1
|
Guo C, Hildick KL, Jiang J, Zhao A, Guo W, Henley JM, Wilkinson KA. SENP3 Promotes an Mff-Primed Bcl-x L -Drp1 Interaction Involved in Cell Death Following Ischemia. Front Cell Dev Biol 2021; 9:752260. [PMID: 34722538 PMCID: PMC8555761 DOI: 10.3389/fcell.2021.752260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/17/2021] [Indexed: 11/13/2022] Open
Abstract
Dysregulation of the mitochondrial fission machinery has been linked to cell death following ischemia. Fission is largely dependent on recruitment of Dynamin-related protein 1 (Drp1) to the receptor Mitochondrial fission factor (Mff) located on the mitochondrial outer membrane (MOM). Drp1 is a target for SUMOylation and its deSUMOylation, mediated by the SUMO protease SENP3, enhances the Drp1-Mff interaction to promote cell death in an oxygen/glucose deprivation (OGD) model of ischemia. Another interacting partner for Drp1 is the Bcl-2 family member Bcl-x L , an important protein in cell death and survival pathways. Here we demonstrate that preventing Drp1 SUMOylation by mutating its SUMO target lysines enhances the Drp1-Bcl-x L interaction in vivo and in vitro. Moreover, SENP3-mediated deSUMOylation of Drp1 promotes the Drp1-Bcl-x L interaction. Our data suggest that Mff primes Drp1 binding to Bcl-x L at the mitochondria and that Mff and Bcl-x L can interact directly, independent of Drp1, through their transmembrane domains. Importantly, SENP3 loss in cells subjected to OGD correlates with reduced Drp1-Bcl-x L interaction, whilst recovery of SENP3 levels in cells subjected to reoxygenation following OGD correlates with increased Drp1-Bcl-x L interaction. Expressing a Bcl-x L mutant with defective Drp1 binding reduces OGD plus reoxygenation-evoked cell death. Taken together, our results indicate that SENP3-mediated deSUMOlyation promotes an Mff-primed Drp1-Bcl-x L interaction that contributes to cell death following ischemia.
Collapse
Affiliation(s)
- Chun Guo
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Keri L Hildick
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Juwei Jiang
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Alice Zhao
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Wenbin Guo
- School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Jeremy M Henley
- School of Biochemistry, University of Bristol, Bristol, United Kingdom.,Faculty of Science, Centre for Neuroscience and Regenerative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
| | - Kevin A Wilkinson
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
2
|
Chen W, Lv L, Nong Z, Chen X, Pan X, Chen C. Hyperbaric oxygen protects against myocardial ischemia‑reperfusion injury through inhibiting mitochondria dysfunction and autophagy. Mol Med Rep 2020; 22:4254-4264. [PMID: 32901878 PMCID: PMC7533464 DOI: 10.3892/mmr.2020.11497] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 06/22/2020] [Indexed: 12/26/2022] Open
Abstract
Our previous study demonstrated that hyperbaric oxygen (HBO) improves heart function predominantly through reducing oxygen stress, modulating energy metabolism and inhibiting cell apoptosis. The present study aimed to investigate the protective effects of HBO on mitochondrial function and autophagy using rats with a ligated left anterior descending artery. The cardioprotective effects of HBO were mainly evaluated using ELISA, fluorescent probes, transmission electron microscopy and reverse transcription-quantitative PCR (RT-qPCR). HBO pretreatment for 14 days (once a day) using a 0.25 MPa chamber improved mitochondrial morphology and decreased the number of autophagic vesicles, as observed using a transmission electron microscope. HBO pretreatment significantly increased the levels of ATP, ADP, energy charge and the opening of the mitochondrial permeability transition pore, but decreased the levels of AMP, cytochrome c and reactive oxygen species. Moreover, HBO pretreatment significantly increased the gene or protein expression levels of eIF4E-binding protein 1, mammalian target of rapamycin (mTOR), mitochondrial DNA, NADH dehydrogenase subunit 1, mitofusin 1 and mitofusin 2, whereas it decreased the gene or protein expression levels of autophagy-related 5 (Atg5), cytochrome c, dynamin-related protein 1 and p53, as determined using RT-qPCR or immunohistochemistry. In conclusion, HBO treatment was observed to protect cardiomyocytes during myocardial ischemia-reperfusion injury (MIRI) by preventing mitochondrial dysfunction and inhibiting autophagy. Thus, these results provide novel evidence to support the use of HBO as a potential agent for the mitigation of MIRI.
