1
|
Hartley B, Bassiouni W, Roczkowsky A, Fahlman R, Schulz R, Julien O. Data-Independent Acquisition Proteomics and N-Terminomics Methods Reveal Alterations in Mitochondrial Function and Metabolism in Ischemic-Reperfused Hearts. J Proteome Res 2024; 23:844-856. [PMID: 38264990 PMCID: PMC10846531 DOI: 10.1021/acs.jproteome.3c00754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 01/25/2024]
Abstract
Myocardial ischemia-reperfusion (IR) (stunning) injury triggers changes in the proteome and degradome of the heart. Here, we utilize quantitative proteomics and comprehensive degradomics to investigate the molecular mechanisms of IR injury in isolated rat hearts. The control group underwent aerobic perfusion, while the IR injury group underwent 20 min of ischemia and 30 min of reperfusion to induce a stunning injury. As MMP-2 activation has been shown to contribute to myocardial injury, hearts also underwent IR injury with ARP-100, an MMP-2-preferring inhibitor, to dissect the contribution of MMP-2 to IR injury. Using data-independent acquisition (DIA) and mass spectroscopy, we quantified 4468 proteins in ventricular extracts, whereby 447 proteins showed significant alterations among the three groups. We then used subtiligase-mediated N-terminomic labeling to identify more than a hundred specific cleavage sites. Among these protease substrates, 15 were identified following IR injury. We identified alterations in numerous proteins involved in mitochondrial function and metabolism following IR injury. Our findings provide valuable insights into the biochemical mechanisms of myocardial IR injury, suggesting alterations in reactive oxygen/nitrogen species handling and generation, fatty acid metabolism, mitochondrial function and metabolism, and cardiomyocyte contraction.
Collapse
Affiliation(s)
- Bridgette Hartley
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Wesam Bassiouni
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Andrej Roczkowsky
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Richard Fahlman
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Richard Schulz
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
- Department
of Pediatrics, University of Alberta, Edmonton T6G 2S2, Canada
| | - Olivier Julien
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| |
Collapse
|
2
|
Bhullar SK, Dhalla NS. Status of Mitochondrial Oxidative Phosphorylation during the Development of Heart Failure. Antioxidants (Basel) 2023; 12:1941. [PMID: 38001794 PMCID: PMC10669359 DOI: 10.3390/antiox12111941] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Mitochondria are specialized organelles, which serve as the "Power House" to generate energy for maintaining heart function. These organelles contain various enzymes for the oxidation of different substrates as well as the electron transport chain in the form of Complexes I to V for producing ATP through the process of oxidative phosphorylation (OXPHOS). Several studies have shown depressed OXPHOS activity due to defects in one or more components of the substrate oxidation and electron transport systems which leads to the depletion of myocardial high-energy phosphates (both creatine phosphate and ATP). Such changes in the mitochondria appear to be due to the development of oxidative stress, inflammation, and Ca2+-handling abnormalities in the failing heart. Although some investigations have failed to detect any changes in the OXPHOS activity in the failing heart, such results appear to be due to a loss of Ca2+ during the mitochondrial isolation procedure. There is ample evidence to suggest that mitochondrial Ca2+-overload occurs, which is associated with impaired mitochondrial OXPHOS activity in the failing heart. The depression in mitochondrial OXPHOS activity may also be due to the increased level of reactive oxygen species, which are formed as a consequence of defects in the electron transport complexes in the failing heart. Various metabolic interventions which promote the generation of ATP have been reported to be beneficial for the therapy of heart failure. Accordingly, it is suggested that depression in mitochondrial OXPHOS activity plays an important role in the development of heart failure.
Collapse
Affiliation(s)
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada;
| |
Collapse
|
3
|
Li Z, Wang H, Zoungrana LI, James A, Slotabec L, Didik S, Fatmi MK, Krause-Hauch M, Lesnefsky EJ, Li J. Administration of metformin rescues age-related vulnerability to ischemic insults through mitochondrial energy metabolism. Biochem Biophys Res Commun 2023; 659:46-53. [PMID: 37031594 PMCID: PMC10190118 DOI: 10.1016/j.bbrc.2023.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Ischemic heart disease (IHD) is the leading cause of death on a global scale. Despite significant advances in the reperfusion treatment of acute myocardial infarction, there is still a significant early mortality rate among the elderly, as angioplasty-achieved reperfusion can exacerbate myocardial damage, leading to severe ischemia/reperfusion (I/R) injury and induce fatal arrhythmias. Mitochondria are a key mediator of ischemic insults; a transient blockade of the electron transport chain (ETC) at complex I during reperfusion can reduce myocardial infarct caused by ischemic insults. The reversible, transient modulation of complex I during early reperfusion is limited by the available of clinically tractable agents. We employed the novel use of acute, high dose metformin to modulate complex I activity during early reperfusion to decrease cardiac injury in the high-risk aged heart. Young (3-6 months) and aged (22-24 months) male and female C57BL/6 J mice were subjected to in vivo regional ischemia for 45 min, followed by metformin (2 mM, i. v.) injection 5 min prior to reperfusion for 24 h. The cardiac functions were measured with echocardiography. A Seahorse XF24 Analyzer was used to ascertain mitochondrial function. Cardiomyocyte sarcomere shortening and calcium transients were measured using the IonOptix Calcium and Contractility System. The results demonstrated that administration of acute, high dose metformin at the onset of reperfusion significantly limited cardiac damage and rescued cardiac dysfunction caused by I/R in both young and aged mice. Importantly, metformin treatment improves contractile functions of isolated cardiomyocytes and maintains mitochondrial integrity under I/R stress conditions. Thus, acute metformin administration at the onset of reperfusion has potential as a mitochondrial-based therapeutic to mitigate reperfusion injury and reduce infarct size in the elderly heart attack patient who remains at greater mortality risk despite reperfusion alone.
Collapse
Affiliation(s)
- Zehui Li
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Hao Wang
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Linda Ines Zoungrana
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Adewale James
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Lily Slotabec
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Steven Didik
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Mohammad Kasim Fatmi
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Meredith Krause-Hauch
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Edward J Lesnefsky
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Cardiology Section, Medical Service, Richmond Department of Veterans Affairs Medical Center, Richmond, VA, USA
| | - Ji Li
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; James A. Haley Veterans' Hospital, Tampa, FL, USA.
| |
Collapse
|
4
|
Ni C, Pan K, Xu J, Long X, Lin F, Nie Y, Yang Y, Yu J. Effects and mechanism of perinatal nonylphenol exposure on cardiac function and myocardial mitochondria in neonatal rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 258:114977. [PMID: 37146387 DOI: 10.1016/j.ecoenv.2023.114977] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 04/18/2023] [Accepted: 05/01/2023] [Indexed: 05/07/2023]
Abstract
BACKGROUND Nonylphenol (NP) is a common environmental endocrine disruptor that is associated with the development of cardiovascular disease. However, the toxic effect of NP on mitochondria in the heart of offspring to exposed individuals remains exclusive. OBJECTIVE To investigate whether perinatal NP exposure causes mitochondrial damage in the hearts of offspring of exposed individuals and determine its mechanism of action through both animal and cell experiments. METHODS AND RESULTS For the in vivo experiment, pregnant rats were randomly divided into four groups: the control group (corn oil, C), low dose group (2.5 mg/kg/day, L-NP group), medium dose group (50 mg/kg/day, M-NP group), and high dose group (100 mg/kg/day, H-NP group), with 12 rats in each group. The NP concentration in the hearts of offspring at PND21 and PND90 increased with the increase of the NP dose. Perinatal NP exposure induced a gradual increase in systolic blood pressure in offspring at PND90. In the H-NP group, there was a high degree of inflammatory cell infiltration, myofibril breaks, inconspicuous or absent nuclei, and pink collagen deposition. At PND90, the membrane integrity of mitochondria in the H-NP group was disrupted, the cristae disorder was aggravated, and there was internal lysis with vacuolation. Compared to the control group, the mitochondrial membrane potential of offspring at PND21 and PND90 was decreased in each of the NP exposure groups. NP exposure decreased the activity of mitochondrial respiratory enzyme complex I (CI) and increased the activity of mitochondrial respiratory enzyme complex IV (CIV) in the offspring. At PND21 and PND90, the mRNA and protein expression levels of cardiac mitochondrial PGC-1α, NRF-1, and TFAM decreased with increasing NP dose in a dose-dependent manner. In the in vitro experiment, H9C2 cells were divided into the following four groups: the blank group, RSV group (15 μg/ml), RSV + NP group (15 μg/ml RSV + 120 mmol/L NP), and NP group (120 mmol/L). With increasing NP concentration, the cell survival rate gradually decreased. Compared to the control, the membrane potential was significantly decreased in the NP group; the protein expression levels of SIRT1, PGC-1α, NRF-1, and TFAM in the NP group were significantly lower. CONCLUSION Perinatal NP exposure caused mitochondrial damage and dysfunction in the offspring of exposed individuals in a dose-dependent manner. This toxic effect may be related to NP-induced mitochondrial pathology in the offspring and the inhibition of both gene and protein expression involved in the PGC-1α/NRF-1/TFAM mitochondrial biogenesis signaling pathway following NP exposure.
Collapse
Affiliation(s)
- Chengyu Ni
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China; Department of Medicine, Hubei College of Chinese Medicine, Jingzhou, Hubei 434020, PR China
| | - Kai Pan
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Jie Xu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Xianping Long
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Guizhou, PR China
| | - FangMei Lin
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Yanling Nie
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Yu Yang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Jie Yu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China.
| |
Collapse
|
5
|
Machado SE, Spangler D, Stacks DA, Darley-Usmar V, Benavides GA, Xie M, Balla J, Zarjou A. Counteraction of Myocardial Ferritin Heavy Chain Deficiency by Heme Oxygenase-1. Int J Mol Sci 2022; 23:8300. [PMID: 35955444 PMCID: PMC9368247 DOI: 10.3390/ijms23158300] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/12/2022] [Accepted: 07/25/2022] [Indexed: 12/13/2022] Open
Abstract
Given the abundance of heme proteins (cytochromes) in the mitochondrion, it is evident that a meticulously orchestrated iron metabolism is essential for cardiac health. Here, we examined the functional significance of myocardial ferritin heavy chain (FtH) in a model of acute myocardial infarction. We report that FtH deletion did not alter either the mitochondrial regulatory and surveillance pathways (fission and fusion) or mitochondrial bioenergetics in response to injury. Furthermore, deletion of myocardial FtH did not affect cardiac function, assessed by measurement of left ventricular ejection fraction, on days 1, 7, and 21 post injury. To identify the modulated pathways providing cardiomyocyte protection coincident with FtH deletion, we performed unbiased transcriptomic analysis. We found that following injury, FtH deletion was associated with upregulation of several genes with anti-ferroptotic properties, including heme oxygenase-1 (HO-1) and the cystine/glutamate anti-porter (Slc7a11). These results suggested that HO-1 overexpression mitigates ferroptosis via upregulation of Slc7a11. Indeed, using transgenic mice with HO-1 overexpression, we demonstrate that overexpressed HO-1 is coupled with increased Slc7a11 expression. In conclusion, we demonstrate that following injury, myocardial FtH deletion leads to a compensatory upregulation in a number of anti-ferroptotic genes, including HO-1. Such HO-1 induction leads to overexpression of Slc7a11 and protects the heart against ischemia-reperfusion-mediated ferroptosis, preserves mitochondrial function, and overall function of the myocardium.
