151
|
Jiang H, Xiao J, Kang B, Zhu X, Xin N, Wang Z. PI3K/SGK1/GSK3β signaling pathway is involved in inhibition of autophagy in neonatal rat cardiomyocytes exposed to hypoxia/reoxygenation by hydrogen sulfide. Exp Cell Res 2015; 345:134-40. [PMID: 26163895 DOI: 10.1016/j.yexcr.2015.07.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 06/19/2015] [Accepted: 07/06/2015] [Indexed: 11/16/2022]
Abstract
Excessive autophagy aggravates myocardial ischemia/reperfusion (IR) injury. Hydrogen sulfide (H2S) has been shown to possess a strong cardioprotective effect due to its anti-necrosis, anti-apoptosis, anti-oxidant and anti-inflammatory properties. Our previous study showed that H2S could also protect the myocardium against IR injury through its anti-autophagy effect in vivo, but the underlying mechanism remains unclear. The aim of the present study was to determine whether PI3K/SGK1/GSK3β signaling pathway was involved in the anti-autophagy effect of H2S against myocardial hypoxia/reoxygenation (HR) injury in vitro. Autophagy was significantly increased in cardiomyocytes subjected to HR, but it was down-regulated by H2S (NaHS donor). Blocking PI3K by LY294002 (a PI3K inhibitor) or knocking down SGK1 by SGK1 siRNA augmented autophagy and attenuated the anti-autophagy effect of H2S. However, blocking GSK3β by tws119 (a GSK3β inhibitor) produced an opposite effect. In addition, while treatment of neonatal rat cardiomyocytes with HR reduced cell viability and augmented cell injury, H2S significantly reversed it. Blocking PI3K or knocking down SGK1 aggravated HR injury and weakened the protective effect of H2S, while blocking GSK3β produced an opposite effect. In conclusion, H2S can inhibit autophagy in neonatal rat cardiomyocytes exposed to H/R and exert a cardioprotective effect at least partly by regulating PI3K/SGK1/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Huan Jiang
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jian Xiao
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Bo Kang
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiaoyan Zhu
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Ni Xin
- Department of Physiology, Second Military Medical University, Shanghai, China.
| | - Zhinong Wang
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
152
|
Ge H, Zhao M, Lee S, Xu Z. Mitochondrial Src tyrosine kinase plays a role in the cardioprotective effect of ischemic preconditioning by modulating complex I activity and mitochondrial ROS generation. Free Radic Res 2015; 49:1210-7. [DOI: 10.3109/10715762.2015.1050013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
153
|
Cardioprotective Signature of Short-Term Caloric Restriction. PLoS One 2015; 10:e0130658. [PMID: 26098549 PMCID: PMC4476723 DOI: 10.1371/journal.pone.0130658] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 05/25/2015] [Indexed: 12/04/2022] Open
Abstract
Objective To understand the molecular pathways underlying the cardiac preconditioning effect of short-term caloric restriction (CR). Background Lifelong CR has been suggested to reduce the incidence of cardiovascular disease through a variety of mechanisms. However, prolonged adherence to a CR life-style is difficult. Here we reveal the pathways that are modulated by short-term CR, which are associated with protection of the mouse heart from ischemia. Methods Male 10-12 wk old C57bl/6 mice were randomly assigned to an ad libitum (AL) diet with free access to regular chow, or CR, receiving 30% less food for 7 days (d), prior to myocardial infarction (MI) via permanent coronary ligation. At d8, the left ventricles (LV) of AL and CR mice were collected for Western blot, mRNA and microRNA (miR) analyses to identify cardioprotective gene expression signatures. In separate groups, infarct size, cardiac hemodynamics and protein abundance of caspase 3 was measured at d2 post-MI. Results This short-term model of CR was associated with cardio-protection, as evidenced by decreased infarct size (18.5±2.4% vs. 26.6±1.7%, N=10/group; P=0.01). mRNA and miR profiles pre-MI (N=5/group) identified genes modulated by short-term CR to be associated with circadian clock, oxidative stress, immune function, apoptosis, metabolism, angiogenesis, cytoskeleton and extracellular matrix (ECM). Western blots pre-MI revealed CR-associated increases in phosphorylated Akt and GSK3ß, reduced levels of phosphorylated AMPK and mitochondrial related proteins PGC-1α, cytochrome C and cyclooxygenase (COX) IV, with no differences in the levels of phosphorylated eNOS or MAPK (ERK1/2; p38). CR regimen was also associated with reduced protein abundance of cleaved caspase 3 in the infarcted heart and improved cardiac function.
Collapse
|
154
|
Magnetic resonance spectroscopy of the ischemic brain under lithium treatment. Link to mitochondrial disorders under stroke. Chem Biol Interact 2015; 237:175-82. [PMID: 26079057 DOI: 10.1016/j.cbi.2015.06.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/22/2015] [Accepted: 06/05/2015] [Indexed: 11/22/2022]
Abstract
Recent evidence suggests that mitochondria are one of the main factors in the pathogenesis in different organs including brain. The pathogenesis after brain damage is caused not only by the change in bioenergetics, but also involves impairment of alternative functions of mitochondria, particularly those related to the control of cell death. In this study we evaluated partial metabolic pathways under the simulation of a stroke by using the occlusion of the middle cerebral artery in rats. The analysis shows that the induced switch to a non-oxidative energy metabolism (glycolysis) due to the block of tissue oxygen supply does not ensure the adequate supply of the tissue with ATP. Moreover, the well-known acidification of the ischemic tissue is not associated with the so-called traditionally and incorrectly considered "lactic acidosis" (the generation of lactate from glucose by itself does not lead to excessive generation of protons), but occurs because of the consumption of tissue ATP under its reduced resynthesis. Incubation of mitochondria isolated from normal rat brain at neutral and slightly acidic pH, mimicking the intracellular pH of normal and ischemic tissues correspondingly, revealed serious changes in mitochondrial bioenergetics, partially reflected in the magnitude of respiratory control and the basal and maximally stimulated respiration rates. Measurement of available metabolites by (1)H MR spectra of normal and ischemia-damaged brains showed a significant increase in lactate and myo-inositol and a moderate decrease in N-acetylaspartate 24h after reperfusion. Remarkably, the administration of lithium chloride in the reperfusion phase normalized the levels of metabolites. Moreover, the introduction of lithium salts (chloride or succinate) in the bloodstream, restored after ischemia, reduced both the size of the ischemia-induced brain damage and the degree of brain swelling. Besides, post-ischemic introduction of lithium salts largely restored the neurological status of the animal.