Collapse
Affiliation(s)
- Wan Chen
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Liwen Lv
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Zhihuan Nong
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Xiaoyu Chen
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Xiaorong Pan
- Department of Hyperbaric Oxygen, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Chunxia Chen
- Department of Hyperbaric Oxygen, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
3
|
Dalal S, Daniels CR, Li Y, Wright GL, Singh M, Singh K. Exogenous ubiquitin attenuates hypoxia/reoxygenation-induced cardiac myocyte apoptosis via the involvement of CXCR4 and modulation of mitochondrial homeostasis. Biochem Cell Biol 2020; 98:492-501. [PMID: 31967865 DOI: 10.1139/bcb-2019-0339] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Exogenous ubiquitin (UB) plays a protective role in β-adrenergic receptor-stimulated and ischemia/reperfusion (I/R)-induced myocardial remodeling. Here, we report that UB treatment inhibits hypoxia/reoxygenation (H/R)-induced apoptosis in adult rat ventricular myocytes (ARVMs). The activation of Akt was elevated, whereas the activation of glycogen synthase kinase-3β was reduced in UB-treated cells post-H/R. The level of oxidative stress was lower, whereas the number of ARVMs with polarized mitochondria was significantly greater in the UB-treated samples. ARVMs express CXCR4 with majority of CXCR4 localized in the membrane fraction. CXCR4 antagonism using AMD3100, and siRNA-mediated knockdown of CXCR4 negated the protective effects of UB. Two mutated UB proteins (unable to bind CXCR4) had no effect on H/R-induced apoptosis, activation of Akt and GSK-3β, or oxidative stress. UB treatment enhanced mitochondrial biogenesis, and inhibition of mitochondrial fission using mdivi1 inhibited H/R-induced apoptosis. Ex vivo, UB treatment significantly decreased infarct size and improved functional recovery of the heart following global I/R. Activation of caspase-9, a key player of the mitochondrial death pathway, was significantly lower in UB-treated hearts post-I/R. UB, most likely acting via CXCR4, plays a protective role in H/R-induced myocyte apoptosis and myocardial I/R injury via modulation of mitochondrial homeostasis and the mitochondrial death pathway of apoptosis.
Collapse
Affiliation(s)
- Suman Dalal
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN 37614, USA
| | - Christopher R Daniels
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Ying Li
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Gary L Wright
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Mahipal Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Krishna Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN 37614, USA.,James H Quillen Veterans Affairs Medical Center, Mountain Home, TN 37684, USA
| |
Collapse
|
4
|
Protective role of the deSUMOylating enzyme SENP3 in myocardial ischemia-reperfusion injury. PLoS One 2019; 14:e0213331. [PMID: 30973885 PMCID: PMC6459529 DOI: 10.1371/journal.pone.0213331] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/02/2019] [Indexed: 12/26/2022] Open
Abstract
Interruption of blood supply to the heart is a leading cause of death and disability. However, the molecular events that occur during heart ischemia, and how these changes prime consequent cell death upon reperfusion, are poorly understood. Protein SUMOylation is a post-translational modification that has been strongly implicated in the protection of cells against a variety of stressors, including ischemia-reperfusion. In particular, the SUMO2/3-specific protease SENP3 has emerged as an important determinant of cell survival after ischemic infarct. Here, we used the Langendorff perfusion model to examine changes in the levels and localisation of SUMOylated target proteins and SENP3 in whole heart. We observed a 50% loss of SENP3 from the cytosolic fraction of hearts after preconditioning, a 90% loss after ischemia and an 80% loss after ischemia-reperfusion. To examine these effects further, we performed ischemia and ischemia-reperfusion experiments in the cardiomyocyte H9C2 cell line. Similar to whole hearts, ischemia induced a decrease in cytosolic SENP3. Furthermore, shRNA-mediated knockdown of SENP3 led to an increase in the rate of cell death upon reperfusion. Together, our results indicate that cardiac ischemia dramatically alter levels of SENP3 and suggest that this may a mechanism to promote cell survival after ischemia-reperfusion in heart.
Collapse
|
5
|
Yang Y, Zhao L, Ma J. Penehyclidine hydrochloride preconditioning provides cardiac protection in a rat model of myocardial ischemia/reperfusion injury via the mechanism of mitochondrial dynamics mechanism. Eur J Pharmacol 2017; 813:130-139. [PMID: 28755984 DOI: 10.1016/j.ejphar.2017.07.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/15/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023]
Abstract
To investigate that penehyclidine hydrochloride (PHC) plays a cardiac protection role in myocardial ischemia/reperfusion injury (IRI) through mitochondrial dynamics mechanism. Rat model of myocardial I/R injury was established by the ligation of left anterior descending coronary artery for 30min followed by 3h perfusion. Before I/R, the rats were pretreated with or without PHC. Cardiac function was measured by echocardiography. The activities/levels of myocardial enzymes, oxidants and antioxidant enzymes were detected. Evans blue/TTC double staining was performed to assess infarct size. Cell apoptosis was evaluated by TUNEL assay. Western blot and real time fluorescent quantitative PCR was performed to analyze the expression of Drp1, Mfn1, Mfn2. Meanwhile, the rats were given a single injection of PHC before I/R. The effects of PHC on myocardial infarct and cardiac function were investigated after 7 days post-reperfusion. Our results showed that PHC pretreatment improved imbalance of mitochondrial dynamics induced by oxidative stressor in IRI. PHC preconditioning alleviated apoptotic rate of cell by improving the imbalance of mitochondrial dynamics in IRI. Meanwhile, we showed that PHC remarkably improved cardiac function, myocardial injury by decreasing infarct size and attenuated levels of myocardial enzyme. Additionally, PHC also exerted long-term cardiac protection in a rat model of I/R injury by decreasing infarct size and improving cardiac function. These results suggested that PHC could efficiently protect the rats against I/R-induced myocardial injury via the mechanism of mitochondrial dynamics.