Collapse
Affiliation(s)
- Sarah E. Machado
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| | - Daryll Spangler
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| | - Delores A. Stacks
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| | - Victor Darley-Usmar
- Mitochondrial Medicine Laboratory, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (V.D.-U.); (G.A.B.)
| | - Gloria A. Benavides
- Mitochondrial Medicine Laboratory, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (V.D.-U.); (G.A.B.)
| | - Min Xie
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - József Balla
- ELKH-UD Vascular Pathophysiology Research Group 11003, Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Abolfazl Zarjou
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| |
Collapse
|
6
|
Hypothermia Prevents Cardiac Dysfunction during Acute Ischemia Reperfusion by Maintaining Mitochondrial Bioenergetics and by Promoting Hexokinase II Binding to Mitochondria. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4476448. [PMID: 35873800 PMCID: PMC9301761 DOI: 10.1155/2022/4476448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/04/2022] [Accepted: 06/14/2022] [Indexed: 11/18/2022]
Abstract
Background Hypothermia (H), cardioplegia (CP), and both combined (HCP) are known to be protective against myocardial ischemia reperfusion (IR) injury. Mitochondria have molecular signaling mechanisms that are associated with both cell survival and cell death. In this study, we investigated the dynamic changes in proapoptotic and prosurvival signaling pathways mediating H, CP, or HCP-induced protection of mitochondrial function after acute myocardial IR injury. Methods Rats were divided into five groups. Each group consists of 3 subgroups based on a specific reperfusion time (5, 20, or 60 min) after a 25-min global ischemia. The time control (TC) groups were not subjected to IR but were perfused with 37 °C Krebs-Ringer's (KR) buffer, containing 4.5 mM K+, in a specific perfusion protocol that corresponded with the duration of each IR protocol. The IR group (control) was perfused for 20 min with KR, followed by 25-min global ischemia, and then KR reperfusion for 5, 20, or 60 min. The treatment groups were exposed to 17 °C H, 37 °C CP (16 mM K+), or HCP (17 °C + CP) for 5 min before ischemia and for 2 min on reperfusion before switching to 37 °C KR perfusion for the remainder of each of the reperfusion times. Cardiac function and mitochondrial redox state (NADH/FAD) were monitored online in the ex vivo hearts before, during, and after ischemia. Mitochondria were isolated at the end of each specified reperfusion time, and changes in O2 consumption, membrane potential (ΔΨm), and Ca2+ retention capacity (CRC) were assessed using complex I and complex II substrates. In another set of hearts, mitochondrial and cytosolic fractions were isolated after a specified reperfusion time to conduct western blot assays to determine hexokinase II (HKII) and Bax binding/translocation to mitochondria, cytosolic pAkt levels, and cytochrome c (Cyto-c) release into the cytosol. Results H and HCP were more protective of mitochondrial integrity and, concomitantly, cardiac function than CP alone; H and HCP improved post-ischemic cardiac function by (1) maintaining mitochondrial bioenergetics, (2) maintaining HKII binding to mitochondria with an increase in pAkt levels, (3) increasing CRC, and (4) decreasing Cyto-c release during reperfusion. Bax translocation/binding to mitochondria was unaffected by any treatment, regardless of cardiac functional recovery. Conclusions Hypothermia preserved mitochondrial function and cardiac function, in part, by maintaining mitochondrial bioenergetics, by retaining HKII binding to mitochondria via upstream pAkt, and by reducing Cyto-c release independently of Bax binding to mitochondria.
Collapse
|
7
|
Role of ranolazine in heart failure: From cellular to clinic perspective. Eur J Pharmacol 2022; 919:174787. [PMID: 35114190 DOI: 10.1016/j.ejphar.2022.174787] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/25/2021] [Accepted: 01/25/2022] [Indexed: 12/17/2022]
Abstract
Ranolazine was approved by the US Food and Drug Administration as an antianginal drug in 2006, and has been used since in certain groups of patients with stable angina. The therapeutic action of ranolazine was initially attributed to inhibitory effects on fatty acids metabolism. As investigations went on, however, it developed that the main beneficial effects of ranolazine arise from its action on the late sodium current in the heart. Since late sodium currents were discovered to be involved in various heart pathologies such as ischemia, arrhythmias, systolic and diastolic dysfunctions, and all these conditions are associated with heart failure, ranolazine has in some way been tested either directly or indirectly on heart failure in numerous experimental and clinical studies. As the heart continuously remodels following any sort of severe injury, the inhibition by ranolazine of the underlying mechanisms of cardiac remodeling including ion disturbances, oxidative stress, inflammation, apoptosis, fibrosis, metabolic dysregulation, and neurohormonal impairment are discussed, along with unresolved issues. A projection of pathologies targeted by ranolazine from cellular level to clinical is provided in this review.
Collapse
|
8
|
Wu C, Zhang Z, Zhang W, Liu X. Mitochondrial dysfunction and mitochondrial therapies in heart failure. Pharmacol Res 2021; 175:106038. [PMID: 34929300 DOI: 10.1016/j.phrs.2021.106038] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/15/2021] [Accepted: 12/15/2021] [Indexed: 12/18/2022]
Abstract
Cardiovascular diseases remain the leading cause of death worldwide in the last decade, accompanied by immense health and economic burdens. Heart failure (HF), as the terminal stage of many cardiovascular diseases, is a common, intractable, and costly medical condition. Despite significant improvements in pharmacologic and device therapies over the years, life expectancy for this disease remains poor. Current therapies have not reversed the trends in morbidity and mortality as expected. Thus, there is an urgent need for novel potential therapeutic agents. Although the pathophysiology of the failing heart is extraordinarily complex, targeting mitochondrial dysfunction can be an effective approach for potential treatment. Increasing evidence has shown that mitochondrial abnormalities, including altered metabolic substrate utilization, impaired mitochondrial oxidative phosphorylation (OXPHOS), increased reactive oxygen species (ROS) formation, and aberrant mitochondrial dynamics, are closely related to HF. Here, we reviewed the findings on the role of mitochondrial dysfunction in HF, along with novel mitochondrial therapeutics and their pharmacological effects.
Collapse
Affiliation(s)
- Chennan Wu
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Zhen Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Weidong Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Xia Liu
- School of Pharmacy, Second Military Medical University, Shanghai, China.
| |
Collapse
|
9
|
Eskaf J, Cleveland WJ, Riess ML. No Direct Postconditioning Effect of Poloxamer 188 on Mitochondrial Function after Ischemia Reperfusion Injury in Rat Isolated Hearts. Int J Mol Sci 2021; 22:4879. [PMID: 34063028 PMCID: PMC8124240 DOI: 10.3390/ijms22094879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 11/17/2022] Open
Abstract
Myocardial infarction is a leading cause for morbidity and mortality worldwide. The only viable treatment for the ischemic insult is timely reperfusion, which further exacerbates myocardial injury. Maintaining mitochondrial function is crucial in preserving cardiomyocyte function in ischemia reperfusion (IR) injury. Poloxamer (P) 188 has been shown to improve cardiac IR injury by improving cellular and mitochondrial function. The aim of this study was to show if P188 postconditioning has direct protective effects on mitochondrial function in the heart. Langendorff prepared rat hearts were subjected to IR injury ex-vivo and reperfused for 10 min with 1 mM P188 vs. vehicle. Cardiac mitochondria were isolated with 1 mM P188 vs. 1 mM polyethylene glycol (PEG) vs. vehicle by differential centrifugation. Mitochondrial function was assessed by adenosine triphosphate synthesis, oxygen consumption, and calcium retention capacity. Mitochondrial function decreased significantly after ischemia and showed mild improvement with reperfusion. P188 did not improve mitochondrial function in the ex-vivo heart, and neither further P188 nor PEG induced direct mitochondrial protection after IR injury in this model.
Collapse
Affiliation(s)
- Josephine Eskaf
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (J.E.); (W.J.C.)
- Department of Anesthesiology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - William J. Cleveland
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (J.E.); (W.J.C.)
| | - Matthias L. Riess
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (J.E.); (W.J.C.)
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Anesthesiology, TVHS VA Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
10
|
Mitochondrial respiratory supercomplexes in mammalian cells: structural versus functional role. J Mol Med (Berl) 2020; 99:57-73. [PMID: 33201259 DOI: 10.1007/s00109-020-02004-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/06/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023]
Abstract
Mitochondria are recognized as the main source of ATP to meet the energy demands of the cell. ATP production occurs by oxidative phosphorylation when electrons are transported through the electron transport chain (ETC) complexes and develop the proton motive force across the inner mitochondrial membrane that is used for ATP synthesis. Studies since the 1960s have been concentrated on the two models of structural organization of ETC complexes known as "solid-state" and "fluid-state" models. However, advanced new techniques such as blue-native gel electrophoresis, mass spectroscopy, and cryogenic electron microscopy for analysis of macromolecular protein complexes provided new data in favor of the solid-state model. According to this model, individual ETC complexes are assembled into macromolecular structures known as respiratory supercomplexes (SCs). A large number of studies over the last 20 years proposed the potential role of SCs to facilitate substrate channeling, maintain the integrity of individual ETC complexes, reduce electron leakage and production of reactive oxygen species, and prevent excessive and random aggregation of proteins in the inner mitochondrial membrane. However, many other studies have challenged the proposed functional role of SCs. Recently, a third model known as the "plasticity" model was proposed that partly reconciles both "solid-state" and "fluid-state" models. According to the "plasticity" model, respiratory SCs can co-exist with the individual ETC complexes. To date, the physiological role of SCs remains unknown, although several studies using tissue samples of patients or animal/cell models of human diseases revealed an associative link between functional changes and the disintegration of SC assembly. This review summarizes and discusses previous studies on the mechanisms and regulation of SC assembly under physiological and pathological conditions.
Collapse
|
11
|
Metabolic Alterations Caused by Defective Cardiolipin Remodeling in Inherited Cardiomyopathies. Life (Basel) 2020; 10:life10110277. [PMID: 33187128 PMCID: PMC7697959 DOI: 10.3390/life10110277] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/21/2022] Open
Abstract
The heart is the most energy-consuming organ in the human body. In heart failure, the homeostasis of energy supply and demand is endangered by an increase in cardiomyocyte workload, or by an insufficiency in energy-providing processes. Energy metabolism is directly associated with mitochondrial redox homeostasis. The production of toxic reactive oxygen species (ROS) may overwhelm mitochondrial and cellular ROS defense mechanisms in case of heart failure. Mitochondria are essential cell organelles and provide 95% of the required energy in the heart. Metabolic remodeling, changes in mitochondrial structure or function, and alterations in mitochondrial calcium signaling diminish mitochondrial energy provision in many forms of cardiomyopathy. The mitochondrial respiratory chain creates a proton gradient across the inner mitochondrial membrane, which couples respiration with oxidative phosphorylation and the preservation of energy in the chemical bonds of ATP. Akin to other mitochondrial enzymes, the respiratory chain is integrated into the inner mitochondrial membrane. The tight association with the mitochondrial phospholipid cardiolipin (CL) ensures its structural integrity and coordinates enzymatic activity. This review focuses on how changes in mitochondrial CL may be associated with heart failure. Dysfunctional CL has been found in diabetic cardiomyopathy, ischemia reperfusion injury and the aging heart. Barth syndrome (BTHS) is caused by an inherited defect in the biosynthesis of cardiolipin. Moreover, a dysfunctional CL pool causes other types of rare inherited cardiomyopathies, such as Sengers syndrome and Dilated Cardiomyopathy with Ataxia (DCMA). Here we review the impact of cardiolipin deficiency on mitochondrial functions in cellular and animal models. We describe the molecular mechanisms concerning mitochondrial dysfunction as an incitement of cardiomyopathy and discuss potential therapeutic strategies.
