1
|
Perry CGR, Sweeney G. Targeting mitochondrial fission for the treatment of myocardial infarction. Eur Heart J 2025:ehaf296. [PMID: 40326356 DOI: 10.1093/eurheartj/ehaf296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/07/2025] Open
Affiliation(s)
- Christopher G R Perry
- School of Kinesiology & Health Science, York University, Toronto, Ontario, Canada
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Gary Sweeney
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
- Department of Biology, York University, 4700 Keele Street, M3J 13P Toronto, Ontario, Canada
| |
Collapse
|
2
|
Hughes MC, Ramos SV, Brahmbhatt AN, Turnbull PC, Polidovitch NN, Garibotti MC, Schlattner U, Hawke TJ, Simpson JA, Backx PH, Perry CG. Mitohormesis during advanced stages of Duchenne muscular dystrophy reveals a redox-sensitive creatine pathway that can be enhanced by the mitochondrial-targeting peptide SBT-20. Redox Biol 2024; 76:103319. [PMID: 39178732 PMCID: PMC11388197 DOI: 10.1016/j.redox.2024.103319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024] Open
Abstract
Mitochondrial creatine kinase (mtCK) regulates the "fast" export of phosphocreatine to support cytoplasmic phosphorylation of ADP to ATP which is more rapid than direct ATP export. Such "creatine-dependent" phosphate shuttling is attenuated in several muscles, including the heart, of the D2.mdx mouse model of Duchenne muscular dystrophy at only 4 weeks of age. However, the degree to which creatine-dependent and -independent systems of phosphate shuttling progressively worsen or potentially adapt in a hormetic manner throughout disease progression remains unknown. Here, we performed a series of proof-of-principle investigations designed to determine how phosphate shuttling pathways worsen or adapt in later disease stages in D2.mdx (12 months of age). We also determined whether changes in creatine-dependent phosphate shuttling are linked to alterations in mtCK thiol redox state. In permeabilized muscle fibres prepared from cardiac left ventricles, we found that 12-month-old male D2.mdx mice have reduced creatine-dependent pyruvate oxidation and elevated complex I-supported H2O2 emission (mH2O2). Surprisingly, creatine-independent ADP-stimulated respiration was increased and mH2O2 was lowered suggesting that impairments in the faster mtCK-mediated phosphocreatine export system resulted in compensation of the alternative slower pathway of ATP export. The apparent impairments in mtCK-dependent bioenergetics occurred independent of mtCK protein content but were related to greater thiol oxidation of mtCK and a more oxidized cellular environment (lower GSH:GSSG). Next, we performed a proof-of-principle study to determine whether creatine-dependent bioenergetics could be enhanced through chronic administration of the mitochondrial-targeting, ROS-lowering tetrapeptide, SBT-20. We found that 12 weeks of daily treatment with SBT-20 (from day 4-∼12 weeks of age) increased respiration and lowered mH2O2 only in the presence of creatine in D2.mdx mice without affecting calcium-induced mitochondrial permeability transition activity. In summary, creatine-dependent mitochondrial bioenergetics are attenuated in older D2.mdx mice in relation to mtCK thiol oxidation that seem to be countered by increased creatine-independent phosphate shuttling as a unique form of mitohormesis. Separate results demonstrate that creatine-dependent bioenergetics can also be enhanced with a ROS-lowering mitochondrial-targeting peptide. These results demonstrate a specific relationship between redox stress and mitochondrial hormetic reprogramming during dystrophin deficiency with proof-of-principle evidence that creatine-dependent bioenergetics could be modified with mitochondrial-targeting small peptide therapeutics.
Collapse
Affiliation(s)
- Meghan C Hughes
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada.
| | - Sofhia V Ramos
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada.
| | - Aditya N Brahmbhatt
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada.
| | - Patrick C Turnbull
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada.
| | - Nazari N Polidovitch
- Department of Biology and the Muscle Health Research Centre, York University, Toronto, ON, Canada.
| | - Madison C Garibotti
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada.
| | - Uwe Schlattner
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), and Institut Universitaire de France, Grenoble, France.
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
| | - Jeremy A Simpson
- Department of Human Health and Nutritional Sciences and Cardiovascular Research Group, University of Guelph, Guelph, ON, Canada; IMPART Team Canada Investigator Network, Saint John, New Brunswick, Canada.
| | - Peter H Backx
- Department of Biology and the Muscle Health Research Centre, York University, Toronto, ON, Canada.
| | - Christopher Gr Perry
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada.
| |
Collapse
|
3
|
Mechanisms of the "No-Reflow" Phenomenon After Acute Myocardial Infarction: Potential Role of Pericytes. JACC Basic Transl Sci 2023; 8:204-220. [PMID: 36908667 PMCID: PMC9998747 DOI: 10.1016/j.jacbts.2022.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 11/20/2022]
Abstract
Pericytes contract during myocardial ischemia resulting in capillary constriction and no reflow. Reversing pericyte contraction pharmacologically reduces no reflow and infarct size. These findings open up an entire new venue of research aimed at altering pericyte function in myocardial ischemia and infarction.