Collapse
|
155
|
|
156
|
Arrázola MS, Silva-Alvarez C, Inestrosa NC. How the Wnt signaling pathway protects from neurodegeneration: the mitochondrial scenario. Front Cell Neurosci 2015; 9:166. [PMID: 25999816 PMCID: PMC4419851 DOI: 10.3389/fncel.2015.00166] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 04/14/2015] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder and is characterized by progressive memory loss and cognitive decline. One of the hallmarks of AD is the overproduction of amyloid-beta aggregates that range from the toxic soluble oligomer (Aβo) form to extracellular accumulations in the brain. Growing evidence indicates that mitochondrial dysfunction is a common feature of neurodegenerative diseases and is observed at an early stage in the pathogenesis of AD. Reports indicate that mitochondrial structure and function are affected by Aβo and can trigger neuronal cell death. Mitochondria are highly dynamic organelles, and the balance between their fusion and fission processes is essential for neuronal function. Interestingly, in AD, the process known as “mitochondrial dynamics” is also impaired by Aβo. On the other hand, the activation of the Wnt signaling pathway has an essential role in synaptic maintenance and neuronal functions, and its deregulation has also been implicated in AD. We have demonstrated that canonical Wnt signaling, through the Wnt3a ligand, prevents the permeabilization of mitochondrial membranes through the inhibition of the mitochondrial permeability transition pore (mPTP), induced by Aβo. In addition, we showed that non-canonical Wnt signaling, through the Wnt5a ligand, protects mitochondria from fission-fusion alterations in AD. These results suggest new approaches by which different Wnt signaling pathways protect neurons in AD, and support the idea that mitochondria have become potential therapeutic targets for the treatment of neurodegenerative disorders. Here we discuss the neuroprotective role of the canonical and non-canonical Wnt signaling pathways in AD and their differential modulation of mitochondrial processes, associated with mitochondrial dysfunction and neurodegeneration.
Collapse
Affiliation(s)
- Macarena S Arrázola
- Facultad de Ciencias Biológicas, Departamento de Biología Celular y Molecular, Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile Santiago, Chile
| | - Carmen Silva-Alvarez
- Facultad de Ciencias Biológicas, Departamento de Biología Celular y Molecular, Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile Santiago, Chile
| | - Nibaldo C Inestrosa
- Facultad de Ciencias Biológicas, Departamento de Biología Celular y Molecular, Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile Santiago, Chile ; Center for Healthy Brain Aging, School of Psychiatry, Faculty of Medicine, University of New South Wales Sydney, NSW, Australia ; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes Punta Arenas, Chile ; Centro UC Síndrome de Down, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
157
|
Kusch A, Schmidt M, Gürgen D, Postpieszala D, Catar R, Hegner B, Davidson MM, Mahmoodzadeh S, Dragun D. 17ß-Estradiol regulates mTORC2 sensitivity to rapamycin in adaptive cardiac remodeling. PLoS One 2015; 10:e0123385. [PMID: 25880554 PMCID: PMC4399939 DOI: 10.1371/journal.pone.0123385] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 02/18/2015] [Indexed: 11/19/2022] Open
Abstract
Adaptive cardiac remodeling is characterized by enhanced signaling of mTORC2 downstream kinase Akt. In females, 17ß-estradiol (E2), as well as Akt contribute essentially to sex-related premenopausal cardioprotection. Pharmacologic mTOR targeting with rapamycin is increasingly used for various clinical indications, yet burdened with clinical heterogeneity in therapy responses. The drug inhibits mTORC1 and less-so mTORC2. In male rodents, rapamycin decreases maladaptive cardiac hypertrophy whereas it leads to detrimental dilative cardiomyopathy in females. We hypothesized that mTOR inhibition could interfere with 17β-estradiol (E2)-mediated sexual dimorphism and adaptive cell growth and tested responses in murine female hearts and cultured female cardiomyocytes. Under physiological in vivo conditions, rapamycin compromised mTORC2 function only in female, but not in male murine hearts. In cultured female cardiomyocytes, rapamycin impaired simultaneously IGF-1 induced activation of both mTOR signaling branches, mTORC1 and mTORC2 only in presence of E2. Use of specific estrogen receptor (ER)α- and ERβ-agonists indicated involvement of both estrogen receptors (ER) in rapamycin effects on mTORC1 and mTORC2. Classical feedback mechanisms common in tumour cells with upregulation of PI3K signaling were not involved. E2 effect on Akt-pS473 downregulation by rapamycin was independent of ERK as shown by sequential mTOR and MEK-inhibition. Furthermore, regulatory mTORC2 complex defining component rictor phosphorylation at Ser1235, known to interfere with Akt-substrate binding to mTORC2, was not altered. Functionally, rapamycin significantly reduced trophic effect of E2 on cell size. In addition, cardiomyocytes with reduced Akt-pS473 under rapamycin treatment displayed decreased SERCA2A mRNA and protein expression suggesting negative functional consequences on cardiomyocyte contractility. Rictor silencing confirmed regulation of SERCA2A expression by mTORC2 in E2-cultured female cardiomyocytes. These data highlight a novel modulatory function of E2 on rapamycin effect on mTORC2 in female cardiomyocytes and regulation of SERCA2A expression by mTORC2. Conceivably, rapamycin abrogates the premenopausal “female advantage”.