Collapse
|
12
|
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
|
13
|
Elkholy SE, Elaidy SM, El-Sherbeeny NA, Toraih EA, El-Gawly HW. Neuroprotective effects of ranolazine versus pioglitazone in experimental diabetic neuropathy: Targeting Nav1.7 channels and PPAR-γ. Life Sci 2020; 250:117557. [PMID: 32184124 DOI: 10.1016/j.lfs.2020.117557] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/22/2020] [Accepted: 03/13/2020] [Indexed: 12/11/2022]
Abstract
Diabetic neuropathy (DN) is a common complication of diabetes mellitus (DM). Pathophysiology of DN includes inflammation and changes in expression and function of voltage-gated sodium channels (Nav) in peripheral nerves; and central reduction of Peroxisome Proliferator Activated Receptor-Gamma (PPAR-γ) expression. AIM This study explored the effect of ranolazine (RN) versus pioglitazone (PIO) in DN induced in rats. The role of sciatic interleukin (IL)-1β, tumor necrosis factor-alpha (TNF)-α, Nav1.7, and spinal PPAR-γ expressions were determined. MATERIALS AND METHODS For induction of Type-2 DM, 40 high fat diet-fed rats were challenged by a single dose of intraperitoneal streptozotocin (30 mg/kg). One week later, oral PIO (10 mg/kg; once daily) or RN (20, 50 and 100 mg/kg; twice daily) were administered for six weeks. Weekly body weight and fasting blood sugar (FBS) were measured. Rats were tested for thermal hyperalgesia and mechanical allodynia. At the end of the experiment, sciatic nerves homogenates were examined for TNF-α and IL-1B levels, and Nav1.7 channel expression. Segments of spinal cords were investigated for the PPAR-γ gene expression. Evaluation of histopathology of sciatic nerves and spinal cords were done. KEY FINDINGS In diabetic rats, PIO and RN individually improved evoked-pain behaviors, reduced sciatic TNF-α and 1L-1B levels; downregulated expressional levels of Nav1.7 channels; and increased the spinal PPAR-γ gene expression. RN in the dose of 100 mg/kg/day showed the most advantageous effects. SIGNIFICANCE RN has neuroprotective effects in Type-2 diabetes-induced DN. Further studies of combined RN-PIO treatment are recommended, especially in diabetic patients with cardiovascular co-morbidity.
Collapse
Affiliation(s)
- Shereen E Elkholy
- Department of Clinical Pharmacology, Faculty of Medicine, Port-Said University, Port-Said, Egypt
| | - Samah M Elaidy
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Nagla A El-Sherbeeny
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Eman A Toraih
- Department of Surgery, Tulane University, School of Medicine, New Orleans, LA, USA; Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt; Center of Excellence of Molecular and Cellular Medicine, Suez Canal University, Ismailia, Egypt
| | - Hoda W El-Gawly
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
14
|
Mitochondrial ROS in myocardial ischemia reperfusion and remodeling. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165768. [PMID: 32173461 DOI: 10.1016/j.bbadis.2020.165768] [Citation(s) in RCA: 252] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022]
Abstract
Despite major progress in interventional and medical treatments, myocardial infarction (MI) and subsequent development of heart failure (HF) are still associated with high mortality. Both during ischemia reperfusion (IR) in the acute setting of MI, as well as in the chronic remodeling process following MI, oxidative stress substantially contributes to cardiac damage. Reactive oxygen species (ROS) generated within mitochondria are particular drivers of mechanisms contributing to IR injury, including induction of mitochondrial permeability transition or oxidative damage of intramitochondrial structures and molecules. But even beyond the acute setting, mechanisms like inflammatory signaling, extracellular remodeling, or pro-apoptotic signaling that contribute to post-infarction remodeling are regulated by mitochondrial ROS. In the current review, we discuss both sources and consequences of mitochondrial ROS during IR and in the chronic setting following MI, thereby emphasizing the potential therapeutic value of attenuating mitochondrial ROS to improve outcome and prognosis for patients suffering MI.
Collapse
|
15
|
Kura B, Szeiffova Bacova B, Kalocayova B, Sykora M, Slezak J. Oxidative Stress-Responsive MicroRNAs in Heart Injury. Int J Mol Sci 2020; 21:E358. [PMID: 31948131 PMCID: PMC6981696 DOI: 10.3390/ijms21010358] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are important molecules in the living organisms as a part of many signaling pathways. However, if overproduced, they also play a significant role in the development of cardiovascular diseases, such as arrhythmia, cardiomyopathy, ischemia/reperfusion injury (e.g., myocardial infarction and heart transplantation), and heart failure. As a result of oxidative stress action, apoptosis, hypertrophy, and fibrosis may occur. MicroRNAs (miRNAs) represent important endogenous nucleotides that regulate many biological processes, including those involved in heart damage caused by oxidative stress. Oxidative stress can alter the expression level of many miRNAs. These changes in miRNA expression occur mainly via modulation of nuclear factor erythroid 2-related factor 2 (Nrf2), sirtuins, calcineurin/nuclear factor of activated T cell (NFAT), or nuclear factor kappa B (NF-κB) pathways. Up until now, several circulating miRNAs have been reported to be potential biomarkers of ROS-related cardiac diseases, including myocardial infarction, hypertrophy, ischemia/reperfusion, and heart failure, such as miRNA-499, miRNA-199, miRNA-21, miRNA-144, miRNA-208a, miRNA-34a, etc. On the other hand, a lot of studies are aimed at using miRNAs for therapeutic purposes. This review points to the need for studying the role of redox-sensitive miRNAs, to identify more effective biomarkers and develop better therapeutic targets for oxidative-stress-related heart diseases.
Collapse
Affiliation(s)
- Branislav Kura
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Barbara Szeiffova Bacova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Barbora Kalocayova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Matus Sykora
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Jan Slezak
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| |
Collapse
|
16
|
Ren RC, Lu X, Zhao YJ, Wei YM, Wang LL, Zhang L, Zhang WT, Zhang C, Zhang XS, Zhao XY. Pentatricopeptide repeat protein DEK40 is required for mitochondrial function and kernel development in maize. JOURNAL OF EXPERIMENTAL BOTANY 2019; 70:6163-6179. [PMID: 31598687 PMCID: PMC6859738 DOI: 10.1093/jxb/erz391] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/15/2019] [Indexed: 05/18/2023]
Abstract
Pentatricopeptide repeat (PPR) proteins are one of the largest protein families, which consists of >400 members in most species. However, the molecular functions of many PPR proteins are still uncharacterized. Here, we isolated a maize mutant, defective kernel 40 (dek40). Positional cloning, and genetic and molecular analyses revealed that DEK40 encodes a new E+ subgroup PPR protein that is localized in the mitochondrion. DEK40 recognizes and directly binds to cox3, nad2, and nad5 transcripts and functions in their processing. In the dek40 mutant, abolishment of the C-to-U editing of cox3-314, nad2-26, and nad5-1916 leads to accumulated reactive oxygen species and promoted programmed cell death in endosperm cells due to the dysfunction of mitochondrial complexes I and IV. Furthermore, RNA sequencing analysis showed that gene expression in some pathways, such as glutathione metabolism and starch biosynthesis, was altered in the dek40 mutant compared with the wild-type control, which might be involved in abnormal development of the maize mutant kernels. Thus, our results provide solid evidence on the molecular mechanism underlying RNA editing by DEK40, and extend our understanding of PPR-E+ type protein in editing functions and kernel development in maize.
Collapse
Affiliation(s)
- Ru Chang Ren
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| | - Xiaoduo Lu
- Institute of Molecular Breeding for Maize, Qilu Normal University, Jinan, China
| | - Ya Jie Zhao
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| | - Yi Ming Wei
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| | - Li Li Wang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| | - Lin Zhang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| | - Wen Ting Zhang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| | - Chunyi Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xian Sheng Zhang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
- Correspondence: or
| | - Xiang Yu Zhao
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
- Correspondence: or
| |
Collapse
|
17
|
Soares ROS, Losada DM, Jordani MC, Évora P, Castro-E-Silva O. Ischemia/Reperfusion Injury Revisited: An Overview of the Latest Pharmacological Strategies. Int J Mol Sci 2019; 20:ijms20205034. [PMID: 31614478 PMCID: PMC6834141 DOI: 10.3390/ijms20205034] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/03/2019] [Accepted: 10/08/2019] [Indexed: 02/08/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) permeates a variety of diseases and is a ubiquitous concern in every transplantation proceeding, from whole organs to modest grafts. Given its significance, efforts to evade the damaging effects of both ischemia and reperfusion are abundant in the literature and they consist of several strategies, such as applying pre-ischemic conditioning protocols, improving protection from preservation solutions, thus providing extended cold ischemia time and so on. In this review, we describe many of the latest pharmacological approaches that have been proven effective against IRI, while also revisiting well-established concepts and presenting recent pathophysiological findings in this ever-expanding field. A plethora of promising protocols has emerged in the last few years. They have been showing exciting results regarding protection against IRI by employing drugs that engage several strategies, such as modulating cell-surviving pathways, evading oxidative damage, physically protecting cell membrane integrity, and enhancing cell energetics.
Collapse
Affiliation(s)
| | - Daniele M Losada
- Department of Anatomic Pathology, Faculty of Medical Sciences, University of Campinas, 13083-970 Campinas, Brazil.
| | - Maria C Jordani
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
| | - Paulo Évora
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
- Department of Gastroenterology, São Paulo Medical School, University of São Paulo, 01246-903 São Paulo, Brazil.
| | - Orlando Castro-E-Silva
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
- Department of Gastroenterology, São Paulo Medical School, University of São Paulo, 01246-903 São Paulo, Brazil.
| |
Collapse
|
18
|
Albrecht M, Zitta K, Groenendaal F, van Bel F, Peeters-Scholte C. Neuroprotective strategies following perinatal hypoxia-ischemia: Taking aim at NOS. Free Radic Biol Med 2019; 142:123-131. [PMID: 30818057 DOI: 10.1016/j.freeradbiomed.2019.02.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/07/2019] [Accepted: 02/19/2019] [Indexed: 12/13/2022]
Abstract
Perinatal asphyxia is characterized by oxygen deprivation and lack of perfusion in the perinatal period, leading to hypoxic-ischemic encephalopathy and sequelae such as cerebral palsy, mental retardation, cerebral visual impairment, epilepsy and learning disabilities. On cellular level PA is associated with a decrease in oxygen and glucose leading to ATP depletion and a compromised mitochondrial function. Upon reoxygenation and reperfusion, the renewed availability of oxygen gives rise to not only restoration of cell function, but also to the activation of multiple detrimental biochemical pathways, leading to secondary energy failure and ultimately, cell death. The formation of reactive oxygen species, nitric oxide and peroxynitrite plays a central role in the development of subsequent neurological damage. In this review we give insight into the pathophysiology of perinatal asphyxia, discuss its clinical relevance and summarize current neuroprotective strategies related to therapeutic hypothermia, ischemic postconditioning and pharmacological interventions. The review will also focus on the possible neuroprotective actions and molecular mechanisms of the selective neuronal and inducible nitric oxide synthase inhibitor 2-iminobiotin that may represent a novel therapeutic agent for the treatment of hypoxic-ischemic encephalopathy, both in combination with therapeutic hypothermia in middle- and high-income countries, as well as stand-alone treatment in low-income countries.
Collapse
Affiliation(s)
- Martin Albrecht
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Karina Zitta
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Floris Groenendaal
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands; Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Frank van Bel
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands; Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cacha Peeters-Scholte
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands; Neurophyxia BV, 's Hertogenbosch, the Netherlands.
| |
Collapse
|
19
|
Stepanova A, Konrad C, Guerrero-Castillo S, Manfredi G, Vannucci S, Arnold S, Galkin A. Deactivation of mitochondrial complex I after hypoxia-ischemia in the immature brain. J Cereb Blood Flow Metab 2019; 39:1790-1802. [PMID: 29629602 PMCID: PMC6727140 DOI: 10.1177/0271678x18770331] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Mortality from perinatal hypoxic-ischemic (HI) brain injury reached 1.15 million worldwide in 2010 and is also a major factor for neurological disability in infants. HI directly influences the oxidative phosphorylation enzyme complexes in mitochondria, but the exact mechanism of HI-reoxygenation response in brain remains largely unresolved. After induction of HI-reoxygenation in postnatal day 10 rats, activities of mitochondrial respiratory chain enzymes were analysed and complexome profiling was performed. The effect of conformational state (active/deactive (A/D) transition) of mitochondrial complex I on H2O2 release was measured simultaneously with mitochondrial oxygen consumption. In contrast to cytochrome c oxidase and succinate dehydrogenase, HI-reoxygenation resulted in inhibition of mitochondrial complex I at 4 h after reoxygenation. Immediately after HI, we observed a robust increase in the content of deactive (D) form of complex I. The D-form is less active in reactive oxygen species (ROS) production via reversed electron transfer, indicating the key role of the deactivation of complex I in ischemia/reoxygenation. We describe a novel mechanism of mitochondrial response to ischemia in the immature brain. HI induced a deactivation of complex I in order to reduce ROS production following reoxygenation. Delayed activation of complex I represents a novel mitochondrial target for pathological-activated therapy.
