ca(2+) mismanagement that leads to depletion of ROS scavengers. Here, we present a computational model to test this hypothesis. Previously published models of ROS production and scavenging were combined and reparameterized to describe ROS regulation in the cellular environment. Extramitochondrial Ca(2+) pulses were applied to simulate frequency-dependent changes in cytosolic Ca(2+). Model results show that decreased mitochondrial Ca(2+)uptake due to mitochondrial Ca(2+) uniporter inhibition (simulating Ru360) or elevated cytosolic Na(+), as in heart failure, leads to a decreased supply of NADH and NADPH upon increasing cellular workload. Oxidation of NADPH leads to oxidation of glutathione (GSH) and increased mitochondrial ROS levels, validating the Ca(2+) mismanagement hypothesis. The model goes on to predict that the ratio of steady-state [H2O2]m during 3Hz pacing to [H2O2]m at rest is highly sensitive to the size of the GSH pool. The largest relative increase in [H2O2]m in response to pacing is shown to occur when the total GSH and GSSG is close to 1 mM, whereas pool sizes below 0.9 mM result in high resting H2O2 levels, a quantitative prediction only possible with a computational model.
Collapse
Affiliation(s)
- Laura D Gauthier
- Institute for Computational Medicine and Department of Biomedical Engineering, The Johns Hopkins University School of Medicine and Whiting School of Engineering, Baltimore, Maryland.
| | - Joseph L Greenstein
- Institute for Computational Medicine and Department of Biomedical Engineering, The Johns Hopkins University School of Medicine and Whiting School of Engineering, Baltimore, Maryland
| | - Brian O'Rourke
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore Maryland
| | - Raimond L Winslow
- Institute for Computational Medicine and Department of Biomedical Engineering, The Johns Hopkins University School of Medicine and Whiting School of Engineering, Baltimore, Maryland
| |
Collapse
|
148
|
Pannala VR, Bazil JN, Camara AKS, Dash RK. A biophysically based mathematical model for the catalytic mechanism of glutathione reductase. Free Radic Biol Med 2013; 65:1385-1397. [PMID: 24120751 PMCID: PMC3870161 DOI: 10.1016/j.freeradbiomed.2013.10.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 09/04/2013] [Accepted: 10/01/2013] [Indexed: 12/11/2022]
Abstract
Glutathione reductase (GR) catalyzes the reduction of oxidized glutathione (GSSG) to reduced glutathione (GSH) using NADPH as the reducing cofactor, and thereby maintains a constant GSH level in the system. GSH scavenges superoxide (O2(*-)) and hydroxyl radicals (OH) nonenzymatically or by serving as an electron donor to several enzymes involved in reactive oxygen species (ROS) detoxification. In either case, GSH oxidizes to GSSG and is subsequently regenerated by the catalytic action of GR. Although the GR kinetic mechanism has been extensively studied under various experimental conditions with variable substrates and products, the catalytic mechanism has not been studied in terms of a mechanistic model that accounts for the effects of the substrates and products on the reaction kinetics. The aim of this study is therefore to develop a comprehensive mathematical model for the catalytic mechanism of GR. We use available experimental data on GR kinetics from various species/sources to develop the mathematical model and estimate the associated model parameters. The model simulations are consistent with the experimental observation that GR operates via both ping-pong and sequential branching mechanisms based on relevant concentrations of its reaction substrate GSSG. Furthermore, we show the observed pH-dependent substrate inhibition of GR activity by GSSG and bimodal behavior of GR activity with pH. The model presents a unique opportunity to understand the effects of products on the kinetics of GR. The model simulations show that under physiological conditions, where both substrates and products are present, the flux distribution depends on the concentrations of both GSSG and NADP(+), with ping-pong flux operating at low levels and sequential flux dominating at higher levels. The kinetic model of GR may serve as a key module for the development of integrated models for ROS-scavenging systems to understand protection of cells under normal and oxidative stress conditions.
