51
|
Wendt MMN, de Oliveira MC, Franco-Salla GB, Castro LS, Parizotto ÂV, Souza Silva FM, Natali MRM, Bersani-Amado CA, Bracht A, Comar JF. Fatty acids uptake and oxidation are increased in the liver of rats with adjuvant-induced arthritis. Biochim Biophys Acta Mol Basis Dis 2018; 1865:696-707. [PMID: 30593897 DOI: 10.1016/j.bbadis.2018.12.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/11/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022]
Abstract
Severe rheumatoid cachexia is associated with pronounced loss of muscle and fat mass in patients with advanced rheumatoid arthritis. This condition is associated with dyslipidemia and predisposition to cardiovascular diseases. Circulating levels of triglycerides (TG) and free fatty acids (FFA) have not yet been consistently defined in severe arthritis. Similarly, the metabolism of these lipids in the arthritic liver has not yet been clarified. Aiming at filling these gaps this study presents a characterization of the circulating lipid profile and of the fatty acids uptake and metabolism in perfused livers of rats with adjuvant-induced arthritis. The levels of TG and total cholesterol were reduced in both serum (10-20%) and liver (20-35%) of arthritic rats. The levels of circulating FFA were 40% higher in arthritic rats, possibly in consequence of cytokine-induced adipose tissue lipolysis. Hepatic uptake and oxidation of palmitic and oleic acids was higher in arthritic livers. The phenomenon results possibly from a more oxidized state of the arthritic liver. Indeed, NADPH/NADP+ and NADH/NAD+ ratios were 30% lower in arthritic livers, which additionally presented higher activities of the citric acid cycle driven by both endogenous and exogenous FFA. The lower levels of circulating and hepatic TG possibly are caused by an increased oxidation associated to a reduced synthesis of fatty acids in arthritic livers. These results reveal that the lipid hepatic metabolism in arthritic rats presents a strong catabolic tendency, a condition that should contribute to the marked cachexia described for arthritic rats and possibly for the severe rheumatoid arthritis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maria R M Natali
- Department of Morphological Sciences, State University of Maringá, PR, Brazil
| | | | - Adelar Bracht
- Department of Biochemistry, State University of Maringá, PR, Brazil
| | - Jurandir F Comar
- Department of Biochemistry, State University of Maringá, PR, Brazil.
| |
Collapse
|
52
|
Young A, Gill R, Mailloux RJ. Protein S-glutathionylation: The linchpin for the transmission of regulatory information on redox buffering capacity in mitochondria. Chem Biol Interact 2018; 299:151-162. [PMID: 30537466 DOI: 10.1016/j.cbi.2018.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/08/2018] [Accepted: 12/07/2018] [Indexed: 01/01/2023]
Abstract
Protein S-glutathionylation reactions are a ubiquitous oxidative modification required to control protein function in response to changes in redox buffering capacity. These reactions are rapid and reversible and are, for the most part, enzymatically mediated by glutaredoxins (GRX) and glutathione S-transferases (GST). Protein S-glutathionylation has been found to control a range of cell functions in response to different physiological cues. Although these reactions occur throughout the cell, mitochondrial proteins seem to be highly susceptible to reversible S-glutathionylation, a feature attributed to the unique physical properties of this organelle. Indeed, mitochondria contain a number of S-glutathionylation targets which includes proteins involved in energy metabolism, solute transport, reactive oxygen species (ROS) production, proton leaks, apoptosis, antioxidant defense, and mitochondrial fission and fusion. Moreover, it has been found that conjugation and removal of glutathione from proteins in mitochondria fulfills a number of important physiological roles and defects in these reactions can have some dire pathological consequences. Here, we provide an updated overview on mitochondrial protein S-glutathionylation reactions and their importance in cell functions and physiology.
Collapse
Affiliation(s)
- Adrian Young
- Department of Biochemistry, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Robert Gill
- Department of Biochemistry, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Ryan J Mailloux
- Department of Biochemistry, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, Canada.
| |
Collapse
|
53
|
Michalak M, Agellon LB. Stress Coping Strategies in the Heart: An Integrated View. Front Cardiovasc Med 2018; 5:168. [PMID: 30519562 PMCID: PMC6258784 DOI: 10.3389/fcvm.2018.00168] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/02/2018] [Indexed: 12/15/2022] Open
Abstract
The heart is made up of an ordered amalgam of cardiac cell types that work together to coordinate four major processes, namely energy production, electrical conductance, mechanical work, and tissue remodeling. Over the last decade, a large body of information has been amassed regarding how different cardiac cell types respond to cellular stress that affect the functionality of their elaborate intracellular membrane networks, the cellular reticular network. In the context of the heart, the manifestations of stress coping strategies likely differ depending on the coping strategy outcomes of the different cardiac cell types, and thus may underlie the development of distinct cardiac disorders. It is not clear whether all cardiac cell types have similar sensitivity to cellular stress, how specific coping response strategies modify their unique roles, and how their metabolic status is communicated to other cells within the heart. Here we discuss our understanding of the roles of specialized cardiac cells that together make the heart function as an organ with the ability to pump blood continuously and follow a regular rhythm.
Collapse
Affiliation(s)
- Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, QC, Canada
| |
Collapse
|
54
|
Cardiac-specific Conditional Knockout of the 18-kDa Mitochondrial Translocator Protein Protects from Pressure Overload Induced Heart Failure. Sci Rep 2018; 8:16213. [PMID: 30385779 PMCID: PMC6212397 DOI: 10.1038/s41598-018-34451-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 10/18/2018] [Indexed: 01/17/2023] Open
Abstract
Heart failure (HF) is characterized by abnormal mitochondrial calcium (Ca2+) handling, energy failure and impaired mitophagy resulting in contractile dysfunction and myocyte death. We have previously shown that the 18-kDa mitochondrial translocator protein of the outer mitochondrial membrane (TSPO) can modulate mitochondrial Ca2+ uptake. Experiments were designed to test the role of the TSPO in a murine pressure-overload model of HF induced by transverse aortic constriction (TAC). Conditional, cardiac-specific TSPO knockout (KO) mice were generated using the Cre-loxP system. TSPO-KO and wild-type (WT) mice underwent TAC for 8 weeks. TAC-induced HF significantly increased TSPO expression in WT mice, associated with a marked reduction in systolic function, mitochondrial Ca2+ uptake, complex I activity and energetics. In contrast, TSPO-KO mice undergoing TAC had preserved ejection fraction, and exhibited fewer clinical signs of HF and fibrosis. Mitochondrial Ca2+ uptake and energetics were restored in TSPO KO mice, associated with decreased ROS, improved complex I activity and preserved mitophagy. Thus, HF increases TSPO expression, while preventing this increase limits the progression of HF, preserves ATP production and decreases oxidative stress, thereby preventing metabolic failure. These findings suggest that pharmacological interventions directed at TSPO may provide novel therapeutics to prevent or treat HF.
Collapse
|
55
|
Trewin AJ, Parker L, Shaw CS, Hiam DS, Garnham A, Levinger I, McConell GK, Stepto NK. Acute HIIE elicits similar changes in human skeletal muscle mitochondrial H2O2 release, respiration, and cell signaling as endurance exercise even with less work. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1003-R1016. [DOI: 10.1152/ajpregu.00096.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
It remains unclear whether high-intensity interval exercise (HIIE) elicits distinct molecular responses to traditional endurance exercise relative to the total work performed. We aimed to investigate the influence of exercise intensity on acute perturbations to skeletal muscle mitochondrial function (respiration and reactive oxygen species) and metabolic and redox signaling responses. In a randomized, repeated measures crossover design, eight recreationally active individuals (24 ± 5 yr; V̇o2peak: 48 ± 11 ml·kg−1·min−1) undertook continuous moderate-intensity [CMIE: 30 min, 50% peak power output (PPO)], high-intensity interval (HIIE: 5 × 4 min, 75% PPO, work matched to CMIE), and low-volume sprint interval (SIE: 4 × 30 s) exercise, ≥7 days apart. Each session included muscle biopsies at baseline, immediately, and 3 h postexercise for high-resolution mitochondrial respirometry ( Jo2) and H2O2 emission ( Jh2o2) and gene and protein expression analysis. Immediately postexercise and irrespective of protocol, Jo2 increased during complex I + II leak/state 4 respiration but Jh2o2 decreased ( P < 0.05). AMP-activated protein kinase and acetyl co-A carboxylase phosphorylation increased ~1.5 and 2.5-fold respectively, while thioredoxin-reductase-1 protein abundance was ~35% lower after CMIE vs. SIE ( P < 0.05). At 3 h postexercise, regardless of protocol, Jo2 was lower during both ADP-stimulated state 3 OXPHOS and uncoupled respiration ( P < 0.05) but Jh2o2 trended higher ( P < 0.08) and PPARGC1A mRNA increased ~13-fold, and peroxiredoxin-1 protein decreased ~35%. In conclusion, intermittent exercise performed at high intensities has similar dynamic effects on muscle mitochondrial function compared with endurance exercise, irrespective of whether total workload is matched. This suggests exercise prescription can accommodate individual preferences while generating comparable molecular signals known to promote beneficial metabolic adaptations.
Collapse
Affiliation(s)
- Adam J. Trewin
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Lewan Parker
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Victoria, Australia
| | - Christopher S. Shaw
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Victoria, Australia
| | - Danielle S. Hiam
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Andrew Garnham
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Victoria, Australia
| | - Itamar Levinger
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science, Department of Medicine, Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Glenn K. McConell
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Nigel K. Stepto
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science, Department of Medicine, Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
- Monash Centre of Health Research and Implementation, School of Public Health and Preventative Medicine, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
56
|
Kembro JM, Cortassa S, Lloyd D, Sollott SJ, Aon MA. Mitochondrial chaotic dynamics: Redox-energetic behavior at the edge of stability. Sci Rep 2018; 8:15422. [PMID: 30337561 PMCID: PMC6194025 DOI: 10.1038/s41598-018-33582-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/25/2018] [Indexed: 12/14/2022] Open
Abstract
Mitochondria serve multiple key cellular functions, including energy generation, redox balance, and regulation of apoptotic cell death, thus making a major impact on healthy and diseased states. Increasingly recognized is that biological network stability/instability can play critical roles in determining health and disease. We report for the first-time mitochondrial chaotic dynamics, characterizing the conditions leading from stability to chaos in this organelle. Using an experimentally validated computational model of mitochondrial function, we show that complex oscillatory dynamics in key metabolic variables, arising at the “edge” between fully functional and pathological behavior, sets the stage for chaos. Under these conditions, a mild, regular sinusoidal redox forcing perturbation triggers chaotic dynamics with main signature traits such as sensitivity to initial conditions, positive Lyapunov exponents, and strange attractors. At the “edge” mitochondrial chaos is exquisitely sensitive to the antioxidant capacity of matrix Mn superoxide dismutase as well as to the amplitude and frequency of the redox perturbation. These results have potential implications both for mitochondrial signaling determining health maintenance, and pathological transformation, including abnormal cardiac rhythms.