Collapse
Affiliation(s)
- Anna Stepanova
- 1 School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, Belfast, UK.,2 Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Csaba Konrad
- 2 Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sergio Guerrero-Castillo
- 3 Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Giovanni Manfredi
- 2 Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Susan Vannucci
- 4 Department of Pediatrics/Newborn Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Susanne Arnold
- 3 Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alexander Galkin
- 1 School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, Belfast, UK.,2 Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
20
|
Li Y, Chen B, Yang X, Zhang C, Jiao Y, Li P, Liu Y, Li Z, Qiao B, Bond Lau W, Ma XL, Du J. S100a8/a9 Signaling Causes Mitochondrial Dysfunction and Cardiomyocyte Death in Response to Ischemic/Reperfusion Injury. Circulation 2019; 140:751-764. [DOI: 10.1161/circulationaha.118.039262] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background:
Myocardial ischemia-reperfusion (MI/R) injury is a significant clinical problem without effective therapy. Unbiased omics approaches may reveal key MI/R mediators to initiate MI/R injury.
Methods:
We used a dynamic transcriptome analysis of mouse heart exposed to various MI/R periods to identify S100a8/a9 as an early mediator. Using loss/gain-of-function approaches to understand the role of S100a8/a9 in MI/R injury, we explored the mechanisms through transcriptome and functional experiment. Dynamic serum S100a8/a9 levels were measured in patients with acute myocardial infarction before and after percutaneous coronary intervention. Patients were prospectively followed for the occurrence of major adverse cardiovascular events.
Results:
S100a8/a9 was identified as the most significantly upregulated gene during the early reperfusion stage. Knockout of S100a9 markedly decreased cardiomyocyte death and improved heart function, whereas hematopoietic overexpression of S100a9 exacerbated MI/R injury. Transcriptome/functional studies revealed that S100a8/a9 caused mitochondrial respiratory dysfunction in cardiomyocytes. Mechanistically, S100a8/a9 downregulated NDUF gene expression with subsequent mitochondrial complex I inhibition via Toll-like receptor 4/Erk–mediated Pparg coactivator 1 alpha/nuclear respiratory factor 1 signaling suppression. Administration of S100a9 neutralizing antibody significantly reduced MI/R injury and improved cardiac function. Finally, we demonstrated that serum S100a8/a9 levels were significantly increased 1 day after percutaneous coronary intervention in patients with acute myocardial infarction, and elevated S100a8/a9 levels were associated with the incidence of major adverse cardiovascular events.
Conclusions:
Our study identified S100a8/a9 as a master regulator causing cardiomyocyte death in the early stage of MI/R injury via the suppression of mitochondrial function. Targeting S100a8/a9-intiated signaling may represent a novel therapeutic intervention against MI/R injury.
Clinical Trial Registration:
URL:
https://www.clinicaltrials.gov
. Unique identifier: NCT03752515
Collapse
Affiliation(s)
- Yulin Li
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (Y. Li, B.C., Z.Y., C.Z., Y.J., P.L., Y. Liu, Z.L., B.Q., J.D.)
| | - Boya Chen
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (Y. Li, B.C., Z.Y., C.Z., Y.J., P.L., Y. Liu, Z.L., B.Q., J.D.)
| | - Xinying Yang
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (Y. Li, B.C., Z.Y., C.Z., Y.J., P.L., Y. Liu, Z.L., B.Q., J.D.)
| | - Congcong Zhang
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (Y. Li, B.C., Z.Y., C.Z., Y.J., P.L., Y. Liu, Z.L., B.Q., J.D.)
| | - Yao Jiao
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (Y. Li, B.C., Z.Y., C.Z., Y.J., P.L., Y. Liu, Z.L., B.Q., J.D.)
| | - Ping Li
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (Y. Li, B.C., Z.Y., C.Z., Y.J., P.L., Y. Liu, Z.L., B.Q., J.D.)
| | - Yan Liu
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (Y. Li, B.C., Z.Y., C.Z., Y.J., P.L., Y. Liu, Z.L., B.Q., J.D.)
| | - Zhenya Li
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (Y. Li, B.C., Z.Y., C.Z., Y.J., P.L., Y. Liu, Z.L., B.Q., J.D.)
| | - Bokang Qiao
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (Y. Li, B.C., Z.Y., C.Z., Y.J., P.L., Y. Liu, Z.L., B.Q., J.D.)
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-l.M.)
| | - Xin-liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-l.M.)
| | - Jie Du
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, China (Y. Li, B.C., Z.Y., C.Z., Y.J., P.L., Y. Liu, Z.L., B.Q., J.D.)
| |
Collapse
|
21
|
Chen Q, Thompson J, Hu Y, Dean J, Lesnefsky EJ. Inhibition of the ubiquitous calpains protects complex I activity and enables improved mitophagy in the heart following ischemia-reperfusion. Am J Physiol Cell Physiol 2019; 317:C910-C921. [PMID: 31411917 DOI: 10.1152/ajpcell.00190.2019] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Activation of calpain 1 (CPN1) and calpain 2 (CPN2) contributes to cardiac injury during ischemia (ISC) and reperfusion (REP). Complex I activity is decreased in heart mitochondria following ISC-REP. CPN1 and CPN2 are ubiquitous calpains that exist in both cytosol (cs)-CPN1 and 2 and mitochondria (mit)-CPN1 and 2. Recent work shows that the complex I subunit (NDUFS7) is a potential substrate of the mit-CPN1. We asked whether ISC-REP led to decreased complex I activity via proteolysis of the NDUFS7 subunit via activation of mit-CPN1 and -2. Activation of cs-CPN1 and -2 decreases mitophagy in hepatocytes following ISC-REP. We asked whether activation of cs-CPN1 and -2 impaired mitophagy in the heart following ISC-REP. Buffer-perfused rat hearts underwent 25 min of global ISC and 30 min of REP. MDL-28170 (MDL; 10 µM) was used to inhibit CPN1 and -2. Cytosol, subsarcolemmal mitochondria (SSM), and interfibrillar mitochondria (IFM) were isolated at the end of heart perfusion. Cardiac ISC-REP led to decreased complex I activity with a decrease in the content of NDUFS7 in both SSM and IFM. ISC-REP also resulted in a decrease in cytosolic beclin-1 content, a key component of the autophagy pathway required to form autophagosomes. MDL treatment protected the contents of cytosolic beclin-1 and mitochondrial NDUFS7 in hearts following ISC-REP. These results support that activation of both cytosolic and mitochondrial calpains impairs mitochondria during cardiac ISC-REP. Mitochondria-localized calpains impair complex I via cleavage of a key subunit. Activation of cytosolic calpains contributes to mitochondrial dysfunction by impairing removal of the impaired mitochondria through depletion of a key component of the mitophagy process.
Collapse
Affiliation(s)
- Qun Chen
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Jeremy Thompson
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Ying Hu
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Joseph Dean
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Edward J Lesnefsky
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia.,Department of Biochemistry and Molecular Biology Virginia Commonwealth University, Richmond, Virginia.,Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia.,McGuire Department of Veterans Affairs Medical Center, Richmond, Virginia
| |
Collapse
|
22
|
Braik A, Lahouel M, Merabet R, Djebar MR, Morin D. Myocardial protection by propolis during prolonged hypothermic preservation. Cryobiology 2019; 88:29-37. [DOI: 10.1016/j.cryobiol.2019.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 11/30/2022]
|
23
|
Ait-Aissa K, Blaszak SC, Beutner G, Tsaih SW, Morgan G, Santos JH, Flister MJ, Joyce DL, Camara AKS, Gutterman DD, Donato AJ, Porter GA, Beyer AM. Mitochondrial Oxidative Phosphorylation defect in the Heart of Subjects with Coronary Artery Disease. Sci Rep 2019; 9:7623. [PMID: 31110224 PMCID: PMC6527853 DOI: 10.1038/s41598-019-43761-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/06/2018] [Indexed: 12/21/2022] Open
Abstract
Coronary artery disease (CAD) is a leading cause of death worldwide and frequently associated with mitochondrial dysfunction. Detailed understanding of abnormalities in mitochondrial function that occur in patients with CAD is lacking. We evaluated mitochondrial damage, energy production, and mitochondrial complex activity in human non-CAD and CAD hearts. Fresh and frozen human heart tissue was used. Cell lysate or mitochondria were isolated using standard techniques. Mitochondrial DNA (mtDNA), NAD + and ATP levels, and mitochondrial oxidative phosphorylation capacity were evaluated. Proteins critical to the regulation of mitochondrial metabolism and function were also evaluated in tissue lysates. PCR analysis revealed an increase in mtDNA lesions and the frequency of mitochondrial common deletion, both established markers for impaired mitochondrial integrity in CAD compared to non-CAD patient samples. NAD+ and ATP levels were significantly decreased in CAD subjects compared to Non-CAD (NAD+ fold change: non-CAD 1.00 ± 0.17 vs. CAD 0.32 ± 0.12* and ATP fold change: non-CAD 1.00 ± 0.294 vs. CAD 0.01 ± 0.001*; N = 15, P < 0.005). We observed decreased respiration control index in CAD tissue and decreased activity of complexes I, II, and III. Expression of ETC complex subunits and respirasome formation were increased; however, elevations in the de-active form of complex I were observed in CAD. We observed a corresponding increase in glycolytic flux, indicated by a rise in pyruvate kinase and lactate dehydrogenase activity, indicating a compensatory increase in glycolysis for cellular energetics. Together, these results indicate a shift in mitochondrial metabolism from oxidative phosphorylation to glycolysis in human hearts subjects with CAD.
Collapse
Affiliation(s)
- Karima Ait-Aissa
- Cardiovascular Center, Department of Medicine, Med College of Wisconsin, Milwaukee, WI, USA.
| | - Scott C Blaszak
- Cardiovascular Center, Department of Medicine, Med College of Wisconsin, Milwaukee, WI, USA
| | - Gisela Beutner
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Shirng-Wern Tsaih
- Department of Physiology, Med College of Wisconsin, Milwaukee, WI, USA
| | - Garrett Morgan
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Janine H Santos
- Genome Integrity and Structural Biology Laboratory, NIHEHS, Raleigh-Durham, NC, USA
| | - Michael J Flister
- Department of Physiology, Med College of Wisconsin, Milwaukee, WI, USA
| | - David L Joyce
- Department of Surgery, Med College of Wisconsin, Milwaukee, WI, USA
| | - Amadou K S Camara
- Department of Physiology, Med College of Wisconsin, Milwaukee, WI, USA.,Department of Anesthesiology, Med College of Wisconsin, Milwaukee, WI, USA
| | - David D Gutterman
- Cardiovascular Center, Department of Medicine, Med College of Wisconsin, Milwaukee, WI, USA
| | - Anthony J Donato
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA.,VA Medical Center-Salt Lake City, GRECC, Salt Lake City, Utah, USA
| | - George A Porter
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA.,Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Medicine (Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Andreas M Beyer
- Cardiovascular Center, Department of Medicine, Med College of Wisconsin, Milwaukee, WI, USA. .,Department of Physiology, Med College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
24
|
The role of cardiolipin concentration and acyl chain composition on mitochondrial inner membrane molecular organization and function. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1039-1052. [PMID: 30951877 DOI: 10.1016/j.bbalip.2019.03.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/19/2019] [Accepted: 03/30/2019] [Indexed: 12/28/2022]
Abstract
Cardiolipin (CL) is a key phospholipid of the mitochondria. A loss of CL content and remodeling of CL's acyl chains is observed in several pathologies. Strong shifts in CL concentration and acyl chain composition would presumably disrupt mitochondrial inner membrane biophysical organization. However, it remains unclear in the literature as to which is the key regulator of mitochondrial membrane biophysical properties. We review the literature to discriminate the effects of CL concentration and acyl chain composition on mitochondrial membrane organization. A widely applicable theme emerges across several pathologies, including cardiovascular diseases, diabetes, Barth syndrome, and neurodegenerative ailments. The loss of CL, often accompanied by increased levels of lyso-CLs, impairs mitochondrial inner membrane organization. Modest remodeling of CL acyl chains is not a major driver of impairments and only in cases of extreme remodeling is there an influence on membrane properties.