Collapse
Affiliation(s)
- Angelika Kusch
- Department of Nephrology and Intensive Care Medicine, Charité—Campus Virchow Klinikum, Universitätsmedizin Berlin, Berlin, Germany
- Center for Cardiovascular Research, Charité, Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| | - Maria Schmidt
- Department of Nephrology and Intensive Care Medicine, Charité—Campus Virchow Klinikum, Universitätsmedizin Berlin, Berlin, Germany
| | - Dennis Gürgen
- Department of Nephrology and Intensive Care Medicine, Charité—Campus Virchow Klinikum, Universitätsmedizin Berlin, Berlin, Germany
- Center for Cardiovascular Research, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Daniel Postpieszala
- Department of Nephrology and Intensive Care Medicine, Charité—Campus Virchow Klinikum, Universitätsmedizin Berlin, Berlin, Germany
| | - Rusan Catar
- Department of Nephrology and Intensive Care Medicine, Charité—Campus Virchow Klinikum, Universitätsmedizin Berlin, Berlin, Germany
- Center for Cardiovascular Research, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Björn Hegner
- Department of Nephrology and Intensive Care Medicine, Charité—Campus Virchow Klinikum, Universitätsmedizin Berlin, Berlin, Germany
- Center for Cardiovascular Research, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Merci M. Davidson
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Shokoufeh Mahmoodzadeh
- Center for Cardiovascular Research, Charité, Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Duska Dragun
- Department of Nephrology and Intensive Care Medicine, Charité—Campus Virchow Klinikum, Universitätsmedizin Berlin, Berlin, Germany
- Center for Cardiovascular Research, Charité, Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
158
|
HOLZEROVÁ K, HLAVÁČKOVÁ M, ŽURMANOVÁ J, BORCHERT G, NECKÁŘ J, KOLÁŘ F, NOVÁK F, NOVÁKOVÁ O. Involvement of PKCε in Cardioprotection Induced by Adaptation to Chronic Continuous Hypoxia. Physiol Res 2015; 64:191-201. [DOI: 10.33549/physiolres.932860] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Continuous normobaric hypoxia (CNH) renders the heart more tolerant to acute ischemia/reperfusion injury. Protein kinase C (PKC) is an important component of the protective signaling pathway, but the contribution of individual PKC isoforms under different hypoxic conditions is poorly understood. The aim of this study was to analyze the expression of PKCε after the adaptation to CNH and to clarify its role in increased cardiac ischemic tolerance with the use of PKCε inhibitory peptide KP-1633. Adult male Wistar rats were exposed to CNH (10 % O2, 3 weeks) or kept under normoxic conditions. The protein level of PKCε and its phosphorylated form was analyzed by Western blot in homogenate, cytosolic and particulate fractions; the expression of PKCε mRNA was measured by RT-PCR. The effect of KP-1633 on cell viability and lactate dehydrogenase (LDH) release was analyzed after 25-min metabolic inhibition followed by 30-min re-energization in freshly isolated left ventricular myocytes. Adaptation to CNH increased myocardial PKCε at protein and mRNA levels. The application of KP-1633 blunted the hypoxia-induced salutary effects on cell viability and LDH release, while control peptide KP-1723 had no effect. This study indicates that PKCε is involved in the cardioprotective mechanism induced by CNH.
Collapse
Affiliation(s)
| | - M. HLAVÁČKOVÁ
- Department of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
159
|
Targeted disruption of PDE3B, but not PDE3A, protects murine heart from ischemia/reperfusion injury. Proc Natl Acad Sci U S A 2015; 112:E2253-62. [PMID: 25877153 DOI: 10.1073/pnas.1416230112] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Although inhibition of cyclic nucleotide phosphodiesterase type 3 (PDE3) has been reported to protect rodent heart against ischemia/reperfusion (I/R) injury, neither the specific PDE3 isoform involved nor the underlying mechanisms have been identified. Targeted disruption of PDE3 subfamily B (PDE3B), but not of PDE3 subfamily A (PDE3A), protected mouse heart from I/R injury in vivo and in vitro, with reduced infarct size and improved cardiac function. The cardioprotective effect in PDE3B(-/-) heart was reversed by blocking cAMP-dependent PKA and by paxilline, an inhibitor of mitochondrial calcium-activated K channels, the opening of which is potentiated by cAMP/PKA signaling. Compared with WT mitochondria, PDE3B(-/-) mitochondria were enriched in antiapoptotic Bcl-2, produced less reactive oxygen species, and more frequently contacted transverse tubules where PDE3B was localized with caveolin-3. Moreover, a PDE3B(-/-) mitochondrial fraction containing connexin-43 and caveolin-3 was more resistant to Ca(2+)-induced opening of the mitochondrial permeability transition pore. Proteomics analyses indicated that PDE3B(-/-) heart mitochondria fractions were enriched in buoyant ischemia-induced caveolin-3-enriched fractions (ICEFs) containing cardioprotective proteins. Accumulation of proteins into ICEFs was PKA dependent and was achieved by ischemic preconditioning or treatment of WT heart with the PDE3 inhibitor cilostamide. Taken together, these findings indicate that PDE3B deletion confers cardioprotective effects because of cAMP/PKA-induced preconditioning, which is associated with the accumulation of proteins with cardioprotective function in ICEFs. To our knowledge, our study is the first to define a role for PDE3B in cardioprotection against I/R injury and suggests PDE3B as a target for cardiovascular therapies.