Collapse
|
4
|
Dai W, Amoedo ND, Perry J, Le Grand B, Boucard A, Carreno J, Zhao L, Brown DA, Rossignol R, Kloner RA. Effects of OP2113 on Myocardial Infarct Size and No Reflow in a Rat Myocardial Ischemia/Reperfusion Model. Cardiovasc Drugs Ther 2022; 36:217-227. [PMID: 33555510 DOI: 10.1007/s10557-020-07113-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE The present study was to determine whether OP2113 could limit myocardial infarction size and the no-reflow phenomenon in a rat myocardial ischemia/reperfusion model. METHODS Rat heart-isolated mitochondria (RHM) were used to investigate mitochondrial respiration and mitochondrial reactive oxygen species (mtROS) generation both in normal conditions and in ischemia/reperfusion-mimicking conditions (using high concentrations of succinate). Human skeletal muscle myoblasts (HSMM) in culture were used to investigate the cellular intermittent deprivation in energy substrates and oxygen as reported in ischemia/reperfusion conditions. In vivo, rats were anesthetized and subjected to 30 min of left coronary artery occlusion followed by 3 h of reperfusion. Rats were randomized to receive OP2113 as an intravenous infusion starting either 5 min prior to coronary artery occlusion (preventive), or 5 min prior to reperfusion (curative), or to receive vehicle starting 5 min prior to coronary artery occlusion. Infusions continued until the end of the study (3 h of reperfusion). RESULTS RHM treated with OP2113 showed a concentration-dependent reduction of succinate-induced mtROS generation. In HSMM cells, OP2113 treatment (5-10 μM) during 48H prevented the reduction in the steady-state level of ATP measured just after reperfusion injuries and decreased the mitochondrial affinity to oxygen. In vivo, myocardial infarct size, expressed as the percentage of the ischemic risk zone, was significantly lower in the OP2113-treated preventive group (44.5 ± 2.9%) versus that in the vehicle group (57.0 ± 3.6%; p < 0.05), with a non-significant trend toward a smaller infarct size in the curative group (50.8 ± 3.9%). The area of no reflow as a percentage of the risk zone was significantly smaller in both the OP2113-treated preventive (28.8 ± 2.4%; p = 0.026 vs vehicle) and curative groups (30.1 ± 2.3%; p = 0.04 vs vehicle) compared with the vehicle group (38.9 ± 3.1%). OP2113 was not associated with any hemodynamic changes. CONCLUSIONS These results suggest that OP2113 is a promising mitochondrial ROS-modulating agent to reduce no-reflow as well as to reduce myocardial infarct size, especially if it is on board early in the course of the infarction. It appears to have benefit on no-reflow even when administered relatively late in the course of ischemia.
Collapse
Affiliation(s)
- Wangde Dai
- HMRI Cardiovascular Research Institute, Huntington Medical Research Institutes, 686 S. Fair Oaks Avenue, Pasadena, CA, 91105, USA.
- Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| | | | - Justin Perry
- Virginia Polytechnic Institute and State University, 1035 ILSB, 1981 Kraft Drive, Virginia Tech Corporate Research Center, Blacksburg, VA, 24060, USA
| | | | | | - Juan Carreno
- HMRI Cardiovascular Research Institute, Huntington Medical Research Institutes, 686 S. Fair Oaks Avenue, Pasadena, CA, 91105, USA
| | - Lifu Zhao
- HMRI Cardiovascular Research Institute, Huntington Medical Research Institutes, 686 S. Fair Oaks Avenue, Pasadena, CA, 91105, USA
| | - David A Brown
- Virginia Polytechnic Institute and State University, 1035 ILSB, 1981 Kraft Drive, Virginia Tech Corporate Research Center, Blacksburg, VA, 24060, USA
| | | | - Robert A Kloner
- HMRI Cardiovascular Research Institute, Huntington Medical Research Institutes, 686 S. Fair Oaks Avenue, Pasadena, CA, 91105, USA
- Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
5
|
Sun L, Xu H, Wang Y, Ma X, Xu Y, Sun F. The mitochondrial-targeted peptide SBT-20 ameliorates inflammation and oxidative stress in chronic renal failure. Aging (Albany NY) 2020; 12:18238-18250. [PMID: 32979258 PMCID: PMC7585075 DOI: 10.18632/aging.103681] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/12/2020] [Indexed: 01/24/2023]
Abstract
Chronic renal failure (CRF) is the final outcome of the development of chronic kidney disease with different causes. Although CRF is a common clinical disease, its pathogenesis remains to be improved. SBT-20 belongs to a class of cell-permeable peptides that target the inner mitochondrial membrane, reduce reactive oxygen species (ROS), normalize electron transport chain function, and ATP generation. Our experiment was to evaluate whether SBT-20 affected the oxidative stress and inflammatory process of CRF. The levels of ROS production, mitochondrial membrane potential, NF- κB-p65, TNF-α, Drp1, and mfn2 were measured before and after SBT-20 treatment. We observed that SBT-20 treatment inhibited H2O2-induced mitochondrial ROS production. SBT-20 could also restore the mitochondrial membrane potential and reduce the elevated levels of NF-κB-p65 and TNF-α in HK-2 cells. In vivo, the renal function of CRF mice recovered after treating with SBT-20, the levels of necrotic cells and inflammation decreased, and the morphology of mitochondria recovered. The results showed that SBT-20 had a protective effect on CRF by reducing oxidative stress, inflammation progression via down-regulating of NF-κB-p65, TNF-α, and Drp1 and upregulating of Mfn2. These data support SBT-20 could be used as a potential preparation for CRF.