Collapse
Affiliation(s)
- Jackelyn M Kembro
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT-CONICET), and Instituto de Ciencia y Tecnología de los Alimentos, Cátedra de Química Biológica, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Velez Sarsfield 1611, Córdoba, X5000HUA, Cordoba, Argentina
| | - Sonia Cortassa
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH. 251 Bayview Boulevard, Baltimore, 21224, MD, USA
| | - David Lloyd
- School of Biosciences, Cardiff University, Main Building, Museum Avenue, Cardiff, CF10 3AT, Wales, UK
| | - Steven J Sollott
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH. 251 Bayview Boulevard, Baltimore, 21224, MD, USA
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH. 251 Bayview Boulevard, Baltimore, 21224, MD, USA.
| |
Collapse
|
57
|
Exercise-Induced Reductive Stress Is a Protective Mechanism against Oxidative Stress in Peripheral Blood Mononuclear Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3053704. [PMID: 30405875 PMCID: PMC6201335 DOI: 10.1155/2018/3053704] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/10/2018] [Indexed: 12/26/2022]
Abstract
Eccentric exercise is a well-studied modality that induces oxidative stress and muscle damage. Furthermore, it promotes inflammatory response in which peripheral blood mononuclear cells (PBMCs) are the major mediators. Although free radicals are necessary in a specific range of concentrations, yet unknown, it remains unclear whether reductive redox status (i.e., increased antioxidant defenses and impaired free radical generation) is beneficial or not. Thus, the aim of the present investigation was to examine the effects of reductive stress and the impact of reduced glutathione (GSH) baseline values on the ability of PBMCs to counteract oxidative stress induced by a potent oxidative agent. PBMCs were isolated from the blood of subjects who performed eccentric exercise and treated with t-BOOH for 24 h. The subjects were clustered in the reductive and the oxidative group on the basis of increased or decreased GSH concentration postexercise compared to preexercise values, respectively. According to our results in PBMCs, lipid peroxidation levels as depicted by thiobarbituric acid reactive substances (TBARS) remained unchanged in the reductive group contrary to the observed enhancement in the oxidative group. In addition, GSH concentration and catalase activity increased in the reductive group, whereas they were not affected in the oxidative group. In conclusion, the effects of an oxidizing agent on the redox status of PBMCs isolated from the blood of athletes after acute eccentric exercise are dependent on the baseline values of GSH in erythrocytes. Otherwise, reductive stress defined by increased GSH levels is a protective mechanism, at least when followed by an oxidative stimulus.
Collapse
|
58
|
Mitochondrial NADH redox potential impacts the reactive oxygen species production of reverse Electron transfer through complex I. J Bioenerg Biomembr 2018; 50:367-377. [PMID: 30136168 DOI: 10.1007/s10863-018-9767-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 08/13/2018] [Indexed: 10/28/2022]
Abstract
There is substantial evidence that Reactive Oxygen Species (ROS) play a major part in cell functioning. Although their harmfulness through oxidative stress is well documented, their role in signaling and sensing as an oxidative signal still needs to be investigated. In most cells, the mitochondrial Electron Transport Chain (ETC) is the primary source of ROS. The production of ROS by reverse electron transfer through complex I has been demonstrated both in an experimental context but also in many pathophysiological situations. Therefore, understanding the mechanisms that regulate this ROS production is of great interest to control its harmful effects. We used nigericin, Pi and valinomycin as tools to modulate the pH gradient (∆pH) and the membrane potential (∆Ψ) of the protonmotive force (∆p) in liver and muscle mitochondria to accurately determine how these parameters control the ROS production. We show that a high ∆Ψ is the "sine qua none" condition for ROS production from the reverse electron transfer (RET) through the complex I. However, a high ∆Ψ is not the only condition governing ROS production. Indeed, using tools that modulate the mitochondrial NADH level, we also demonstrate that ROS production is directly related to the mitochondrial redox potential when the membrane potential is almost stable.
Collapse
|
59
|
Subtropical hibernation in juvenile tegu lizards (Salvator merianae): insights from intestine redox dynamics. Sci Rep 2018; 8:9368. [PMID: 29921981 PMCID: PMC6008456 DOI: 10.1038/s41598-018-27263-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/21/2018] [Indexed: 12/11/2022] Open
Abstract
Juvenile tegu lizards (Salvator merianae) experience gradual and mild temperature changes from autumn to winter in their habitat. This tropical/subtropical reptile enter a state of dormancy, with an 80% reduction in metabolic rate, that remains almost constant during winter. The redox metabolism in non-mammalian vertebrates that hibernate under such distinguished conditions is poorly understood. We analyzed the redox metabolism in the intestine of juvenile tegus during different stages of their first annual cycle. The effect of food deprivation (in spring) was also studied to compare with fasting during hibernation. Both winter dormancy and food deprivation caused decreases in reduced glutathione levels and glutathione transferase activity. While glutathione peroxidase and glutathione transferase activities decreased during winter dormancy, as well as glutathione (GSH) levels, other antioxidant enzymes (catalase, superoxide dismutase and glutathione reductase) remained unchanged. Notably, levels of disulfide glutathione (GSSG) were 2.1-fold higher in late autumn, when animals were in the process of depressing metabolism towards hibernation. This increased “oxidative tonus” could be due to a disruption in NADPH-dependent antioxidant systems. In dormancy, GSSG and lipid hydroperoxides were diminished by 60–70%. The results suggest that the entrance into hibernation is the main challenge for the redox homeostasis in the intestine of juvenile tegus.
Collapse
|
60
|
Marei WFA, De Bie J, Mohey-Elsaeed O, Wydooghe E, Bols PEJ, Leroy JLMR. Alpha-linolenic acid protects the developmental capacity of bovine cumulus-oocyte complexes matured under lipotoxic conditions in vitro. Biol Reprod 2018; 96:1181-1196. [PMID: 28520897 DOI: 10.1093/biolre/iox046] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/17/2017] [Indexed: 12/22/2022] Open
Abstract
Elevated concentrations of free fatty acids (FFAs), predominantly palmitic, stearic, and oleic acids (PSO), exert detrimental effects on oocyte developmental competence. This study examined the effects of omega-3 alpha-linolenic acid (ALA) during in vitro oocyte maturation (IVM) in the presence of PSO on subsequent embryo development and quality, and the cellular mechanisms that might be involved. Bovine cumulus-oocyte complexes (COCs) were supplemented during IVM with ALA (50 μM), PSO (425 μM), or PSO+ALA. Compared with FFA-free controls (P < 0.05), PSO increased embryo fragmentation and decreased good quality embryos on day 2 postfertilization. Day 7 blastocyst rate was also reduced. Day 8 blastocysts had lower cell counts and higher apoptosis but normal metabolic profile. In the PSO group, cumulus cell (CC) expansion was inhibited with an increased CC apoptosis while COC metabolism was not affected. Mitochondrial inner membrane potential (MMP; JC-1 staining) was reduced in the CCs and oocytes. Heat shock protein 70 (HSP70) but not glucose-regulated protein 78 kDa (GRP78, known as BiP; an endoplasmic reticulum stress marker) was upregulated in the CCs. Higher reactive oxygen species levels (DCHFDA staining) were detected in the oocytes. In contrast, adding ALA in the presence of PSO normalized embryo fragmentation, cleavage, blastocyst rates, and blastocyst quality compared to controls (P > 0.05). Combined treatment with ALA also reduced CC apoptosis, partially recovered CC expansion, abrogated the reduction in MMP in the CCs but not in the oocytes, and reduced BiP and HSP70 expression in CCs, compared with PSO only (P < 0.05). In conclusion, ALA supplementation protected oocyte developmental capacity under lipotoxic conditions mainly by protecting cumulus cell viability.
Collapse
Affiliation(s)
- Waleed F A Marei
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium.,Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Jessie De Bie
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Omnia Mohey-Elsaeed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Eline Wydooghe
- Department of Reproduction, Obstetrics, and Herd Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Peter E J Bols
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Jo L M R Leroy
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
61
|
Debashree B, Kumar M, Keshava Prasad TS, Natarajan A, Christopher R, Nalini A, Bindu PS, Gayathri N, Srinivas Bharath MM. Mitochondrial dysfunction in human skeletal muscle biopsies of lipid storage disorder. J Neurochem 2018; 145:323-341. [PMID: 29424033 DOI: 10.1111/jnc.14318] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/28/2018] [Accepted: 01/30/2018] [Indexed: 02/06/2023]
Abstract
Mitochondria regulate the balance between lipid metabolism and storage in the skeletal muscle. Altered lipid transport, metabolism and storage influence the bioenergetics, redox status and insulin signalling, contributing to cardiac and neurological diseases. Lipid storage disorders (LSDs) are neurological disorders which entail intramuscular lipid accumulation and impaired mitochondrial bioenergetics in the skeletal muscle causing progressive myopathy with muscle weakness. However, the mitochondrial changes including molecular events associated with impaired lipid storage have not been completely understood in the human skeletal muscle. We carried out morphological and biochemical analysis of mitochondrial function in muscle biopsies of human subjects with LSDs (n = 7), compared to controls (n = 10). Routine histology, enzyme histochemistry and ultrastructural analysis indicated altered muscle cell morphology and mitochondrial structure. Protein profiling of the muscle mitochondria from LSD samples (n = 5) (vs. control, n = 5) by high-throughput mass spectrometric analysis revealed that impaired metabolic processes could contribute to mitochondrial dysfunction and ensuing myopathy in LSDs. We propose that impaired fatty acid and respiratory metabolism along with increased membrane permeability, elevated lipolysis and altered cristae entail mitochondrial dysfunction in LSDs. Some of these mechanisms were unique to LSD apart from others that were common to dystrophic and inflammatory muscle pathologies. Many differentially regulated mitochondrial proteins in LSD are linked with other human diseases, indicating that mitochondrial protection via targeted drugs could be a treatment modality in LSD and related metabolic diseases. Cover Image for this Issue: doi: 10.1111/jnc.14177.
Collapse
Affiliation(s)
- Bandopadhyay Debashree
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Manish Kumar
- Institute of Bioinformatics, Bangalore, Karnataka, India.,Manipal University, Manipal, Karnataka, India
| | - Thottethodi Subrahmanya Keshava Prasad
- Institute of Bioinformatics, Bangalore, Karnataka, India.,Center for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore, Karnataka, India
| | - Archana Natarajan
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Rita Christopher
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Parayil Sankaran Bindu
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Narayanappa Gayathri
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | | |
Collapse
|
62
|
Metabolic Reprogramming and Redox Signaling in Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:241-260. [PMID: 29047090 DOI: 10.1007/978-3-319-63245-2_14] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pulmonary hypertension is a complex disease of the pulmonary vasculature, which in severe cases terminates in right heart failure. Complex remodeling of pulmonary arteries comprises the central issue of its pathology. This includes extensive proliferation, apoptotic resistance and inflammation. As such, the molecular and cellular features of pulmonary hypertension resemble hallmark characteristics of cancer cell behavior. The vascular remodeling derives from significant metabolic changes in resident cells, which we describe in detail. It affects not only cells of pulmonary artery wall, but also its immediate microenvironment involving cells of immune system (i.e., macrophages). Thus aberrant metabolism constitutes principle component of the cancer-like theory of pulmonary hypertension. The metabolic changes in pulmonary artery cells resemble the cancer associated Warburg effect, involving incomplete glucose oxidation through aerobic glycolysis with depressed mitochondrial catabolism enabling the fueling of anabolic reactions with amino acids, nucleotides and lipids to sustain proliferation. Macrophages also undergo overlapping but distinct metabolic reprogramming inducing specific activation or polarization states that enable their participation in the vascular remodeling process. Such metabolic synergy drives chronic inflammation further contributing to remodeling. Enhanced glycolytic flux together with suppressed mitochondrial bioenergetics promotes the accumulation of reducing equivalents, NAD(P)H. We discuss the enzymes and reactions involved. The reducing equivalents modulate the regulation of proteins using NAD(P)H as the transcriptional co-repressor C-terminal binding protein 1 cofactor and significantly impact redox status (through GSH, NAD(P)H oxidases, etc.), which together act to control the phenotype of the cells of pulmonary arteries. The altered mitochondrial metabolism changes its redox poise, which together with enhanced NAD(P)H oxidase activity and reduced enzymatic antioxidant activity promotes a pro-oxidative cellular status. Herein we discuss all described metabolic changes along with resultant alterations in redox status, which result in excessive proliferation, apoptotic resistance, and inflammation, further leading to pulmonary arterial wall remodeling and thus establishing pulmonary artery hypertension pathology.