Collapse
|
160
|
Abstract
Reperfusion is mandatory to salvage ischemic myocardium from infarction, but reperfusion per se contributes to injury and ultimate infarct size. Therefore, cardioprotection beyond that by timely reperfusion is needed to reduce infarct size and improve the prognosis of patients with acute myocardial infarction. The conditioning phenomena provide such cardioprotection, insofar as brief episodes of coronary occlusion/reperfusion preceding (ischemic preconditioning) or following (ischemic postconditioning) sustained myocardial ischemia with reperfusion reduce infarct size. Even ischemia/reperfusion in organs remote from the heart provides cardioprotection (remote ischemic conditioning). The present review characterizes the signal transduction underlying the conditioning phenomena, including their physical and chemical triggers, intracellular signal transduction, and effector mechanisms, notably in the mitochondria. Cardioprotective signal transduction appears as a highly concerted spatiotemporal program. Although the translation of ischemic postconditioning and remote ischemic conditioning protocols to patients with acute myocardial infarction has been fairly successful, the pharmacological recruitment of cardioprotective signaling has been largely disappointing to date.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
161
|
Rasola A, Bernardi P. Reprint of "The mitochondrial permeability transition pore and its adaptive responses in tumor cells". Cell Calcium 2015; 58:18-26. [PMID: 25828565 DOI: 10.1016/j.ceca.2015.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/06/2014] [Accepted: 10/07/2014] [Indexed: 02/07/2023]
Abstract
This review covers recent progress on the nature of the mitochondrial permeability transition pore (PTP) – a key effector in the mitochondrial pathways to cell death – and on the adaptive responses of tumor cells that desensitize the PTP to Ca(2+) and reactive oxygen species (ROS), thereby playing an important role in the resistance of tumors to cell death. The discovery that the PTP forms from dimers of F-ATP synthase; and the definition of the Ca(2+)- and ROS-dependent signaling pathways affecting the transition of the F-ATP synthase from an energy-conserving to an energy-dissipating device open new perspectives for therapeutic intervention in cancer cells.
Collapse
Affiliation(s)
- Andrea Rasola
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Italy.
| | - Paolo Bernardi
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Italy.
| |
Collapse
|
162
|
Bonora M, Wieckowski MR, Chinopoulos C, Kepp O, Kroemer G, Galluzzi L, Pinton P. Molecular mechanisms of cell death: central implication of ATP synthase in mitochondrial permeability transition. Oncogene 2015; 34:1475-86. [PMID: 24727893 DOI: 10.1038/onc.2014.96] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 02/20/2014] [Accepted: 02/27/2014] [Indexed: 12/14/2022]
Abstract
The term mitochondrial permeability transition (MPT) is commonly used to indicate an abrupt increase in the permeability of the inner mitochondrial membrane to low molecular weight solutes. Widespread MPT has catastrophic consequences for the cell, de facto marking the boundary between cellular life and death. MPT results indeed in the structural and functional collapse of mitochondria, an event that commits cells to suicide via regulated necrosis or apoptosis. MPT has a central role in the etiology of both acute and chronic diseases characterized by the loss of post-mitotic cells. Moreover, cancer cells are often relatively insensitive to the induction of MPT, underlying their increased resistance to potentially lethal cues. Thus, intense efforts have been dedicated not only at the understanding of MPT in mechanistic terms, but also at the development of pharmacological MPT modulators. In this setting, multiple mitochondrial and extramitochondrial proteins have been suspected to critically regulate the MPT. So far, however, only peptidylprolyl isomerase F (best known as cyclophilin D) appears to constitute a key component of the so-called permeability transition pore complex (PTPC), the supramolecular entity that is believed to mediate MPT. Here, after reviewing the structural and functional features of the PTPC, we summarize recent findings suggesting that another of its core components is represented by the c subunit of mitochondrial ATP synthase.
Collapse
Affiliation(s)
- M Bonora
- Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, Interdisciplinary Centre for the Study of Inflammation (ICSI), University of Ferrara, Ferrara, Italy
| | - M R Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - C Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - O Kepp
- 1] Equipe 11 labelisée par la Ligue Nationale contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France [2] Université Paris Descartes/Paris 5, Sorbonne Paris Cité, Paris, France [3] Metabolomics and Cell Biology platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
| | - G Kroemer
- 1] Equipe 11 labelisée par la Ligue Nationale contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France [2] Université Paris Descartes/Paris 5, Sorbonne Paris Cité, Paris, France [3] Metabolomics and Cell Biology platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France [4] Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - L Galluzzi
- 1] Equipe 11 labelisée par la Ligue Nationale contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France [2] Université Paris Descartes/Paris 5, Sorbonne Paris Cité, Paris, France [3] Gustave Roussy Comprehensive Cancer Center, Villejuif, France
| | - P Pinton
- Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, Interdisciplinary Centre for the Study of Inflammation (ICSI), University of Ferrara, Ferrara, Italy
| |
Collapse
|
163
|