Collapse
Affiliation(s)
- Lina Sun
- Department of Nephrology, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Haiping Xu
- Department of Nephrology, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Yunfei Wang
- Department of Cardiology, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Xiaoying Ma
- Department of Nephrology, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Yan Xu
- Department of Nephrology, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Fuyun Sun
- Department of Nephrology, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| |
Collapse
|
6
|
Allen ME, Pennington ER, Perry JB, Dadoo S, Makrecka-Kuka M, Dambrova M, Moukdar F, Patel HD, Han X, Kidd GK, Benson EK, Raisch TB, Poelzing S, Brown DA, Shaikh SR. The cardiolipin-binding peptide elamipretide mitigates fragmentation of cristae networks following cardiac ischemia reperfusion in rats. Commun Biol 2020; 3:389. [PMID: 32680996 PMCID: PMC7368046 DOI: 10.1038/s42003-020-1101-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 06/23/2020] [Indexed: 01/05/2023] Open
Abstract
Mitochondrial dysfunction contributes to cardiac pathologies. Barriers to new therapies include an incomplete understanding of underlying molecular culprits and a lack of effective mitochondria-targeted medicines. Here, we test the hypothesis that the cardiolipin-binding peptide elamipretide, a clinical-stage compound under investigation for diseases of mitochondrial dysfunction, mitigates impairments in mitochondrial structure-function observed after rat cardiac ischemia-reperfusion. Respirometry with permeabilized ventricular fibers indicates that ischemia-reperfusion induced decrements in the activity of complexes I, II, and IV are alleviated with elamipretide. Serial block face scanning electron microscopy used to create 3D reconstructions of cristae ultrastructure reveals that disease-induced fragmentation of cristae networks are improved with elamipretide. Mass spectrometry shows elamipretide did not protect against the reduction of cardiolipin concentration after ischemia-reperfusion. Finally, elamipretide improves biophysical properties of biomimetic membranes by aggregating cardiolipin. The data suggest mitochondrial structure-function are interdependent and demonstrate elamipretide targets mitochondrial membranes to sustain cristae networks and improve bioenergetic function.
Collapse
Affiliation(s)
- Mitchell E Allen
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA
| | - Edward Ross Pennington
- Department of Biochemistry and Molecular Biology, East Carolina University, Greenville, NC, USA
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Justin B Perry
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA
| | - Sahil Dadoo
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Maija Dambrova
- Latvian Institute for Organic Synthesis Riga Latvia, Norwich, UK
| | - Fatiha Moukdar
- Department of Physiology, East Carolina University, Greenville, NC, USA
| | - Hetal D Patel
- Department of Physiology, East Carolina University, Greenville, NC, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX, USA
| | - Grahame K Kidd
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
- Renovo Neural Inc, Cleveland, OH, USA
| | | | - Tristan B Raisch
- Virginia Tech Faculty of Health Sciences, Roanoke, VA, USA
- Fralin Biomedical Research Institute at Virginia Tech Carillion, Roanoke, VA, USA
| | - Steven Poelzing
- Virginia Tech Faculty of Health Sciences, Roanoke, VA, USA
- Fralin Biomedical Research Institute at Virginia Tech Carillion, Roanoke, VA, USA
- Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, USA
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - David A Brown
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA
- Virginia Tech Faculty of Health Sciences, Roanoke, VA, USA
- Virginia Tech Center for Drug Discovery, Blacksburg, VA, USA
- Virginia Tech Metabolism Core Virginia Tech, Blacksburg, VA, USA
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
7
|
Cardioprotective effects of idebenone do not involve ROS scavenging: Evidence for mitochondrial complex I bypass in ischemia/reperfusion injury. J Mol Cell Cardiol 2019; 135:160-171. [DOI: 10.1016/j.yjmcc.2019.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022]
|
8
|
Goswami I, Perry JB, Allen ME, Brown DA, von Spakovsky MR, Verbridge SS. Influence of Pulsed Electric Fields and Mitochondria-Cytoskeleton Interactions on Cell Respiration. Biophys J 2019; 114:2951-2964. [PMID: 29925031 DOI: 10.1016/j.bpj.2018.04.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/11/2018] [Accepted: 04/23/2018] [Indexed: 12/25/2022] Open
Abstract
Pulsed electric fields with microsecond pulse width (μsPEFs) are used clinically; namely, irreversible electroporation/Nanoknife is used for soft tissue tumor ablation. The μsPEF pulse parameters used in irreversible electroporation (0.5-1 kV/cm, 80-100 pulses, ∼100 μs each, 1 Hz frequency) may cause an internal field to develop within the cell because of the disruption of the outer cell membrane and subsequent penetration of the electric field. An internal field may disrupt voltage-sensitive mitochondria, although the research literature has been relatively unclear regarding whether such disruptions occur with μsPEFs. This investigation reports the influence of clinically used μsPEF parameters on mitochondrial respiration in live cells. Using a high-throughput Agilent Seahorse machine, it was observed that μsPEF exposure comprising 80 pulses with amplitudes of 600 or 700 V/cm did not alter mitochondrial respiration in 4T1 cells measured after overnight postexposure recovery. To record alterations in mitochondrial function immediately after μsPEF exposure, high-resolution respirometry was used to measure the electron transport chain state via responses to glutamate-malate and ADP and mitochondrial membrane potential via response to carbonyl cyanide-p-trifluoromethoxyphenylhydrazone. In addition to measuring immediate mitochondrial responses to μsPEF exposure, measurements were also made on cells permeabilized using digitonin and those with compromised cytoskeleton due to actin depolymerization via treatment with the drug latrunculin B. The former treatment was used as a control to tease out the effects of plasma membrane permeabilization, whereas the latter was used to investigate indirect effects on the mitochondria that may occur if μsPEFs impact the cytoskeleton on which the mitochondria are anchored. Based on the results, it was concluded that within the pulse parameters tested, μsPEFs alone do not hinder mitochondrial physiology but can be used to impact the mitochondria upon compromising the actin. Mitochondrial susceptibility to μsPEF after actin depolymerization provides, to our knowledge, a novel avenue for cancer therapeutics.