Collapse
|
63
|
Vakrou S, Fukunaga R, Foster DB, Sorensen L, Liu Y, Guan Y, Woldemichael K, Pineda-Reyes R, Liu T, Tardiff JC, Leinwand LA, Tocchetti CG, Abraham TP, O'Rourke B, Aon MA, Abraham MR. Allele-specific differences in transcriptome, miRNome, and mitochondrial function in two hypertrophic cardiomyopathy mouse models. JCI Insight 2018; 3:94493. [PMID: 29563334 DOI: 10.1172/jci.insight.94493] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 02/14/2018] [Indexed: 01/06/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) stems from mutations in sarcomeric proteins that elicit distinct biophysical sequelae, which in turn may yield radically different intracellular signaling and molecular pathologic profiles. These signaling events remain largely unaddressed by clinical trials that have selected patients based on clinical HCM diagnosis, irrespective of genotype. In this study, we determined how two mouse models of HCM differ, with respect to cellular/mitochondrial function and molecular biosignatures, at an early stage of disease. We show that hearts from young R92W-TnT and R403Q-αMyHC mutation-bearing mice differ in their transcriptome, miRNome, intracellular redox environment, mitochondrial antioxidant defense mechanisms, and susceptibility to mitochondrial permeability transition pore opening. Pathway analysis of mRNA-sequencing data and microRNA profiles indicate that R92W-TnT mutants exhibit a biosignature consistent with activation of profibrotic TGF-β signaling. Our results suggest that the oxidative environment and mitochondrial impairment in young R92W-TnT mice promote activation of TGF-β signaling that foreshadows a pernicious phenotype in young individuals. Of the two mutations, R92W-TnT is more likely to benefit from anti-TGF-β signaling effects conferred by angiotensin receptor blockers and may be responsive to mitochondrial antioxidant strategies in the early stage of disease. Molecular and functional profiling may therefore serve as aids to guide precision therapy for HCM.
Collapse
Affiliation(s)
- Styliani Vakrou
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ryuya Fukunaga
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - D Brian Foster
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Lars Sorensen
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Yamin Liu
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| | - Yufan Guan
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kirubel Woldemichael
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Roberto Pineda-Reyes
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ting Liu
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Jill C Tardiff
- Department of Internal Medicine and Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Leslie A Leinwand
- Department of Molecular, Cellular, and Developmental Biology and the BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Carlo G Tocchetti
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Theodore P Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| | - Brian O'Rourke
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Miguel A Aon
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - M Roselle Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| |
Collapse
|
64
|
Varricchi G, Ameri P, Cadeddu C, Ghigo A, Madonna R, Marone G, Mercurio V, Monte I, Novo G, Parrella P, Pirozzi F, Pecoraro A, Spallarossa P, Zito C, Mercuro G, Pagliaro P, Tocchetti CG. Antineoplastic Drug-Induced Cardiotoxicity: A Redox Perspective. Front Physiol 2018; 9:167. [PMID: 29563880 PMCID: PMC5846016 DOI: 10.3389/fphys.2018.00167] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/20/2018] [Indexed: 12/28/2022] Open
Abstract
Antineoplastic drugs can be associated with several side effects, including cardiovascular toxicity (CTX). Biochemical studies have identified multiple mechanisms of CTX. Chemoterapeutic agents can alter redox homeostasis by increasing the production of reactive oxygen species (ROS) and reactive nitrogen species RNS. Cellular sources of ROS/RNS are cardiomyocytes, endothelial cells, stromal and inflammatory cells in the heart. Mitochondria, peroxisomes and other subcellular components are central hubs that control redox homeostasis. Mitochondria are central targets for antineoplastic drug-induced CTX. Understanding the mechanisms of CTX is fundamental for effective cardioprotection, without compromising the efficacy of anticancer treatments. Type 1 CTX is associated with irreversible cardiac cell injury and is typically caused by anthracyclines and conventional chemotherapeutic agents. Type 2 CTX, associated with reversible myocardial dysfunction, is generally caused by biologicals and targeted drugs. Although oxidative/nitrosative reactions play a central role in CTX caused by different antineoplastic drugs, additional mechanisms involving directly and indirectly cardiomyocytes and inflammatory cells play a role in cardiovascular toxicities. Identification of cardiologic risk factors and an integrated approach using molecular, imaging, and clinical data may allow the selection of patients at risk of developing chemotherapy-related CTX. Although the last decade has witnessed intense research related to the molecular and biochemical mechanisms of CTX of antineoplastic drugs, experimental and clinical studies are urgently needed to balance safety and efficacy of novel cancer therapies.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, Naples, Italy
| | - Pietro Ameri
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Christian Cadeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Rosalinda Madonna
- Institute of Cardiology, Center of Excellence on Aging, Università degli Studi “G. d'Annunzio” Chieti – Pescara, Chieti, Italy
- Department of Internal Medicine, Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, University of Texas Health Science Center, Houston, TX, United States
| | - Giancarlo Marone
- Section of Hygiene, Department of Public Health, University of Naples Federico II, Naples, Italy
- Monaldi Hospital Pharmacy, Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Ines Monte
- Department of General Surgery and Medical-Surgery Specialities, University of Catania, Catania, Italy
| | - Giuseppina Novo
- U.O.C. Magnetic Resonance Imaging, Fondazione Toscana G. Monasterio C.N.R., Pisa, Italy
| | - Paolo Parrella
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Flora Pirozzi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Antonio Pecoraro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Paolo Spallarossa
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Concetta Zito
- Division of Clinical and Experimental Cardiology, Department of Medicine and Pharmacology, Policlinico “G. Martino” University of Messina, Messina, Italy
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
65
|
D'Alessandro A, El Kasmi KC, Plecitá-Hlavatá L, Ježek P, Li M, Zhang H, Gupte SA, Stenmark KR. Hallmarks of Pulmonary Hypertension: Mesenchymal and Inflammatory Cell Metabolic Reprogramming. Antioxid Redox Signal 2018; 28. [PMID: 28637353 PMCID: PMC5737722 DOI: 10.1089/ars.2017.7217] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE The molecular events that promote the development of pulmonary hypertension (PH) are complex and incompletely understood. The complex interplay between the pulmonary vasculature and its immediate microenvironment involving cells of immune system (i.e., macrophages) promotes a persistent inflammatory state, pathological angiogenesis, and fibrosis that are driven by metabolic reprogramming of mesenchymal and immune cells. Recent Advancements: Consistent with previous findings in the field of cancer metabolism, increased glycolytic rates, incomplete glucose and glutamine oxidation to support anabolism and anaplerosis, altered lipid synthesis/oxidation ratios, increased one-carbon metabolism, and activation of the pentose phosphate pathway to support nucleoside synthesis are but some of the key metabolic signatures of vascular cells in PH. In addition, metabolic reprogramming of macrophages is observed in PH and is characterized by distinct features, such as the induction of specific activation or polarization states that enable their participation in the vascular remodeling process. CRITICAL ISSUES Accumulation of reducing equivalents, such as NAD(P)H in PH cells, also contributes to their altered phenotype both directly and indirectly by regulating the activity of the transcriptional co-repressor C-terminal-binding protein 1 to control the proliferative/inflammatory gene expression in resident and immune cells. Further, similar to the role of anomalous metabolism in mitochondria in cancer, in PH short-term hypoxia-dependent and long-term hypoxia-independent alterations of mitochondrial activity, in the absence of genetic mutation of key mitochondrial enzymes, have been observed and explored as potential therapeutic targets. FUTURE DIRECTIONS For the foreseeable future, short- and long-term metabolic reprogramming will become a candidate druggable target in the treatment of PH. Antioxid. Redox Signal. 28, 230-250.
Collapse
Affiliation(s)
- Angelo D'Alessandro
- 1 Department of Biochemistry and Molecular Genetics, University of Colorado - Denver , Colorado
| | - Karim C El Kasmi
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado.,3 Department of Pediatric Gastroenterology, University of Colorado - Denver , Colorado
| | - Lydie Plecitá-Hlavatá
- 4 Department of Mitochondrial Physiology, Institute of Physiology , Czech Academy of Sciences, Prague, Czech Republic
| | - Petr Ježek
- 4 Department of Mitochondrial Physiology, Institute of Physiology , Czech Academy of Sciences, Prague, Czech Republic
| | - Min Li
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| | - Hui Zhang
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| | - Sachin A Gupte
- 5 Department of Pharmacology, School of Medicine, New York Medical College , Valhalla, New York
| | - Kurt R Stenmark
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| |
Collapse
|
66
|
Lau GY, Richards JG. Interspecific variation in brain mitochondrial complex I and II capacity and ROS emission in marine sculpins. J Exp Biol 2018; 222:jeb.189407. [DOI: 10.1242/jeb.189407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/29/2018] [Indexed: 12/29/2022]
Abstract
Environmental hypoxia presents a metabolic challenge for animals because it inhibits mitochondrial respiration and can lead to the generation of reactive oxygen species (ROS). We investigated the interplay between O2 use for aerobic respiration and ROS generation among sculpin fishes (Cottidae, Actinopterygii) that are known to vary in whole-animal hypoxia tolerance. We hypothesized that mitochondria from hypoxia tolerant sculpins would show more efficient O2 use with a higher phosphorylation efficiency and lower ROS emission. We showed that brain mitochondria from more hypoxia tolerant sculpins had lower complex I and higher complex II flux capacities compared with less hypoxia tolerant sculpins, but these differences were not related to variation in phosphorylation efficiency (ADP/O) or mitochondrial coupling (respiratory control ratio). The hypoxia tolerant sculpin had higher mitochondrial H2O2 emission per O2 consumed (H2O2/O2) under oligomycin-induced state 4 conditions compared to less hypoxia tolerant sculpin. An in vitro redox challenge experiment revealed species differences in how well mitochondria defend their glutathione redox status when challenged with high levels of reduced glutathione, but the redox challenge elicited the same H2O2/O2 in all species. Furthermore, in vitro anoxia-recovery lowered absolute H2O2 emission (H2O2/mg mitochondrial protein) in all species and negatively impacted state 3 respiration rates in some species, but the responses were not related to hypoxia tolerance. Overall, we clearly demonstrate a relationship between hypoxia tolerance and complex I and II flux capacities in sculpins, but the differences in complex flux capacity do not appear to be directly related to variation in ROS metabolism.
