101
|
Langdon-Jones EE, Lloyd D, Hayes AJ, Wainwright SD, Mottram HJ, Coles SJ, Horton PN, Pope SJA. Alkynyl-naphthalimide Fluorophores: Gold Coordination Chemistry and Cellular Imaging Applications. Inorg Chem 2015; 54:6606-15. [DOI: 10.1021/acs.inorgchem.5b00954] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Emily E. Langdon-Jones
- School of Chemistry, Main Building, Cardiff University, Cardiff CF10 3AT, United Kingdom
| | - David Lloyd
- School of Biosciences, Main Building, Cardiff University, Cardiff CF10 3AT, United Kingdom
| | - Anthony J. Hayes
- School of Biosciences, Main Building, Cardiff University, Cardiff CF10 3AT, United Kingdom
| | - Shane D. Wainwright
- School of Biosciences, Main Building, Cardiff University, Cardiff CF10 3AT, United Kingdom
| | - Huw J. Mottram
- School of Pharmacy,
Redwood Building, Cardiff University, Cardiff CF10 3NB, United Kingdom
| | - Simon J. Coles
- UK National Crystallographic Service, Chemistry,
Faculty of Natural and Environmental Sciences, University of Southampton, Highfield,
Southampton SO17 1BJ, United Kingdom
| | - Peter N. Horton
- UK National Crystallographic Service, Chemistry,
Faculty of Natural and Environmental Sciences, University of Southampton, Highfield,
Southampton SO17 1BJ, United Kingdom
| | - Simon J. A. Pope
- School of Chemistry, Main Building, Cardiff University, Cardiff CF10 3AT, United Kingdom
| |
Collapse
|
102
|
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
|
103
|
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
|
104
|
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
|
105
|
Sung MM, Hamza SM, Dyck JRB. Myocardial metabolism in diabetic cardiomyopathy: potential therapeutic targets. Antioxid Redox Signal 2015; 22:1606-30. [PMID: 25808033 DOI: 10.1089/ars.2015.6305] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Cardiovascular complications in diabetes are particularly serious and represent the primary cause of morbidity and mortality in diabetic patients. Despite early observations of cardiac dysfunction in diabetic humans, cardiomyopathy unique to diabetes has only recently been recognized. RECENT ADVANCES Research has focused on understanding the pathogenic mechanisms underlying the initiation and development of diabetic cardiomyopathy. Emerging data highlight the importance of altered mitochondrial function as a major contributor to cardiac dysfunction in diabetes. Mitochondrial dysfunction occurs by several mechanisms involving altered cardiac substrate metabolism, lipotoxicity, impaired cardiac insulin and glucose homeostasis, impaired cellular and mitochondrial calcium handling, oxidative stress, and mitochondrial uncoupling. CRITICAL ISSUES Currently, treatment is not specifically tailored for diabetic patients with cardiac dysfunction. Given the multifactorial development and progression of diabetic cardiomyopathy, traditional treatments such as anti-diabetic agents, as well as cellular and mitochondrial fatty acid uptake inhibitors aimed at shifting the balance of cardiac metabolism from utilizing fat to glucose may not adequately target all aspects of this condition. Thus, an alternative treatment such as resveratrol, which targets multiple facets of diabetes, may represent a safe and promising supplement to currently recommended clinical therapy and lifestyle changes. FUTURE DIRECTIONS Elucidation of the mechanisms underlying the initiation and progression of diabetic cardiomyopathy is essential for development of effective and targeted treatment strategies. Of particular interest is the investigation of alternative therapies such as resveratrol, which can function as both preventative and mitigating agents in the management of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Miranda M Sung
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
| | - Shereen M Hamza
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
| | - Jason R B Dyck
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
| |
Collapse
|
106
|
Roul D, Recchia FA. Metabolic alterations induce oxidative stress in diabetic and failing hearts: different pathways, same outcome. Antioxid Redox Signal 2015; 22:1502-14. [PMID: 25836025 PMCID: PMC4449624 DOI: 10.1089/ars.2015.6311] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
SIGNIFICANCE Several authors have proposed a link between altered cardiac energy substrate metabolism and reactive oxygen species (ROS) generation. A cogent evidence of this association has been found in diabetic cardiomyopathy (dCM); however, experimental findings in animal models of heart failure (HF) and in human myocardium also seem to support the coexistence of the two alterations in HF. CRITICAL ISSUES Two important questions remain open: whether pathological changes in metabolism play an important role in enhancing oxidative stress and whether there is a common pathway linking altered substrate utilization and activation of ROS-generating enzymes, independently of the underlying cardiac pathology. In this regard, the comparison between dCM and HF is intriguing, in that these pathological conditions display very different cardiac metabolic phenotypes. RECENT ADVANCES Our literature review on this topic indicates that a vast body of knowledge is now available documenting the relationship between the metabolism of energy substrates and ROS generation in dCM. In some cases, biochemical mechanisms have been identified. On the other hand, only a few and relatively recent studies have explored this phenomenon in HF and their conclusions are not consistent. FUTURE DIRECTIONS Better methods of investigation, especially in vivo, will be necessary to test whether the metabolic fate of certain substrates is causally linked to ROS production. If successful, these studies will place a new emphasis on the potential clinical relevance of metabolic modulators, which might indirectly mitigate cardiac oxidative stress in dCM, HF, and, possibly, in other pathological conditions.
Collapse
Affiliation(s)
- David Roul
- 1Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Fabio A Recchia
- 1Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania.,2Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| |
Collapse
|
107
|
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
|
108
|
Hellfritsch J, Kirsch J, Schneider M, Fluege T, Wortmann M, Frijhoff J, Dagnell M, Fey T, Esposito I, Kölle P, Pogoda K, Angeli JPF, Ingold I, Kuhlencordt P, Östman A, Pohl U, Conrad M, Beck H. Knockout of mitochondrial thioredoxin reductase stabilizes prolyl hydroxylase 2 and inhibits tumor growth and tumor-derived angiogenesis. Antioxid Redox Signal 2015; 22:938-50. [PMID: 25647640 PMCID: PMC4376289 DOI: 10.1089/ars.2014.5889] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AIMS Mitochondrial thioredoxin reductase (Txnrd2) is a central player in the control of mitochondrial hydrogen peroxide (H2O2) abundance by serving as a direct electron donor to the thioredoxin-peroxiredoxin axis. In this study, we investigated the impact of targeted disruption of Txnrd2 on tumor growth. RESULTS Tumor cells with a Txnrd2 deficiency failed to activate hypoxia-inducible factor-1α (Hif-1α) signaling; it rather caused PHD2 accumulation, Hif-1α degradation and decreased vascular endothelial growth factor (VEGF) levels, ultimately leading to reduced tumor growth and tumor vascularization. Increased c-Jun NH2-terminal Kinase (JNK) activation proved to be the molecular link between the loss of Txnrd2, an altered mitochondrial redox balance with compensatory upregulation of glutaredoxin-2, and elevated PHD2 expression. INNOVATION Our data provide compelling evidence for a yet-unrecognized mitochondrial Txnrd-driven, regulatory mechanism that ultimately prevents cellular Hif-1α accumulation. In addition, simultaneous targeting of both the mitochondrial thioredoxin and glutathione systems was used as an efficient therapeutic approach in hindering tumor growth. CONCLUSION This work demonstrates an unexpected regulatory link between mitochondrial Txnrd and the JNK-PHD2-Hif-1α axis, which highlights how the loss of Txnrd2 and the resulting altered mitochondrial redox balance impairs tumor growth as well as tumor-related angiogenesis. Furthermore, it opens a new avenue for a therapeutic approach to hinder tumor growth by the simultaneous targeting of both the mitochondrial thioredoxin and glutathione systems.
Collapse
Affiliation(s)
- Juliane Hellfritsch
- 1 Walter Brendel Centre of Experimental Medicine, Munich Heart Alliance, Ludwig-Maximilians-University , Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Thiopurines induce oxidative stress in T-lymphocytes: a proteomic approach. Mediators Inflamm 2015; 2015:434825. [PMID: 25873760 PMCID: PMC4385670 DOI: 10.1155/2015/434825] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/06/2014] [Accepted: 09/09/2014] [Indexed: 01/24/2023] Open
Abstract
Thiopurines are extensively used immunosuppressants for the treatment of inflammatory bowel disease (IBD). The polymorphism of thiopurine S-methyltransferase (TPMT) influences thiopurine metabolism and therapy outcome. We used a TPMT knockdown (kd) model of human Jurkat T-lymphocytes cells to study the effects of treatment with 6-mercaptopurine (6-MP) and 6-thioguanine (6-TG) on proteome and phosphoproteome. We identified thirteen proteins with altered expression and nine proteins with altered phosphorylation signals. Three proteins (THIO, TXD17, and GSTM3) with putative functions in cellular oxidative stress responses were altered by 6-TG treatment and another protein PRDX3 was differentially phosphorylated in TPMT kd cells. Furthermore, reactive oxygen species (ROS) assay results were consistent with a significant induction of oxidative stress by both TPMT knockdown and thiopurine treatments. Immunoblot analyses showed treatment altered expression of key antioxidant enzymes (i.e., SOD2 and catalase) in both wt and kd groups, while SOD1 was downregulated by 6-TG treatment and TPMT knockdown. Collectively, increased oxidative stress might be a mechanism involved in thiopurine induced cytotoxicity and adverse effects (i.e., hepatotoxicity) and an antioxidant cotherapy might help to combat this. Results highlight the significance of oxidative stress in thiopurines' actions and could have important implications for the treatment of IBD patients.