Collapse
Affiliation(s)
- Ishan Goswami
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Justin B Perry
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Mitchell E Allen
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - David A Brown
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Michael R von Spakovsky
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Scott S Verbridge
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Virginia.
| |
Collapse
|
9
|
Yu H, Kalogeris T, Korthuis RJ. Reactive species-induced microvascular dysfunction in ischemia/reperfusion. Free Radic Biol Med 2019; 135:182-197. [PMID: 30849489 PMCID: PMC6503659 DOI: 10.1016/j.freeradbiomed.2019.02.031] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/26/2019] [Accepted: 02/26/2019] [Indexed: 12/13/2022]
Abstract
Vascular endothelial cells line the inner surface of the entire cardiovascular system as a single layer and are involved in an impressive array of functions, ranging from the regulation of vascular tone in resistance arteries and arterioles, modulation of microvascular barrier function in capillaries and postcapillary venules, and control of proinflammatory and prothrombotic processes, which occur in all segments of the vascular tree but can be especially prominent in postcapillary venules. When tissues are subjected to ischemia/reperfusion (I/R), the endothelium of resistance arteries and arterioles, capillaries, and postcapillary venules become dysfunctional, resulting in impaired endothelium-dependent vasodilator and enhanced endothelium-dependent vasoconstrictor responses along with increased vulnerability to thrombus formation, enhanced fluid filtration and protein extravasation, and increased blood-to-interstitium trafficking of leukocytes in these functionally distinct segments of the microcirculation. The number of capillaries open to flow upon reperfusion also declines as a result of I/R, which impairs nutritive perfusion. All of these pathologic microvascular events involve the formation of reactive species (RS) derived from molecular oxygen and/or nitric oxide. In addition to these effects, I/R-induced RS activate NLRP3 inflammasomes, alter connexin/pannexin signaling, provoke mitochondrial fission, and cause release of microvesicles in endothelial cells, resulting in deranged function in arterioles, capillaries, and venules. It is now apparent that this microvascular dysfunction is an important determinant of the severity of injury sustained by parenchymal cells in ischemic tissues, as well as being predictive of clinical outcome after reperfusion therapy. On the other hand, RS production at signaling levels promotes ischemic angiogenesis, mediates flow-induced dilation in patients with coronary artery disease, and instigates the activation of cell survival programs by conditioning stimuli that render tissues resistant to the deleterious effects of prolonged I/R. These topics will be reviewed in this article.
Collapse
Affiliation(s)
- Hong Yu
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Ted Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA; Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO 65211, USA.
| |
Collapse
|
10
|
Hughes MC, Ramos SV, Turnbull PC, Edgett BA, Huber JS, Polidovitch N, Schlattner U, Backx PH, Simpson JA, Perry CGR. Impairments in left ventricular mitochondrial bioenergetics precede overt cardiac dysfunction and remodelling in Duchenne muscular dystrophy. J Physiol 2019; 598:1377-1392. [PMID: 30674086 DOI: 10.1113/jp277306] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 01/22/2019] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Ninety-eight per cent of patients with Duchenne muscular dystrophy (DMD) develop cardiomyopathy, with 40% developing heart failure. While increased propensity for mitochondrial induction of cell death has been observed in left ventricle, it remains unknown whether this is linked to impaired mitochondrial respiratory control and elevated H2 O2 emission prior to the onset of cardiomyopathy. Classic mouse models of DMD demonstrate hyper-regeneration in skeletal muscle which may mask mitochondrial abnormalities. Using a model with less regenerative capacity that is more akin to DMD patients, we observed elevated left ventricular mitochondrial H2 O2 and impaired oxidative phosphorylation in the absence of cardiac remodelling or overt cardiac dysfunction at 4 weeks. These impairments were associated with dysfunctions at complex I, governance by ADP and creatine-dependent phosphate shuttling, which results in a less efficient response to energy demands. Mitochondria may be a therapeutic target for the treatment of cardiomyopathy in DMD. ABSTRACT In Duchenne muscular dystrophy (DMD), mitochondrial dysfunction is predicted as a response to numerous cellular stressors, yet the contribution of mitochondria to the onset of cardiomyopathy remains unknown. To resolve this uncertainty, we designed in vitro assessments of mitochondrial bioenergetics to model mitochondrial control parameters that influence cardiac function. Both left ventricular mitochondrial responsiveness to the central bioenergetic controller ADP and the ability of creatine to facilitate mitochondrial-cytoplasmic phosphate shuttling were assessed. These measurements were performed in D2.B10-DMDmdx /2J mice - a model that demonstrates skeletal muscle atrophy and weakness due to limited regenerative capacities and cardiomyopathy more akin to people with DMD than classic models. At 4 weeks of age, there was no evidence of cardiac remodelling or cardiac dysfunction despite impairments in ADP-stimulated respiration and ADP attenuation of H2 O2 emission. These impairments were seen at both submaximal and maximal ADP concentrations despite no reductions in mitochondrial content markers. The ability of creatine to enhance ADP's control of mitochondrial bioenergetics was also impaired, suggesting an impairment in mitochondrial creatine kinase-dependent phosphate shuttling. Susceptibly to permeability transition pore opening and the subsequent activation of cell death pathways remained unchanged. Mitochondrial H2 O2 emission was elevated despite no change in markers of irreversible oxidative damage, suggesting alternative redox signalling mechanisms should be explored. These findings demonstrate that selective mitochondrial dysfunction precedes the onset of overt cardiomyopathy in D2.mdx mice, suggesting that improving mitochondrial bioenergetics by restoring ADP, creatine-dependent phosphate shuttling and complex I should be considered for treating DMD patients.