Collapse
Affiliation(s)
- Gigi Y. Lau
- Department of Zoology, The University of British Columbia, 6270 University Boulevard, Vancouver, B.C., Canada, V6T 1Z4
| | - Jeffrey G. Richards
- Department of Zoology, The University of British Columbia, 6270 University Boulevard, Vancouver, B.C., Canada, V6T 1Z4
| |
Collapse
|
67
|
Wollenman LC, Vander Ploeg MR, Miller ML, Zhang Y, Bazil JN. The effect of respiration buffer composition on mitochondrial metabolism and function. PLoS One 2017; 12:e0187523. [PMID: 29091971 PMCID: PMC5665555 DOI: 10.1371/journal.pone.0187523] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/20/2017] [Indexed: 11/19/2022] Open
Abstract
Functional studies on isolated mitochondria critically rely on the right choice of respiration buffer. Differences in buffer composition can lead to dramatically different respiration rates leading to difficulties in comparing prior studies. The ideal buffer facilities high ADP-stimulated respiratory rates and minimizes substrate transport effects so that the ability to distinguish between various treatments and conditions is maximal. In this study, we analyzed a variety of respiration buffers and substrate combinations to determine the optimal conditions to support mitochondrial function through ADP-stimulated respiration and uncoupled respiration using FCCP. The buffers consisted of a standard KCl based buffer (B1) and three modified buffers with chloride replaced by the K-lactobionate, sucrose, and the antioxidant taurine (B2) or K-gluconate (B3). The fourth buffer (B4) was identical to B2 except that K-lactobionate was replaced with K-gluconate. The substrate combinations consisted of metabolites that utilize different pathways of mitochondrial metabolism. To test mitochondrial function, we used isolated cardiac guinea pig mitochondria and measured oxygen consumption for three respiratory states using an Oroboros Oxygraph-2k. These states were the leak state (energized mitochondria in the absence of adenylates), ADP-stimulated state (energized mitochondria in the presence of saturating ADP concentrations), and uncoupled state (energized mitochondria in the presence of FCCP). On average across all substrate combinations, buffers B2, B3, and B4 had an increase of 16%, 26%, and 35% for the leak state, ADP-simulated state, and uncoupled state, respectively, relative to rates using B1. The common feature distinguishing these buffers from B1 is the notable lack of high chloride concentrations. Based on the respiratory rate metrics obtained with the substrate combinations, we conclude that the adenine nucleotide translocase, the dicarboxylate carrier, and the alpha-ketoglutarate exchanger are partially inhibited by chloride. Therefore, when the goal is to maximize ADP-stimulated respiration, buffers containing K-lactobionate or K-gluconate are superior choices compared to the standard KCl-based buffers.
Collapse
Affiliation(s)
- Lucas C. Wollenman
- Department of Physiology, Michigan State University, East Lansing, MI, United States of America
- Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Matthew R. Vander Ploeg
- Department of Physiology, Michigan State University, East Lansing, MI, United States of America
| | - Mackinzie L. Miller
- Biomedical Laboratory Diagnostics, Michigan State University, East Lansing, MI, United States of America
- Nephrology and Hypertension, Henry Ford Hospital, Detroit, MI, United States of America
| | - Yizhu Zhang
- Department of Physiology, Michigan State University, East Lansing, MI, United States of America
| | - Jason N. Bazil
- Department of Physiology, Michigan State University, East Lansing, MI, United States of America
| |
Collapse
|
68
|
Kuksal N, Chalker J, Mailloux RJ. Progress in understanding the molecular oxygen paradox - function of mitochondrial reactive oxygen species in cell signaling. Biol Chem 2017; 398:1209-1227. [PMID: 28675747 DOI: 10.1515/hsz-2017-0160] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/27/2017] [Indexed: 11/15/2022]
Abstract
The molecular oxygen (O2) paradox was coined to describe its essential nature and toxicity. The latter characteristic of O2 is associated with the formation of reactive oxygen species (ROS), which can damage structures vital for cellular function. Mammals are equipped with antioxidant systems to fend off the potentially damaging effects of ROS. However, under certain circumstances antioxidant systems can become overwhelmed leading to oxidative stress and damage. Over the past few decades, it has become evident that ROS, specifically H2O2, are integral signaling molecules complicating the previous logos that oxyradicals were unfortunate by-products of oxygen metabolism that indiscriminately damage cell structures. To avoid its potential toxicity whilst taking advantage of its signaling properties, it is vital for mitochondria to control ROS production and degradation. H2O2 elimination pathways are well characterized in mitochondria. However, less is known about how H2O2 production is controlled. The present review examines the importance of mitochondrial H2O2 in controlling various cellular programs and emerging evidence for how production is regulated. Recently published studies showing how mitochondrial H2O2 can be used as a secondary messenger will be discussed in detail. This will be followed with a description of how mitochondria use S-glutathionylation to control H2O2 production.
Collapse
|
69
|
Oscillations of ultra-weak photon emission from cancer and non-cancer cells stressed by culture medium change and TNF-α. Sci Rep 2017; 7:11249. [PMID: 28900100 PMCID: PMC5596028 DOI: 10.1038/s41598-017-10949-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 06/29/2017] [Indexed: 12/16/2022] Open
Abstract
Cells spontaneously emit photons in the UV to visible/near-infrared range (ultra-weak photon emission, UPE). Perturbations of the cells’ state cause changes in UPE (evoked UPE). The aim of the present study was to analyze the evoked UPE dynamics of cells caused by two types of cell perturbations (stressors): (i) a cell culture medium change, and (ii) application of the pro-inflammatory cytokine tumor necrosis factor alpha (TNF-α). Four types of human cell lines were used (squamous cell carcinoma cells, A431; adenocarcinomic alveolar basal epithelial cells, A549; p53-deficient keratinocytes, HaCaT, and cervical cancer cells, HeLa). In addition to the medium change, TNF-α was applied at different concentrations (5, 10, 20, and 40 ng/mL) and UPE measurements were performed after incubation times of 0, 30, 60, 90 min, 2, 5, 12, 24, 48 h. It was observed that (i) the change of cell culture medium (without added TNF-α) induces a cell type-specific transient increase in UPE with the largest UPE increase observed in A549 cells, (ii) the addition of TNF-α induces a cell type-specific and dose-dependent change in UPE, and (iii) stressed cell cultures in general exhibit oscillatory UPE changes.
Collapse
|
70
|
Mitochondrial transition ROS spike (mTRS) results from coordinated activities of complex I and nicotinamide nucleotide transhydrogenase. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:955-965. [PMID: 28866380 DOI: 10.1016/j.bbabio.2017.08.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 08/20/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
Abstract
Mitochondria exhibit suppressed ATP production, membrane potential (∆Ψmt) polarization and reactive oxygen species (ROS) bursts during some cellular metabolic transitions. Although mitochondrial ROS release is influenced by ∆Ψmt and respiratory state, the relationship between these properties remains controversial primarily because they have not been measured simultaneously. We developed a multiparametric method for probing mitochondrial function that allowed precise characterization of the temporal relationship between ROS, ∆Ψmt and respiration. We uncovered a previously unknown spontaneous ROS spike - termed mitochondrial transition ROS spike (mTRS) - associated with re-polarization of ∆Ψmt that occurs at the transition between mitochondrial energy states. Pharmacological inhibition of complex CI (CI), nicotinamide nucleotide transhydrogenase (NNT) and antioxidant system significantly decreased the ability of mitochondria to exhibit mTRS. NADH levels followed a similar trend to that of ROS during the mTRS, providing a link between CI and NNT in mTRS regulation. We show that (i) mTRS is enhanced by simultaneous activation of CI and complex II (CII); (ii) CI is the principal origin of mTRS; (iii) NNT regulates mTRS via NADH- and ∆Ψmt-dependent mechanisms; (iv) mTRS is not a pH spike; and (v), mTRS changes in amplitude under stress conditions and its occurrence can be a signature of mitochondrial health. Collectively, we uncovered and characterized the biophysical properties and mechanisms of mTRS, and propose it as a potential diagnostic tool for CI-related dysfunctions, and as a biomarker of mitochondrial functional integrity.
Collapse
|
71
|
Kanaan GN, Harper ME. Cellular redox dysfunction in the development of cardiovascular diseases. Biochim Biophys Acta Gen Subj 2017; 1861:2822-2829. [PMID: 28778485 DOI: 10.1016/j.bbagen.2017.07.027] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/21/2017] [Accepted: 07/30/2017] [Indexed: 12/22/2022]
Abstract
To meet its exceptionally high energy demands, the heart relies largely on fatty acid oxidation, which then drives the oxidative phosphorylation system in mitochondria. Each day, this system produces about 6kg of ATP to sustain heart function. Fatty acid oxidation is sometimes associated with high rates of mitochondrial reactive oxygen species (ROS) production. By definition, ROS are singlet electron intermediates formed during the partial reduction of oxygen to water and they include radical and non-radical intermediates like superoxide, hydrogen peroxide and hydroxyl radical. Superoxide can also interact with nitric oxide to produce peroxynitrite that in turn can give rise to other radical or non-radical reactive nitrogen species (RNS) like nitrogen dioxide, dinitrogen trioxide and others. While mitochondrial and cellular functions can be impaired by ROS if they accumulate, under normal physiological conditions ROS are important signaling molecules in the cardiovascular system. A fine balance between ROS production and antioxidant systems, including glutathione redox, is essential in the heart; otherwise the ensuing damage can contribute to pathogenic processes, which can culminate in endothelial dysfunction, atherosclerosis, hypertension, cardiac hypertrophy, arrhythmias, myocardial ischemia/reperfusion damage, and heart failure. Here we provide a succinct review of recent findings.
Collapse
Affiliation(s)
- Georges N Kanaan
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
72
|
Nishida M, Nishimura A, Matsunaga T, Motohashi H, Kasamatsu S, Akaike T. Redox regulation of electrophilic signaling by reactive persulfides in cardiac cells. Free Radic Biol Med 2017; 109:132-140. [PMID: 28109891 DOI: 10.1016/j.freeradbiomed.2017.01.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 01/14/2017] [Accepted: 01/15/2017] [Indexed: 12/30/2022]
Abstract
Maintaining a redox balance by means of precisely controlled systems that regulate production, and elimination, and metabolism of electrophilic substances (electrophiles) is essential for normal cardiovascular function. Electrophilic signaling is mainly regulated by endogenous electrophiles that are generated from reactive oxygen species, nitric oxide, and the derivative reactive species of nitric oxide during stress responses, as well as by exogenous electrophiles including compounds in foods and environmental pollutants. Among electrophiles formed endogenously, 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP) has unique cell signaling functions, and pathways for its biosynthesis, signaling mechanism, and metabolism in cells have been clarified. Reactive persulfide species such as cysteine persulfides and polysulfides that are endogenously produced in cells are likely to be involved in 8-nitro-cGMP metabolism. These new aspects of redox biology may stimulate innovative and multidisciplinary research in cardiovascular physiology and pathophysiology. In our review, we focus on the redox-dependent regulation of electrophilic signaling via reduction and metabolism of electrophiles by reactive persulfides in cardiac cells, and we include suggestions for a new therapeutic strategy for cardiovascular disease.