Collapse
|
25
|
Malyala S, Zhang Y, Strubbe JO, Bazil JN. Calcium phosphate precipitation inhibits mitochondrial energy metabolism. PLoS Comput Biol 2019; 15:e1006719. [PMID: 30615608 PMCID: PMC6336351 DOI: 10.1371/journal.pcbi.1006719] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 01/17/2019] [Accepted: 12/17/2018] [Indexed: 11/19/2022] Open
Abstract
Early studies have shown that moderate levels of calcium overload can cause lower oxidative phosphorylation rates. However, the mechanistic interpretations of these findings were inadequate. And while the effect of excessive calcium overload on mitochondrial function is well appreciated, there has been little to no reports on the consequences of low to moderate calcium overload. To resolve this inadequacy, mitochondrial function from guinea pig hearts was quantified using several well-established methods including high-resolution respirometry and spectrofluorimetry and analyzed using mathematical modeling. We measured key mitochondrial variables such as respiration, mitochondrial membrane potential, buffer calcium, and substrate effects for a range of mitochondrial calcium loads from near zero to levels approaching mitochondrial permeability transition. In addition, we developed a computer model closely mimicking the experimental conditions and used this model to design experiments capable of eliminating many hypotheses generated from the data analysis. We subsequently performed those experiments and determined why mitochondrial ADP-stimulated respiration is significantly lowered during calcium overload. We found that when calcium phosphate levels, not matrix free calcium, reached sufficient levels, complex I activity is inhibited, and the rate of ATP synthesis is reduced. Our findings suggest that calcium phosphate granules form physical barriers that isolate complex I from NADH, disrupt complex I activity, or destabilize cristae and inhibit NADH-dependent respiration. Mitochondrial calcium handling has been studied for nearly half a century. As we understand it today, low concentrations (1–10 nmol/mg mitochondria) of calcium exert beneficial effects on energy transduction. And high concentrations (>500 nmol/mg mitochondria) lead to respiratory uncoupling and membrane damage. But relatively little is known about the effect of moderate calcium concentrations (10–500 nmol/mg mitochondria) on mitochondrial function. At these concentrations, mitochondrial membrane integrity remains intact and energized, while ATP synthesis becomes significantly impaired. Prior studies have postulated several possible mechanisms, but the precise consequence of calcium overload on mitochondrial ATP production remained obscure. In this study, we combine experimental and computational approaches to show that calcium phosphate precipitation, as opposed to matrix free calcium, inhibits respiratory function at complex I just enough to limit proton pumping during oxidative phosphorylation and decrease ATP synthesis rates.
Collapse
Affiliation(s)
- Sathyavani Malyala
- Department of Physiology, Michigan State University, East Lansing, MI, United States of America
| | - Yizhu Zhang
- Department of Physiology, Michigan State University, East Lansing, MI, United States of America
| | - Jasiel O. Strubbe
- 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
- * E-mail:
| |
Collapse
|
26
|
MacDougall G, Anderton RS, Mastaglia FL, Knuckey NW, Meloni BP. Mitochondria and neuroprotection in stroke: Cationic arginine-rich peptides (CARPs) as a novel class of mitochondria-targeted neuroprotective therapeutics. Neurobiol Dis 2018; 121:17-33. [PMID: 30218759 DOI: 10.1016/j.nbd.2018.09.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/26/2018] [Accepted: 09/11/2018] [Indexed: 01/11/2023] Open
Abstract
Stroke is the second leading cause of death globally and represents a major cause of devastating long-term disability. Despite sustained efforts to develop clinically effective neuroprotective therapies, presently there is no clinically available neuroprotective agent for stroke. As a central mediator of neurodamaging events in stroke, mitochondria are recognised as a critical neuroprotective target, and as such, provide a focus for developing mitochondrial-targeted therapeutics. In recent years, cationic arginine-rich peptides (CARPs) have been identified as a novel class of neuroprotective agent with several demonstrated mechanisms of action, including their ability to target mitochondria and exert positive effects on the organelle. This review provides an overview on neuronal mitochondrial dysfunction in ischaemic stroke pathophysiology and highlights the potential beneficial effects of CARPs on mitochondria in the ischaemic brain following stroke.
Collapse
Affiliation(s)
- Gabriella MacDougall
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Perron Institute for Neurological and Translational Science, Nedlands, Australia; School of Heath Sciences, and Institute for Health Research, The University Notre Dame Australia, Fremantle, Australia.
| | - Ryan S Anderton
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Perron Institute for Neurological and Translational Science, Nedlands, Australia; School of Heath Sciences, and Institute for Health Research, The University Notre Dame Australia, Fremantle, Australia
| | - Frank L Mastaglia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Perron Institute for Neurological and Translational Science, Nedlands, Australia
| | - Neville W Knuckey
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Perron Institute for Neurological and Translational Science, Nedlands, Australia; Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Bruno P Meloni
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Perron Institute for Neurological and Translational Science, Nedlands, Australia; Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia
| |
Collapse
|
27
|
Paradies G, Paradies V, Ruggiero FM, Petrosillo G. Mitochondrial bioenergetics and cardiolipin alterations in myocardial ischemia-reperfusion injury: implications for pharmacological cardioprotection. Am J Physiol Heart Circ Physiol 2018; 315:H1341-H1352. [PMID: 30095969 DOI: 10.1152/ajpheart.00028.2018] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mitochondrial dysfunction plays a central role in myocardial ischemia-reperfusion (I/R) injury. Increased reactive oxygen species production, impaired electron transport chain activity, aberrant mitochondrial dynamics, Ca2+ overload, and opening of the mitochondrial permeability transition pore have been proposed as major contributory factors to mitochondrial dysfunction during myocardial I/R injury. Cardiolipin (CL), a mitochondria-specific phospholipid, plays a pivotal role in multiple mitochondrial bioenergetic processes, including respiration and energy conversion, in mitochondrial morphology and dynamics as well as in several steps of the apoptotic process. Changes in CL levels, species composition, and degree of oxidation may have deleterious consequences for mitochondrial function with important implications in a variety of pathophysiological conditions, including myocardial I/R injury. In this review, we focus on the role played by CL alterations in mitochondrial dysfunction in myocardial I/R injury. Pharmacological strategies to prevent myocardial injury during I/R targeting mitochondrial CL are also examined.
Collapse
Affiliation(s)
- Giuseppe Paradies
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari , Bari , Italy
| | | | - Francesca Maria Ruggiero
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari , Bari , Italy
| | - Giuseppe Petrosillo
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, National Research Council , Bari , Italy
| |
Collapse
|
28
|
Ding H, Xu XW, Wang H, Xiao L, Zhao L, Duan GL, Li XR, Ma ZX, Chen HP. DJ-1 plays an obligatory role in the cardioprotection of delayed hypoxic preconditioning against hypoxia/reoxygenation-induced oxidative stress through maintaining mitochondrial complex I activity. Cell Biochem Funct 2018; 36:147-154. [DOI: 10.1002/cbf.3326] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/13/2018] [Accepted: 01/22/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Hao Ding
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Xing-Wang Xu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Huan Wang
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Lin Xiao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Le Zhao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Guang-Ling Duan
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Xiao-Ran Li
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Zhao-Xia Ma
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - He-Ping Chen
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| |
Collapse
|
29
|
Zhang Y, Li XR, Zhao L, Duan GL, Xiao L, Chen HP. DJ-1 preserving mitochondrial complex I activity plays a critical role in resveratrol–mediated cardioprotection against hypoxia/reoxygenation–induced oxidative stress. Biomed Pharmacother 2018; 98:545-552. [DOI: 10.1016/j.biopha.2017.12.094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 01/03/2023] Open
|
30
|
la Cour MF, Mehrvar S, Kim J, Martin A, Zimmerman MA, Hong JC, Ranji M. Optical imaging for the assessment of hepatocyte metabolic state in ischemia and reperfusion injuries. BIOMEDICAL OPTICS EXPRESS 2017; 8:4419-4426. [PMID: 29082074 PMCID: PMC5654789 DOI: 10.1364/boe.8.004419] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/10/2017] [Accepted: 08/13/2017] [Indexed: 05/09/2023]
Abstract
Deterioration in mitochondrial function leads to hepatic ischemia and reperfusion injury (IRI) in liver surgery and transplantation. 3D optical cryoimaging was used to measure the levels of mitochondrial coenzymes NADH and FAD, and their redox ratio (NADH/FAD) gave a quantitative marker for hepatocyte oxidative stress during IRI. Using a rat model, five groups were compared: control, ischemia for 60 or 90 minutes (Isc60, Isc90), ischemia for 60 or 90 minutes followed by reperfusion of 24 hours (IRI60, IRI90). Ischemia alone did not cause a significant increase in the redox ratio; however, the redox ratio in both IRI60 and IRI90 groups was significantly decreased by 29% and 71%, respectively. A significant correlation was observed between the redox ratio and other markers of injury such as serum aminotransferase levels and the tissue ATP level. The mitochondrial redox state can be successfully measured using optical cryoimaging as a quantitative marker of hepatic IR injury.
Collapse
Affiliation(s)
- Mette F. la Cour
- Biopotonics Lab, Electrical Engineering Department, University of Wisconsin - Milwaukee, 3200 N Cramer St, Milwaukee, WI 53211, USA
- Both contributed equally and are therefore first authors
| | - Shima Mehrvar
- Biopotonics Lab, Electrical Engineering Department, University of Wisconsin - Milwaukee, 3200 N Cramer St, Milwaukee, WI 53211, USA
- Both contributed equally and are therefore first authors
| | - Joohyun Kim
- Division of Transplant Surgery, Department of Surgery, Medical College of Wisconsin, 9200 W. Wisconsin Avenue, Suite E5700, Milwaukee, WI 53226, USA
| | - Alicia Martin
- Division of Transplant Surgery, Department of Surgery, Medical College of Wisconsin, 9200 W. Wisconsin Avenue, Suite E5700, Milwaukee, WI 53226, USA
| | - Michael A. Zimmerman
- Division of Transplant Surgery, Department of Surgery, Medical College of Wisconsin, 9200 W. Wisconsin Avenue, Suite E5700, Milwaukee, WI 53226, USA
| | - Johnny C. Hong
- Division of Transplant Surgery, Department of Surgery, Medical College of Wisconsin, 9200 W. Wisconsin Avenue, Suite E5700, Milwaukee, WI 53226, USA
| | - Mahsa Ranji
- Biopotonics Lab, Electrical Engineering Department, University of Wisconsin - Milwaukee, 3200 N Cramer St, Milwaukee, WI 53211, USA
| |
Collapse
|
31
|
Matsuura TR, Bartos JA, Tsangaris A, Shekar KC, Olson MD, Riess ML, Bienengraeber M, Aufderheide TP, Neumar RW, Rees JN, McKnite SH, Dikalova AE, Dikalov SI, Douglas HF, Yannopoulos D. Early Effects of Prolonged Cardiac Arrest and Ischemic Postconditioning during Cardiopulmonary Resuscitation on Cardiac and Brain Mitochondrial Function in Pigs. Resuscitation 2017; 116:8-15. [PMID: 28408349 PMCID: PMC5552370 DOI: 10.1016/j.resuscitation.2017.03.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 11/21/2022]
Abstract
BACKGROUND Out-of-hospital cardiac arrest (CA) is a prevalent medical crisis resulting in severe injury to the heart and brain and an overall survival of less than 10%. Mitochondrial dysfunction is predicted to be a key determinant of poor outcomes following prolonged CA. However, the onset and severity of mitochondrial dysfunction during CA and cardiopulmonary resuscitation (CPR) is not fully understood. Ischemic postconditioning (IPC), controlled pauses during the initiation of CPR, has been shown to improve cardiac function and neurologically favorable outcomes after 15min of CA. We tested the hypothesis that mitochondrial dysfunction develops during prolonged CA and can be rescued with IPC during CPR (IPC-CPR). METHODS A total of 63 swine were randomized to no ischemia (Naïve), 19min of ventricular fibrillation (VF) CA without CPR (Untreated VF), or 15min of CA with 4min of reperfusion with either standard CPR (S-CPR) or IPC-CPR. Mitochondria were isolated from the heart and brain to quantify respiration, rate of ATP synthesis, and calcium retention capacity (CRC). Reactive oxygen species (ROS) production was quantified from fresh frozen heart and brain tissue. RESULTS Compared to Naïve, Untreated VF induced cardiac and brain ROS overproduction concurrent with decreased mitochondrial respiratory coupling and CRC, as well as decreased cardiac ATP synthesis. Compared to Untreated VF, S-CPR attenuated brain ROS overproduction but had no other effect on mitochondrial function in the heart or brain. Compared to Untreated VF, IPC-CPR improved cardiac mitochondrial respiratory coupling and rate of ATP synthesis, and decreased ROS overproduction in the heart and brain. CONCLUSIONS Fifteen minutes of VF CA results in diminished mitochondrial respiration, ATP synthesis, CRC, and increased ROS production in the heart and brain. IPC-CPR attenuates cardiac mitochondrial dysfunction caused by prolonged VF CA after only 4min of reperfusion, suggesting that IPC-CPR is an effective intervention to reduce cardiac injury. However, reperfusion with both CPR methods had limited effect on mitochondrial function in the brain, emphasizing an important physiological divergence in post-arrest recovery between those two vital organs.