Collapse
Affiliation(s)
- Venkat R Pannala
- Biotechnology and Bioengineering Center and Department of Physiology and
| | - Jason N Bazil
- Biotechnology and Bioengineering Center and Department of Physiology and
| | - Amadou K S Camara
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ranjan K Dash
- Biotechnology and Bioengineering Center and Department of Physiology and.
| |
Collapse
|
149
|
Cortassa S, O'Rourke B, Aon MA. Redox-optimized ROS balance and the relationship between mitochondrial respiration and ROS. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2013; 1837:287-95. [PMID: 24269780 DOI: 10.1016/j.bbabio.2013.11.007] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 11/07/2013] [Accepted: 11/11/2013] [Indexed: 01/01/2023]
Abstract
The Redox-Optimized ROS Balance [R-ORB] hypothesis postulates that the redox environment [RE] is the main intermediary between mitochondrial respiration and reactive oxygen species [ROS]. According to R-ORB, ROS emission levels will attain a minimum vs. RE when respiratory rate (VO2) reaches a maximum following ADP stimulation, a tenet that we test herein in isolated heart mitochondria under forward electron transport [FET]. ROS emission increased two-fold as a function of changes in the RE (~400 to ~900mV·mM) in state 4 respiration elicited by increasing glutamate/malate (G/M). In G/M energized mitochondria, ROS emission decreases two-fold for RE ~500 to ~300mV·mM in state 3 respiration at increasing ADP. Stressed mitochondria released higher ROS, that was only weakly dependent on RE under state 3. As a function of VO2, the ROS dependence on RE was strong between ~550 and ~350mV·mM, when VO2 is maximal, primarily due to changes in glutathione redox potential. A similar dependence was observed with stressed mitochondria, but over a significantly more oxidized RE and ~3-fold higher ROS emission overall, as compared with non-stressed controls. We conclude that under non-stressful conditions mitochondrial ROS efflux decreases when the RE becomes less reduced within a range in which VO2 is maximal. These results agree with the R-ORB postulate that mitochondria minimize ROS emission as they maximize VO2 and ATP synthesis. This relationship is altered quantitatively, but not qualitatively, by oxidative stress although stressed mitochondria exhibit diminished energetic performance and increased ROS release.
Collapse
Affiliation(s)
- Sonia Cortassa
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian O'Rourke
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Miguel A Aon
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
150
|
Kato T, Zhou X, Ma Y. Possible involvement of nitric oxide and reactive oxygen species in glucose deprivation-induced activation of transcription factor rst2. PLoS One 2013; 8:e78012. [PMID: 24155978 PMCID: PMC3796501 DOI: 10.1371/journal.pone.0078012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 09/07/2013] [Indexed: 11/18/2022] Open
Abstract
Glucose is one of the most important sources of cellular nutrition and glucose deprivation induces various cellular responses. In Schizosaccharomyces pombe, zinc finger protein Rst2 is activated upon glucose deprivation, and regulates gene expression via the STREP (stress response element of Schizosaccharomyces pombe) motif. However, the activation mechanism of Rst2 is not fully understood. We monitored Rst2 transcriptional activity in living cells using a Renilla luciferase reporter system. Hydrogen peroxide (H2O2) enhanced Rst2 transcriptional activity upon glucose deprivation and free radical scavenger inhibited Rst2 transcriptional activity upon glucose deprivation. In addition, deletion of the trx2 (+) gene encoding mitochondrial thioredoxin enhanced Rst2 transcriptional activity. Notably, nitric oxide (NO) generators enhanced Rst2 transcriptional activity upon glucose deprivation as well as under glucose-rich conditions. Furthermore, NO specific scavenger inhibited Rst2 transcriptional activity upon glucose deprivation. Altogether, our data suggest that NO and reactive oxygen species may be involved in the activation of transcription factor Rst2.
Collapse
Affiliation(s)
- Toshiaki Kato
- Division of Molecular Pharmacology and Pharmacogenomics, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Xin Zhou
- Division of Molecular Pharmacology and Pharmacogenomics, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- The First Affiliated Hospital of Liaoning Medical University, Jinzhou City, Liaoning Province, China
| | - Yan Ma
- Division of Molecular Pharmacology and Pharmacogenomics, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- * E-mail:
| |
Collapse
|