McCubrey JA, Steelman LS, Bertrand FE, Davis NM, Sokolosky M, Abrams SL, Montalto G, D'Assoro AB, Libra M, Nicoletti F, Maestro R, Basecke J, Rakus D, Gizak A, Demidenko ZN, Cocco L, Martelli AM, Cervello M. GSK-3 as potential target for therapeutic intervention in cancer. Oncotarget 2015; 5:2881-911. [PMID: 24931005 PMCID: PMC4102778 DOI: 10.18632/oncotarget.2037] [Citation(s) in RCA: 377] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The serine/threonine kinase glycogen synthase kinase-3 (GSK-3) was initially identified and studied in the regulation of glycogen synthesis. GSK-3 functions in a wide range of cellular processes. Aberrant activity of GSK-3 has been implicated in many human pathologies including: bipolar depression, Alzheimer's disease, Parkinson's disease, cancer, non-insulin-dependent diabetes mellitus (NIDDM) and others. In some cases, suppression of GSK-3 activity by phosphorylation by Akt and other kinases has been associated with cancer progression. In these cases, GSK-3 has tumor suppressor functions. In other cases, GSK-3 has been associated with tumor progression by stabilizing components of the beta-catenin complex. In these situations, GSK-3 has oncogenic properties. While many inhibitors to GSK-3 have been developed, their use remains controversial because of the ambiguous role of GSK-3 in cancer development. In this review, we will focus on the diverse roles that GSK-3 plays in various human cancers, in particular in solid tumors. Recently, GSK-3 has also been implicated in the generation of cancer stem cells in various cell types. We will also discuss how this pivotal kinase interacts with multiple signaling pathways such as: PI3K/PTEN/Akt/mTORC1, Ras/Raf/MEK/ERK, Wnt/beta-catenin, Hedgehog, Notch and others.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology,Brody School of Medicine at East Carolina University Greenville, NC 27858 USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Zirpoli H, Abdillahi M, Quadri N, Ananthakrishnan R, Wang L, Rosario R, Zhu Z, Deckelbaum RJ, Ramasamy R. Acute administration of n-3 rich triglyceride emulsions provides cardioprotection in murine models after ischemia-reperfusion. PLoS One 2015; 10:e0116274. [PMID: 25559887 PMCID: PMC4283969 DOI: 10.1371/journal.pone.0116274] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 12/08/2014] [Indexed: 11/19/2022] Open
Abstract
Dietary n-3 fatty acids (FAs) may reduce cardiovascular disease risk. We questioned whether acute administration of n-3 rich triglyceride (TG) emulsions could preserve cardiac function and decrease injury after ischemia/reperfusion (I/R) insult. We used two different experimental models: in vivo, C57BL/6 mice were exposed to acute occlusion of the left anterior descending coronary artery (LAD), and ex-vivo, C57BL/6 murine hearts were perfused using Langendorff technique (LT). In the LAD model, mice treated with n-3 TG emulsion (1.5g/kg body weight), immediately after ischemia and 1h later during reperfusion, significantly reduced infarct size and maintained cardiac function (p<0.05). In the LT model, administration of n-3 TG emulsion (300mgTG/100ml) during reperfusion significantly improved functional recovery (p<0.05). In both models, lactate dehydrogenase (LDH) levels, as a marker of injury, were significantly reduced by n-3 TG emulsion. To investigate the mechanisms by which n-3 FAs protects hearts from I/R injury, we investigated changes in key pathways linked to cardioprotection. In the ex-vivo model, we showed that n-3 FAs increased phosphorylation of AKT and GSK3β proteins (p<0.05). Acute n-3 TG emulsion treatment also increased Bcl-2 protein level and reduced an autophagy marker, Beclin-1 (p<0.05). Additionally, cardioprotection by n-3 TG emulsion was linked to changes in PPARγ protein expression (p<0.05). Rosiglitazone and p-AKT inhibitor counteracted the positive effect of n-3 TG; GSK3β inhibitor plus n-3 TG significantly inhibited LDH release. We conclude that acute n-3 TG injection during reperfusion provides cardioprotection. This may prove to be a novel acute adjunctive reperfusion therapy after treating patients with myocardial infarction.
Collapse
Affiliation(s)
- Hylde Zirpoli
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Mariane Abdillahi
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Nosirudeen Quadri
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Radha Ananthakrishnan
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Lingjie Wang
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Rosa Rosario
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Zhengbin Zhu
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Richard J. Deckelbaum
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- * E-mail: (R. Ramasamy); (RJD)
| | - Ravichandran Ramasamy
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- * E-mail: (R. Ramasamy); (RJD)
| |
Collapse
|
165
|
Alam MR, Baetz D, Ovize M. Cyclophilin D and myocardial ischemia-reperfusion injury: a fresh perspective. J Mol Cell Cardiol 2015; 78:80-9. [PMID: 25281838 DOI: 10.1016/j.yjmcc.2014.09.026] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 09/23/2014] [Accepted: 09/25/2014] [Indexed: 01/06/2023]
Abstract
Reperfusion is characterized by a deregulation of ion homeostasis and generation of reactive oxygen species that enhance the ischemia-related tissue damage culminating in cell death. The mitochondrial permeability transition pore (mPTP) has been established as an important mediator of ischemia-reperfusion (IR)-induced necrotic cell death. Although a handful of proteins have been proposed to contribute in mPTP induction, cyclophilin D (CypD) remains its only bona fide regulatory component. In this review we summarize existing knowledge on the involvement of CypD in mPTP formation in general and its relevance to cardiac IR injury in specific. Moreover, we provide insights of recent advancements on additional functions of CypD depending on its interaction partners and post-translational modifications. Finally we emphasize the therapeutic strategies targeting CypD in myocardial IR injury. This article is part of a Special Issue entitled "Mitochondria: From Basic Mitochondrial Biology to Cardiovascular Disease".