Collapse
|
110
|
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
|
111
|
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: 141] [Impact Index Per Article: 14.1] [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
|
112
|
Korge P, Calmettes G, Weiss JN. Increased reactive oxygen species production during reductive stress: The roles of mitochondrial glutathione and thioredoxin reductases. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:514-25. [PMID: 25701705 DOI: 10.1016/j.bbabio.2015.02.012] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 02/09/2015] [Accepted: 02/12/2015] [Indexed: 01/22/2023]
Abstract
Both extremes of redox balance are known to cause cardiac injury, with mounting evidence revealing that the injury induced by both oxidative and reductive stress is oxidative in nature. During reductive stress, when electron acceptors are expected to be mostly reduced, some redox proteins can donate electrons to O2 instead, which increases reactive oxygen species (ROS) production. However, the high level of reducing equivalents also concomitantly enhances ROS scavenging systems involving redox couples such as NADPH/NADP+ and GSH/GSSG. Here our objective was to explore how reductive stress paradoxically increases net mitochondrial ROS production despite the concomitant enhancement of ROS scavenging systems. Using recombinant enzymes and isolated permeabilized cardiac mitochondria, we show that two normally antioxidant matrix NADPH reductases, glutathione reductase and thioredoxin reductase, generate H2O2 by leaking electrons from their reduced flavoprotein to O2 when electron flow is impaired by inhibitors or because of limited availability of their natural electron acceptors, GSSG and oxidized thioredoxin. The spillover of H2O2 under these conditions depends on H2O2 reduction by peroxiredoxin activity, which may regulate redox signaling in response to endogenous or exogenous factors. These findings may explain how ROS production during reductive stress overwhelms ROS scavenging capability, generating the net mitochondrial ROS spillover causing oxidative injury. These enzymes could potentially be targeted to increase cancer cell death or modulate H2O2-induced redox signaling to protect the heart against ischemia/reperfusion damage.
Collapse
Affiliation(s)
- Paavo Korge
- UCLA Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Department of Medicine (Cardiology), David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - Guillaume Calmettes
- UCLA Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Department of Medicine (Cardiology), David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - James N Weiss
- UCLA Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Department of Medicine (Cardiology), David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States.
| |
Collapse
|
113
|
Hoffman NE, Miller BA, Wang J, Elrod JW, Rajan S, Gao E, Song J, Zhang XQ, Hirschler-Laszkiewicz I, Shanmughapriya S, Koch WJ, Feldman AM, Madesh M, Cheung JY. Ca²⁺ entry via Trpm2 is essential for cardiac myocyte bioenergetics maintenance. Am J Physiol Heart Circ Physiol 2015; 308:H637-50. [PMID: 25576627 DOI: 10.1152/ajpheart.00720.2014] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ubiquitously expressed Trpm2 channel limits oxidative stress and preserves mitochondrial function. We first demonstrated that intracellular Ca(2+) concentration increase after Trpm2 activation was due to direct Ca(2+) influx and not indirectly via reverse Na(+)/Ca(2+) exchange. To elucidate whether Ca(2+) entry via Trpm2 is required to maintain cellular bioenergetics, we injected adenovirus expressing green fluorescent protein (GFP), wild-type (WT) Trpm2, and loss-of-function (E960D) Trpm2 mutant into left ventricles of global Trpm2 knockout (gKO) or WT hearts. Five days post-injection, gKO-GFP heart slices had higher reactive oxygen species (ROS) levels but lower oxygen consumption rate (OCR) than WT-GFP heart slices. Trpm2 but not E960D decreased ROS and restored OCR in gKO hearts back to normal levels. In gKO myocytes expressing Trpm2 or its mutants, Trpm2 but not E960D reduced the elevated mitochondrial superoxide (O2(.-)) levels in gKO myocytes. After hypoxia-reoxygenation (H/R), Trpm2 but not E906D or P1018L (inactivates Trpm2 current) lowered O2(.-) levels in gKO myocytes and only in the presence of extracellular Ca(2+), indicating sustained Ca(2+) entry is necessary for Trpm2-mediated preservation of mitochondrial function. After ischemic-reperfusion (I/R), cardiac-specific Trpm2 KO hearts exhibited lower maximal first time derivative of LV pressure rise (+dP/dt) than WT hearts in vivo. After doxorubicin treatment, Trpm2 KO mice had worse survival and lower +dP/dt. We conclude 1) cardiac Trpm2-mediated Ca(2+) influx is necessary to maintain mitochondrial function and protect against H/R injury; 2) Ca(2+) influx via cardiac Trpm2 confers protection against H/R and I/R injury by reducing mitochondrial oxidants; and 3) Trpm2 confers protection in doxorubicin cardiomyopathy.
Collapse
Affiliation(s)
- Nicholas E Hoffman
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; Department of Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Barbara A Miller
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - JuFang Wang
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - John W Elrod
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Sudasan Rajan
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; Department of Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Erhe Gao
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Jianliang Song
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Xue-Qian Zhang
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | | | - Santhanam Shanmughapriya
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; Department of Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Walter J Koch
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Arthur M Feldman
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Muniswamy Madesh
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; Department of Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Joseph Y Cheung
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania; Division of Nephrology, Temple University School of Medicine, Philadelphia, Pennsylvania;
| |
Collapse
|
114
|
Selenium-Functionalized Molecules (SeFMs) as Potential Drugs and Nutritional Supplements. TOPICS IN MEDICINAL CHEMISTRY 2015. [DOI: 10.1007/7355_2015_87] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
115
|
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
|
116
|
Goichberg P, Chang J, Liao R, Leri A. Cardiac stem cells: biology and clinical applications. Antioxid Redox Signal 2014; 21:2002-17. [PMID: 24597850 PMCID: PMC4208604 DOI: 10.1089/ars.2014.5875] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Heart disease is the primary cause of death in the industrialized world. Cardiac failure is dictated by an uncompensated reduction in the number of viable and fully functional cardiomyocytes. While current pharmacological therapies alleviate the symptoms associated with cardiac deterioration, heart transplantation remains the only therapy for advanced heart failure. Therefore, there is a pressing need for novel therapeutic modalities. Cell-based therapies involving cardiac stem cells (CSCs) constitute a promising emerging approach for the replenishment of the lost tissue and the restoration of cardiac contractility. RECENT ADVANCES CSCs reside in the adult heart and govern myocardial homeostasis and repair after injury by producing new cardiomyocytes and vascular structures. In the last decade, different classes of immature cells expressing distinct stem cell markers have been identified and characterized in terms of their growth properties, differentiation potential, and regenerative ability. Phase I clinical trials, employing autologous CSCs in patients with ischemic cardiomyopathy, are being completed with encouraging results. CRITICAL ISSUES Accumulating evidence concerning the role of CSCs in heart regeneration imposes a reconsideration of the mechanisms of cardiac aging and the etiology of heart failure. Deciphering the molecular pathways that prevent activation of CSCs in their environment and understanding the processes that affect CSC survival and regenerative function with cardiac pathologies, commonly accompanied by alterations in redox conditions, are of great clinical importance. FUTURE DIRECTIONS Further investigations of CSC biology may be translated into highly effective and novel therapeutic strategies aiming at the enhancement of the endogenous healing capacity of the diseased heart.