Collapse
Affiliation(s)
- Timothy R Matsuura
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Jason A Bartos
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Adamantios Tsangaris
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | | | - Matthew D Olson
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Matthias L Riess
- Department of Anesthesiology, TVHS VA Medical Center, Nashville, TN, USA; Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Martin Bienengraeber
- Departments of Anesthesiology and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Tom P Aufderheide
- Department of Emergency Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Robert W Neumar
- Department of Emergency Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer N Rees
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Scott H McKnite
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Anna E Dikalova
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sergey I Dikalov
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hunter F Douglas
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Demetris Yannopoulos
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
32
|
Camara AKS, Zhou Y, Wen PC, Tajkhorshid E, Kwok WM. Mitochondrial VDAC1: A Key Gatekeeper as Potential Therapeutic Target. Front Physiol 2017; 8:460. [PMID: 28713289 PMCID: PMC5491678 DOI: 10.3389/fphys.2017.00460] [Citation(s) in RCA: 254] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/16/2017] [Indexed: 12/23/2022] Open
Abstract
Mitochondria are the key source of ATP that fuels cellular functions, and they are also central in cellular signaling, cell division and apoptosis. Dysfunction of mitochondria has been implicated in a wide range of diseases, including neurodegenerative and cardiac diseases, and various types of cancer. One of the key proteins that regulate mitochondrial function is the voltage-dependent anion channel 1 (VDAC1), the most abundant protein on the outer membrane of mitochondria. VDAC1 is the gatekeeper for the passages of metabolites, nucleotides, and ions; it plays a crucial role in regulating apoptosis due to its interaction with apoptotic and anti-apoptotic proteins, namely members of the Bcl-2 family of proteins and hexokinase. Therefore, regulation of VDAC1 is crucial not only for metabolic functions of mitochondria, but also for cell survival. In fact, multiple lines of evidence have confirmed the involvement of VDAC1 in several diseases. Consequently, modulation or dysregulation of VDAC1 function can potentially attenuate or exacerbate pathophysiological conditions. Understanding the role of VDAC1 in health and disease could lead to selective protection of cells in different tissues and diverse diseases. The purpose of this review is to discuss the role of VDAC1 in the pathogenesis of diseases and as a potentially effective target for therapeutic management of various pathologies.
Collapse
Affiliation(s)
- Amadou K S Camara
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, United States.,Cardiovascular Center, Medical College of WisconsinMilwaukee, WI, United States
| | - YiFan Zhou
- Department of Assay Development, HD BiosciencesShanghai, China
| | - Po-Chao Wen
- Department of Biochemistry, Beckman Institute for Advanced Science and Technology, Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-ChampaignUrbana, IL, United States
| | - Emad Tajkhorshid
- Department of Biochemistry, Beckman Institute for Advanced Science and Technology, Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-ChampaignUrbana, IL, United States
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, United States.,Cardiovascular Center, Medical College of WisconsinMilwaukee, WI, United States.,Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukee, WI, United States
| |
Collapse
|
33
|
Azam MA, Zamiri N, Massé S, Kusha M, Lai PFH, Nair GK, Tan NS, Labos C, Nanthakumar K. Effects of Late Sodium Current Blockade on Ventricular Refibrillation in a Rabbit Model. Circ Arrhythm Electrophysiol 2017; 10:CIRCEP.116.004331. [PMID: 28314848 DOI: 10.1161/circep.116.004331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 01/19/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND After defibrillation of initial ventricular fibrillation (VF), it is crucial to prevent refibrillation to ensure successful resuscitation outcomes. Inability of the late Na+ current to inactivate leads to intracellular Ca2+ dysregulation and arrhythmias. Our aim was to determine the effects of ranolazine and GS-967, inhibitors of the late Na+ current, on ventricular refibrillation. METHODS AND RESULTS Long-duration VF was induced electrically in Langendorff-perfused rabbit hearts (n=22) and terminated with a defibrillator after 6 minutes. Fibrillating hearts were randomized into 3 groups: treatment with ranolazine, GS-967, or nontreated controls. In the treated groups, hearts were perfused with ranolazine or GS-967 at 2 minutes of VF. In control experiments, perfusion solution was supplemented with isotonic saline in lieu of a drug. Inducibility of refibrillation was assessed after initial long-duration VF by attempting to reinduce VF. Sustained refibrillation was successful in fewer ranolazine-treated (29.17%; P=0.005) or GS-967-treated (45.83%, P=0.035) hearts compared with that in nontreated control hearts (84.85%). In GS-967-treated hearts, significantly more spontaneous termination of initial long-duration VF was observed (66.67%; P=0.01). Ca2+ transient duration was reduced in ranolazine-treated hearts compared with that in controls (P=0.05) and also Ca2+ alternans (P=0.03). CONCLUSIONS Late Na+ current inhibition during long-duration VF reduces the susceptibility to subsequent refibrillation, partially by mitigating dysregulation of intracellular Ca2+. These results suggest the potential therapeutic use of ranolazine and GS-967 and call for further testing in cardiac arrest models.
Collapse
Affiliation(s)
- Mohammed Ali Azam
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Nima Zamiri
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Stéphane Massé
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Marjan Kusha
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Patrick F H Lai
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Govind K Nair
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Nigel S Tan
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Christopher Labos
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Kumaraswamy Nanthakumar
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.).
| |
Collapse
|
34
|
Parodi-Rullán RM, Chapa-Dubocq X, Rullán PJ, Jang S, Javadov S. High Sensitivity of SIRT3 Deficient Hearts to Ischemia-Reperfusion Is Associated with Mitochondrial Abnormalities. Front Pharmacol 2017; 8:275. [PMID: 28559847 PMCID: PMC5432544 DOI: 10.3389/fphar.2017.00275] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/02/2017] [Indexed: 12/31/2022] Open
Abstract
Aim: Sirtuins are NAD+-dependent deacetylases that regulate cell metabolism through protein acetylation/deacetylation, and SIRT3 is the major deacetylase among mitochondrial isoforms. Here, we elucidated the possible role of acetylation of cyclophilin D, a key regulator of the mitochondrial permeability transition pore (mPTP), in mitochondria-mediated cardiac dysfunction induced by ischemia-reperfusion (IR) in wild type (WT) and SIRT3 knockout (SIRT3-/-) mice. Materials and Methods: Isolated and Langendorff-mode perfused hearts of WT and SIRT3-/- mice were subjected to 25-min global ischemia followed by 60-min of reperfusion in the presence or absence of the mPTP inhibitor, sanglifehrin A (SfA). Results: Analysis of mitochondrial sirtuins demonstrated that SIRT3 deficiency upregulated SIRT4 with no effect on SIRT5 expression. Hearts of SIRT3-/- mice exhibited significantly less recovery of cardiac function at the end of IR compared to WT mice. Intact (non-perfused) SIRT3-/- hearts exhibited an increased rate of Ca2+-induced swelling in mitochondria as an indicator of mPTP opening. However, there was no difference in mPTP opening and cyclophilin D acetylation between WT and SIRT3-/- hearts subjected to IR injury. Ca2+-stimulated H2O2 production was significantly higher in SIRT3-/- mitochondria that was prevented by SfA. Superoxide dismutase activity was lower in SIRT3-/- heart mitochondria subjected to IR which correlated with an increase in protein carbonylation. However, mitochondrial DNA integrity was not affected in SIRT3-/- hearts after IR. Conclusion: SIRT3 deficiency exacerbates cardiac dysfunction during post-ischemic recovery, and increases mPTP opening and ROS generation without oxidative damage to mitochondrial proteins and DNA.
Collapse
Affiliation(s)
- Rebecca M Parodi-Rullán
- Department of Physiology, University of Puerto Rico School of Medicine, San JuanPR, United States
| | - Xavier Chapa-Dubocq
- Department of Physiology, University of Puerto Rico School of Medicine, San JuanPR, United States
| | - Pedro J Rullán
- Department of Physiology, University of Puerto Rico School of Medicine, San JuanPR, United States
| | - Sehwan Jang
- Department of Physiology, University of Puerto Rico School of Medicine, San JuanPR, United States
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San JuanPR, United States
| |
Collapse
|
35
|
Moreno A, Kuzmiak-Glancy S, Jaimes R, Kay MW. Enzyme-dependent fluorescence recovery of NADH after photobleaching to assess dehydrogenase activity of isolated perfused hearts. Sci Rep 2017; 7:45744. [PMID: 28361886 PMCID: PMC5374639 DOI: 10.1038/srep45744] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/02/2017] [Indexed: 01/09/2023] Open
Abstract
Reduction of NAD+ by dehydrogenase enzymes to form NADH is a key component of cellular metabolism. In cellular preparations and isolated mitochondria suspensions, enzyme-dependent fluorescence recovery after photobleaching (ED-FRAP) of NADH has been shown to be an effective approach for measuring the rate of NADH production to assess dehydrogenase enzyme activity. Our objective was to demonstrate how dehydrogenase activity could be assessed within the myocardium of perfused hearts using NADH ED-FRAP. This was accomplished using a combination of high intensity UV pulses to photobleach epicardial NADH. Replenishment of epicardial NADH fluorescence was then imaged using low intensity UV illumination. NADH ED-FRAP parameters were optimized to deliver 23.8 mJ of photobleaching light energy at a pulse width of 6 msec and a duty cycle of 50%. These parameters provided repeatable measurements of NADH production rate during multiple metabolic perturbations, including changes in perfusate temperature, electromechanical uncoupling, and acute ischemia/reperfusion injury. NADH production rate was significantly higher in every perturbation where the energy demand was either higher or uncompromised. We also found that NADH production rate remained significantly impaired after 10 min of reperfusion after global ischemia. Overall, our results indicate that myocardial NADH ED-FRAP is a useful optical non-destructive approach for assessing dehydrogenase activity.
Collapse
Affiliation(s)
- Angel Moreno
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
| | - Sarah Kuzmiak-Glancy
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
| | - Rafael Jaimes
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
| | - Matthew W Kay
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
| |
Collapse
|
36
|
Mitochondrial Cx43 hemichannels contribute to mitochondrial calcium entry and cell death in the heart. Basic Res Cardiol 2017; 112:27. [PMID: 28364353 DOI: 10.1007/s00395-017-0618-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/30/2017] [Indexed: 10/19/2022]
Abstract
Mitochondrial connexin 43 (Cx43) plays a key role in cardiac cytoprotection caused by repeated exposure to short periods of non-lethal ischemia/reperfusion, a condition known as ischemic preconditioning. Cx43 also forms calcium (Ca2+)-permeable hemichannels that may potentially lead to mitochondrial Ca2+ overload and cell death. Here, we studied the role of Cx43 in facilitating mitochondrial Ca2+ entry and investigated its downstream consequences. To that purpose, we used various connexin-targeting peptides interacting with extracellular (Gap26) and intracellular (Gap19, RRNYRRNY) Cx43 domains, and tested their effect on mitochondrial dye- and Ca2+-uptake, electrophysiological properties of plasmalemmal and mitochondrial Cx43 channels, and cell injury/cell death. Our results in isolated mice cardiac subsarcolemmal mitochondria indicate that Cx43 forms hemichannels that contribute to Ca2+ entry and may trigger permeability transition and cell injury/death. RRNYRRNY displayed the strongest effects in all assays and inhibited plasma membrane as well as mitochondrial Cx43 hemichannels. RRNYRRNY also strongly reduced the infarct size in ex vivo cardiac ischemia-reperfusion studies. These results indicate that Cx43 contributes to mitochondrial Ca2+ homeostasis and is involved in triggering cell injury/death pathways that can be inhibited by RRNYRRNY peptide.