Collapse
Affiliation(s)
- Motohiro Nishida
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8787, Japan; Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; PRESTO, Japan Science and Technology Agency (JST), Kawaguchi 332-0012, Japan.
| | - Akiyuki Nishimura
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
| | - Tetsuro Matsunaga
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Hozumi Motohashi
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Shingo Kasamatsu
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Takaaki Akaike
- Department of Environmental Health Sciences and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.
| |
Collapse
|
73
|
Mitochondrial health, the epigenome and healthspan. Clin Sci (Lond) 2017; 130:1285-305. [PMID: 27358026 DOI: 10.1042/cs20160002] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/03/2016] [Indexed: 02/07/2023]
Abstract
Food nutrients and metabolic supply-demand dynamics constitute environmental factors that interact with our genome influencing health and disease states. These gene-environment interactions converge at the metabolic-epigenome-genome axis to regulate gene expression and phenotypic outcomes. Mounting evidence indicates that nutrients and lifestyle strongly influence genome-metabolic functional interactions determining disease via altered epigenetic regulation. The mitochondrial network is a central player of the metabolic-epigenome-genome axis, regulating the level of key metabolites [NAD(+), AcCoA (acetyl CoA), ATP] acting as substrates/cofactors for acetyl transferases, kinases (e.g. protein kinase A) and deacetylases (e.g. sirtuins, SIRTs). The chromatin, an assembly of DNA and nucleoproteins, regulates the transcriptional process, acting at the epigenomic interface between metabolism and the genome. Within this framework, we review existing evidence showing that preservation of mitochondrial network function is directly involved in decreasing the rate of damage accumulation thus slowing aging and improving healthspan.
Collapse
|
74
|
Kurz FT, Aon MA, O'Rourke B, Armoundas AA. Functional Implications of Cardiac Mitochondria Clustering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:1-24. [PMID: 28551779 PMCID: PMC7003720 DOI: 10.1007/978-3-319-55330-6_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The spatio-temporal organization of mitochondria in cardiac myocytes facilitates myocyte-wide, cluster-bound, mitochondrial inner membrane potential oscillatory depolarizations, commonly triggered by metabolic or oxidative stressors. Local intermitochondrial coupling can be mediated by reactive oxygen species (ROS) that activate inner membrane pores to initiate a ROS-induced-ROS-release process that produces synchronized limit cycle oscillations of mitochondrial clusters within the whole mitochondrial network. The network's dynamic organization, structure and function can be assessed by quantifying dynamic local coupling constants and dynamic functional clustering coefficients, both providing information about the network's response to external stimuli. In addition to its special organization, the mitochondrial network of cardiac myocytes exhibits substrate-sensitive coupling constants and clustering coefficients. The myocyte's ability to form functional clusters of synchronously oscillating mitochondria is sensitive to conditions such as substrate availability (e.g., glucose, pyruvate, β-hydroxybutyrate), antioxidant status, respiratory chain activity, or history of oxidative challenge (e.g., ischemia-reperfusion). This underscores the relevance of quantitative methods to characterize the network's functional status as a way to assess the myocyte's resilience to pathological stressors.
Collapse
Affiliation(s)
- Felix T Kurz
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.
- Massachusetts General Hospital, Cardiovascular Research Center, Harvard Medical School, Charlestown, MA, USA.
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Antonis A Armoundas
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
75
|
Llano-Diez M, Sinclair J, Yamada T, Zong M, Fauconnier J, Zhang SJ, Katz A, Jardemark K, Westerblad H, Andersson DC, Lanner JT. The Role of Reactive Oxygen Species in β-Adrenergic Signaling in Cardiomyocytes from Mice with the Metabolic Syndrome. PLoS One 2016; 11:e0167090. [PMID: 27907040 PMCID: PMC5131978 DOI: 10.1371/journal.pone.0167090] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/07/2016] [Indexed: 12/20/2022] Open
Abstract
The metabolic syndrome is associated with prolonged stress and hyperactivity of the sympathetic nervous system and afflicted subjects are prone to develop cardiovascular disease. Under normal conditions, the cardiomyocyte response to acute β-adrenergic stimulation partly depends on increased production of reactive oxygen species (ROS). Here we investigated the interplay between beta-adrenergic signaling, ROS and cardiac contractility using freshly isolated cardiomyocytes and whole hearts from two mouse models with the metabolic syndrome (high-fat diet and ob/ob mice). We hypothesized that cardiomyocytes of mice with the metabolic syndrome would experience excessive ROS levels that trigger cellular dysfunctions. Fluorescent dyes and confocal microscopy were used to assess mitochondrial ROS production, cellular Ca2+ handling and contractile function in freshly isolated adult cardiomyocytes. Immunofluorescence, western blot and enzyme assay were used to study protein biochemistry. Unexpectedly, our results point towards decreased cardiac ROS signaling in a stable, chronic phase of the metabolic syndrome because: β-adrenergic-induced increases in the amplitude of intracellular Ca2+ signals were insensitive to antioxidant treatment; mitochondrial ROS production showed decreased basal rate and smaller response to β-adrenergic stimulation. Moreover, control hearts and hearts with the metabolic syndrome showed similar basal levels of ROS-mediated protein modification, but only control hearts showed increases after β-adrenergic stimulation. In conclusion, in contrast to the situation in control hearts, the cardiomyocyte response to acute β-adrenergic stimulation does not involve increased mitochondrial ROS production in a stable, chronic phase of the metabolic syndrome. This can be seen as a beneficial adaptation to prevent excessive ROS levels.
Collapse
Affiliation(s)
- Monica Llano-Diez
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Jon Sinclair
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Takashi Yamada
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Mei Zong
- Karolinska University Hospital, Rheumatology unit, CMM, Stockholm Sweden
| | - Jeremy Fauconnier
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Shi-Jin Zhang
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Abram Katz
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Kent Jardemark
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Håkan Westerblad
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | | | - Johanna T. Lanner
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
76
|
Jones IV AR, Meshulam T, Oliveira MF, Burritt N, Corkey BE. Extracellular Redox Regulation of Intracellular Reactive Oxygen Generation, Mitochondrial Function and Lipid Turnover in Cultured Human Adipocytes. PLoS One 2016; 11:e0164011. [PMID: 27741233 PMCID: PMC5065187 DOI: 10.1371/journal.pone.0164011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 09/19/2016] [Indexed: 12/04/2022] Open
Abstract
Background Many tissues play an important role in metabolic homeostasis and the development of diabetes and obesity. We hypothesized that the circulating redox metabolome is a master metabolic regulatory system that impacts all organs and modulates reactive oxygen species (ROS) production, lipid peroxidation, energy production and changes in lipid turnover in many cells including adipocytes. Methods Differentiated human preadipocytes were exposed to the redox couples, lactate (L) and pyruvate (P), β–hydroxybutyrate (βOHB) and acetoacetate (Acoc), and the thiol-disulfides cysteine/ cystine (Cys/CySS) and GSH/GSSG for 1.5–4 hours. ROS measurements were done with CM-H2DCFDA. Lipid peroxidation (LPO) was assessed by a modification of the thiobarbituric acid method. Lipolysis was measured as glycerol release. Lipid synthesis was measured as 14C-glucose incorporated into lipid. Respiration was assessed using the SeaHorse XF24 analyzer and the proton leak was determined from the difference in respiration with oligomycin and antimycin A. Results Metabolites with increasing oxidation potentials (GSSG, CySS, Acoc) increased adipocyte ROS. In contrast, P caused a decrease in ROS compared with L. Acoc also induced a significant increase in both LPO and lipid synthesis. L and Acoc increased lipolysis. βOHB increased respiration, mainly due to an increased proton leak. GSSG, when present throughout 14 days of differentiation significantly increased fat accumulation, but not when added later. Conclusions We demonstrated that in human adipocytes changes in the external redox state impacted ROS production, LPO, energy efficiency, lipid handling, and differentiation. A more oxidized state generally led to increased ROS, LPO and lipid turnover and more reduction led to increased respiration and a proton leak. However, not all of the redox couples were the same suggesting compartmentalization. These data are consistent with the concept of the circulating redox metabolome as a master metabolic regulatory system.
Collapse
Affiliation(s)
- Albert R. Jones IV
- Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Tova Meshulam
- Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Marcus F. Oliveira
- Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, Brazil
| | - Nathan Burritt
- Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Barbara E. Corkey
- Obesity Research Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
77
|
Fernandes J, Hao L, Bijli KM, Chandler JD, Orr M, Hu X, Jones DP, Go YM. From the Cover: Manganese Stimulates Mitochondrial H2O2 Production in SH-SY5Y Human Neuroblastoma Cells Over Physiologic as well as Toxicologic Range. Toxicol Sci 2016; 155:213-223. [PMID: 27701121 DOI: 10.1093/toxsci/kfw196] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Manganese (Mn) is an abundant redox-active metal with well-characterized mitochondrial accumulation and neurotoxicity due to excessive exposures. Mn is also an essential co-factor for the mitochondrial antioxidant protein, superoxide dismutase-2 (SOD2), and the range for adequate intake established by the Institute of Medicine Food and Nutrition Board is 20% of the interim guidance value for toxicity by the Agency for Toxic Substances and Disease Registry, leaving little margin for safety. To study toxic mechanisms over this critical dose range, we treated human neuroblastoma SH-SY5Y cells with a series of MnCl2 concentrations (from 0 to 100 μM) and measured cellular content to compare to human brain Mn content. Concentrations ≤10 μM gave cellular concentrations comparable to literature values for normal human brain, whereas concentrations ≥50 μM resulted in values comparable to brains from individuals with toxic Mn exposures. Cellular oxygen consumption rate increased as a function of Mn up to 10 μM and decreased with Mn dose ≥50 μM. Over this range, Mn had no effect on superoxide production as measured by aconitase activity or MitoSOX but increased H2O2 production as measured by MitoPY1. Consistent with increased production of H2O2, SOD2 activity, and steady-state oxidation of total thiol increased with increasing Mn. These findings have important implications for Mn toxicity by re-directing attention from superoxide anion radical to H2O2-dependent mechanisms and to investigation over the entire physiologic range to toxicologic range. Additionally, the results show that controlled Mn exposure provides a useful cell manipulation for toxicological studies of mitochondrial H2O2 signaling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
78
|
Wang W, Gong G, Wang X, Wei-LaPierre L, Cheng H, Dirksen R, Sheu SS. Mitochondrial Flash: Integrative Reactive Oxygen Species and pH Signals in Cell and Organelle Biology. Antioxid Redox Signal 2016; 25:534-49. [PMID: 27245241 PMCID: PMC5035371 DOI: 10.1089/ars.2016.6739] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
SIGNIFICANCE Recent breakthroughs in mitochondrial research have advanced, reshaped, and revolutionized our view of the role of mitochondria in health and disease. These discoveries include the development of novel tools to probe mitochondrial biology, the molecular identification of mitochondrial functional proteins, and the emergence of new concepts and mechanisms in mitochondrial function regulation. The discovery of "mitochondrial flash" activity has provided unique insights not only into real-time visualization of individual mitochondrial redox and pH dynamics in live cells but has also advanced understanding of the excitability, autonomy, and integration of mitochondrial function in vivo. RECENT ADVANCES The mitochondrial flash is a transient and stochastic event confined within an individual mitochondrion and is observed in a wide range of organisms from plants to Caenorhabditis elegans to mammals. As flash events involve multiple transient concurrent changes within the mitochondrion (e.g., superoxide, pH, and membrane potential), a number of different mitochondrial targeted fluorescent indicators can detect flash activity. Accumulating evidence indicates that flash events reflect integrated snapshots of an intermittent mitochondrial process arising from mitochondrial respiration chain activity associated with the transient opening of the mitochondrial permeability transition pore. CRITICAL ISSUES We review the history of flash discovery, summarize current understanding of flash biology, highlight controversies regarding the relative roles of superoxide and pH signals during a flash event, and bring forth the integration of both signals in flash genesis. FUTURE DIRECTIONS Investigations using flash as a biomarker and establishing its role in cell signaling pathway will move the field forward. Antioxid. Redox Signal. 25, 534-549.