Collapse
Affiliation(s)
- Muhammad Rizwan Alam
- INSERM U1060, CarMeN Laboratory, Claude Bernard Lyon 1 University, F-69373 Lyon, France
| | - Delphine Baetz
- INSERM U1060, CarMeN Laboratory, Claude Bernard Lyon 1 University, F-69373 Lyon, France
| | - Michel Ovize
- INSERM U1060, CarMeN Laboratory, Claude Bernard Lyon 1 University, F-69373 Lyon, France; Hospices Civils de Lyon, Hôpital Louis Pradel, Service d'Explorations Fonctionnelles Cardiovasculaires & CIC de Lyon, F-69394 Lyon, France.
| |
Collapse
|
166
|
Zaman J, Jeddi S, Ghasemi A. The effects of ischemic postconditioning on myocardial function and nitric oxide metabolites following ischemia-reperfusion in hyperthyroid rats. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:481-7. [PMID: 25598662 PMCID: PMC4296037 DOI: 10.4196/kjpp.2014.18.6.481] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/28/2014] [Accepted: 10/27/2014] [Indexed: 11/27/2022]
Abstract
Ischemic postconditioning (IPost) could decrease ischemia-reperfusion (IR) injury. It has not yet reported whether IPost is useful when ischemic heart disease is accompanied with co-morbidities like hyperthyroidism. The aim of this study was to examine the effect of IPost on myocardial IR injury in hyperthyroid male rats. Hyperthyroidism was induced with administration of thyroxine in drinking water (12 mg/L) over a period of 21 days. After thoracotomy, the hearts of control and hyperthyroid rats were perfused in the Langendorff apparatus and subjected to 30 minutes global ischemia, followed by 120 minutes reperfusion; IPost, intermittent early reperfusion, was induced instantly following ischemia. In control rats, IPost significantly improved the left ventricular developed pressure (LVDP) and ±dp/dt during reperfusion (p<0.05); however it had no effect in hyperthyroid rats. In addition, hyperthyroidism significantly increased basal NOx (nitrate+nitrite) content in serum (125.5±5.4 µmol/L vs. 102.8±3.7 µmol/L; p< 0.05) and heart (34.9±4.1 µmol/L vs. 19.9±1.94 µmol/L; p<0.05). In hyperthyroid groups, heart NOx concentration significantly increased after IR and IPost, whereas in the control groups, heart NOx were significantly higher after IR and lower after IPost (p< 0.05). IPost reduced infarct size (p<0.05) only in control groups. In hyperthyroid group subjected to IPost, aminoguanidine, an inducible nitric oxide (NO) inhibitor, significantly reduced both the infarct size and heart NOx concentrations. In conclusion, unlike normal rats, IPost cycles following reperfusion does not provide cardioprotection against IR injury in hyperthyroid rats; an effect that may be due to NO overproduction because it is restored by iNOS inhibition.
Collapse
Affiliation(s)
- Jalal Zaman
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran. ; Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran
| | - Sajjad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran. ; Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran. ; Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19395-4763, Iran
| |
Collapse
|
167
|
Bice JS, Baxter GF. Postconditioning signalling in the heart: mechanisms and translatability. Br J Pharmacol 2014; 172:1933-46. [PMID: 25303373 DOI: 10.1111/bph.12976] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/29/2014] [Accepted: 10/05/2014] [Indexed: 12/15/2022] Open
Abstract
The protective effect of ischaemic postconditioning (short cycles of reperfusion and reocclusion of a previously occluded vessel) was identified over a decade ago commanding intense interest as an approach for modifying reperfusion injury which contributes to infarct size in acute myocardial infarction. Elucidation of the major mechanisms of postconditioning has identified potential pharmacological targets for limitation of reperfusion injury. These include ligands for membrane-associated receptors, activators of phosphokinase survival signalling pathways and inhibitors of the mitochondrial permeability transition pore. In experimental models, numerous agents that target these mechanisms have shown promise as postconditioning mimetics. Nevertheless, clinical studies of ischaemic postconditioning and pharmacological postconditioning mimetics are equivocal. The majority of experimental research is conducted in animal models which do not fully portray the complexity of risk factors and comorbidities with which patients present and which we now know modify the signalling pathways recruited in postconditioning. Cohort size and power, patient selection, and deficiencies in clinical infarct size estimation may all represent major obstacles to assessing the therapeutic efficacy of postconditioning. Furthermore, chronic treatment of these patients with drugs like ACE inhibitors, statins and nitrates may modify signalling, inhibiting the protective effect of postconditioning mimetics, or conversely induce a maximally protected state wherein no further benefit can be demonstrated. Arguably, successful translation of postconditioning cannot occur until all of these issues are addressed, that is, experimental investigation requires more complex models that better reflect the clinical setting, while clinical investigation requires bigger trials with appropriate patient selection and standardization of clinical infarct size measurements.
Collapse
Affiliation(s)
- Justin S Bice
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | | |
Collapse
|
168
|
Rasola A, Bernardi P. The mitochondrial permeability transition pore and its adaptive responses in tumor cells. Cell Calcium 2014; 56:437-45. [PMID: 25454774 PMCID: PMC4274314 DOI: 10.1016/j.ceca.2014.10.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/06/2014] [Accepted: 10/07/2014] [Indexed: 01/12/2023]
Abstract
This review covers recent progress on the nature of the mitochondrial permeability transition pore (PTP) - a key effector in the mitochondrial pathways to cell death - and on the adaptive responses of tumor cells that desensitize the PTP to Ca(2+) and reactive oxygen species (ROS), thereby playing an important role in the resistance of tumors to cell death. The discovery that the PTP forms from dimers of F-ATP synthase; and the definition of the Ca(2+)- and ROS-dependent signaling pathways affecting the transition of the F-ATP synthase from an energy-conserving to an energy-dissipating device open new perspectives for therapeutic intervention in cancer cells.