Collapse
Affiliation(s)
- Yanli Yang
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| | - Liyun Zhao
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Jun Ma
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| |
Collapse
|
6
|
PEDF Inhibits the Activation of NLRP3 Inflammasome in Hypoxia Cardiomyocytes through PEDF Receptor/Phospholipase A2. Int J Mol Sci 2016; 17:ijms17122064. [PMID: 27973457 PMCID: PMC5187864 DOI: 10.3390/ijms17122064] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/26/2016] [Accepted: 12/02/2016] [Indexed: 12/23/2022] Open
Abstract
The nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome has been linked to sterile inflammation, which is involved in ischemic injury in myocardial cells. Pigment epithelium-derived factor (PEDF) is a multifunctional secreted glycoprotein with many biological activities, such as anti-inflammatory, antioxidant and anti-angiogenic properties. However, it is not known whether and how PEDF acts to regulate the activation of the NLRP3 inflammasome in cardiomyocytes. In the present study, we used the neonatal cardiomyocytes models of ischemia-like conditions to evaluate the mitochondrial fission and the activation of the NLRP3 inflammasome. We also determined the mechanism by which PEDF inhibits hypoxia-induced activation of the NLRP3 inflammasome. We found that PEDF decreased the activation of the NLRP3 inflammasome in neonatal cardiomyocytes through pigment epithelial-derived factor receptor/calcium-independent phospholipase A2 (PEDFR/iPLA2). Meanwhile, PEDF reduced Drp1-induced mitochondrial fission and mitochondrial fission-induced mitochondrial DNA (mtDNA), as well as mitochondrial reactive oxygen species (mtROS) release into cytosol through PEDFR/iPLA2. We also found that PEDF inhibited mitochondrial fission-induced NLRP3 inflammasome activation. Furthermore, previous research has found that endogenous cytosolic mtDNA and mtROS can serve as activators of NLRP3 inflammasome activity. Therefore, we hypothesized that PEDF can protect against hypoxia-induced activation of the NLRP3 inflammasome by inhibiting mitochondrial fission though PEDFR/iPLA2.
Collapse
|
7
|
Billur D, Tuncay E, Okatan EN, Olgar Y, Durak AT, Degirmenci S, Can B, Turan B. Interplay Between Cytosolic Free Zn 2+ and Mitochondrion Morphological Changes in Rat Ventricular Cardiomyocytes. Biol Trace Elem Res 2016; 174:177-188. [PMID: 27107885 DOI: 10.1007/s12011-016-0704-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/12/2016] [Indexed: 12/11/2022]
Abstract
The Zn2+ in cardiomyocytes is buffered by structures near T-tubulus and/or sarcoplasmic/endoplasmic reticulum (S(E)R) while playing roles as either an antioxidant or a toxic agent, depending on the concentration. Therefore, we aimed first to examine a direct effect of ZnPO4 (extracellular exposure) or Zn2+ pyrithione (ZnPT) (intracellular exposure) application on the structure of the mitochondrion in ventricular cardiomyocytes by using histological investigations. The light microscopy data demonstrated that Zn2+ exposure induced marked increases on cellular surface area, an indication of hypertrophy, in a concentration-dependent manner. Furthermore, a whole-cell patch-clamp measurement of cell capacitance also supported the hypertrophy in the cells. We observed marked increases in mitochondrial matrix/cristae area and matrix volume together with increased lysosome numbers in ZnPO4- or ZnPT-incubated cells by using transmission electron microscopy, again in a concentration-dependent manner. Furthermore, we observed notable clustering and vacuolated mitochondrion, markedly disrupted and damaged myofibrils, and electron-dense small granules in Zn2+-exposed cells together with some implications of fission-fusion defects in the mitochondria. Moreover, we observed marked depolarization in mitochondrial membrane potential during 1-μM ZnPT minute applications by using confocal microscopy. We also showed that 1-μM ZnPT incubation induced significant increases in the phosphorylation levels of GSK3β (Ser21 and Ser9), Akt (Ser473), and NFκB (Ser276 and Thr254) together with increased expression levels in ER stress proteins such as GRP78 and calregulin. Furthermore, a new key player at ER-mitochondria sites, promyelocytic leukemia protein (PML) level, was markedly increased in ZnPT-incubated cells. As a summary, our present data suggest that increased cytosolic free Zn2+ can induce marked alterations in mitochondrion morphology as well as depolarization in mitochondrion membrane potential and changes in some cytosolic signaling proteins as well as a defect in ER-mitochondria cross talk.
Collapse
Affiliation(s)
- Deniz Billur
- Department of Histology-Embryology, Faculty of Medicine, Ankara University, 06100, Ankara, Turkey
| | - Erkan Tuncay
- Department of Biophysics, Faculty of Medicine, Ankara University, 06100, Ankara, Turkey
| | - Esma Nur Okatan
- Department of Biophysics, Faculty of Medicine, Ankara University, 06100, Ankara, Turkey
| | - Yusuf Olgar
- Department of Biophysics, Faculty of Medicine, Ankara University, 06100, Ankara, Turkey
| | - Aysegul Toy Durak
- Department of Biophysics, Faculty of Medicine, Ankara University, 06100, Ankara, Turkey
| | - Sinan Degirmenci
- Department of Biophysics, Faculty of Medicine, Ankara University, 06100, Ankara, Turkey
| | - Belgin Can
- Department of Histology-Embryology, Faculty of Medicine, Ankara University, 06100, Ankara, Turkey
| | - Belma Turan
- Department of Biophysics, Faculty of Medicine, Ankara University, 06100, Ankara, Turkey.