Collapse
Affiliation(s)
- Polina Goichberg
- Departments of Anesthesia and Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | | | | | | |
Collapse
|
117
|
Ziolo MT, Houser SR. Abnormal Ca(2+) cycling in failing ventricular myocytes: role of NOS1-mediated nitroso-redox balance. Antioxid Redox Signal 2014; 21:2044-59. [PMID: 24801117 PMCID: PMC4208612 DOI: 10.1089/ars.2014.5873] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
SIGNIFICANCE Heart failure (HF) results from poor heart function and is the leading cause of death in Western society. Abnormalities of Ca(2+) handling at the level of the ventricular myocyte are largely responsible for much of the poor heart function. RECENT ADVANCES Although studies have unraveled numerous mechanisms for the abnormal Ca(2+) handling, investigations over the past decade have indicated that much of the contractile dysfunction and adverse remodeling that occurs in HF involves oxidative stress. CRITICAL ISSUES Regrettably, antioxidant therapy has been an immense disappointment in clinical trials. Thus, redox signaling is being reassessed to elucidate why antioxidants failed to treat HF. FUTURE DIRECTIONS A recently identified aspect of redox signaling (specifically the superoxide anion radical) is its interaction with nitric oxide, known as the nitroso-redox balance. There is a large nitroso-redox imbalance with HF, and we suggest that correcting this imbalance may be able to restore myocyte contraction and improve heart function.
Collapse
Affiliation(s)
- Mark T Ziolo
- 1 Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University , Columbus, Ohio
| | | |
Collapse
|
118
|
Cho GS, Fernandez L, Kwon C. Regenerative medicine for the heart: perspectives on stem-cell therapy. Antioxid Redox Signal 2014; 21:2018-31. [PMID: 25133793 PMCID: PMC4208610 DOI: 10.1089/ars.2014.6063] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Despite decades of progress in cardiovascular biology and medicine, heart disease remains the leading cause of death, and there is no cure for the failing heart. Since heart failure is mostly caused by loss or dysfunction of cardiomyocytes (CMs), replacing dead or damaged CMs with new CMs might be an ideal way to reverse the disease. However, the adult heart is composed mainly of terminally differentiated CMs that have no significant self-regeneration capacity. RECENT ADVANCES Stem cells have tremendous regenerative potential and, thus, current cardiac regenerative research has focused on developing stem cell sources to repair damaged myocardium. CRITICAL ISSUES In this review, we examine the potential sources of cells that could be used for heart therapies, including embryonic stem cells and induced pluripotent stem cells, as well as alternative methods for activating the endogenous regenerative mechanisms of the heart via transdifferentiation and cell reprogramming. We also discuss the current state of knowledge of cell purification, delivery, and retention. FUTURE DIRECTIONS Efforts are underway to improve the current stem cell strategies and methodologies, which will accelerate the development of innovative stem-cell therapies for heart regeneration.
Collapse
Affiliation(s)
- Gun-Sik Cho
- Division of Cardiology, Department of Medicine, Institute for Cell Engineering, Johns Hopkins University , Baltimore, Maryland
| | | | | |
Collapse
|
119
|
Salloum FN, Sturz GR, Yin C, Rehman S, Hoke NN, Kukreja RC, Xi L. Beetroot juice reduces infarct size and improves cardiac function following ischemia-reperfusion injury: Possible involvement of endogenous H2S. Exp Biol Med (Maywood) 2014; 240:669-81. [PMID: 25361774 DOI: 10.1177/1535370214558024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/22/2014] [Indexed: 11/16/2022] Open
Abstract
Ingestion of high dietary nitrate in the form of beetroot juice (BRJ) has been shown to exert antihypertensive effects in humans through increasing cyclic guanosine monophosphate (cGMP) levels. Since enhanced cGMP protects against myocardial ischemia-reperfusion (I/R) injury through upregulation of hydrogen sulfide (H2S), we tested the hypothesis that BRJ protects against I/R injury via H2S. Adult male CD-1 mice received either regular drinking water or those dissolved with BRJ powder (10 g/L, containing ∼ 0.7 mM nitrate). Seven days later, the hearts were explanted for molecular analyses. Subsets of mice were subjected to I/R injury by occlusion of the left coronary artery for 30 min and reperfusion for 24 h. A specific inhibitor of H2S producing enzyme--cystathionine-γ-lyase (CSE), DL-propargylglycine (PAG, 50 mg/kg) was given i.p. 30 min before ischemia. Myocardial infarct size was significantly reduced in BRJ-fed mice (15.8 ± 3.2%) versus controls (46.5 ± 3.5%, mean ± standard error [SE], n = 6/group, P < .05). PAG completely blocked the infarct-limiting effect of BRJ. Moreover, BRJ significantly preserved ventricular function following I/R. Myocardial levels of H2S and its putative protein target--vascular endothelial growth factor receptor 2 (VEGFR2) were significantly increased by BRJ intake, whereas CSE mRNA and protein content did not change. Interestingly, the BRJ-induced cardioprotection was not associated with elevated blood nitrate-nitrite levels following I/R nor induction of cardiac peroxiredoxin 5, a mitochondrial antioxidant enzyme previously linked to nitrate-induced cardioprotection. We conclude that BRJ ingestion protects against post-I/R myocardial infarction and ventricular dysfunction possibly through CSE-mediated endogenous H2S generation. BRJ could be a promising natural and inexpensive nutraceutical supplement to reduce cardiac I/R injury in patients.
Collapse
Affiliation(s)
- Fadi N Salloum
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Gregory R Sturz
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Chang Yin
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Shabina Rehman
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Nicholas N Hoke
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Rakesh C Kukreja
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Lei Xi
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
120
|
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
|
121
|
Alleman RJ, Katunga LA, Nelson MAM, Brown DA, Anderson EJ. The "Goldilocks Zone" from a redox perspective-Adaptive vs. deleterious responses to oxidative stress in striated muscle. Front Physiol 2014; 5:358. [PMID: 25278906 PMCID: PMC4166897 DOI: 10.3389/fphys.2014.00358] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/02/2014] [Indexed: 01/17/2023] Open
Abstract
Consequences of oxidative stress may be beneficial or detrimental in physiological systems. An organ system's position on the “hormetic curve” is governed by the source and temporality of reactive oxygen species (ROS) production, proximity of ROS to moieties most susceptible to damage, and the capacity of the endogenous cellular ROS scavenging mechanisms. Most importantly, the resilience of the tissue (the capacity to recover from damage) is a decisive factor, and this is reflected in the disparate response to ROS in cardiac and skeletal muscle. In myocytes, a high oxidative capacity invariably results in a significant ROS burden which in homeostasis, is rapidly neutralized by the robust antioxidant network. The up-regulation of key pathways in the antioxidant network is a central component of the hormetic response to ROS. Despite such adaptations, persistent oxidative stress over an extended time-frame (e.g., months to years) inevitably leads to cumulative damages, maladaptation and ultimately the pathogenesis of chronic diseases. Indeed, persistent oxidative stress in heart and skeletal muscle has been repeatedly demonstrated to have causal roles in the etiology of heart disease and insulin resistance, respectively. Deciphering the mechanisms that underlie the divergence between adaptive and maladaptive responses to oxidative stress remains an active area of research for basic scientists and clinicians alike, as this would undoubtedly lead to novel therapeutic approaches. Here, we provide an overview of major types of ROS in striated muscle and the divergent adaptations that occur in response to them. Emphasis is placed on highlighting newly uncovered areas of research on this topic, with particular focus on the mitochondria, and the diverging roles that ROS play in muscle health (e.g., exercise or preconditioning) and disease (e.g., cardiomyopathy, ischemia, metabolic syndrome).
Collapse
Affiliation(s)
- Rick J Alleman
- Departments of Physiology, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA
| | - Lalage A Katunga
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Margaret A M Nelson
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - David A Brown
- Departments of Physiology, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA
| | - Ethan J Anderson
- East Carolina Diabetes and Obesity Institute, East Carolina University Greenville, NC, USA ; Pharmacology and Toxicology, Brody School of Medicine, East Carolina University Greenville, NC, USA
| |
Collapse
|
122
|
Vakrou S, Abraham MR. Hypertrophic cardiomyopathy: a heart in need of an energy bar? Front Physiol 2014; 5:309. [PMID: 25191275 PMCID: PMC4137386 DOI: 10.3389/fphys.2014.00309] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/30/2014] [Indexed: 01/08/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) has been recently recognized as the most common inherited cardiovascular disorder, affecting 1 in 500 adults worldwide. HCM is characterized by myocyte hypertrophy resulting in thickening of the ventricular wall, myocyte disarray, interstitial and/or replacement fibrosis, decreased ventricular cavity volume and diastolic dysfunction. HCM is also the most common cause of sudden death in the young. A large proportion of patients diagnosed with HCM have mutations in sarcomeric proteins. However, it is unclear how these mutations lead to the cardiac phenotype, which is variable even in patients carrying the same causal mutation. Abnormalities in calcium cycling, oxidative stress, mitochondrial dysfunction and energetic deficiency have been described constituting the basis of therapies in experimental models of HCM and HCM patients. This review focuses on evidence supporting the role of cellular metabolism and mitochondria in HCM.