Collapse
|
37
|
Dudek J, Maack C. Barth syndrome cardiomyopathy. Cardiovasc Res 2017; 113:399-410. [PMID: 28158532 DOI: 10.1093/cvr/cvx014] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/19/2016] [Accepted: 01/04/2017] [Indexed: 02/02/2023] Open
Abstract
Barth syndrome (BTHS) is an inherited form of cardiomyopathy, caused by a mutation within the gene encoding the mitochondrial transacylase tafazzin. Tafazzin is involved in the biosynthesis of the unique phospholipid cardiolipin (CL), which is almost exclusively found in mitochondrial membranes. CL directly interacts with a number of essential protein complexes in the mitochondrial membranes including the respiratory chain, mitochondrial metabolite carriers, and proteins, involved in shaping mitochondrial morphology. Here we describe, how in BTHS CL deficiency causes changes in the morphology of mitochondria, structural changes in the respiratory chain, decreased respiration, and increased generation of reactive oxygen species. A large number of cellular and animal models for BTHS have been established to elucidate how mitochondrial dysfunction induces sarcomere disorganization and reduced contractility, resulting in dilated cardiomyopathy in vivo.
Collapse
Affiliation(s)
- Jan Dudek
- Department of Cellular Biochemistry, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Christoph Maack
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg/Saar, Germany
| |
Collapse
|
38
|
Ronchi C, Torre E, Rizzetto R, Bernardi J, Rocchetti M, Zaza A. Late sodium current and intracellular ionic homeostasis in acute ischemia. Basic Res Cardiol 2017; 112:12. [PMID: 28101642 DOI: 10.1007/s00395-017-0602-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 01/03/2017] [Indexed: 11/25/2022]
Abstract
Blockade of the late Na+ current (I NaL) protects from ischemia/reperfusion damage; nevertheless, information on changes in I NaL during acute ischemia and their effect on intracellular milieu is missing. I NaL, cytosolic Na+ and Ca2+ activities (Nacyt, Cacyt) were measured in isolated rat ventricular myocytes during 7 min of simulated ischemia (ISC); in all the conditions tested, effects consistently exerted by ranolazine (RAN) and tetrodotoxin (TTX) were interpreted as due to I NaL blockade. The results indicate that I NaL was enhanced during ISC in spite of changes in action potential (AP) contour; I NaL significantly contributed to Nacyt rise, but only marginally to Cacyt rise. The impact of I NaL on Cacyt was markedly enhanced by blockade of the sarcolemmal(s) Na+/Ca2+ exchanger (NCX) and was due to the presence of (Na+-sensitive) Ca2+ efflux through mitochondrial NCX (mNCX). sNCX blockade increased Cacyt and decreased Nacyt, thus indicating that, throughout ISC, sNCX operated in the forward mode, in spite of the substantial Nacyt increment. Thus, a robust Ca2+ source, other than sNCX and including mitochondria, contributed to Cacyt during ISC. Most, but not all, of RAN effects were shared by TTX. (1) The paradigm that attributes Cacyt accumulation during acute ischemia to decrease/reversal of sNCX transport may not be of general applicability; (2) I NaL is enhanced during ISC, when the effect of Nacyt on mitochondrial Ca2+ transport may substantially contribute to I NaL impact on Cacyt; (3) RAN may act mostly, but not exclusively, through I NaL blockade during ISC.
Collapse
Affiliation(s)
- Carlotta Ronchi
- Department of Biotechnologies and Biosciences, University Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Eleonora Torre
- Department of Biotechnologies and Biosciences, University Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Riccardo Rizzetto
- Department of Biotechnologies and Biosciences, University Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Joyce Bernardi
- Department of Biotechnologies and Biosciences, University Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Marcella Rocchetti
- Department of Biotechnologies and Biosciences, University Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Antonio Zaza
- Department of Biotechnologies and Biosciences, University Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy.
| |
Collapse
|
39
|
Zou D, Geng N, Chen Y, Ren L, Liu X, Wan J, Guo S, Wang S. Ranolazine improves oxidative stress and mitochondrial function in the atrium of acetylcholine-CaCl2 induced atrial fibrillation rats. Life Sci 2016; 156:7-14. [DOI: 10.1016/j.lfs.2016.05.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 05/08/2016] [Accepted: 05/17/2016] [Indexed: 12/19/2022]
|
40
|
Ranji M, Motlagh MM, Salehpour F, Sepehr R, Heisner JS, Dash RK, Camara AKS. Optical Cryoimaging Reveals a Heterogeneous Distribution of Mitochondrial Redox State in ex vivo Guinea Pig Hearts and Its Alteration During Ischemia and Reperfusion. IEEE JOURNAL OF TRANSLATIONAL ENGINEERING IN HEALTH AND MEDICINE-JTEHM 2016; 4:1800210. [PMID: 27574574 PMCID: PMC4993131 DOI: 10.1109/jtehm.2016.2570219] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/21/2016] [Accepted: 04/26/2016] [Indexed: 12/16/2022]
Abstract
Oxidation of substrates to generate ATP in mitochondria is mediated by redox reactions of NADH and FADH2. Cardiac ischemia and reperfusion (IR) injury compromises mitochondrial oxidative phosphorylation. We hypothesize that IR alters the metabolic heterogeneity of mitochondrial redox state of the heart that is only evident in the 3-D optical cryoimaging of the perfused heart before, during, and after IR. The study involved four groups of hearts: time control (TC: heart perfusion without IR), global ischemia (Isch), global ischemia followed by reperfusion (IR) and TC with PCP (a mitochondrial uncoupler) perfusion. Mitochondrial NADH and FAD autofluorescence signals were recorded spectrofluorometrically online in guinea pig ex vivo-perfused hearts in the Langendorff mode. At the end of each specified protocol, hearts were rapidly removed and snap frozen in liquid N2 for later 3-D optical cryoimaging of the mitochondrial NADH, FAD, and NADH/FAD redox ratio (RR). The TC hearts revealed a heterogeneous spatial distribution of NADH, FAD, and RR. Ischemia and IR altered the spatial distribution and caused an overall increase and decrease in the RR by 55% and 64%, respectively. Uncoupling with PCP resulted in the lowest level of the RR (73% oxidation) compared with TC. The 3-D optical cryoimaging of the heart provides novel insights into the heterogeneous distribution of mitochondrial NADH, FAD, RR, and metabolism from the base to the apex during ischemia and IR. This 3-D information of the mitochondrial redox state in the normal and ischemic heart was not apparent in the dynamic spectrofluorometric data.
Collapse
Affiliation(s)
- Mahsa Ranji
- Department of Electrical EngineeringUniversity of Wisconsin-MilwaukeeMilwaukeeWI53211USA; Biotechnology and Bioengineering CenterMedical College of WisconsinMilwaukeeWI53226USA
| | | | - Fahimeh Salehpour
- Department of Electrical Engineering University of Wisconsin-Milwaukee Milwaukee WI 53211 USA
| | - Reyhaneh Sepehr
- Department of Electrical Engineering University of Wisconsin-Milwaukee Milwaukee WI 53211 USA
| | - James S Heisner
- Department of Anesthesiology Medical College of Wisconsin Milwaukee WI 53226 USA
| | - Ranjan K Dash
- Department of PhysiologyMedical College of WisconsinMilwaukeeWI53226USA; Biotechnology and Bioengineering CenterMedical College of WisconsinMilwaukeeWI53226USA; Cardiovascular Research CenterMedical College of WisconsinMilwaukeeWI53226USA
| | - Amadou K S Camara
- Cardiovascular Research CenterMedical College of WisconsinMilwaukeeWI53226USA; Department of AnesthesiologyMedical College of WisconsinMilwaukeeWI53226USA
| |
Collapse
|
41
|
The Role of Oxidative Stress in Myocardial Ischemia and Reperfusion Injury and Remodeling: Revisited. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1656450. [PMID: 27313825 PMCID: PMC4897712 DOI: 10.1155/2016/1656450] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/11/2016] [Accepted: 05/03/2016] [Indexed: 01/11/2023]
Abstract
Oxidative and reductive stress are dual dynamic phases experienced by the cells undergoing adaptation towards endogenous or exogenous noxious stimulus. The former arises due to the imbalance between the reactive oxygen species production and antioxidant defenses, while the latter is due to the aberrant increase in the reducing equivalents. Mitochondrial malfunction is the common denominator arising from the aberrant functioning of the rheostat that maintains the homeostasis between oxidative and reductive stress. Recent experimental evidences suggest that the maladaptation during oxidative stress could play a pivotal role in the pathophysiology of major cardiovascular diseases such as myocardial infraction, atherosclerosis, and diabetic cardiovascular complications. In this review we have discussed the role of oxidative and reductive stress pathways in the pathogenesis of myocardial ischemia/reperfusion injury and diabetic cardiomyopathy (DCM). Furthermore, we have provided impetus for the development of subcellular organelle targeted antioxidant drug therapy for thwarting the deterioration of the failing myocardium in the aforementioned cardiovascular conditions.
Collapse
|
42
|
The Role of Mitochondrial Reactive Oxygen Species in Cardiovascular Injury and Protective Strategies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8254942. [PMID: 27200148 PMCID: PMC4856919 DOI: 10.1155/2016/8254942] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/14/2022]
Abstract
Ischaemia/reperfusion (I/R) injury of the heart represents a major health burden mainly associated with acute coronary syndromes. While timely coronary reperfusion has become the established routine therapy in patients with ST-elevation myocardial infarction, the restoration of blood flow into the previously ischaemic area is always accompanied by myocardial injury. The central mechanism involved in this phenomenon is represented by the excessive generation of reactive oxygen species (ROS). Besides their harmful role when highly generated during early reperfusion, minimal ROS formation during ischaemia and/or at reperfusion is critical for the redox signaling of cardioprotection. In the past decades, mitochondria have emerged as the major source of ROS as well as a critical target for cardioprotective strategies at reperfusion. Mitochondria dysfunction associated with I/R myocardial injury is further described and ultimately analyzed with respect to its role as source of both deleterious and beneficial ROS. Furthermore, the contribution of ROS in the highly investigated field of conditioning strategies is analyzed. In the end, the vascular sources of mitochondria-derived ROS are briefly reviewed.
Collapse
|
43
|
Dolinsky VW, Cole LK, Sparagna GC, Hatch GM. Cardiac mitochondrial energy metabolism in heart failure: Role of cardiolipin and sirtuins. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1544-54. [PMID: 26972373 DOI: 10.1016/j.bbalip.2016.03.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/03/2016] [Accepted: 03/04/2016] [Indexed: 01/19/2023]
Abstract
Mitochondrial oxidation of fatty acids accounts for the majority of cardiac ATP production in the heart. Fatty acid utilization by cardiac mitochondria is controlled at the level of fatty acid uptake, lipid synthesis, mobilization and mitochondrial import and oxidation. Consequently defective mitochondrial function appears to be central to the development of heart failure. Cardiolipin is a key mitochondrial phospholipid required for the activity of the electron transport chain. In heart failure, loss of cardiolipin and tetralinoleoylcardiolipin helps to fuel the generation of excessive reactive oxygen species that are a by-product of inefficient mitochondrial electron transport chain complexes I and III. In this vicious cycle, reactive oxygen species generate lipid peroxides and may, in turn, cause oxidation of cardiolipin catalyzed by cytochrome c leading to cardiomyocyte apoptosis. Hence, preservation of cardiolipin and mitochondrial function may be keys to the prevention of heart failure development. In this review, we summarize cardiac energy metabolism and the important role that fatty acid uptake and metabolism play in this process and how defects in these result in heart failure. We highlight the key role that cardiolipin and sirtuins play in cardiac mitochondrial β-oxidation. In addition, we review the potential of pharmacological modulation of cardiolipin through the polyphenolic molecule resveratrol as a sirtuin-activator in attenuating mitochondrial dysfunction. Finally, we provide novel experimental evidence that resveratrol treatment increases cardiolipin in isolated H9c2 cardiac myocytes and tetralinoleoylcardiolipin in the heart of the spontaneously hypertensive rat and hypothesize that this leads to improvement in mitochondrial function. This article is part of a Special Issue entitled: Heart Lipid Metabolism edited by G.D. Lopaschuk.