Collapse
Affiliation(s)
- Wang Wang
- 1 Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington , Seattle, Washington
| | - Guohua Gong
- 1 Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington , Seattle, Washington
| | - Xianhua Wang
- 2 State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University , Beijing, China
| | - Lan Wei-LaPierre
- 3 Department of Pharmacology and Physiology, University of Rochester Medical Center , Rochester, New York
| | - Heping Cheng
- 2 State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University , Beijing, China
| | - Robert Dirksen
- 3 Department of Pharmacology and Physiology, University of Rochester Medical Center , Rochester, New York
| | - Shey-Shing Sheu
- 4 Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
79
|
Swain L, Kesemeyer A, Meyer-Roxlau S, Vettel C, Zieseniss A, Güntsch A, Jatho A, Becker A, Nanadikar MS, Morgan B, Dennerlein S, Shah AM, El-Armouche A, Nikolaev VO, Katschinski DM. Redox Imaging Using Cardiac Myocyte-Specific Transgenic Biosensor Mice. Circ Res 2016; 119:1004-1016. [PMID: 27553648 DOI: 10.1161/circresaha.116.309551] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 08/22/2016] [Indexed: 01/09/2023]
Abstract
RATIONALE Changes in redox potentials of cardiac myocytes are linked to several cardiovascular diseases. Redox alterations are currently mostly described qualitatively using chemical sensors, which however do not allow quantifying redox potentials, lack specificity, and the possibility to analyze subcellular domains. Recent advances to quantitatively describe defined redox changes include the application of genetically encoded redox biosensors. OBJECTIVE Establishment of mouse models, which allow the quantification of the glutathione redox potential (EGSH) in the cytoplasm and the mitochondrial matrix of isolated cardiac myocytes and in Langendorff-perfused hearts based on the use of the redox-sensitive green fluorescent protein 2, coupled to the glutaredoxin 1 (Grx1-roGFP2). METHODS AND RESULTS We generated transgenic mice with cardiac myocyte-restricted expression of Grx1-roGFP2 targeted either to the mitochondrial matrix or to the cytoplasm. The response of the roGFP2 toward H2O2, diamide, and dithiothreitol was titrated and used to determine the EGSH in isolated cardiac myocytes and in Langendorff-perfused hearts. Distinct EGSH were observed in the cytoplasm and the mitochondrial matrix. Stimulation of the cardiac myocytes with isoprenaline, angiotensin II, or exposure to hypoxia/reoxygenation additionally underscored that these compartments responded independently. A compartment-specific response was also observed 3 to 14 days after myocardial infarction. CONCLUSIONS We introduce redox biosensor mice as a new tool, which allows quantification of defined alterations of EGSH in the cytoplasm and the mitochondrial matrix in cardiac myocytes and can be exploited to answer questions in basic and translational cardiovascular research.
Collapse
Affiliation(s)
- Lija Swain
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Andrea Kesemeyer
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Stefanie Meyer-Roxlau
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Christiane Vettel
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Anke Zieseniss
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Annemarie Güntsch
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Aline Jatho
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Andreas Becker
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Maithily S Nanadikar
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Bruce Morgan
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Sven Dennerlein
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Ajay M Shah
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Ali El-Armouche
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Viacheslav O Nikolaev
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.)
| | - Dörthe M Katschinski
- From the Institute of Cardiovascular Physiology, Georg August University Göttingen, Germany (L.S., A.K., A.Z., A.G., A.J., A.B., M.S.N., D.M.K.); Institute of Pharmacology, Technical University Dresden, Germany (S.M.-R., A.E.-A.); Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.V.); Cellular Biochemistry, Department of Biology, University of Kaiserslautern, Germany (B.M.); Department of Cellular Biochemistry, University Medical Center Göttingen, Germany (S.D.); Cardiovascular Division, King's College London, British Heart Foundation Centre, United Kingdom (A.M.S.); and German Center for Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); and Institute of Experimental Cardiovascular Research, Hamburg, Germany (V.O.N.).
| |
Collapse
|
80
|
Abstract
A sufficiently complex set of molecules, if subject to perturbation, will self-organize and show emergent behaviour. If such a system can take on information it will become subject to natural selection. This could explain how self-replicating molecules evolved into life and how intelligence arose. A pivotal step in this evolutionary process was of course the emergence of the eukaryote and the advent of the mitochondrion, which both enhanced energy production per cell and increased the ability to process, store and utilize information. Recent research suggest that from its inception life embraced quantum effects such as 'tunnelling' and 'coherence' while competition and stressful conditions provided a constant driver for natural selection. We believe that the biphasic adaptive response to stress described by hormesis-a process that captures information to enable adaptability, is central to this whole process. Critically, hormesis could improve mitochondrial quantum efficiency, improving the ATP/ROS ratio, whereas inflammation, which is tightly associated with the aging process, might do the opposite. This all suggests that to achieve optimal health and healthy aging, one has to sufficiently stress the system to ensure peak mitochondrial function, which itself could reflect selection of optimum efficiency at the quantum level.
Collapse
Affiliation(s)
- Alistair V W Nunn
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London W1W 6UW, U.K.
| | - Geoffrey W Guy
- GW Pharmaceuticals, Porton Down, Salisbury, Wiltshire SP4 0JQ, U.K
| | - Jimmy D Bell
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London W1W 6UW, U.K
| |
Collapse
|
81
|
Wüst RCI, de Vries HJ, Wintjes LT, Rodenburg RJ, Niessen HWM, Stienen GJM. Mitochondrial complex I dysfunction and altered NAD(P)H kinetics in rat myocardium in cardiac right ventricular hypertrophy and failure. Cardiovasc Res 2016; 111:362-72. [PMID: 27402402 DOI: 10.1093/cvr/cvw176] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 06/16/2016] [Indexed: 01/31/2023] Open
Abstract
AIMS In cardiac hypertrophy (CH) and heart failure (HF), alterations occur in mitochondrial enzyme content and activities but the origin and implications of these changes for mitochondrial function need to be resolved. METHODS AND RESULTS Right ventricular CH or HF was induced by monocrotaline injection, which causes pulmonary artery hypertension, in rats. Results were compared with saline injection (CON). NAD(P)H and FAD autofluorescence were recorded in thin intact cardiac trabeculae during transitions in stimulation frequency, to assess mitochondrial complex I and complex II function, respectively. Oxygen consumption, mitochondrial morphology, protein content, and enzymatic activity were assessed. NAD(P)H autofluorescence upon an increase in stimulation frequency showed a rapid decline followed by a slow recovery. FAD autofluorescence followed a similar time course, but in opposite direction. The amplitude of the early rapid change in NAD(P)H autofluorescence was severely depressed in CH and HF compared with CON. The rapid changes in FAD autofluorescence in CH and HF were reduced to a lesser extent. Complex I-coupled respiration showed an ∼3.5-fold reduction in CH and HF; complex II-coupled respiration was depressed two-fold in HF. Western blot analyses revealed modest reductions in complex I protein content in CH and HF and in complex I activity in supercomplexes in HF. Mitochondrial volume density was similar, but mitochondrial remodelling was evident from changes in ultrastructure and fusion/fission indices in CH and HF. CONCLUSION These results suggest that the alterations in mitochondrial function observed in right ventricular CH and HF can be mainly attributed to complex I dysfunction.
Collapse
Affiliation(s)
- Rob C I Wüst
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, O
- 2 Building, De Boelelaan 1118, Amsterdam 1081 HV, The Netherlands
| | - Heder J de Vries
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, O
- 2 Building, De Boelelaan 1118, Amsterdam 1081 HV, The Netherlands
| | - Liesbeth T Wintjes
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Richard J Rodenburg
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans W M Niessen
- Department of Pathology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam 1081 HV, The Netherlands
| | - Ger J M Stienen
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, O
- 2 Building, De Boelelaan 1118, Amsterdam 1081 HV, The Netherlands Faculty of Science, Department of Physics and Astronomy, VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
82
|
Murphy E, Ardehali H, Balaban RS, DiLisa F, Dorn GW, Kitsis RN, Otsu K, Ping P, Rizzuto R, Sack MN, Wallace D, Youle RJ. Mitochondrial Function, Biology, and Role in Disease: A Scientific Statement From the American Heart Association. Circ Res 2016; 118:1960-91. [PMID: 27126807 PMCID: PMC6398603 DOI: 10.1161/res.0000000000000104] [Citation(s) in RCA: 303] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cardiovascular disease is a major leading cause of morbidity and mortality in the United States and elsewhere. Alterations in mitochondrial function are increasingly being recognized as a contributing factor in myocardial infarction and in patients presenting with cardiomyopathy. Recent understanding of the complex interaction of the mitochondria in regulating metabolism and cell death can provide novel insight and therapeutic targets. The purpose of this statement is to better define the potential role of mitochondria in the genesis of cardiovascular disease such as ischemia and heart failure. To accomplish this, we will define the key mitochondrial processes that play a role in cardiovascular disease that are potential targets for novel therapeutic interventions. This is an exciting time in mitochondrial research. The past decade has provided novel insight into the role of mitochondria function and their importance in complex diseases. This statement will define the key roles that mitochondria play in cardiovascular physiology and disease and provide insight into how mitochondrial defects can contribute to cardiovascular disease; it will also discuss potential biomarkers of mitochondrial disease and suggest potential novel therapeutic approaches.
Collapse
|
83
|
O'Brien KM. New Lessons from an Old Fish: What Antarctic Icefishes May Reveal about the Functions of Oxygen-Binding Proteins. Integr Comp Biol 2016; 56:531-41. [PMID: 27252192 DOI: 10.1093/icb/icw062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The loss of expression of the oxygen-binding protein hemoglobin (Hb) in the family Channichthyidae (suborder Notothenioidei) of Antarctic fishes is considered a disaptation that has persisted because of the unusual conditions prevailing in the Southern Ocean during the evolution of the family. The loss of expression of the intracellular oxygen-binding protein myoglobin (Mb) in heart ventricles is more of a conundrum because it occurred at four points during the radiation of the family, suggesting weakened selective pressure maintaining expression of the protein. Yet, studies have shown that when present, Mb enhances function. Here, I discuss potential reasons for weakened selective pressure maintaining Mb expression in light of the multiple functions proposed for Mb. Additionally, I discuss results from recent studies exploring the possibility that the loss of Hb and Mb may be advantageous because it reduces the production of reactive oxygen species, levels of oxidized proteins, and the energetic costs associated with replacing oxidatively damaged proteins.