| |
Collapse
|
8
|
Forini F, Nicolini G, Iervasi G. Mitochondria as key targets of cardioprotection in cardiac ischemic disease: role of thyroid hormone triiodothyronine. Int J Mol Sci 2015; 16:6312-36. [PMID: 25809607 PMCID: PMC4394534 DOI: 10.3390/ijms16036312] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/10/2015] [Accepted: 03/12/2015] [Indexed: 12/30/2022] Open
Abstract
Ischemic heart disease is the major cause of mortality and morbidity worldwide. Early reperfusion after acute myocardial ischemia has reduced short-term mortality, but it is also responsible for additional myocardial damage, which in the long run favors adverse cardiac remodeling and heart failure evolution. A growing body of experimental and clinical evidence show that the mitochondrion is an essential end effector of ischemia/reperfusion injury and a major trigger of cell death in the acute ischemic phase (up to 48–72 h after the insult), the subacute phase (from 72 h to 7–10 days) and chronic stage (from 10–14 days to one month after the insult). As such, in recent years scientific efforts have focused on mitochondria as a target for cardioprotective strategies in ischemic heart disease and cardiomyopathy. The present review discusses recent advances in this field, with special emphasis on the emerging role of the biologically active thyroid hormone triiodothyronine (T3).
Collapse
Affiliation(s)
- Francesca Forini
- CNR Institute of Clinical Physiology, Via G. Moruzzi 1, Pisa 56124, Italy.
| | - Giuseppina Nicolini
- CNR Institute of Clinical Physiology, Via G. Moruzzi 1, Pisa 56124, Italy.
- Tuscany Region G. Monasterio Foundation, Via G. Moruzzi 1, Pisa 56124, Italy.
| | - Giorgio Iervasi
- CNR Institute of Clinical Physiology, Via G. Moruzzi 1, Pisa 56124, Italy.
| |
Collapse
|
9
|
Qipshidze-Kelm N, Piell KM, Solinger JC, Cole MP. Co-treatment with conjugated linoleic acid and nitrite protects against myocardial infarction. Redox Biol 2013; 2:1-7. [PMID: 24363996 PMCID: PMC3863133 DOI: 10.1016/j.redox.2013.10.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 10/29/2013] [Accepted: 10/29/2013] [Indexed: 12/22/2022] Open
Abstract
According to the CDC, the most common type of heart disease is coronary artery disease, which commonly leads to myocardial infarction (MI). Therapeutic approaches to lessen the resulting cardiovascular injury associated with MI are limited. Recently, MicroRNAs (miRNAs) have been shown to act as negative regulators of gene expression by inhibiting mRNA translation and/or stimulating mRNA degradation. A single miRNA can modulate physiological or disease phenotypes by regulating whole functional systems. Importantly, miRNAs can regulate cardiac function, thereby modulating heart muscle contraction, heart growth and morphogenesis. MicroRNA-499 (miRNA-499) is a cardiac-specific miRNA that when elevated causes cardiomyocyte hypertrophy, in turn preventing cardiac dysfunction during MI. Previous studies revealed that combination treatment with conjugated linoleic acid (cLA) and nitrite preserved cardiovascular function in mice. Therefore, it was hypothesized that cLA and nitrite may regulate miRNA-499, thus providing cardiac protection during MI. To test this hypothesis, 12-week old mice were treated with cLA (10 mg/kg/d-via osmotic mini-pump) or cLA and nitrite (50 ppm-drinking water) 3 days prior to MI (ligation of the left anterior descending artery). Echocardiography and pressure–volume (PV)-loop analysis revealed that cLA and nitrite-treated MI mice had improved heart function (10 days following MI) compared to untreated MI mice. Treatment with cLA and nitrite significantly induced levels of miRNA-499 compared to untreated MI mice. In addition, treatment with cLA and nitrite abolished MI-induced protein expression of p53 and dynamin-related protein-1 (DRP-1). Moreover, the antioxidant enzyme expression of heme oxygenase-1 (HO-1) was elevated in MI mice treated with cLA and nitrite compared to untreated MI mice. Confocal imaging on heart tissue confirmed expression the levels of HO-1 and p53. Taken together, these results suggest that therapeutic treatment with cLA and nitrite may provide significant protection during MI through regulation of both cardiac specific miRNA-499 and upregulation of phase 2 antioxidant enzyme expression. Co-treatment with cLA and nitrite increases cardiac specific miRNA-499, leading to cardioprotection in MI. MI-induced p53 and DRP-1 expression is abolished with cLA and nitrite treatment. HO-1 expression following treatment with cLA and nitrite is cardioprotective. Modulation of miR-499 may represent a therapeutic approach to treat apoptosis-related cardiac disease, including MI.