Collapse
Affiliation(s)
- Styliani Vakrou
- Division of Cardiology, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| | - M Roselle Abraham
- Division of Cardiology, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| |
Collapse
|
123
|
Kaludercic N, Deshwal S, Di Lisa F. Reactive oxygen species and redox compartmentalization. Front Physiol 2014; 5:285. [PMID: 25161621 PMCID: PMC4130307 DOI: 10.3389/fphys.2014.00285] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/11/2014] [Indexed: 01/01/2023] Open
Abstract
Reactive oxygen species (ROS) formation and signaling are of major importance and regulate a number of processes in physiological conditions. A disruption in redox status regulation, however, has been associated with numerous pathological conditions. In recent years it has become increasingly clear that oxidative and reductive modifications are confined in a spatio-temporal manner. This makes ROS signaling similar to that of Ca(2+) or other second messengers. Some subcellular compartments are more oxidizing (such as lysosomes or peroxisomes) whereas others are more reducing (mitochondria, nuclei). Moreover, although more reducing, mitochondria are especially susceptible to oxidation, most likely due to the high number of exposed thiols present in that compartment. Recent advances in the development of redox probes allow specific measurement of defined ROS in different cellular compartments in intact living cells or organisms. The availability of these tools now allows simultaneous spatio-temporal measurements and correlation between ROS generation and organelle and/or cellular function. The study of ROS compartmentalization and microdomains will help elucidate their role in physiology and disease. Here we will examine redox probes currently available and how ROS generation may vary between subcellular compartments. Furthermore, we will discuss ROS compartmentalization in physiological and pathological conditions focusing our attention on mitochondria, since their vulnerability to oxidative stress is likely at the basis of several diseases.
Collapse
Affiliation(s)
- Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR) Padova, Italy
| | - Soni Deshwal
- Department of Biomedical Sciences, University of Padova Padova, Italy
| | - Fabio Di Lisa
- Neuroscience Institute, National Research Council of Italy (CNR) Padova, Italy ; Department of Biomedical Sciences, University of Padova Padova, Italy
| |
Collapse
|
124
|
Zhang H, Du Y, Zhang X, Lu J, Holmgren A. Glutaredoxin 2 reduces both thioredoxin 2 and thioredoxin 1 and protects cells from apoptosis induced by auranofin and 4-hydroxynonenal. Antioxid Redox Signal 2014; 21:669-81. [PMID: 24295294 PMCID: PMC4098818 DOI: 10.1089/ars.2013.5499] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
AIMS Mitochondrial thioredoxin (Trx) is critical for defense against oxidative stress-induced cell apoptosis. To date, mitochondrial thioredoxin reductase (TrxR) is the only known enzyme catalyzing Trx2 reduction in mitochondria. However, TrxR is sensitive to inactivation by exo/endogenous electrophiles, for example, 4-hydroxynonenal (HNE). In this study, we characterized the mitochondrial glutaredoxin 2 (Grx2) system as a backup for the mitochondrial TrxR. Meanwhile, as Grx2 is also present in the cytosol/nucleus of certain cancer cell lines, the reducing activity of Grx2 on Trx1 was also tested. RESULTS Glutathione alone could reduce oxidized Trx2, and the presence of physiological concentrations of Grx2 markedly increased the reaction rate. HeLa cells with Grx2 overexpression (particularly in the mitochondria) exhibited higher viabilities than the wild-type cells after treatment with TrxR inhibitors (Auranofin or HNE), whereas knockdown of Grx2 sensitized the cells to TrxR inhibitors. Accordingly, Grx2 overexpression in the mitochondria had protected Trx2 from oxidation by HNE treatment, whereas Grx2 knockdown had sensitized Trx2 to oxidation. On the other hand, Grx2 reduced Trx1 with similar activities as that of Trx2. Overexpression of Grx2 in the cytosol had protected Trx1 from oxidation, indicating a supportive role of Grx2 in the cytosolic redox balance of cancer cells. INNOVATION This work explores the reductase activity of Grx2 on Trx2/1, and demonstrates the physiological importance of the activity by using in vivo redox western blot assays. CONCLUSION Grx2 system could help to keep Trx2/1 reduced during an oxidative stress, thereby contributing to the anti-apoptotic signaling.
Collapse
Affiliation(s)
- Huihui Zhang
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institute , Stockholm, Sweden
| | | | | | | | | |
Collapse
|
125
|
Sag CM, Santos CX, Shah AM. Redox regulation of cardiac hypertrophy. J Mol Cell Cardiol 2014; 73:103-11. [DOI: 10.1016/j.yjmcc.2014.02.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 01/31/2014] [Accepted: 02/03/2014] [Indexed: 02/07/2023]
|
126
|
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: 164] [Impact Index Per Article: 14.9] [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
|
127
|
Isakov E, Weisman-Shomer P, Benhar M. Suppression of the pro-inflammatory NLRP3/interleukin-1β pathway in macrophages by the thioredoxin reductase inhibitor auranofin. Biochim Biophys Acta Gen Subj 2014; 1840:3153-61. [PMID: 25065288 DOI: 10.1016/j.bbagen.2014.07.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 07/01/2014] [Accepted: 07/17/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND The thioredoxin/thioredoxin reductase system, which is best known for its essential role in antioxidant defense and redox homeostasis, is increasingly implicated in the regulation of multiple cellular signaling pathways. In the present study, we asked if the thioredoxin system in macrophages might regulate toll-like receptor 4 (TLR4)-dependent gene expression and consequent responses. METHODS Using microarray analysis we analyzed the effect of auranofin, a highly potent and specific inhibitor of thioredoxin reductase, on the transcriptional program activated in J774 macrophages by the TLR4 agonist, lipopolysaccharide (LPS). We used quantitative real-time PCR (qPCR), Western blotting, ELISA and cytotoxicity assays to confirm and extend the microarray results. RESULTS Global transcriptional profiling revealed that macrophage treatment with auranofin exerted a selective effect on LPS-induced gene expression, suppressing the induction of a small number of genes. Interestingly, among these suppressed genes were three members of the interleukin-1 (IL-1) family of genes, among which IL-1β was most affected. qPCR analyses confirmed the repressive effects of auranofin on IL-1 genes. In addition, qPCR and Western blot analyses showed that auranofin impaired TLR4-dependent induction of the inflammasome receptor NLRP3, which plays a critical role in IL-1β processing. Consistent with these findings, inflammasome-dependent release of IL-1β from stimulated macrophages was suppressed by auranofin as was inflammasome-mediated cell death. CONCLUSIONS Our findings suggest a regulatory role for the thioredoxin system in macrophage inflammatory signaling. Inhibition of the thioredoxin system in macrophages exerts an anti-inflammatory effect by repressing the activation of the NLRP3/IL-1β pathway.
Collapse
Affiliation(s)
- Elina Isakov
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Pnina Weisman-Shomer
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Moran Benhar
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
128
|
Chen Q, Szczepanek K, Hu Y, Thompson J, Lesnefsky EJ. A deficiency of apoptosis inducing factor (AIF) in Harlequin mouse heart mitochondria paradoxically reduces ROS generation during ischemia-reperfusion. Front Physiol 2014; 5:271. [PMID: 25101006 PMCID: PMC4106194 DOI: 10.3389/fphys.2014.00271] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/30/2014] [Indexed: 12/30/2022] Open
Abstract
Background and Aims: AIF (apoptosis inducing factor) is a flavin and NADH containing protein located within mitochondria required for optimal function of the respiratory chain. AIF may function as an antioxidant within mitochondria, yet when released from mitochondria it activates caspase-independent cell death. The Harlequin (Hq) mouse has a markedly reduced content of AIF, providing an experimental model to query if the main role of AIF in the exacerbation of cell death is enhanced mitochondrial generation of reactive oxygen species (ROS) or the activation of cell death programs. We asked if the ROS generation is altered in Hq heart mitochondria at baseline or following ischemia-reperfusion (IR). Methods: Buffer perfused mouse hearts underwent 30 min ischemia and 30 min reperfusion. Mitochondrial function including oxidative phosphorylation and H2O2 generation was measured. Immunoblotting was used to determine the contents of AIF and PAR [poly(ADP-ribose)] in cell fractions. Results: There were no differences in the release of H2O2 between wild type (WT) and Hq heart mitochondria at baseline. IR increased H2O2 generation from WT but not from Hq mitochondria compared to corresponding time controls. The complex I activity was decreased in WT but not in Hq mice following IR. The relocation of AIF from mitochondria to nucleus was increased in WT but not in Hq mice. IR activated PARP-1 only in WT mice. Cell injury was decreased in the Hq mouse heart following in vitro IR. Conclusion: A deficiency of AIF within mitochondria does not increase ROS production during IR, indicating that AIF functions less as an antioxidant within mitochondria. The decreased cardiac injury in Hq mouse heart accompanied by less AIF translocation to the nucleus suggests that AIF relocation, rather than the AIF content within mitochondria, contributes to cardiac injury during IR.