Collapse
Affiliation(s)
- Kristin M O'Brien
- University of Alaska Fairbanks, Institute of Arctic Biology, Department of Biology and Wildlife, Fairbanks, AK 99775, USA
| |
Collapse
|
84
|
Alleman RJ, Tsang AM, Ryan TE, Patteson DJ, McClung JM, Spangenburg EE, Shaikh SR, Neufer PD, Brown DA. Exercise-induced protection against reperfusion arrhythmia involves stabilization of mitochondrial energetics. Am J Physiol Heart Circ Physiol 2016; 310:H1360-70. [PMID: 26945082 PMCID: PMC4888539 DOI: 10.1152/ajpheart.00858.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/26/2016] [Indexed: 11/22/2022]
Abstract
Mitochondria influence cardiac electrophysiology through energy- and redox-sensitive ion channels in the sarcolemma, with the collapse of energetics believed to be centrally involved in arrhythmogenesis. This study was conducted to determine if preservation of mitochondrial membrane potential (ΔΨm) contributes to the antiarrhythmic effect of exercise. We utilized perfused hearts, isolated myocytes, and isolated mitochondria exposed to metabolic challenge to determine the effects of exercise on cardiac mitochondria. Hearts from sedentary (Sed) and exercised (Ex; 10 days of treadmill running) Sprague-Dawley rats were perfused on a two-photon microscope stage for simultaneous measurement of ΔΨm and ECG. After ischemia-reperfusion, the collapse of ΔΨm was commensurate with the onset of arrhythmia. Exercise preserved ΔΨm and decreased the incidence of fibrillation/tachycardia (P < 0.05). Our findings in intact hearts were corroborated in isolated myocytes exposed to in vitro hypoxia-reoxygenation, with Ex rats demonstrating enhanced redox control and sustained ΔΨm during reoxygenation. Finally, we induced anoxia-reoxygenation in isolated mitochondria using high-resolution respirometry with simultaneous measurement of respiration and H2O2 Mitochondria from Ex rats sustained respiration with lower rates of H2O2 emission than Sed rats. Exercise helps sustain postischemic mitochondrial bioenergetics and redox homeostasis, which is associated with preserved ΔΨm and protection against reperfusion arrhythmia. The reduction of fatal ventricular arrhythmias through exercise-induced mitochondrial adaptations indicates that mitochondrial therapeutics may be an effective target for the treatment of heart disease.
Collapse
Affiliation(s)
- Rick J Alleman
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Alvin M Tsang
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Terence E Ryan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Daniel J Patteson
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Espen E Spangenburg
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - Saame Raza Shaikh
- East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - P Darrell Neufer
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| | - David A Brown
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and
| |
Collapse
|
85
|
Novel Perspectives in Redox Biology and Pathophysiology of Failing Myocytes: Modulation of the Intramyocardial Redox Milieu for Therapeutic Interventions-A Review Article from the Working Group of Cardiac Cell Biology, Italian Society of Cardiology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6353469. [PMID: 26881035 PMCID: PMC4736421 DOI: 10.1155/2016/6353469] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 11/16/2015] [Indexed: 12/11/2022]
Abstract
The prevalence of heart failure (HF) is still increasing worldwide, with enormous human, social, and economic costs, in spite of huge efforts in understanding pathogenetic mechanisms and in developing effective therapies that have transformed this syndrome into a chronic disease. Myocardial redox imbalance is a hallmark of this syndrome, since excessive reactive oxygen and nitrogen species can behave as signaling molecules in the pathogenesis of hypertrophy and heart failure, leading to dysregulation of cellular calcium handling, of the contractile machinery, of myocardial energetics and metabolism, and of extracellular matrix deposition. Recently, following new interesting advances in understanding myocardial ROS and RNS signaling pathways, new promising therapeutical approaches with antioxidant properties are being developed, keeping in mind that scavenging ROS and RNS tout court is detrimental as well, since these molecules also play a role in physiological myocardial homeostasis.
Collapse
|
86
|
Vendrov AE, Vendrov KC, Smith A, Yuan J, Sumida A, Robidoux J, Runge MS, Madamanchi NR. NOX4 NADPH Oxidase-Dependent Mitochondrial Oxidative Stress in Aging-Associated Cardiovascular Disease. Antioxid Redox Signal 2015; 23:1389-409. [PMID: 26054376 PMCID: PMC4692134 DOI: 10.1089/ars.2014.6221] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIMS Increased oxidative stress and vascular inflammation are implicated in increased cardiovascular disease (CVD) incidence with age. We and others demonstrated that NOX1/2 NADPH oxidase inhibition, by genetic deletion of p47phox, in Apoe(-/-) mice decreases vascular reactive oxygen species (ROS) generation and atherosclerosis in young age. The present study examined whether NOX1/2 NADPH oxidases are also pivotal to aging-associated CVD. RESULTS Both aged (16 months) Apoe(-/-) and Apoe(-/-)/p47phox(-/-) mice had increased atherosclerotic lesion area, aortic stiffness, and systolic dysfunction compared with young (4 months) cohorts. Cellular and mitochondrial ROS (mtROS) levels were significantly higher in aortic wall and vascular smooth muscle cells (VSMCs) from aged wild-type and p47phox(-/-) mice. VSMCs from aged mice had increased mitochondrial protein oxidation and dysfunction and increased vascular cell adhesion molecule 1 expression, which was abrogated with (2-(2,2,6,6-Tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (MitoTEMPO) treatment. NOX4 expression was increased in the vasculature and mitochondria of aged mice and its suppression with shRNA in VSMCs from aged mice decreased mtROS levels and improved function. Increased mtROS levels were associated with enhanced mitochondrial NOX4 expression in aortic VSMCs from aged subjects, and NOX4 expression levels in arterial wall correlated with age and atherosclerotic severity. Aged Apoe(-/-) mice treated with MitoTEMPO and 2-(2-chlorophenyl)-4-methyl-5-(pyridin-2-ylmethyl)-1H-pyrazolo[4,3-c]pyridine-3,6(2H,5H)-dione had decreased vascular ROS levels and atherosclerosis and preserved vascular and cardiac function. INNOVATION AND CONCLUSION These data suggest that NOX4, but not NOX1/2, and mitochondrial oxidative stress are mediators of CVD in aging under hyperlipidemic conditions. Regulating NOX4 activity/expression and using mitochondrial antioxidants are potential approaches to reducing aging-associated CVD.
Collapse
Affiliation(s)
- Aleksandr E Vendrov
- 1 Department of Medicine, McAllister Heart Institute, University of North Carolina , Chapel Hill, North Carolina
| | - Kimberly C Vendrov
- 2 Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina
| | - Alberto Smith
- 3 Cardiovascular Division, Academic Department of Surgery, National Institute for Health Research Biomedical Research Center at Guy's and St Thomas' National Health Service Foundation Trust , King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Jinling Yuan
- 1 Department of Medicine, McAllister Heart Institute, University of North Carolina , Chapel Hill, North Carolina
| | - Arihiro Sumida
- 1 Department of Medicine, McAllister Heart Institute, University of North Carolina , Chapel Hill, North Carolina
| | - Jacques Robidoux
- 4 Department of Pharmacology and Toxicology, The East Carolina Diabetes and Obesity Institute, East Carolina University , Greenville, North Carolina
| | - Marschall S Runge
- 1 Department of Medicine, McAllister Heart Institute, University of North Carolina , Chapel Hill, North Carolina
| | - Nageswara R Madamanchi
- 1 Department of Medicine, McAllister Heart Institute, University of North Carolina , Chapel Hill, North Carolina
| |
Collapse
|
87
|
Lorenz E, Schmacht M, Stahl U, Senz M. Enhanced incorporation yield of cysteine for glutathione overproduction by fed-batch fermentation of Saccharomyces cerevisiae. J Biotechnol 2015; 216:131-9. [DOI: 10.1016/j.jbiotec.2015.10.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 10/08/2015] [Accepted: 10/19/2015] [Indexed: 02/07/2023]
|
88
|
Naquet P, Giessner C, Galland F. Metabolic adaptation of tissues to stress releases metabolites influencing innate immunity. Curr Opin Immunol 2015; 38:30-8. [PMID: 26605965 DOI: 10.1016/j.coi.2015.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/03/2015] [Accepted: 10/21/2015] [Indexed: 12/11/2022]
Abstract
Recent developments have demonstrated that metabolic rewiring imposed by adaptation of tissues to stress leads to the release of various metabolites which directly or indirectly impact innate immune responses and inflammation. Some metabolites can behave as second messengers and leave local cues in tissues. Immune cells which infiltrate stressed tissues reorient their metabolism to cope with these microenvironmental cues while preserving their effector functions in tissues.
Collapse
Affiliation(s)
- Philippe Naquet
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France.
| | - Caroline Giessner
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France
| | - Franck Galland
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France
| |
Collapse
|
89
|
Bartz RR, Suliman HB, Piantadosi CA. Redox mechanisms of cardiomyocyte mitochondrial protection. Front Physiol 2015; 6:291. [PMID: 26578967 PMCID: PMC4620408 DOI: 10.3389/fphys.2015.00291] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 10/02/2015] [Indexed: 12/30/2022] Open
Abstract
Oxidative and nitrosative stress are primary contributors to the loss of myocardial tissue in insults ranging from ischemia/reperfusion injury from coronary artery disease and heart transplantation to sepsis-induced myocardial dysfunction and drug-induced myocardial damage. This cell damage caused by oxidative and nitrosative stress leads to mitochondrial protein, DNA, and lipid modifications, which inhibits energy production and contractile function, potentially leading to cell necrosis and/or apoptosis. However, cardiomyocytes have evolved an elegant set of redox-sensitive mechanisms that respond to and contain oxidative and nitrosative damage. These responses include the rapid induction of antioxidant enzymes, mitochondrial DNA repair mechanisms, selective mitochondrial autophagy (mitophagy), and mitochondrial biogenesis. Coordinated cytoplasmic to nuclear cell-signaling and mitochondrial transcriptional responses to the presence of elevated cytoplasmic oxidant production, e.g., H2O2, allows nuclear translocation of the Nfe2l2 transcription factor and up-regulation of downstream cytoprotective genes such as heme oxygenase-1 which generates physiologic signals, such as CO that up-regulates Nfe212 gene transcription. Simultaneously, a number of other DNA binding transcription factors are expressed and/or activated under redox control, such as Nuclear Respiratory Factor-1 (NRF-1), and lead to the induction of genes involved in both intracellular and mitochondria-specific repair mechanisms. The same insults, particularly those related to vascular stress and inflammation also produce elevated levels of nitric oxide, which also has mitochondrial protein thiol-protective functions and induces mitochondrial biogenesis through cyclic GMP-dependent and perhaps other pathways. This brief review provides an overview of these pathways and interconnected cardiac repair mechanisms.