Collapse
Affiliation(s)
- Meghan C Hughes
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Sofhia V Ramos
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Patrick C Turnbull
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Brittany A Edgett
- Department of Human Health and Nutritional Sciences and Cardiovascular Research Group, University of Guelph, Guelph, ON, Canada.,Department of Pharmacology, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada.,IMPART Team Canada Investigator Network, Saint John, New Brunswick, Canada
| | - Jason S Huber
- Department of Human Health and Nutritional Sciences and Cardiovascular Research Group, University of Guelph, Guelph, ON, Canada
| | - Nazari Polidovitch
- Department of Biology and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Uwe Schlattner
- Laboratory of Fundamental and Applied Bioenergetics (LBFA) and SFR Environmental and Systems Biology (BEeSy), University Grenoble Alpes, Grenoble, France
| | - Peter H Backx
- Department of Biology and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Jeremy A Simpson
- Department of Human Health and Nutritional Sciences and Cardiovascular Research Group, University of Guelph, Guelph, ON, Canada.,IMPART Team Canada Investigator Network, Saint John, New Brunswick, Canada
| | - Christopher G R Perry
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| |
Collapse
|
11
|
Bitner BF, Ray JD, Kener KB, Herring JA, Tueller JA, Johnson DK, Tellez Freitas CM, Fausnacht DW, Allen ME, Thomson AH, Weber KS, McMillan RP, Hulver MW, Brown DA, Tessem JS, Neilson AP. Common gut microbial metabolites of dietary flavonoids exert potent protective activities in β-cells and skeletal muscle cells. J Nutr Biochem 2018; 62:95-107. [PMID: 30286378 DOI: 10.1016/j.jnutbio.2018.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/20/2018] [Accepted: 09/11/2018] [Indexed: 01/06/2023]
Abstract
Flavonoids are dietary compounds with potential anti-diabetes activities. Many flavonoids have poor bioavailability and thus low circulating concentrations. Unabsorbed flavonoids are metabolized by the gut microbiota to smaller metabolites, which are more bioavailable than their precursors. The activities of these metabolites may be partly responsible for associations between flavonoids and health. However, these activities remain poorly understood. We investigated bioactivities of flavonoid microbial metabolites [hippuric acid (HA), homovanillic acid (HVA), and 5-phenylvaleric acid (5PVA)] in primary skeletal muscle and β-cells compared to a native flavonoid [(-)-epicatechin, EC]. In muscle, EC was the most potent stimulator of glucose oxidation, while 5PVA and HA simulated glucose metabolism at 25 μM, and all compounds preserved mitochondrial function after insult. However, EC and the metabolites did not uncouple mitochonndrial respiration, with the exception of 5PVA at10 μM. In β-cells, all metabolites more potently enhanced glucose-stimulated insulin secretion (GSIS) compared to EC. Unlike EC, the metabolites appear to enhance GSIS without enhancing β-cell mitochondrial respiration or increasing expression of mitochondrial electron transport chain components, and with varying effects on β-cell insulin content. The present results demonstrate the activities of flavonoid microbial metabolites for preservation of β-cell function and glucose utilization. Additionally, our data suggest that metabolites and native compounds may act by distinct mechanisms, suggesting complementary and synergistic activities in vivo which warrant further investigation. This raises the intriguing prospect that bioavailability of native dietary flavonoids may not be as critical of a limiting factor to bioactivity as previously thought.