Collapse
Affiliation(s)
- Qun Chen
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University Richmond, VA, USA
| | - Karol Szczepanek
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University Richmond, VA, USA
| | - Ying Hu
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University Richmond, VA, USA
| | - Jeremy Thompson
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University Richmond, VA, USA
| | - Edward J Lesnefsky
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University Richmond, VA, USA ; Department of Biochemistry and Molecular Biology, Virginia Commonwealth University Richmond, VA, USA ; McGuire Department of Veterans Affairs Medical Center Richmond, VA, USA
| |
Collapse
|
129
|
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
|
130
|
Jan YH, Heck DE, Dragomir AC, Gardner CR, Laskin DL, Laskin JD. Acetaminophen reactive intermediates target hepatic thioredoxin reductase. Chem Res Toxicol 2014; 27:882-94. [PMID: 24661219 PMCID: PMC4033643 DOI: 10.1021/tx5000443] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Acetaminophen (APAP) is metabolized in the liver to N-acetyl-p-benzoquinone imine (NAPQI), an electrophilic metabolite known to bind liver proteins resulting in hepatotoxicity. Mammalian thioredoxin reductase (TrxR) is a cellular antioxidant containing selenocysteine (Sec) in its C-terminal redox center, a highly accessible target for electrophilic modification. In the present study, we determined if NAPQI targets TrxR. Hepatotoxicity induced by APAP treatment of mice (300 mg/kg, i.p.) was associated with a marked inhibition of both cytosolic TrxR1 and mitochondrial TrxR2 activity. Maximal inhibition was detected at 1 and 6 h post-APAP for TrxR1 and TrxR2, respectively. In purified rat liver TrxR1, enzyme inactivation was correlated with the metabolic activation of APAP by cytochrome P450, indicating that enzyme inhibition was due to APAP-reactive metabolites. NAPQI was also found to inhibit TrxR1. NADPH-reduced TrxR1 was significantly more sensitive to NAPQI (IC50 = 0.023 μM) than the oxidized enzyme (IC50 = 1.0 μM) or a human TrxR1 Sec498Cys mutant enzyme (IC50 = 17 μM), indicating that cysteine and selenocysteine residues in the redox motifs of TrxR are critical for enzyme inactivation. This is supported by our findings that alkylation of reduced TrxR with biotin-conjugated iodoacetamide, which selectively reacts with selenol or thiol groups on proteins, was inhibited by NAPQI. LC-MS/MS analysis confirmed that NAPQI modified cysteine 59, cysteine 497, and selenocysteine 498 residues in the redox centers of TrxR, resulting in enzyme inhibition. In addition to disulfide reduction, TrxR is also known to mediate chemical redox cycling. We found that menadione redox cycling by TrxR was markedly less sensitive to NAPQI than disulfide reduction, suggesting that TrxR mediates these reactions via distinct mechanisms. These data demonstrate that APAP-reactive metabolites target TrxR, suggesting an additional mechanism by which APAP induces oxidative stress and hepatotoxicity.
Collapse
Affiliation(s)
- Yi-Hua Jan
- Department of Environmental and Occupational Medicine, Rutgers University-Robert Wood Johnson Medical School , Piscataway, New Jersey 08854, United States
| | | | | | | | | | | |
Collapse
|
131
|
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: 108] [Impact Index Per Article: 9.8] [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
|
132
|
Role of 8-nitro-cGMP and its redox regulation in cardiovascular electrophilic signaling. J Mol Cell Cardiol 2014; 73:10-7. [PMID: 24530900 DOI: 10.1016/j.yjmcc.2014.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/03/2014] [Accepted: 02/05/2014] [Indexed: 12/11/2022]
Abstract
Structural and morphological changes of the cardiovascular systems (cardiovascular remodeling) are a major clinical outcome of cardiovascular diseases. Many lines of evidences have implied that transfiguration of reduction/oxidation (redox) homeostasis due to excess production of reactive oxygen species (ROS) and/or ROS-derived electrophilic metabolites (electrophiles) is the main cause of cardiovascular remodeling. Gasotransmitters, such as nitric oxide (NO) and endogenous electrophiles, are considered major bioactive species and have been extensively studied in the context of physiological and pathological cardiovascular events. We have recently found that hydrogen sulfide-related reactive species function as potent nucleophiles to eliminate electrophilic modification of signaling proteins induced by NO-derived electrophilic byproducts (e.g., 8-nitroguanosine 3',5'-cyclic monophosphate and nitro-oleic acid). In this review, we discuss the current understanding of redox control of cardiovascular pathophysiology by electrophiles and nucleophiles. We propose that modulation of electrophile-mediated post-translational modification of protein cysteine thiols may be a new therapeutic strategy of cardiovascular diseases. This article is part of a Special Issue entitled "Redox Signalling in the Cardiovascular System".
Collapse
|
133
|
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]
|
134
|
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
|
135
|
Li J, O W, Li W, Jiang ZG, Ghanbari HA. Oxidative stress and neurodegenerative disorders. Int J Mol Sci 2013; 14:24438-75. [PMID: 24351827 PMCID: PMC3876121 DOI: 10.3390/ijms141224438] [Citation(s) in RCA: 290] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/27/2013] [Accepted: 12/06/2013] [Indexed: 12/14/2022] Open
Abstract
Living cells continually generate reactive oxygen species (ROS) through the respiratory chain during energetic metabolism. ROS at low or moderate concentration can play important physiological roles. However, an excessive amount of ROS under oxidative stress would be extremely deleterious. The central nervous system (CNS) is particularly vulnerable to oxidative stress due to its high oxygen consumption, weakly antioxidative systems and the terminal-differentiation characteristic of neurons. Thus, oxidative stress elicits various neurodegenerative diseases. In addition, chemotherapy could result in severe side effects on the CNS and peripheral nervous system (PNS) of cancer patients, and a growing body of evidence demonstrates the involvement of ROS in drug-induced neurotoxicities as well. Therefore, development of antioxidants as neuroprotective drugs is a potentially beneficial strategy for clinical therapy. In this review, we summarize the source, balance maintenance and physiologic functions of ROS, oxidative stress and its toxic mechanisms underlying a number of neurodegenerative diseases, and the possible involvement of ROS in chemotherapy-induced toxicity to the CNS and PNS. We ultimately assess the value for antioxidants as neuroprotective drugs and provide our comments on the unmet needs.
Collapse
Affiliation(s)
- Jie Li
- Department of Geratology, First Hospital of Jilin University, Changchun, Jilin 130021, China; E-Mail:
| | - Wuliji O
- College of Pharmacology, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia 028000, China; E-Mail:
| | - Wei Li
- Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130021, China; E-Mail:
| | - Zhi-Gang Jiang
- Panacea Pharmaceuticals, Inc., Gaithersburg, MD 20877, USA; E-Mail:
| | | |
Collapse
|
136
|
Fisher-Wellman KH, Gilliam LAA, Lin CT, Cathey BL, Lark DS, Darrell Neufer P. Mitochondrial glutathione depletion reveals a novel role for the pyruvate dehydrogenase complex as a key H2O2-emitting source under conditions of nutrient overload. Free Radic Biol Med 2013; 65:1201-1208. [PMID: 24056031 PMCID: PMC3965186 DOI: 10.1016/j.freeradbiomed.2013.09.008] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 08/26/2013] [Accepted: 09/13/2013] [Indexed: 11/25/2022]
Abstract
Once regarded as a "by-product" of aerobic metabolism, the production of superoxide/H2O2 is now understood to be a highly specialized and extensively regulated process responsible for exerting control over a vast number of thiol-containing proteins, collectively referred to as the redox-sensitive proteome. Although disruptions within this process, secondary to elevated peroxide exposure, have been linked to disease, the sources and mechanisms regulating increased peroxide burden remain poorly defined and as such are difficult to target using pharmacotherapy. Here we identify the pyruvate dehydrogenase complex (PDC) as a key source of H2O2 within skeletal muscle mitochondria under conditions of depressed glutathione redox buffering integrity. Treatment of permeabilized myofibers with varying concentrations of the glutathione-depleting agent 1-chloro-2,4-dinitrobenzene led to a dose-dependent increase in pyruvate-supported JH2O2 emission (the flux of H2O2 diffusing out of the mitochondrial matrix into the surrounding assay medium), with emission rates eventually rising to exceed those of all substrate combinations tested. This striking sensitivity to glutathione depletion was observed in permeabilized fibers prepared from multiple species and was specific to PDC. Physiological oxidation of the cellular glutathione pool after high-fat feeding in rodents was found to elevate PDC JH2O2 emission, as well as increasing the sensitivity of the complex to GSH depletion. These findings reveal PDC as a potential major site of H2O2 production that is extremely sensitive to mitochondrial glutathione redox status.