Collapse
Affiliation(s)
- Andrea Rasola
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Italy.
| | - Paolo Bernardi
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Italy.
| |
Collapse
|
169
|
22nd annual meeting of Chinese Society of Anesthesiology. Br J Anaesth 2014. [DOI: 10.1093/bja/aeu337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
170
|
Halestrap AP, Pereira GC, Pasdois P. The role of hexokinase in cardioprotection - mechanism and potential for translation. Br J Pharmacol 2014; 172:2085-100. [PMID: 25204670 PMCID: PMC4386983 DOI: 10.1111/bph.12899] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 08/21/2014] [Accepted: 08/28/2014] [Indexed: 12/22/2022] Open
Abstract
Mitochondrial permeability transition pore (mPTP) opening plays a critical role in cardiac reperfusion injury and its prevention is cardioprotective. Tumour cell mitochondria usually have high levels of hexokinase isoform 2 (HK2) bound to their outer mitochondrial membranes (OMM) and HK2 binding to heart mitochondria has also been implicated in resistance to reperfusion injury. HK2 dissociates from heart mitochondria during ischaemia, and the extent of this correlates with the infarct size on reperfusion. Here we review the mechanisms and regulations of HK2 binding to mitochondria and how this inhibits mPTP opening and consequent reperfusion injury. Major determinants of HK2 dissociation are the elevated glucose‐6‐phosphate concentrations and decreased pH in ischaemia. These are modulated by the myriad of signalling pathways implicated in preconditioning protocols as a result of a decrease in pre‐ischaemic glycogen content. Loss of mitochondrial HK2 during ischaemia is associated with permeabilization of the OMM to cytochrome c, which leads to greater reactive oxygen species production and mPTP opening during reperfusion. Potential interactions between HK2 and OMM proteins associated with mitochondrial fission (e.g. Drp1) and apoptosis (B‐cell lymphoma 2 family members) in these processes are examined. Also considered is the role of HK2 binding in stabilizing contact sites between the OMM and the inner membrane. Breakage of these during ischaemia is proposed to facilitate cytochrome c loss during ischaemia while increasing mPTP opening and compromising cellular bioenergetics during reperfusion. We end by highlighting the many unanswered questions and discussing the potential of modulating mitochondrial HK2 binding as a pharmacological target. Linked Articles This article is part of a themed section on Conditioning the Heart – Pathways to Translation. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue‐8
Collapse
Affiliation(s)
- Andrew P Halestrap
- School of Biochemistry and Bristol CardioVascular, University of Bristol, Bristol, UK
| | | | | |
Collapse
|
171
|
Hsieh SR, Cheng WC, Su YM, Chiu CH, Liou YM. Molecular targets for anti-oxidative protection of green tea polyphenols against myocardial ischemic injury. Biomedicine (Taipei) 2014; 4:23. [PMID: 25520936 PMCID: PMC4264984 DOI: 10.7603/s40681-014-0023-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 10/14/2014] [Indexed: 12/20/2022] Open
Abstract
Ischemic heart disease is the leading cause of death worldwide. An improved understanding of the mechanisms involved in myocardial injury would allow intervention downstream in the pathway where certain drugs including natural products could be efficiently applied to target the end effectors of the cell death pathway. Green tea polyphenols (GTPs) have potent anti-oxidative capabilities, which may account for their beneficial effects in preventing oxidative stress associated with ischemia injury. Although studies have provided convincing evidence to support the protective effects of GTPs in cardiovascular system, the potential end effectors that mediate cardiac protection are only beginning to be addressed. Proteomics analyses widely used to identify the protein targets for many cardiovascular diseases have advanced the discovery of the signaling mechanism for GTPs-mediated cardio-protection. This review focuses on putative triggers, mediators, and end effectors for the GTPs-mediated cardio-protection signaling pathways engaged in myocardial ischemia crisis, allowing a promising natural product to be used for ameliorating oxidative stress associated with ischemic heart diseases.
Collapse
Affiliation(s)
- Shih-Rong Hsieh
- Department of Cardiovascular Surgery, Taichung Veterans General Hospital, 407 Taichung, Taiwan
| | - Wei-Chen Cheng
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, 300 Hsinchu, Taiwan
| | - Yi-Min Su
- Department of Life Sciences, National Chung-Hsing University, 402 No. 250, Kuokang Road, Taichung, Taiwan
| | - Chun-Hwei Chiu
- Department of Life Sciences, National Chung-Hsing University, 402 No. 250, Kuokang Road, Taichung, Taiwan
| | - Ying-Ming Liou
- Department of Life Sciences, National Chung-Hsing University, 402 No. 250, Kuokang Road, Taichung, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, 402 Taichung, Taiwan
| |
Collapse
|
172
|
Affiliation(s)
- Jennifer L Hall
- Lillehei Heart Institute, Division of Cardiology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
173
|
Zheng X, Zu L, Becker L, Cai ZP. Ischemic preconditioning inhibits mitochondrial permeability transition pore opening through the PTEN/PDE4 signaling pathway. Cardiology 2014; 129:163-73. [PMID: 25301476 DOI: 10.1159/000363646] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 05/14/2014] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Ischemic preconditioning (IPC) induces cardioprotection against ischemia-reperfusion (IR) injury by inhibiting the mitochondrial permeability transition pore (mPTP). Here, we tested the hypothesis that IPC-induced cardioprotection is mediated by the phosphatase PTEN and PDE4 (phosphodiesterase 4). METHODS Isolated hearts from wild-type mice (WT, n = 110) and myocyte-specific PTEN-knockout mice (PKO, n = 94) were exposed to IPC or control conditions followed by IR. Subcellular fractionation was performed by sucrose gradient ultracentrifugation. RESULTS IPC limited myocardial infarct size (IS) in WT mice. The PDE4 inhibitor rolipram abolished the protective effect of IPC. However, small IS was found in PKO hearts after IR, and IPC did not decrease IS but enlarged it in PKO hearts. IPC promoted PDE4D localization to caveolin-3-enriched fractions in WT mice by increasing Akt levels at the caveolae. In PKO hearts, basal PDE4D levels were elevated at the caveolae, and IPC decreased PDE4D levels. Consistent with the subcellular PDE4D protein levels and its activity, elevation in intracellular Ca(2+) levels in the ischemic heart and opening of mPTP after IR were inhibited by IPC in WT mice, but not by IPC in PKO mice. CONCLUSIONS IPC inhibits mPTP opening by regulating the PTEN/PDE4 signaling pathway.