Collapse
Affiliation(s)
- Benjamin F Bitner
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, S243 ESC, Provo, UT 84602
| | - Jason D Ray
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, S243 ESC, Provo, UT 84602
| | - Kyle B Kener
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, S243 ESC, Provo, UT 84602
| | - Jacob A Herring
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, S243 ESC, Provo, UT 84602; Department of Microbiology and Molecular Biology, Brigham Young University, 3137 LSB, Provo, UT 84602
| | - Josie A Tueller
- Department of Microbiology and Molecular Biology, Brigham Young University, 3137 LSB, Provo, UT 84602
| | - Deborah K Johnson
- Department of Microbiology and Molecular Biology, Brigham Young University, 3137 LSB, Provo, UT 84602
| | - Claudia M Tellez Freitas
- Department of Microbiology and Molecular Biology, Brigham Young University, 3137 LSB, Provo, UT 84602
| | - Dane W Fausnacht
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, 1981 Kraft Dr., Blacksburg, VA 24060
| | - Mitchell E Allen
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, 1981 Kraft Dr., Blacksburg, VA 24060
| | - Alexander H Thomson
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, 1981 Kraft Dr., Blacksburg, VA 24060
| | - K Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, 3137 LSB, Provo, UT 84602
| | - Ryan P McMillan
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, 1981 Kraft Dr., Blacksburg, VA 24060; Metabolic Phenotyping Core Facility, Virginia Tech, 1981 Kraft Dr., Blacksburg, VA 24060
| | - Matthew W Hulver
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, 1981 Kraft Dr., Blacksburg, VA 24060; Metabolic Phenotyping Core Facility, Virginia Tech, 1981 Kraft Dr., Blacksburg, VA 24060
| | - David A Brown
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, 1981 Kraft Dr., Blacksburg, VA 24060; Metabolic Phenotyping Core Facility, Virginia Tech, 1981 Kraft Dr., Blacksburg, VA 24060; Virginia Tech Center for Drug Discovery, 800 West Campus Dr. Room 3111, Blacksburg, VA 24061
| | - Jeffery S Tessem
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, S243 ESC, Provo, UT 84602
| | - Andrew P Neilson
- Department of Food Science and Technology, Virginia Tech, 1981 Kraft Dr., Blacksburg, VA 24060.
| |
Collapse
|
12
|
Combined Therapy with SS31 and Mitochondria Mitigates Myocardial Ischemia-Reperfusion Injury in Rats. Int J Mol Sci 2018; 19:ijms19092782. [PMID: 30223594 PMCID: PMC6164143 DOI: 10.3390/ijms19092782] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/09/2018] [Accepted: 09/13/2018] [Indexed: 02/04/2023] Open
Abstract
Myocardial ischemia-reperfusion (IR) injury contributes to adverse cardiac outcomes after myocardial ischemia, cardiac surgery, or circulatory arrest. In this study, we evaluated the ability of combined SS31-mitochondria (Mito) therapy to protect heart cells from myocardial IR injury. Adult male SD rats (n = 8/each group) were randomized: group 1 (sham-operated control), group 2 (IR, 30-min ischemia/72 h reperfusion), group 3 (IR-SS31 (2 mg intra-peritoneal injection at 30 min/24 h/48 h after IR)), group 4 (IR-mitochondria (2 mg/derived from donor liver/intra-venous administration/30 min after IR procedure)), and group 5 (IR-SS31-mitochondria). In H9C2 cells, SS31 suppressed menadione-induced oxidative-stress markers (NOX-1, NOX-2, oxidized protein) while it increased SIRT1/SIRT3 expression and ATP levels. In adult male rats 72 h after IR, left ventricular ejection fraction (LVEF) was highest in sham-operated control animals and lowest in the IR group. LVEF was also higher in IR rats treated with SS31-Mito than untreated IR rats or those treated with Mito or SS31 alone. Areas of fibrosis/collagen-deposition showed the opposite pattern. Likewise, levels of oxidative-stress markers (NOX-1, NOX-2, oxidized protein), inflammatory markers (MMP-9, CD11, IL-1β, TNF-α), apoptotic markers (mitochondrial-Bax, cleaved-caspase-3, PARP), fibrosis markers (p-Smad3, TGF-β), DNA-damage (γ-H2AX), sarcomere-length, and pressure/volume overload markers (BNP, β-MHC) all showed a pattern opposite that of LVEF. Conversely, anti-apoptotic (BMP-2, Smad1/5) and energy integrity (PGC-1α/mitochondrial cytochrome-C) markers exhibited a pattern identical to that of LVEF. This study demonstrates that the combined SS31-Mito therapy is superior to either therapy alone for protecting myocardium from IR injury and indicates that the responsible mechanisms involved increased SIRT1/SIRT3 expression, which suppresses inflammation and oxidative stress and protects mitochondrial integrity.
Collapse
|
13
|
Abstract
The no-reflow phenomenon refers to the observation that when an organ is made ischemic by occlusion of a large artery supplying it, restoration of patency in that artery does not restore perfusion to the microvasculature supplying the parenchyma of that organ. This has been observed after prolonged arterial occlusions in the heart (30–90 min), brain, skin, and kidney. In experimental models, zones of no reflow in the heart are characterized by ultrastructural microvascular damage, including focal endothelial swelling obstructing the lumen of small vessels. Blood elements such as neutrophil plugs, platelets, and stacking of erythrocytes have also been implicated. No reflow is associated with poor healing of the myocardial infarction. In patients, no reflow is associated with a poor clinical outcome independent of infarct size, suggesting that therapy for no reflow may be an important approach to improving outcome for ST elevation myocardial infarction. No reflow occurs after reperfusion of experimental cerebral ischemia and may be observed after only 5-min episodes of ischemia. Aggregation of blood elements may play a greater role than in cardiac no reflow. No reflow in the brain may involve cortical spreading depression with disturbed local vascular control and high, vasculotonic levels of extracellular K+ concentration, postischemic swelling in endothelial cells and abutting end feet of pericytes, pericyte contraction and death, interstitial edema with collapse of cerebral capillaries, and inflammatory reaction. New guidelines suggesting that reperfusion for stroke may be considered as late as 24 h after the onset of symptoms suggest that clinicians may be seeing more no reflow in the future.