Collapse
Affiliation(s)
- Natia Qipshidze-Kelm
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40202, United States ; Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40202, United States
| | - Kellianne M Piell
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40202, United States
| | - Jane C Solinger
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40202, United States
| | - Marsha P Cole
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40202, United States ; Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40202, United States
| |
Collapse
|
10
|
Abstract
SIGNIFICANCE Mitochondria are dynamic organelles capable of changing their shape and distribution by undergoing either fission or fusion. Changes in mitochondrial dynamics, which is under the control of specific mitochondrial fission and fusion proteins, have been implicated in cell division, embryonic development, apoptosis, autophagy, and metabolism. Although the machinery for modulating mitochondrial dynamics is present in the cardiovascular system, its function there has only recently been investigated. In this article, we review the emerging role of mitochondrial dynamics in cardiovascular health and disease. RECENT ADVANCES Changes in mitochondrial dynamics have been implicated in vascular smooth cell proliferation, cardiac development and differentiation, cardiomyocyte hypertrophy, myocardial ischemia-reperfusion injury, cardioprotection, and heart failure. CRITICAL ISSUES Many of the experimental studies investigating mitochondrial dynamics in the cardiovascular system have been confined to cardiac cell lines, vascular cells, or neonatal cardiomyocytes, in which mitochondria are distributed throughout the cytoplasm and are free to move. However, in the adult heart where mitochondrial movements are restricted by their tightly-packed distribution along myofibrils or beneath the subsarcolemma, the relevance of mitochondrial dynamics is less obvious. The investigation of transgenic mice deficient in cardiac mitochondrial fission or fusion proteins should help elucidate the role of mitochondrial dynamics in the adult heart. FUTURE DIRECTIONS Investigating the role of mitochondrial dynamics in cardiovascular health and disease should result in the identification of novel therapeutic targets for treating patients with cardiovascular disease, the leading cause of death and disability globally.
Collapse
Affiliation(s)
- Sang-Bing Ong
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, San Diego, CA, USA
| | | | | |
Collapse
|
11
|
The metastasis-promoting S100A4 protein confers neuroprotection in brain injury. Nat Commun 2013; 3:1197. [PMID: 23149742 DOI: 10.1038/ncomms2202] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 10/15/2012] [Indexed: 12/31/2022] Open
Abstract
Identification of novel pro-survival factors in the brain is paramount for developing neuroprotective therapies. The multifunctional S100 family proteins have important roles in many human diseases and are also upregulated by brain injury. However, S100 functions in the nervous system remain unclear. Here we show that the S100A4 protein, mostly studied in cancer, is overexpressed in the damaged human and rodent brain and released from stressed astrocytes. Genetic deletion of S100A4 exacerbates neuronal loss after brain trauma or excitotoxicity, increasing oxidative cell damage and downregulating the neuroprotective protein metallothionein I+II. We identify two neurotrophic motifs in S100A4 and show that these motifs are neuroprotective in animal models of brain trauma. Finally, we find that S100A4 rescues neurons via the Janus kinase/STAT pathway and, partially, the interleukin-10 receptor. Our data introduce S100A4 as a therapeutic target in neurodegeneration, and raise the entire S100 family as a potentially important factor in central nervous system injury.
Collapse
|
12
|
Autophagy upregulation promotes survival and attenuates doxorubicin-induced cardiotoxicity. Biochem Pharmacol 2012; 85:124-34. [PMID: 23107818 DOI: 10.1016/j.bcp.2012.10.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 10/04/2012] [Accepted: 10/08/2012] [Indexed: 01/12/2023]
Abstract
This study evaluated whether the manipulation of autophagy could attenuate the cardiotoxic effects of doxorubicin (DXR) in vitro as well as in a tumour-bearing mouse model of acute doxorubicin-induced cardiotoxicity. We examined the effect of an increase or inhibition of autophagy in combination with DXR on apoptosis, reactive oxygen species (ROS) production and mitochondrial function. H9C2 rat cardiac myoblasts were pre-treated with bafilomycin A1 (autophagy inhibitor, 10 nM) or rapamycin (autophagy inducer, 50 μM) followed by DXR treatment (3 μM). The augmentation of autophagy with rapamycin in the presence of DXR substantially ameliorated the detrimental effects induced by DXR. This combination treatment demonstrated improved cell viability, decreased apoptosis and ROS production and enhanced mitochondrial function. To corroborate these findings, GFP-LC3 mice were inoculated with a mouse breast cancer cell line (EO771). Following the appearance of tumours, animals were either treated with one injection of rapamycin (4 mg/kg) followed by two injections of DXR (10 mg/kg). Mice were then sacrificed and their hearts rapidly excised and utilized for biochemical and histological analyses. The combination treatment, rather than the combinants alone, conferred a cardioprotective effect. These hearts expressed down-regulation of the pro-apoptotic protein caspase-3 and cardiomyocyte cross-sectional area was preserved. These results strongly indicate that the co-treatment strategy with rapamycin can attenuate the cardiotoxic effects of DXR in a tumour-bearing mouse model.