Collapse
Affiliation(s)
- Raquel R Bartz
- Department of Anesthesiology, Duke University School of Medicine Durham, NC, USA ; Department of Medicine, Duke University School of Medicine Durham, NC, USA
| | - Hagir B Suliman
- Department of Anesthesiology, Duke University School of Medicine Durham, NC, USA ; Department of Pathology, Duke University School of Medicine Durham, NC, USA
| | - Claude A Piantadosi
- Department of Anesthesiology, Duke University School of Medicine Durham, NC, USA ; Department of Medicine, Duke University School of Medicine Durham, NC, USA ; Department of Pathology, Duke University School of Medicine Durham, NC, USA ; Durham Veterans Affairs Hospital Durham, NC, USA
| |
Collapse
|
90
|
Granger DN, Kvietys PR. Reperfusion injury and reactive oxygen species: The evolution of a concept. Redox Biol 2015; 6:524-551. [PMID: 26484802 PMCID: PMC4625011 DOI: 10.1016/j.redox.2015.08.020] [Citation(s) in RCA: 936] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 12/11/2022] Open
Abstract
Reperfusion injury, the paradoxical tissue response that is manifested by blood flow-deprived and oxygen-starved organs following the restoration of blood flow and tissue oxygenation, has been a focus of basic and clinical research for over 4-decades. While a variety of molecular mechanisms have been proposed to explain this phenomenon, excess production of reactive oxygen species (ROS) continues to receive much attention as a critical factor in the genesis of reperfusion injury. As a consequence, considerable effort has been devoted to identifying the dominant cellular and enzymatic sources of excess ROS production following ischemia-reperfusion (I/R). Of the potential ROS sources described to date, xanthine oxidase, NADPH oxidase (Nox), mitochondria, and uncoupled nitric oxide synthase have gained a status as the most likely contributors to reperfusion-induced oxidative stress and represent priority targets for therapeutic intervention against reperfusion-induced organ dysfunction and tissue damage. Although all four enzymatic sources are present in most tissues and are likely to play some role in reperfusion injury, priority and emphasis has been given to specific ROS sources that are enriched in certain tissues, such as xanthine oxidase in the gastrointestinal tract and mitochondria in the metabolically active heart and brain. The possibility that multiple ROS sources contribute to reperfusion injury in most tissues is supported by evidence demonstrating that redox-signaling enables ROS produced by one enzymatic source (e.g., Nox) to activate and enhance ROS production by a second source (e.g., mitochondria). This review provides a synopsis of the evidence implicating ROS in reperfusion injury, the clinical implications of this phenomenon, and summarizes current understanding of the four most frequently invoked enzymatic sources of ROS production in post-ischemic tissue. Reperfusion injury is implicated in a variety of human diseases and disorders. Evidence implicating ROS in reperfusion injury continues to grow. Several enzymes are candidate sources of ROS in post-ischemic tissue. Inter-enzymatic ROS-dependent signaling enhances the oxidative stress caused by I/R. .
Collapse
Affiliation(s)
- D Neil Granger
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, United States.
| | - Peter R Kvietys
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
91
|
Kehrer JP, Klotz LO. Free radicals and related reactive species as mediators of tissue injury and disease: implications for Health. Crit Rev Toxicol 2015; 45:765-98. [DOI: 10.3109/10408444.2015.1074159] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
92
|
Salin K, Auer SK, Rey B, Selman C, Metcalfe NB. Variation in the link between oxygen consumption and ATP production, and its relevance for animal performance. Proc Biol Sci 2015; 282:20151028. [PMID: 26203001 PMCID: PMC4528520 DOI: 10.1098/rspb.2015.1028] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/24/2015] [Indexed: 12/17/2022] Open
Abstract
It is often assumed that an animal's metabolic rate can be estimated through measuring the whole-organism oxygen consumption rate. However, oxygen consumption alone is unlikely to be a sufficient marker of energy metabolism in many situations. This is due to the inherent variability in the link between oxidation and phosphorylation; that is, the amount of adenosine triphosphate (ATP) generated per molecule of oxygen consumed by mitochondria (P/O ratio). In this article, we describe how the P/O ratio can vary within and among individuals, and in response to a number of environmental parameters, including diet and temperature. As the P/O ratio affects the efficiency of cellular energy production, its variability may have significant consequences for animal performance, such as growth rate and reproductive output. We explore the adaptive significance of such variability and hypothesize that while a reduction in the P/O ratio is energetically costly, it may be associated with advantages in terms of somatic maintenance through reduced production of reactive oxygen species. Finally, we discuss how considering variation in mitochondrial efficiency, together with whole-organism oxygen consumption, can permit a better understanding of the relationship between energy metabolism and life history for studies in evolutionary ecology.
Collapse
Affiliation(s)
- Karine Salin
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Sonya K Auer
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Benjamin Rey
- Laboratoire de Biométrie et Biologie Évolutive, UMR 5558, CNRS, Université de Lyon 1, Lyon, France Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Colin Selman
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Neil B Metcalfe
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
93
|
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: 2.9] [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
|
94
|
Hafstad AD, Boardman N, Aasum E. How exercise may amend metabolic disturbances in diabetic cardiomyopathy. Antioxid Redox Signal 2015; 22:1587-605. [PMID: 25738326 PMCID: PMC4449627 DOI: 10.1089/ars.2015.6304] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
SIGNIFICANCE Over-nutrition and sedentary lifestyle has led to a worldwide increase in obesity, insulin resistance, and type 2 diabetes (T2D) associated with an increased risk of development of cardiovascular disorders. Diabetic cardiomyopathy, independent of hypertension or coronary disease, is induced by a range of systemic changes and may through multiple processes result in functional and structural cardiac derangements. The pathogenesis of this cardiomyopathy is complex and multifactorial, and it will eventually lead to reduced cardiac working capacity and increased susceptibility to ischemic injury. RECENT ADVANCES Metabolic disturbances such as altered lipid handling and substrate utilization, decreased mechanical efficiency, mitochondrial dysfunction, disturbances in nonoxidative glucose pathways, and increased oxidative stress are hallmarks of diabetic cardiomyopathy. Interestingly, several of these disturbances are found to precede the development of cardiac dysfunction. CRITICAL ISSUES Exercise training is effective in the prevention and treatment of obesity and T2D. In addition to its beneficial influence on diabetes/obesity-related systemic changes, it may also amend many of the metabolic disturbances characterizing the diabetic myocardium. These changes are due to both indirect effects, exercise-mediated systemic changes, and direct effects originating from the high contractile activity of the heart during physical training. FUTURE DIRECTIONS Revealing the molecular mechanisms behind the beneficial effects of exercise training is of considerable scientific value to generate evidence-based therapy and in the development of new treatment strategies.
Collapse
Affiliation(s)
- Anne D Hafstad
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Neoma Boardman
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Ellen Aasum
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
95
|
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
|
96
|
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: 4.0] [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
|
97
|
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: 3.8] [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
|
98
|
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: 49] [Impact Index Per Article: 5.4] [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
|
99
|
Yang KC, Kyle JW, Makielski JC, Dudley SC. Mechanisms of sudden cardiac death: oxidants and metabolism. Circ Res 2015; 116:1937-55. [PMID: 26044249 PMCID: PMC4458707 DOI: 10.1161/circresaha.116.304691] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 02/09/2015] [Indexed: 02/07/2023]
Abstract
Ventricular arrhythmia is the leading cause of sudden cardiac death (SCD). Deranged cardiac metabolism and abnormal redox state during cardiac diseases foment arrhythmogenic substrates through direct or indirect modulation of cardiac ion channel/transporter function. This review presents current evidence on the mechanisms linking metabolic derangement and excessive oxidative stress to ion channel/transporter dysfunction that predisposes to ventricular arrhythmias and SCD. Because conventional antiarrhythmic agents aiming at ion channels have proven challenging to use, targeting arrhythmogenic metabolic changes and redox imbalance may provide novel therapeutics to treat or prevent life-threatening arrhythmias and SCD.
Collapse
Affiliation(s)
- Kai-Chien Yang
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.)
| | - John W Kyle
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.)
| | - Jonathan C Makielski
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.).
| | - Samuel C Dudley
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.).
| |
Collapse
|
100
|
Rovenko BM, Kubrak OI, Gospodaryov DV, Yurkevych IS, Sanz A, Lushchak OV, Lushchak VI. Restriction of glucose and fructose causes mild oxidative stress independently of mitochondrial activity and reactive oxygen species in Drosophila melanogaster. Comp Biochem Physiol A Mol Integr Physiol 2015; 187:27-39. [PMID: 25941153 DOI: 10.1016/j.cbpa.2015.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/17/2015] [Accepted: 04/23/2015] [Indexed: 01/25/2023]
Abstract
Our recent study showed different effects of glucose and fructose overconsumption on the development of obese phenotypes in Drosophila. Glucose induced glucose toxicity due to the increase in circulating glucose, whereas fructose was more prone to induce obesity promoting accumulation of reserve lipids and carbohydrates (Rovenko et al., Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2015, 180, 75-85). Searching for mechanisms responsible for these phenotypes in this study, we analyzed mitochondrial activity, mitochondrial density, mtROS production, oxidative stress markers and antioxidant defense in fruit flies fed 0.25%, 4% and 10% glucose or fructose. It is shown that there is a complex interaction between dietary monosaccharide concentrations, mitochondrial activity and oxidative modifications to proteins and lipids. Glucose at high concentration (10%) reduced mitochondrial protein density and consequently respiration in flies, while fructose did not affect these parameters. The production of ROS by mitochondria did not reflect activities of mitochondrial complexes. Moreover, there was no clear connection between mtROS production and antioxidant defense or between antioxidant defense and developmental survival, shown in our previous study (Rovenko et al., Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2015, 180, 75-85). Instead, mtROS and antioxidant machinery cooperated to maintain a redox state that determined survival rates, and paradoxically, pro-oxidant conditions facilitated larva survival independently of the type of carbohydrate. It seems that in this complex system glucose controls the amount of oxidative modification regulating mitochondrial activity, while fructose regulates steady-state mRNA levels of antioxidant enzymes.
Collapse
Affiliation(s)
- Bohdana M Rovenko
- Department of Biochemistry and Biotechnology, Vassyl Stefanyk Precarpathian National University, Ivano-Frankivsk 76018, Ukraine
| | - Olga I Kubrak
- Department of Biochemistry and Biotechnology, Vassyl Stefanyk Precarpathian National University, Ivano-Frankivsk 76018, Ukraine
| | - Dmytro V Gospodaryov
- Department of Biochemistry and Biotechnology, Vassyl Stefanyk Precarpathian National University, Ivano-Frankivsk 76018, Ukraine
| | - Ihor S Yurkevych
- Department of Biochemistry and Biotechnology, Vassyl Stefanyk Precarpathian National University, Ivano-Frankivsk 76018, Ukraine
| | - Alberto Sanz
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle-Upon-Tyne NE4 5PL, UK; Newcastle University Institute for Ageing, Newcastle University, Newcastle-Upon-Tyne NE4 5PL, UK
| | - Oleh V Lushchak
- Department of Biochemistry and Biotechnology, Vassyl Stefanyk Precarpathian National University, Ivano-Frankivsk 76018, Ukraine.
| | - Volodymyr I Lushchak
- Department of Biochemistry and Biotechnology, Vassyl Stefanyk Precarpathian National University, Ivano-Frankivsk 76018, Ukraine.
| |
Collapse
|