Collapse
Affiliation(s)
- Kelsey H Fisher-Wellman
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27858, USA; Department of Physiology, East Carolina University, Greenville, NC 27858, USA.
| | - Laura A A Gilliam
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27858, USA; Department of Physiology, East Carolina University, Greenville, NC 27858, USA
| | - Chien-Te Lin
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27858, USA; Department of Physiology, East Carolina University, Greenville, NC 27858, USA
| | - Brook L Cathey
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27858, USA; Department of Physiology, East Carolina University, Greenville, NC 27858, USA
| | - Daniel S Lark
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27858, USA; Department of Kinesiology, East Carolina University, Greenville, NC 27858, USA
| | - P Darrell Neufer
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27858, USA; Department of Physiology, East Carolina University, Greenville, NC 27858, USA; Department of Kinesiology, East Carolina University, Greenville, NC 27858, USA
| |
Collapse
|
137
|
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
|
138
|
Dikalov SI, Nazarewicz RR. Angiotensin II-induced production of mitochondrial reactive oxygen species: potential mechanisms and relevance for cardiovascular disease. Antioxid Redox Signal 2013; 19:1085-94. [PMID: 22443458 PMCID: PMC3771548 DOI: 10.1089/ars.2012.4604] [Citation(s) in RCA: 244] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
SIGNIFICANCE The role of reactive oxygen species (ROS) in angiotensin II (AngII) induced endothelial dysfunction, cardiovascular and renal remodeling, inflammation, and fibrosis has been well documented. The molecular mechanisms of AngII pathophysiological activity involve the stimulation of NADPH oxidases, which produce superoxide and hydrogen peroxide. AngII also increases the production of mitochondrial ROS, while the inhibition of AngII improves mitochondrial function; however, the specific molecular mechanisms of the stimulation of mitochondrial ROS is not clear. RECENT ADVANCES Interestingly, the overexpression of mitochondrial thioredoxin 2 or mitochondrial superoxide dismutase attenuates AngII-induced hypertension, which demonstrates the importance of mitochondrial ROS in AngII-mediated cardiovascular diseases. CRITICAL ISSUES Although mitochondrial ROS plays an important role in normal physiological cell signaling, AngII, high glucose, high fat, or hypoxia may cause the overproduction of mitochondrial ROS, leading to the feed-forward redox stimulation of NADPH oxidases. This vicious cycle may contribute to the development of pathological conditions and facilitate organ damage in hypertension, atherosclerosis, and diabetes. FUTURE DIRECTIONS The development of antioxidant strategies specifically targeting mitochondria could be therapeutically beneficial in these disease conditions.
Collapse
Affiliation(s)
- Sergey I Dikalov
- Free Radicals in Medicine Core, Division of Clinical Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee
| | | |
Collapse
|
139
|
Kembro JM, Aon MA, Winslow RL, O'Rourke B, Cortassa S. Integrating mitochondrial energetics, redox and ROS metabolic networks: a two-compartment model. Biophys J 2013; 104:332-43. [PMID: 23442855 DOI: 10.1016/j.bpj.2012.11.3808] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Revised: 11/14/2012] [Accepted: 11/27/2012] [Indexed: 12/18/2022] Open
Abstract
To understand the mechanisms involved in the control and regulation of mitochondrial reactive oxygen species (ROS) levels, a two-compartment computational mitochondrial energetic-redox (ME-R) model accounting for energetic, redox, and ROS metabolisms is presented. The ME-R model incorporates four main redox couples (NADH/NAD(+), NADPH/NADP(+), GSH/GSSG, Trx(SH)(2)/TrxSS). Scavenging systems-glutathione, thioredoxin, superoxide dismutase, catalase-are distributed in mitochondrial matrix and extra-matrix compartments, and transport between compartments of ROS species (superoxide: O(2)(⋅-), hydrogen peroxide: H(2)O(2)), and GSH is also taken into account. Model simulations are compared with experimental data obtained from isolated heart mitochondria. The ME-R model is able to simulate: i), the shape and order of magnitude of H(2)O(2) emission and dose-response kinetics observed after treatment with inhibitors of the GSH or Trx scavenging systems and ii), steady and transient behavior of ΔΨ(m) and NADH after single or repetitive pulses of substrate- or uncoupler-elicited energetic-redox transitions. The dynamics of the redox environment in both compartments is analyzed with the model following substrate addition. The ME-R model represents a useful computational tool for exploring ROS dynamics, the role of compartmentation in the modulation of the redox environment, and how redox regulation participates in the control of mitochondrial function.
Collapse
Affiliation(s)
- Jackelyn M Kembro
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore Maryland, USA
| | | | | | | | | |
Collapse
|
140
|
Garlid AO, Jaburek M, Jacobs JP, Garlid KD. Mitochondrial reactive oxygen species: which ROS signals cardioprotection? Am J Physiol Heart Circ Physiol 2013; 305:H960-8. [PMID: 23913710 DOI: 10.1152/ajpheart.00858.2012] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mitochondria are the major effectors of cardioprotection by procedures that open the mitochondrial ATP-sensitive potassium channel (mitoKATP), including ischemic and pharmacological preconditioning. MitoKATP opening leads to increased reactive oxygen species (ROS), which then activate a mitoKATP-associated PKCε, which phosphorylates mitoKATP and leaves it in a persistent open state (Costa AD, Garlid KD. Am J Physiol Heart Circ Physiol 295, H874-H882, 2008). The ROS responsible for this effect is not known. The present study focuses on superoxide (O2(·-)), hydrogen peroxide (H2O2), and hydroxyl radical (HO(·)), each of which has been proposed as the signaling ROS. Feedback activation of mitoKATP provides an ideal setting for studying endogenous ROS signaling. Respiring rat heart mitochondria were preincubated with ATP and diazoxide, together with an agent being tested for interference with this process, either by scavenging ROS or by blocking ROS transformations. The mitochondria were then assayed to determine whether or not the persistent phosphorylated open state was achieved. Dimethylsulfoxide (DMSO), dimethylformamide (DMF), deferoxamine, Trolox, and bromoenol lactone each interfered with formation of the ROS-dependent open state. Catalase did not interfere with this step. We also found that DMF blocked cardioprotection by both ischemic preconditioning and diazoxide. The lack of a catalase effect and the inhibitory effects of agents acting downstream of HO(·) excludes H2O2 as the endogenous signaling ROS. Taken together, the results support the conclusion that the ROS message is carried by a downstream product of HO(·) and that it is probably a product of phospholipid oxidation.
Collapse
Affiliation(s)
- Anders O Garlid
- Department of Biology, Portland State University, Portland, Oregon; and
| | | | | | | |
Collapse
|
141
|
Stromal response to prostate cancer: nanotechnology-based detection of thioredoxin-interacting protein partners distinguishes prostate cancer associated stroma from that of benign prostatic hyperplasia. PLoS One 2013; 8:e60562. [PMID: 23762225 PMCID: PMC3675098 DOI: 10.1371/journal.pone.0060562] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 05/14/2013] [Indexed: 11/28/2022] Open
Abstract
Histological staining of reactive stroma has been shown to be a predictor of biochemical recurrence in prostate cancer, however, molecular markers of the stromal response to prostate cancer have not yet been fully delineated. The objective of this study was to determine whether or not the stromal biomarkers detected with a thioredoxin-targeted nanodevice could be used to distinguish the stroma associated with benign prostatic hyperplasia from that associated with PCA. In this regard, we recently demonstrated that a thioredoxin-targeted nanodevice selectively binds to reactive stroma in frozen prostate tumor tissue sections. To accomplish this, random frozen prostate tissue sections from each of 35 patients who underwent resection were incubated with the nanodevice and graded for fluorescent intensity. An adjacent section from each case was stained with Hematoxylin & Eosin to confirm the diagnosis. Select cases were stained with Masson's Trichrome or immunohistochemically using antibodies to thioredoxin reductase 1, thioredoxin reductase 2 or peroxiredoxin 1. Our results demonstrate that the graded intensity of nanodevice binding to the stroma associated with PCA was significantly higher (p = 0.0127) than that of benign prostatic hyperplasia using the t-test. Immunohistochemical staining of adjacent sections in representative cases showed that none of the two commonly studied thioredoxin interacting protein partners mirrored the fluorescence pattern seen with the nanodevice. However, thioredoxin reductase 2 protein was clearly shown to be a biomarker of prostate cancer-associated reactive stroma whose presence distinguishes the stroma associated with benign prostatic hyperplasia from that associated with prostate cancer. We conclude that the signal detected by the nanodevice, in contrast to individual targets detected with antibodies used in this study, originates from multiple thioredoxin interacting protein partners that distinguish the M2 neutrophil and macrophage associated inflammatory response in prostate cancer-associated stroma from the CD4+ T-Lymphocyte linked inflammation in benign prostatic hyperplasia.