Collapse
|
13
|
RETRACTED: Dynamic changes of mitochondrial fission proteins after transient cerebral ischemia in mice. Brain Res 2012; 1456:94-9. [DOI: 10.1016/j.brainres.2012.03.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 02/09/2012] [Accepted: 03/19/2012] [Indexed: 11/19/2022]
|
14
|
Liu W, Tian F, Kurata T, Morimoto N, Abe K. Dynamic changes of mitochondrial fusion and fission proteins after transient cerebral ischemia in mice. J Neurosci Res 2012; 90:1183-9. [DOI: 10.1002/jnr.23016] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Revised: 11/22/2011] [Accepted: 12/02/2011] [Indexed: 01/16/2023]
|
15
|
Hausenloy DJ, Lecour S, Yellon DM. Reperfusion injury salvage kinase and survivor activating factor enhancement prosurvival signaling pathways in ischemic postconditioning: two sides of the same coin. Antioxid Redox Signal 2011; 14:893-907. [PMID: 20615076 DOI: 10.1089/ars.2010.3360] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The discovery of ischemic postconditioning (IPost) has rejuvenated the field of cardioprotection. As an interventional strategy to be applied at the onset of myocardial reperfusion, the transition of IPost from a bench-side curiosity to potential clinical therapy has been impressively rapid. Its existence also confirms the existence of lethal myocardial reperfusion injury in man, suggesting that 40%-50% of the final reperfused myocardial infarct may actually be due to myocardial reperfusion injury. Intensive analysis of the signal transduction pathways underlying IPost has identified similarities with the signaling pathways underlying its preischemic counterpart, ischemic preconditioning. In this article, the reperfusion injury salvage kinase pathway and the more recently described survivor activating factor enhancement pathway, two apparently distinct signaling pathways that actually interact to convey the IPost stimulus from the cell surface to the mitochondria, where many of the prosurvival and death signals appear to converge. The elucidation of the reperfusion signaling pathways underlying IPost may result in the identification of novel pharmacological targets for cardioprotection.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, United Kingdom.
| | | | | |
Collapse
|
16
|
Ong SB, Hausenloy DJ. Mitochondrial morphology and cardiovascular disease. Cardiovasc Res 2010; 88:16-29. [PMID: 20631158 PMCID: PMC2936127 DOI: 10.1093/cvr/cvq237] [Citation(s) in RCA: 248] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 07/06/2010] [Accepted: 07/07/2010] [Indexed: 01/14/2023] Open
Abstract
Mitochondria are dynamic and are able to interchange their morphology between elongated interconnected mitochondrial networks and a fragmented disconnected arrangement by the processes of mitochondrial fusion and fission, respectively. Changes in mitochondrial morphology are regulated by the mitochondrial fusion proteins (mitofusins 1 and 2, and optic atrophy 1) and the mitochondrial fission proteins (dynamin-related peptide 1 and mitochondrial fission protein 1) and have been implicated in a variety of biological processes including embryonic development, metabolism, apoptosis, and autophagy, although the majority of studies have been largely confined to non-cardiac cells. Despite the unique arrangement of mitochondria in the adult heart, emerging data suggest that changes in mitochondrial morphology may be relevant to various aspects of cardiovascular biology-these include cardiac development, the response to ischaemia-reperfusion injury, heart failure, diabetes mellitus, and apoptosis. Interestingly, the machinery required for altering mitochondrial shape in terms of the mitochondrial fusion and fission proteins are all present in the adult heart, but their physiological function remains unclear. In this article, we review the current developments in this exciting new field of mitochondrial biology, the implications for cardiovascular physiology, and the potential for discovering novel therapeutic strategies for treating cardiovascular disease.
Collapse
Affiliation(s)
| | - Derek J. Hausenloy
- The Hatter Cardiovascular Institute, University College London Hospital and Medical School, 67 Chenies Mews, London WC1E 6HX, UK
| |
Collapse
|
17
|
Ong SB, Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation 2010; 121:2012-22. [PMID: 20421521 DOI: 10.1161/circulationaha.109.906610] [Citation(s) in RCA: 783] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Whether alterations in mitochondrial morphology affect the susceptibility of the heart to ischemia/reperfusion injury is unknown. We hypothesized that modulating mitochondrial morphology protects the heart against ischemia/reperfusion injury. METHODS AND RESULTS In response to ischemia, mitochondria in HL-1 cells (a cardiac-derived cell line) undergo fragmentation, a process that is dependent on the mitochondrial fission protein dynamin-related protein 1 (Drp1). Transfection of HL-1 cells with the mitochondrial fusion proteins mitofusin 1 or 2 or with Drp1(K38A), a dominant-negative mutant form of Drp1, increased the percentage of cells containing elongated mitochondria (65+/-4%, 69+/-5%, and 63+/-6%, respectively, versus 46+/-6% in control: n=80 cells per group; P<0.05), decreased mitochondrial permeability transition pore sensitivity (by 2.4+/-0.5-, 2.3+/-0.7-, and 2.4+/-0.3-fold, respectively; n=80 cells per group; P<0.05), and reduced cell death after simulated ischemia/reperfusion injury (11.6+/-3.9%, 16.2+/-3.9%, and 12.1+/-2.9%, respectively, versus 41.8+/-4.1% in control: n=320 cells per group; P<0.05). Treatment of HL-1 cells with mitochondrial division inhibitor-1, a pharmacological inhibitor of Drp1, replicated these beneficial effects. Interestingly, elongated interfibrillar mitochondria were identified in the adult rodent heart with confocal and electron microscopy, and in vivo treatment with mitochondrial division inhibitor-1 increased the percentage of elongated mitochondria from 3.6+/-0.5% to 14.5+/-2.8% (P=0.023). Finally, treatment of adult murine cardiomyocytes with mitochondrial division inhibitor-1 reduced cell death and inhibited mitochondrial permeability transition pore opening after simulated ischemia/reperfusion injury, and in vivo treatment with mitochondrial division inhibitor-1 reduced myocardial infarct size in mice subject to coronary artery occlusion and reperfusion (21.0+/-2.2% with mitochondrial division inhibitor-1 versus 48.0+/-4.5% in control; n=6 animals per group; P<0.05). CONCLUSIONS Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury, suggesting a novel pharmacological strategy for cardioprotection.