Collapse
Affiliation(s)
- Robert A. Kloner
- Huntington Medical Research Institutes, Pasadena, California
- Cardiovascular Division, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Kevin S. King
- Huntington Medical Research Institutes, Pasadena, California
| | - Michael G. Harrington
- Huntington Medical Research Institutes, Pasadena, California
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
14
|
Kloner RA, Dai W, Hale SL. No-Reflow Phenomenon. A New Target for Therapy of Acute Myocardial Infarction Independent of Myocardial Infarct Size. J Cardiovasc Pharmacol Ther 2018; 23:273-276. [DOI: 10.1177/1074248418764467] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Robert A. Kloner
- Cardiovascular Research Institute, Huntington Medical Research Institutes, Pasadena, CA, USA
- Department of Medicine, Keck School of Medicine at University of Southern California, Los Angeles, CA, USA
| | - Wangde Dai
- Cardiovascular Research Institute, Huntington Medical Research Institutes, Pasadena, CA, USA
- Department of Medicine, Keck School of Medicine at University of Southern California, Los Angeles, CA, USA
| | - Sharon L. Hale
- Cardiovascular Research Institute, Huntington Medical Research Institutes, Pasadena, CA, USA
| |
Collapse
|
15
|
Zhang J, Culp ML, Craver JG, Darley-Usmar V. Mitochondrial function and autophagy: integrating proteotoxic, redox, and metabolic stress in Parkinson's disease. J Neurochem 2018; 144:691-709. [PMID: 29341130 PMCID: PMC5897151 DOI: 10.1111/jnc.14308] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/04/2018] [Accepted: 01/09/2018] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is a movement disorder with widespread neurodegeneration in the brain. Significant oxidative, reductive, metabolic, and proteotoxic alterations have been observed in PD postmortem brains. The alterations of mitochondrial function resulting in decreased bioenergetic health is important and needs to be further examined to help develop biomarkers for PD severity and prognosis. It is now becoming clear that multiple hits on metabolic and signaling pathways are likely to exacerbate PD pathogenesis. Indeed, data obtained from genetic and genome association studies have implicated interactive contributions of genes controlling protein quality control and metabolism. For example, loss of key proteins that are responsible for clearance of dysfunctional mitochondria through a process called mitophagy has been found to cause PD, and a significant proportion of genes associated with PD encode proteins involved in the autophagy-lysosomal pathway. In this review, we highlight the evidence for the targeting of mitochondria by proteotoxic, redox and metabolic stress, and the role autophagic surveillance in maintenance of mitochondrial quality. Furthermore, we summarize the role of α-synuclein, leucine-rich repeat kinase 2, and tau in modulating mitochondrial function and autophagy. Among the stressors that can overwhelm the mitochondrial quality control mechanisms, we will discuss 4-hydroxynonenal and nitric oxide. The impact of autophagy is context depend and as such can have both beneficial and detrimental effects. Furthermore, we highlight the potential of targeting mitochondria and autophagic function as an integrated therapeutic strategy and the emerging contribution of the microbiome to PD susceptibility.
Collapse
Affiliation(s)
- Jianhua Zhang
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
- Department of Veterans Affairs, Birmingham VA Medical Center
| | - M Lillian Culp
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
| | - Jason G Craver
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
| | - Victor Darley-Usmar
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
| |
Collapse
|
16
|
Kloner RA, Brown DA, Csete M, Dai W, Downey JM, Gottlieb RA, Hale SL, Shi J. New and revisited approaches to preserving the reperfused myocardium. Nat Rev Cardiol 2017; 14:679-693. [PMID: 28748958 PMCID: PMC5991096 DOI: 10.1038/nrcardio.2017.102] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Early coronary artery reperfusion improves outcomes for patients with ST-segment elevation myocardial infarction (STEMI), but morbidity and mortality after STEMI remain unacceptably high. The primary deficits seen in these patients include inadequate pump function, owing to rapid infarction of muscle in the first few hours of treatment, and adverse remodelling of the heart in the months that follow. Given that attempts to further reduce myocardial infarct size beyond early reperfusion in clinical trials have so far been disappointing, effective therapies are still needed to protect the reperfused myocardium. In this Review, we discuss several approaches to preserving the reperfused heart, such as therapies that target the mechanisms involved in mitochondrial bioenergetics, pyroptosis, and autophagy, as well as treatments that harness the cardioprotective properties of inhaled anaesthetic agents. We also discuss potential therapies focused on correcting the no-reflow phenomenon and its effect on healing and adverse left ventricular remodelling.