Collapse
|
142
|
Dröse S. Differential effects of complex II on mitochondrial ROS production and their relation to cardioprotective pre- and postconditioning. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2013; 1827:578-87. [DOI: 10.1016/j.bbabio.2013.01.004] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 01/04/2013] [Accepted: 01/09/2013] [Indexed: 11/30/2022]
|
143
|
Fisher-Wellman KH, Mattox TA, Thayne K, Katunga LA, La Favor JD, Neufer PD, Hickner RC, Wingard CJ, Anderson EJ. Novel role for thioredoxin reductase-2 in mitochondrial redox adaptations to obesogenic diet and exercise in heart and skeletal muscle. J Physiol 2013; 591:3471-86. [PMID: 23613536 DOI: 10.1113/jphysiol.2013.254193] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Increased fatty acid availability and oxidative stress are physiological consequences of exercise (Ex) and a high-fat, high-sugar (HFHS) diet. Despite these similarities, the global effects of Ex are beneficial, whereas HFHS diets are largely deleterious to the cardiovascular system. The reasons for this disparity are multifactorial and incompletely understood. We hypothesized that differences in redox adaptations following HFHS diet in comparison to exercise may underlie this disparity, particularly in mitochondria. Our objective in this study was to determine mechanisms by which heart and skeletal muscle (red gastrocnemius, RG) mitochondria experience differential redox adaptations to 12 weeks of HFHS diet and/or exercise training (Ex) in rats. Surprisingly, both HFHS feeding and Ex led to contrasting effects in heart and RG, in that mitochondrial H2O2 decreased in heart but increased in RG following both HFHS diet and Ex, in comparison to sedentary animals fed a control diet. These differences were determined to be due largely to increased antioxidant/anti-inflammatory enzymes in the heart following the HFHS diet, which did not occur in RG. Specifically, upregulation of mitochondrial thioredoxin reductase-2 occurred with both HFHS and Ex in the heart, but only with Ex in RG, and systematic evaluation of this enzyme revealed that it is critical for suppressing mitochondrial H2O2 during fatty acid oxidation. These findings are novel and important in that they illustrate the unique ability of the heart to adapt to oxidative stress imposed by HFHS diet, in part through upregulation of thioredoxin reductase-2. Furthermore, upregulation of thioredoxin reductase-2 plays a critical role in preserving the mitochondrial redox status in the heart and skeletal muscle with exercise.
Collapse
|
144
|
Bleier L, Dröse S. Superoxide generation by complex III: from mechanistic rationales to functional consequences. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2012; 1827:1320-31. [PMID: 23269318 DOI: 10.1016/j.bbabio.2012.12.002] [Citation(s) in RCA: 241] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 12/05/2012] [Accepted: 12/12/2012] [Indexed: 01/21/2023]
Abstract
Apart from complex I (NADH:ubiquinone oxidoreductase) the mitochondrial cytochrome bc1 complex (complex III; ubiquinol:cytochrome c oxidoreductase) has been identified as the main producer of superoxide and derived reactive oxygen species (ROS) within the mitochondrial respiratory chain. Mitochondrial ROS are generally linked to oxidative stress, aging and other pathophysiological settings like in neurodegenerative diseases. However, ROS produced at the ubiquinol oxidation center (center P, Qo site) of complex III seem to have additional physiological functions as signaling molecules during cellular processes like the adaptation to hypoxia. The molecular mechanism of superoxide production that is mechanistically linked to the electron bifurcation during ubiquinol oxidation is still a matter of debate. Some insight comes from extensive kinetic studies with mutated complexes from yeast and bacterial cytochrome bc1 complexes. This review is intended to bridge the gap between those mechanistic studies and investigations on complex III ROS in cellular signal transduction and highlights factors that impact superoxide generation. This article is part of a Special Issue entitled: Respiratory complex III and related bc complexes.
Collapse
Affiliation(s)
- Lea Bleier
- Molecular Bioenergetics Group, Medical School, Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| | | |
Collapse
|
145
|
Tocchetti CG, Caceres V, Stanley BA, Xie C, Shi S, Watson WH, O’Rourke B, Spadari-Bratfisch RC, Cortassa S, Akar FG, Paolocci N, Aon MA. GSH or palmitate preserves mitochondrial energetic/redox balance, preventing mechanical dysfunction in metabolically challenged myocytes/hearts from type 2 diabetic mice. Diabetes 2012; 61:3094-105. [PMID: 22807033 PMCID: PMC3501888 DOI: 10.2337/db12-0072] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In type 2 diabetes, hyperglycemia and increased sympathetic drive may alter mitochondria energetic/redox properties, decreasing the organelle's functionality. These perturbations may prompt or sustain basal low-cardiac performance and limited exercise capacity. Yet the precise steps involved in this mitochondrial failure remain elusive. Here, we have identified dysfunctional mitochondrial respiration with substrates of complex I, II, and IV and lowered thioredoxin-2/glutathione (GSH) pools as the main processes accounting for impaired state 4→3 energetic transition shown by mitochondria from hearts of type 2 diabetic db/db mice upon challenge with high glucose (HG) and the β-agonist isoproterenol (ISO). By mimicking clinically relevant conditions in type 2 diabetic patients, this regimen triggers a major overflow of reactive oxygen species (ROS) from mitochondria that directly perturbs cardiac electro-contraction coupling, ultimately leading to heart dysfunction. Exogenous GSH or, even more so, the fatty acid palmitate rescues basal and β-stimulated function in db/db myocyte/heart preparations exposed to HG/ISO. This occurs because both interventions provide the reducing equivalents necessary to counter mitochondrial ROS outburst and energetic failure. Thus, in the presence of poor glycemic control, the diabetic patient's inability to cope with increased cardiac work demand largely stems from mitochondrial redox/energetic disarrangements that mutually influence each other, leading to myocyte or whole-heart mechanical dysfunction.
Collapse
Affiliation(s)
- Carlo G. Tocchetti
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Viviane Caceres
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Brian A. Stanley
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chaoqin Xie
- Cardiovascular Research Center, Division of Cardiology, Mount Sinai School of Medicine, New York, New York
| | - Sa Shi
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Walter H. Watson
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, Kentucky
| | - Brian O’Rourke
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Sonia Cortassa
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Fadi G. Akar
- Cardiovascular Research Center, Division of Cardiology, Mount Sinai School of Medicine, New York, New York
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Dipartimento di Medicina Clinica e Sperimentale, Universita di Perugia, Perugia, Italy
| | - Miguel A. Aon
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Corresponding author: Miguel A. Aon,
| |
Collapse
|
146
|
Lopert P, Day BJ, Patel M. Thioredoxin reductase deficiency potentiates oxidative stress, mitochondrial dysfunction and cell death in dopaminergic cells. PLoS One 2012; 7:e50683. [PMID: 23226354 PMCID: PMC3511321 DOI: 10.1371/journal.pone.0050683] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 10/25/2012] [Indexed: 01/06/2023] Open
Abstract
Mitochondria are considered major generators of cellular reactive oxygen species (ROS) which are implicated in the pathogenesis of neurodegenerative diseases such as Parkinson's disease (PD). We have recently shown that isolated mitochondria consume hydrogen peroxide (H₂O₂) in a substrate- and respiration-dependent manner predominantly via the thioredoxin/peroxiredoxin (Trx/Prx) system. The goal of this study was to determine the role of Trx/Prx system in dopaminergic cell death. We asked if pharmacological and lentiviral inhibition of the Trx/Prx system sensitized dopaminergic cells to mitochondrial dysfunction, increased steady-state H₂O₂ levels and death in response to toxicants implicated in PD. Incubation of N27 dopaminergic cells or primary rat mesencephalic cultures with the Trx reductase (TrxR) inhibitor auranofin in the presence of sub-toxic concentrations of parkinsonian toxicants paraquat; PQ or 6-hydroxydopamine; 6OHDA (for N27 cells) resulted in a synergistic increase in H₂O₂ levels and subsequent cell death. shRNA targeting the mitochondrial thioredoxin reductase (TrxR2) in N27 cells confirmed the effects of pharmacological inhibition. A synergistic decrease in maximal and reserve respiratory capacity was observed in auranofin treated cells and TrxR2 deficient cells following incubation with PQ or 6OHDA. Additionally, TrxR2 deficient cells showed decreased basal mitochondrial oxygen consumption rates. These data demonstrate that inhibition of the mitochondrial Trx/Prx system sensitizes dopaminergic cells to mitochondrial dysfunction, increased steady-state H₂O₂, and cell death. Therefore, in addition to their role in the production of cellular H₂O₂ the mitochondrial Trx/Prx system serve as a major sink for cellular H₂O₂ and its disruption may contribute to dopaminergic pathology associated with PD.