Collapse
Affiliation(s)
- Sang-Bing Ong
- The Hatter Cardiovascular Institute, University College London Hospital, UK
| | | | | | | | | | | |
Collapse
|
18
|
Suppression of hippocampal TRPM7 protein prevents delayed neuronal death in brain ischemia. Nat Neurosci 2009; 12:1300-7. [PMID: 19734892 DOI: 10.1038/nn.2395] [Citation(s) in RCA: 228] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 08/11/2009] [Indexed: 11/09/2022]
Abstract
Cardiac arrest victims may experience transient brain hypoperfusion leading to delayed death of hippocampal CA1 neurons and cognitive impairment. We prevented this in adult rats by inhibiting the expression of transient receptor potential melastatin 7 (TRPM7), a transient receptor potential channel that is essential for embryonic development, is necessary for cell survival and trace ion homeostasis in vitro, and whose global deletion in mice is lethal. TRPM7 was suppressed in CA1 neurons by intrahippocampal injections of viral vectors bearing shRNA specific for TRPM7. This had no ill effect on animal survival, neuronal and dendritic morphology, neuronal excitability, or synaptic plasticity, as exemplified by robust long-term potentiation (LTP). However, TRPM7 suppression made neurons resistant to ischemic death after brain ischemia and preserved neuronal morphology and function. Also, it prevented ischemia-induced deficits in LTP and preserved performance in fear-associated and spatial-navigational memory tasks. Thus, regional suppression of TRPM7 is feasible, well tolerated and inhibits delayed neuronal death in vivo.
Collapse
|
19
|
Hausenloy DJ, Ong SB, Yellon DM. The mitochondrial permeability transition pore as a target for preconditioning and postconditioning. Basic Res Cardiol 2009; 104:189-202. [PMID: 19242644 DOI: 10.1007/s00395-009-0010-x] [Citation(s) in RCA: 201] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 01/25/2009] [Accepted: 01/27/2009] [Indexed: 12/19/2022]
Abstract
The experimental evidence supporting the mitochondrial permeability transition pore (mPTP) as a major mediator of lethal myocardial reperfusion injury and therefore a critical target for cardioprotection is persuasive. Although, its molecular identity eludes investigators, it is generally accepted that mitochondrial cyclophilin-D, the target for the inhibitory effects of cyclosporine-A on the mPTP, is a regulatory component of the mPTP. Animal myocardial infarction studies and a recent clinical proof-of-concept study have demonstrated that pharmacologically inhibiting its opening at the onset of myocardial reperfusion reduces myocardial infarct size in the region of 30-50%. Interestingly, the inhibition of mPTP opening at this time appears to underpin the infarct-limiting effects of the endogenous cardioprotective strategies of ischemic preconditioning (IPC) and postconditioning (IPost). However, the mechanism underlying this inhibitory action of IPC and IPost on mPTP opening is unclear. The objectve of this review article will be to explore the potential mechanisms which link IPC and IPost to mPTP inhibition in the reperfused heart.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Institute and Center for Cardiology, University College London Hospitals and Medical School, Grafton Way, London, UK.
| | | | | |
Collapse
|
20
|
Cardioprotection: a radical view Free radicals in pre and postconditioning. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1787:781-93. [PMID: 19248760 DOI: 10.1016/j.bbabio.2009.02.008] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 02/12/2009] [Accepted: 02/13/2009] [Indexed: 12/13/2022]
Abstract
A series of brief (a few minutes) ischemia/reperfusion cycles (ischemic preconditioning, IP) limits myocardial injury produced by a subsequent prolonged period of coronary artery occlusion and reperfusion. Postconditioning (PostC), which is a series of brief (a few seconds) reperfusion/ischemia cycles at reperfusion onset, attenuates also ischemia/reperfusion injury. In recent years the main idea has been that reactive oxygen species (ROS) play an essential, though double-edged, role in cardioprotection: they may participate in reperfusion injury or may play a role as signaling elements of protection in the pre-ischemic phase. It has been demonstrated that preconditioning triggering is redox-sensitive, using either ROS scavengers or ROS generators. We have shown that nitroxyl triggers preconditioning via pro-oxidative, and/or nitrosative stress-related mechanism(s). Several metabolites, including acetylcholine, bradykinin, opioids and phenylephrine, trigger preconditioning-like protection via a mitochondrial K(ATP)-ROS-dependent mechanism. Intriguingly, and contradictory to the above mentioned theory of ROS as an obligatory part of reperfusion-induced damage, some studies suggest the possibility that some ROS at low concentrations could protect ischemic hearts against reperfusion injury. Yet, we demonstrated that ischemic PostC is also a cardioprotective phenomenon that requires the intervention of redox signaling to be protective. Emerging evidence suggests that in a preconditioning scenario a redox signal is required during the first few minutes of myocardial reperfusion following the index ischemic period. Intriguingly, the ROS signaling in the early reperfusion appear crucial to both preconditioning- and postconditioning-induced protection. Therefore, our and others' results suggest that the role of ROS in reperfusion may be reconsidered as they are not only deleterious.