Collapse
Affiliation(s)
- Robert A Kloner
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Division of Cardiovascular Medicine and Department of Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, California 90033, USA
| | - David A Brown
- Department of Human Nutrition, Foods, and Exercise, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
- Virginia Tech Center for Drug Discovery, Virginia Tech, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
- Virginia Tech Metabolic Phenotyping Core, Virginia Tech, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
| | - Marie Csete
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Department of Anesthesiology, Keck School of Medicine, University of Southern California, Los Angeles, California 90017, USA
| | - Wangde Dai
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Division of Cardiovascular Medicine and Department of Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, California 90033, USA
| | - James M Downey
- Department of Physiology and Cell Biology, University of South Alabama, 5851 USA Drive North, Mobile, Alabama 36688, USA
| | - Roberta A Gottlieb
- Department of Medicine, Barbra Streisand Women's Heart Center, Heart Institute of Cedars-Sinai, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, California 90048, USA
| | - Sharon L Hale
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
| | - Jianru Shi
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Division of Cardiovascular Medicine and Department of Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, California 90033, USA
| |
Collapse
|
17
|
Andrienko TN, Pasdois P, Pereira GC, Ovens MJ, Halestrap AP. The role of succinate and ROS in reperfusion injury - A critical appraisal. J Mol Cell Cardiol 2017; 110:1-14. [PMID: 28689004 PMCID: PMC5678286 DOI: 10.1016/j.yjmcc.2017.06.016] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/14/2017] [Accepted: 06/30/2017] [Indexed: 12/20/2022]
Abstract
We critically assess the proposal that succinate-fuelled reverse electron flow (REF) drives mitochondrial matrix superoxide production from Complex I early in reperfusion, thus acting as a key mediator of ischemia/reperfusion (IR) injury. Real-time surface fluorescence measurements of NAD(P)H and flavoprotein redox state suggest that conditions are unfavourable for REF during early reperfusion. Furthermore, rapid loss of succinate accumulated during ischemia can be explained by its efflux rather than oxidation. Moreover, succinate accumulation during ischemia is not attenuated by ischemic preconditioning (IP) despite powerful cardioprotection. In addition, measurement of intracellular reactive oxygen species (ROS) during reperfusion using surface fluorescence and mitochondrial aconitase activity detected major increases in ROS only after mitochondrial permeability transition pore (mPTP) opening was first detected. We conclude that mPTP opening is probably triggered initially by factors other than ROS, including increased mitochondrial [Ca2+]. However, IP only attenuates [Ca2+] increases later in reperfusion, again after initial mPTP opening, implying that IP regulates mPTP opening through additional mechanisms. One such is mitochondria-bound hexokinase 2 (HK2) which dissociates from mitochondria during ischemia in control hearts but not those subject to IP. Indeed, there is a strong correlation between the extent of HK2 loss from mitochondria during ischemia and infarct size on subsequent reperfusion. Mechanisms linking HK2 dissociation to mPTP sensitisation remain to be fully established but several related processes have been implicated including VDAC1 oligomerisation, the stability of contact sites between the inner and outer membranes, cristae morphology, Bcl-2 family members and mitochondrial fission proteins such as Drp1.
Collapse
Affiliation(s)
- Tatyana N Andrienko
- School of Biochemistry and The Bristol Heart Institute, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Philippe Pasdois
- School of Biochemistry and The Bristol Heart Institute, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Gonçalo C Pereira
- School of Biochemistry and The Bristol Heart Institute, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Matthew J Ovens
- School of Biochemistry and The Bristol Heart Institute, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Andrew P Halestrap
- School of Biochemistry and The Bristol Heart Institute, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK.
| |
Collapse
|
18
|
Affiliation(s)
- Robert A. Kloner
- From the Cardiovascular Research Institute, Huntington Medical Research Institutes, Pasadena, CA; and Cardiovascular Division, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA
| |
Collapse
|
19
|
Powers SK. Exercise: Teaching myocytes new tricks. J Appl Physiol (1985) 2017; 123:460-472. [PMID: 28572498 DOI: 10.1152/japplphysiol.00418.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 05/26/2017] [Accepted: 05/28/2017] [Indexed: 12/31/2022] Open
Abstract
Endurance exercise training promotes numerous cellular adaptations in both cardiac myocytes and skeletal muscle fibers. For example, exercise training fosters changes in mitochondrial function due to increased mitochondrial protein expression and accelerated mitochondrial turnover. Additionally, endurance exercise training alters the abundance of numerous cytosolic and mitochondrial proteins in both cardiac and skeletal muscle myocytes, resulting in a protective phenotype in the active fibers; this exercise-induced protection of cardiac and skeletal muscle fibers is often referred to as "exercise preconditioning." As few as 3-5 consecutive days of endurance exercise training result in a preconditioned cardiac phenotype that is sheltered against ischemia-reperfusion-induced injury. Similarly, endurance exercise training results in preconditioned skeletal muscle fibers that are resistant to a variety of stresses (e.g., heat stress, exercise-induced oxidative stress, and inactivity-induced atrophy). Many studies have probed the mechanisms responsible for exercise-induced preconditioning of cardiac and skeletal muscle fibers; these studies are important, because they provide an improved understanding of the biochemical mechanisms responsible for exercise-induced preconditioning, which has the potential to lead to innovative pharmacological therapies aimed at minimizing stress-induced injury to cardiac and skeletal muscle. This review summarizes the development of exercise-induced protection of cardiac myocytes and skeletal muscle fibers and highlights the putative mechanisms responsible for exercise-induced protection in the heart and skeletal muscles.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| |
Collapse
|