Collapse
Affiliation(s)
- Pamela Lopert
- Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Brian J. Day
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Manisha Patel
- Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
147
|
Abstract
Pyridine nucleotides (PNs), such as NAD(H) and NADP(H), mediate electron transfer in many catabolic and anabolic processes. In general, NAD(+) and NADP(+) receive electrons to become NADH and NADPH by coupling with catabolic processes. These electrons are utilized for biologically essential reactions such as ATP production, anabolism and cellular oxidation-reduction (redox) regulation. Thus, in addition to ATP, NADH and NADPH could be defined as high-energy intermediates and "molecular units of currency" in energy transfer. We discuss the significance of PNs as energy/electron transporters and signal transducers, in regulating cell death and/or survival processes. In the first part of this review, we describe the role of NADH and NADPH as electron donors for NADPH oxidases (Noxs), glutathione (GSH), and thioredoxin (Trx) systems in cellular redox regulation. Noxs produce superoxide/hydrogen peroxide yielding oxidative environment, whereas GSH and Trx systems protect against oxidative stress. We then describe the role of NAD(+) and NADH as signal transducers through NAD(+)-dependent enzymes such as PARP-1 and Sirt1. PARP-1 is activated by damaged DNA in order to repair the DNA, which attenuates energy production through NAD(+) consumption; Sirt1 is activated by an increased NAD(+)/NADH ratio to facilitate signal transduction for metabolic adaption as well as stress responses. We conclude that PNs serve as an important interface for distinct cellular responses, including stress response, energy metabolism, and cell survival/death.
Collapse
Affiliation(s)
- Shin-Ichi Oka
- Cardiovascular Research Institute, UMDNJ-Newark, 185 S Orange Ave, MSB G609, Newark, NJ 07103, USA
| | | | | |
Collapse
|
148
|
Zhang Y, Tocchetti CG, Krieg T, Moens AL. Oxidative and nitrosative stress in the maintenance of myocardial function. Free Radic Biol Med 2012; 53:1531-40. [PMID: 22819981 DOI: 10.1016/j.freeradbiomed.2012.07.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Revised: 06/12/2012] [Accepted: 07/11/2012] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) are generated by several different cellular sources, and their accumulation within the myocardium is widely considered to cause harmful oxidative stress. On the other hand, their role as second messengers has gradually emerged. The equilibrium of the nitroso/redox balance between reactive nitrogen species and ROS is crucial for the health of cardiomyocytes. This review provides a comprehensive overview of sources of oxidative stress in cardiac myocytes and describes the role of the nitroso/redox balance in cardiac pathophysiology. Although the exact mechanism of ROS production by nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Nox's) is not completely understood, Nox2 and Nox4 have particularly important roles within the myocardium. Increasing evidence suggests that Nox2 produces superoxide and Nox4 generates only hydrogen peroxide. We also discuss the key role of nitric oxide synthases (NOSs) in the maintenance of the nitroso/redox balance: uncoupled endothelial NOS has been suggested to shift from nitric oxide to ROS production, contributing to increased oxidative stress within the myocardium. Furthermore, we highlight the importance of sequentially targeting and/or regulating the specific sources of oxidative and nitrosative stress to prevent and/or reverse myocardial dysfunction. Inhibition of NADPH oxidase-dependent ROS is considered to be a potential strategy for treatment of cardiomyopathy. Neither in vivo nor clinical data are available for NADPH oxidase inhibitors. Specifically targeting the mitochondria with the antioxidant MitoQ would be a very promising translation approach, because it could prevent mitochondrial permeability transition pore opening when ROS are produced during heart reperfusion. Enhancing NO signaling could also be a promising therapeutic approach against myocardial dysfunction.
Collapse
Affiliation(s)
- Yixuan Zhang
- Department of Cardiology, Maastricht University Medical Center, Cardiovascular Research Institute Maastricht, 6202 AZ Maastricht, The Netherlands
| | | | | | | |
Collapse
|
149
|
Madeira JM, Gibson DL, Kean WF, Klegeris A. The biological activity of auranofin: implications for novel treatment of diseases. Inflammopharmacology 2012; 20:297-306. [PMID: 22965242 DOI: 10.1007/s10787-012-0149-1] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 08/22/2012] [Indexed: 11/30/2022]
Abstract
More than 30 years ago, auranofin was developed for the treatment of rheumatoid arthritis as a substitution for the injectable gold compounds aurothiomalate and aurothioglucose. Both the ease of oral administration over intramuscular injections and more potent anti-inflammatory effects in vitro made auranofin seem like an excellent substitute for the traditional injectable gold compounds. Despite efficacy in the treatment of both rheumatoid arthritis and psoriasis, currently, auranofin is seldom used as a treatment for patients with rheumatoid arthritis as more novel anti-rheumatic medications have become available. Despite the decline in its clinical applications, research on auranofin has continued as it shows promise in the treatment of several different diseases. In recent years, advances in technology have allowed researchers to use molecular techniques to identify novel mechanisms of action of auranofin. Additionally, researchers are discovering potential new applications of auranofin. Dual inhibition of inflammatory pathways and thiol redox enzymes by auranofin makes it a new candidate for cancer therapy and treating microbial infections. This review will summarize recently obtained data on the mechanisms of action of auranofin, and potential new applications of auranofin in the treatment of various diseases, including several types of leukaemia, carcinomas, and parasitic, bacterial, and viral infections.
Collapse
Affiliation(s)
- J M Madeira
- Department of Biology, Irving K. Barber School of Arts and Sciences, University of British Columbia Okanagan Campus, Kelowna, BC, Canada
| | | | | | | |
Collapse
|
150
|
Cacho-Valadez B, Muñoz-Lobato F, Pedrajas JR, Cabello J, Fierro-González JC, Navas P, Swoboda P, Link CD, Miranda-Vizuete A. The characterization of the Caenorhabditis elegans mitochondrial thioredoxin system uncovers an unexpected protective role of thioredoxin reductase 2 in β-amyloid peptide toxicity. Antioxid Redox Signal 2012; 16:1384-400. [PMID: 22220943 PMCID: PMC3329951 DOI: 10.1089/ars.2011.4265] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIM Functional in vivo studies on the mitochondrial thioredoxin system are hampered by the embryonic or larval lethal phenotypes displayed by murine or Drosophila knock-out models. Thus, the access to alternative metazoan knock-out models for the mitochondrial thioredoxin system is of critical importance. RESULTS We report here the characterization of the mitochondrial thioredoxin system of Caenorhabditis elegans that is composed of the genes trx-2 and trxr-2. We demonstrate that the proteins thioredoxin 2 (TRX-2) and thioredoxin reductase 2 (TRXR-2) localize to the mitochondria of several cells and tissues of the nematode and that trx-2 and trxr-2 are upregulated upon induction of the mitochondrial unfolded protein response. Surprisingly, C. elegans trx-2 (lof ) and trxr-2 (null) single and double mutants are viable and display similar growth rates as wild-type controls. Moreover, the lack of the mitochondrial thioredoxin system does not affect longevity, reactive oxygen species production or the apoptotic program. Interestingly, we found a protective role of TRXR-2 in a transgenic nematode model of Alzheimer's disease (AD) that expresses human β-amyloid peptide and causes an age-dependent progressive paralysis. Hence, trxr-2 downregulation enhanced the paralysis phenotype, while a strong decrease of β-amyloid peptide and amyloid deposits occurred when TRXR-2 was overexpressed. INNOVATION C. elegans provides the first viable metazoan knock-out model for the mitochondrial thioredoxin system and identifies a novel role of this system in β-amyloid peptide toxicity and AD. CONCLUSION The nematode strains characterized in this work make C. elegans an ideal model organism to study the pathophysiology of the mitochondrial thioredoxin system at the level of a complete organism.
Collapse
Affiliation(s)
- Briseida Cacho-Valadez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC), Depto. de Fisiología, Anatomía y Biología Celular, Universidad Pablo de Olavide, Sevilla, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|