Collapse
|
21
|
Murray J, Capaldi RA. Screening for the metabolic basis of neurodegeneration: developing a focused proteomic approach. Ann N Y Acad Sci 2009; 1147:348-57. [PMID: 19076456 DOI: 10.1196/annals.1427.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Metabolism is controlled by a complex system of transcriptional events and posttranslational modifications stimulated by substrate and metabolite availability. It is becoming clear that neurodegenerative diseases are a symptom of a deficiency in the regulation or execution of metabolic reactions. Mitochondria, as the central organelles in metabolic regulation as well as the chief generators of reactive species, clearly have a role to play in the etiology of neurodegenerative conditions. We are developing antibody-based capture arrays to determine multiple parameters of key mitochondrial proteins. Parameters include enzyme activity, quantity, oxidative modification (including nitrative and oxidative stress), and regulation (phosphorylation and acetylation). At this time the core of this array focuses on the enzymes of oxidative phosphorylation. We continue to expand this array as antibodies for enzyme isolation and modification detection become available. Here we demonstrate the use of this array by analyzing the proteomic differences in oxidative phosphorylation enzymes between human heart and liver tissues, cells grown in media promoting aerobic versus anaerobic metabolism, and the catalytic/proteomic effects of mitochondria exposed to oxidative stress.
Collapse
|
22
|
Leung AM, Redlak MJ, Miller TA. Aspirin-induced mucosal cell death in human gastric cells: role of a caspase-independent mechanism. Dig Dis Sci 2009; 54:28-35. [PMID: 18612824 DOI: 10.1007/s10620-008-0321-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Accepted: 05/06/2008] [Indexed: 12/09/2022]
Abstract
Aspirin and other nonsteroidal anti-inflammatory drugs (NSAIDs) are commonly used for the treatment of pain and inflammation. Their use may result in gastroduodenal side effects, such as gastric irritation and ulcer formation. Although various strategies have been employed to minimize these adverse effects induced by NSAIDs, effective therapeutic targeting of this problem has been prevented by an incomplete understanding of the mechanisms underlying their pathogenesis. This study was undertaken to determine the role that non-caspase-mediated apoptosis plays in inducing cellular injury and death in gastric mucosa exposed to aspirin. We proposed that the responsible mechanism was through mitochondrial failure, increased mitochondrial membrane permeability, and translocation of the intramitochondrial protein apoptosis-inducing factor (AIF). Human gastric adenocarcinoma mucosal cells (AGS cells) received no pretreatment or were preincubated with caspase inhibitors for 30 min. Cells were then treated with 40 mM aspirin for 2-4 h. Apoptosis was assessed by measuring the DNA-histone complex formation. Cell viability was determined by an acridine orange-ethidium bromide (EtBr) assay. The activation of AIF was evaluated by both Western blotting of the cytosol and mitochondrial extracts as well as by visualization and staining using fluorescence microscopy. Results showed that caspase inhibitor preincubation decreased DNA-histone complex formation when compared to aspirin treatment alone. Based on light microscope visualization, however, we determined that caspase inhibitor preincubation was unable to prevent AGS cell damage and death. These findings were confirmed by the acridine orange-EtBr test, which showed decreased cell viability with caspase inhibitor preincubation and aspirin treatment. We then tested whether non-caspase-mediated cell death occurred through an AIF mitochondrial pathway using Western blotting and fluorescence microscopy to determine AIF activation. The results showed that untreated cells had AIF localized to the mitochondria and cytosol. With 40 mM ASA at 4 h, translocation of AIF from the mitochondria to the nucleus occurred, showing activation. Caspase inhibition with z-VAD was unable to prevent AIF localization to the nucleus and subsequently unable to prevent cell death. Our results indicate that ASA in the presence of caspase inhibitors causes gastric mucosal cell death through a caspase-independent pathway suggestive of apoptosis-like programmed cell death. Effective therapeutic targeting of aspirin-induced apoptosis likely requires inhibition of both mitochondrial and caspase-mediated pathways.
Collapse
Affiliation(s)
- Anna M Leung
- Department of Surgery, Medical College of Virginia Campus of Virginia Commonwealth University, P.O. Box 980645, Richmond, Virginia 23298-0568, USA
| | | | | |
Collapse
|
23
|
Schwartz DR, Sack MN. Targeting the mitochondria to augment myocardial protection. Curr Opin Pharmacol 2008; 8:160-5. [PMID: 18243790 PMCID: PMC2361131 DOI: 10.1016/j.coph.2007.12.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 12/07/2007] [Accepted: 12/10/2007] [Indexed: 11/28/2022]
Abstract
The dynamic regulation of the structure, function and turnover of mitochondria is recognized as an immutable control node maintaining cellular integrity and homeostasis. The term 'mitohormesis' has recently been coined to describe the adaptive reprogramming of mitochondrial biology in response to low levels of metabolic substrate deprivation to augment subsequent mitochondrial and cellular tolerance to biological stress. Disruption of these regulatory programs gives rise to cardiovascular and neurodegenerative diseases, and augmentation or fine-tuning of these programs may ameliorate mitochondrial and global cellular stress tolerance. This is in part via the regulation of reactive oxygen species, calcium homeostasis, and in response to caloric restriction, the capacity to augment DNA repair. The objective of this manuscript is to briefly review these regulatory programs and to postulate novel therapeutic approaches with the primary goal of modulating mitochondria to enhance tolerance to cardiac ischemic stress.
Collapse
Affiliation(s)
- Daniel R Schwartz
- Translational Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD 20892-1454, United States
| | | |
Collapse
|