Collapse
Affiliation(s)
- Vernon W Dolinsky
- Department of Pharmacology & Therapeutics, Faculty of Health Sciences, University of Manitoba, Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba (CHRIM), Canada
| | - Laura K Cole
- Department of Pharmacology & Therapeutics, Faculty of Health Sciences, University of Manitoba, Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba (CHRIM), Canada
| | - Genevieve C Sparagna
- Department of Medicine, Division of Cardiology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Grant M Hatch
- Department of Pharmacology & Therapeutics, Faculty of Health Sciences, University of Manitoba, Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba (CHRIM), Canada; Department of Biochemistry and Medical Genetics, Faculty of Health Sciences, Center for Research and Treatment of Atherosclerosis, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
44
|
Aldasoro M, Guerra-Ojeda S, Aguirre-Rueda D, Mauricio MD, Vila JM, Marchio P, Iradi A, Aldasoro C, Jorda A, Obrador E, Valles SL. Effects of Ranolazine on Astrocytes and Neurons in Primary Culture. PLoS One 2016; 11:e0150619. [PMID: 26950436 PMCID: PMC4780741 DOI: 10.1371/journal.pone.0150619] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 02/17/2016] [Indexed: 12/15/2022] Open
Abstract
Ranolazine (Rn) is an antianginal agent used for the treatment of chronic angina pectoris when angina is not adequately controlled by other drugs. Rn also acts in the central nervous system and it has been proposed for the treatment of pain and epileptic disorders. Under the hypothesis that ranolazine could act as a neuroprotective drug, we studied its effects on astrocytes and neurons in primary culture. We incubated rat astrocytes and neurons in primary cultures for 24 hours with Rn (10-7, 10-6 and 10-5 M). Cell viability and proliferation were measured using trypan blue exclusion assay, MTT conversion assay and LDH release assay. Apoptosis was determined by Caspase 3 activity assay. The effects of Rn on pro-inflammatory mediators IL-β and TNF-α was determined by ELISA technique, and protein expression levels of Smac/Diablo, PPAR-γ, Mn-SOD and Cu/Zn-SOD by western blot technique. In cultured astrocytes, Rn significantly increased cell viability and proliferation at any concentration tested, and decreased LDH leakage, Smac/Diablo expression and Caspase 3 activity indicating less cell death. Rn also increased anti-inflammatory PPAR-γ protein expression and reduced pro-inflammatory proteins IL-1 β and TNFα levels. Furthermore, antioxidant proteins Cu/Zn-SOD and Mn-SOD significantly increased after Rn addition in cultured astrocytes. Conversely, Rn did not exert any effect on cultured neurons. In conclusion, Rn could act as a neuroprotective drug in the central nervous system by promoting astrocyte viability, preventing necrosis and apoptosis, inhibiting inflammatory phenomena and inducing anti-inflammatory and antioxidant agents.
Collapse
Affiliation(s)
- Martin Aldasoro
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Sol Guerra-Ojeda
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | | | | | - Jose Mª Vila
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Patricia Marchio
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Antonio Iradi
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Constanza Aldasoro
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Adrian Jorda
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Elena Obrador
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Soraya L. Valles
- Department of Physiology, School of Medicine, University of Valencia, Spain
- * E-mail:
| |
Collapse
|
45
|
Ahmad S, Ahmad A, Hendry-Hofer TB, Loader JE, Claycomb WC, Mozziconacci O, Schöneich C, Reisdorph N, Powell RL, Chandler JD, Day BJ, Veress LA, White CW. Sarcoendoplasmic reticulum Ca(2+) ATPase. A critical target in chlorine inhalation-induced cardiotoxicity. Am J Respir Cell Mol Biol 2016; 52:492-502. [PMID: 25188881 DOI: 10.1165/rcmb.2014-0005oc] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Autopsy specimens from human victims or experimental animals that die due to acute chlorine gas exposure present features of cardiovascular pathology. We demonstrate acute chlorine inhalation-induced reduction in heart rate and oxygen saturation in rats. Chlorine inhalation elevated chlorine reactants, such as chlorotyrosine and chloramine, in blood plasma. Using heart tissue and primary cardiomyocytes, we demonstrated that acute high-concentration chlorine exposure in vivo (500 ppm for 30 min) caused decreased total ATP content and loss of sarcoendoplasmic reticulum calcium ATPase (SERCA) activity. Loss of SERCA activity was attributed to chlorination of tyrosine residues and oxidation of an important cysteine residue, cysteine-674, in SERCA, as demonstrated by immunoblots and mass spectrometry. Using cardiomyocytes, we found that chlorine-induced cell death and damage to SERCA could be decreased by thiocyanate, an important biological antioxidant, and by genetic SERCA2 overexpression. We also investigated a U.S. Food and Drug Administration-approved drug, ranolazine, used in treatment of cardiac diseases, and previously shown to stabilize SERCA in animal models of ischemia-reperfusion. Pretreatment with ranolazine or istaroxime, another SERCA activator, prevented chlorine-induced cardiomyocyte death. Further investigation of responsible mechanisms showed that ranolazine- and istaroxime-treated cells preserved mitochondrial membrane potential and ATP after chlorine exposure. Thus, these studies demonstrate a novel critical target for chlorine in the heart and identify potentially useful therapies to mitigate toxicity of acute chlorine exposure.
Collapse
Affiliation(s)
- Shama Ahmad
- 1 Pediatric Airway Research Center, Department of Pediatrics, University of Colorado, Denver, Aurora, Colorado
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
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: 1011] [Impact Index Per Article: 101.1] [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
|
47
|
Paradies G, Paradies V, Ruggiero FM, Petrosillo G. Cardiolipin alterations and mitochondrial dysfunction in heart ischemia/reperfusion injury. ACTA ACUST UNITED AC 2015. [DOI: 10.2217/clp.15.31] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
48
|
Cosyns SMR, Shiva S, Lefebvre RA. Protective effect of exogenous nitrite in postoperative ileus. Br J Pharmacol 2015; 172:4864-74. [PMID: 26227770 PMCID: PMC4621985 DOI: 10.1111/bph.13255] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 07/09/2015] [Accepted: 07/15/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE As the pathogenesis of postoperative ileus (POI) involves inflammation and oxidative stress, comparable to ischaemia/reperfusion injury which can be ameliorated with nitrite, we investigated whether nitrite can protect against POI and explored the mechanisms involved. EXPERIMENTAL APPROACH We used intestinal manipulation (IM) of the small intestine to induce POI in C57BL/6J mice. Sodium nitrite (48 nmol) was administered intravenously just before IM. Intestinal transit was assessed using fluorescent imaging. Bethanechol-stimulated jejunal circular muscle contractions were measured in organ baths. Inflammatory parameters, neutrophil infiltration, inducible NOS (iNOS) activity, reactive oxygen species (ROS) levels, mitochondrial complex I activity and cGMP were measured in the intestinal muscularis. KEY RESULTS Pre-treatment with nitrite markedly improved the delay in intestinal transit and restored the reduced intestinal contractility observed 24 h following IM. This was accompanied by reduced protein levels of TNF-α, IL-6 and the chemokine CCL2, along with reduced iNOS activity and ROS levels. The associated neutrophil influx at 24 h was not influenced by nitrite. IM reduced mitochondrial complex I activity and cGMP levels; treatment with nitrite increased cGMP levels. Pre-treatment with the NO scavenger carboxy-PTIO or the soluble guanylyl cyclase inhibitor ODQ abolished nitrite-induced protective effects. CONCLUSIONS AND IMPLICATIONS Exogenous nitrite deserves further investigation as a possible treatment for POI. Nitrite-induced protection of POI in mice was dependent on NO and this effect was not related to inhibition of mitochondrial complex I, but did involve activation of soluble guanylyl cyclase.
Collapse
Affiliation(s)
- S M R Cosyns
- Heymans Institute of Pharmacology, Ghent University, Ghent, Belgium
| | - S Shiva
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - R A Lefebvre
- Heymans Institute of Pharmacology, Ghent University, Ghent, Belgium
| |
Collapse
|
49
|
Mu HN, Li Q, Pan CS, Liu YY, Yan L, Hu BH, Sun K, Chang X, Zhao XR, Fan JY, Han JY. Caffeic acid attenuates rat liver reperfusion injury through sirtuin 3-dependent regulation of mitochondrial respiratory chain. Free Radic Biol Med 2015; 85:237-49. [PMID: 25960048 DOI: 10.1016/j.freeradbiomed.2015.04.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 04/21/2015] [Accepted: 04/29/2015] [Indexed: 02/07/2023]
Abstract
Sirtuin 3 (Sirt3) plays critical roles in regulating mitochondrial oxidative metabolism. However, whether Sirt3 is involved in liver ischemia and reperfusion (I/R) injury remains elusive. Caffeic acid (CA) is a natural antioxidant derived from Salvia miltiorrhiza. Whether CA protects against liver I/R injury through regulating Sirt3 and the mitochondrial respiratory chain (MRC) is unclear. This study investigated the effect of CA on liver I/R injury, microcirculatory disturbance, and potential mechanisms, particularly focusing on Sirt3-dependent MRC. Liver I/R of male Sprague-Dawley rats was established by occlusion of portal area vessels for 30 min followed by 120 min of reperfusion. CA (15 mg/kg/h) was continuously infused via the femoral vein starting 30 min before ischemia. After I/R, Sirt3 expression, and MRC activity decreased, acetylation of NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 9 and succinate dehydrogenase complex, subunit A, flavoprotein variant provoked, and the liver microcirculatory disturbance and injury were observed. Treatment with CA attenuated liver injury, inhibited Sirt3 down-expression, and up-regulated MRC activity. CA attenuated rat liver microcirculatory disturbance and oxidative injury through regulation of Sirt3 and the mitochondrial respiratory chain.
Collapse
Affiliation(s)
- Hong-Na Mu
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China; Key Laboratory of Stasis and Phlegm of State Administration of Traditional Chinese Medicine, Beijing, China
| | - Quan Li
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China; Key Laboratory of Stasis and Phlegm of State Administration of Traditional Chinese Medicine, Beijing, China
| | - Chun-Shui Pan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China; Key Laboratory of Stasis and Phlegm of State Administration of Traditional Chinese Medicine, Beijing, China
| | - Yu-Ying Liu
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China; Key Laboratory of Stasis and Phlegm of State Administration of Traditional Chinese Medicine, Beijing, China
| | - Li Yan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China; Key Laboratory of Stasis and Phlegm of State Administration of Traditional Chinese Medicine, Beijing, China
| | - Bai-He Hu
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China; Key Laboratory of Stasis and Phlegm of State Administration of Traditional Chinese Medicine, Beijing, China
| | - Kai Sun
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China; Key Laboratory of Stasis and Phlegm of State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xin Chang
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China; Key Laboratory of Stasis and Phlegm of State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xin-Rong Zhao
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China; Key Laboratory of Stasis and Phlegm of State Administration of Traditional Chinese Medicine, Beijing, China
| | - Jing-Yu Fan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China; Key Laboratory of Stasis and Phlegm of State Administration of Traditional Chinese Medicine, Beijing, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China; Key Laboratory of Stasis and Phlegm of State Administration of Traditional Chinese Medicine, Beijing, China.
| |
Collapse
|
50
|
Endogenic oxidative stress response contributes to glutathione over-accumulation in mutant Saccharomyces cerevisiae Y518. Appl Microbiol Biotechnol 2015; 99:7069-78. [DOI: 10.1007/s00253-015-6629-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/14/2015] [Accepted: 04/18/2015] [Indexed: 12/16/2022]
|