Collapse
Affiliation(s)
- Xiaoxu Zheng
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | | | | | | |
Collapse
|
174
|
Nduhirabandi F, Huisamen B, Strijdom H, Blackhurst D, Lochner A. Short-term melatonin consumption protects the heart of obese rats independent of body weight change and visceral adiposity. J Pineal Res 2014; 57:317-32. [PMID: 25187154 DOI: 10.1111/jpi.12171] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 08/29/2014] [Indexed: 12/17/2022]
Abstract
Chronic melatonin treatment has been shown to prevent the harmful effects of diet-induced obesity and reduce myocardial susceptibility to ischaemia-reperfusion injury (IRI). However, the exact mechanism whereby it exerts its beneficial actions on the heart in obesity/insulin resistance remains unknown. Herein, we investigated the effects of relatively short-term melatonin treatment on the heart in a rat model of diet-induced obesity. Control and diet-induced obese Wistar rats (fed a high calorie diet for 20 wk) were each subdivided into three groups receiving drinking water with or without melatonin (4 mg/kg/day) for the last 6 or 3 wk of experimentation. A number of isolated hearts were perfused in the working mode, subjected to regional or global ischaemia-reperfusion; others were nonperfused. Metabolic parameters, myocardial infarct sizes (IFS), baseline and postischaemic activation of PKB/Akt, ERK42/44, GSK-3β and STAT-3 were determined. Diet-induced obesity caused increases in body weight gain, visceral adiposity, fasting blood glucose, serum insulin and triglyceride (TG) levels with a concomitant cardiac hypertrophy, large postischaemic myocardial IFSs and a reduced cardiac output. Melatonin treatment (3 and 6 wk) decreased serum insulin levels and the HOMA index (P < 0.05) with no effect on weight gain (after 3 wk), visceral adiposity, serum TG and glucose levels. It increased serum adiponectin levels, reduced myocardial IFSs in both groups and activated baseline myocardial STAT-3 and PKB/Akt, ERK42/44 and GSK-3β during reperfusion. Overall, short-term melatonin administration to obese/insulin resistant rats reduced insulin resistance and protected the heart against ex vivo myocardial IRI independently of body weight change and visceral adiposity.
Collapse
Affiliation(s)
- Frederic Nduhirabandi
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | | | | | | | | |
Collapse
|
175
|
Tanno M, Kuno A, Ishikawa S, Miki T, Kouzu H, Yano T, Murase H, Tobisawa T, Ogasawara M, Horio Y, Miura T. Translocation of glycogen synthase kinase-3β (GSK-3β), a trigger of permeability transition, is kinase activity-dependent and mediated by interaction with voltage-dependent anion channel 2 (VDAC2). J Biol Chem 2014; 289:29285-96. [PMID: 25187518 DOI: 10.1074/jbc.m114.563924] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Glycogen synthase kinase-3β (GSK-3β) is a major positive regulator of the mitochondrial permeability transition pore (mPTP), a principle trigger of cell death, under the condition of oxidative stress. However, the mechanism by which cytosolic GSK-3β translocates to mitochondria, promoting mPTP opening, remains unclear. Here we addressed this issue by analyses of the effect of site-directed mutations in GSK-3β on mitochondrial translocation and protein/protein interactions upon oxidative stress. H9c2 cardiomyoblasts were transfected with GFP-tagged GSK-3β (WT), a mutant GSK-3β insensitive to inhibitory phosphorylation (S9A), or kinase-deficient GSK-3β (K85R). Time lapse observation revealed that WT and S9A translocated from the cytosol to the mitochondria more promptly than did K85R after exposure to oxidative stress. H2O2 increased the density of nine spots on two-dimensional gel electrophoresis of anti-GSK-3β-immunoprecipitates by more than 3-fold. MALDI-TOF/MS analysis revealed that one of the spots contained voltage-dependent anion channel 2 (VDAC2). Knockdown of VDAC2, but not VDAC1 or VDAC3, by siRNA attenuated both the mitochondrial translocation of GSK-3β and mPTP opening under stress conditions. The mitochondrial translocation of GSK-3β was attenuated also when Lys-15, but not Arg-4 or Arg-6, in the N-terminal domain of GSK-3β was replaced with alanine. The oxidative stress-induced mitochondrial translocation of GSK-3β was associated with an increase in cell death, which was suppressed by lithium chloride (LiCl), a GSK-3β inhibitor. These results demonstrate that GSK-3β translocates from the cytosol to mitochondria in a kinase activity- and VDAC2-dependent manner in which an N-terminal domain of GSK-3β may function as a mitochondrial targeting sequence.
Collapse
Affiliation(s)
- Masaya Tanno
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine and
| | - Atsushi Kuno
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine and Pharmacology, Sapporo Medical University School of Medicine, S1 W16, Chuo-ku, Sapporo 060-8543, Japan
| | - Satoko Ishikawa
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine and
| | - Takayuki Miki
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine and
| | - Hidemichi Kouzu
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine and
| | - Toshiyuki Yano
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine and
| | - Hiromichi Murase
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine and
| | - Toshiyuki Tobisawa
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine and
| | - Makoto Ogasawara
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine and
| | - Yoshiyuki Horio
- Pharmacology, Sapporo Medical University School of Medicine, S1 W16, Chuo-ku, Sapporo 060-8543, Japan
| | - Tetsuji Miura
- From the Departments of Cardiovascular, Renal, and Metabolic Medicine and
| |
Collapse
|
176
|
Zorov DB, Juhaszova M, Sollott SJ. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev 2014; 94:909-50. [PMID: 24987008 DOI: 10.1152/physrev.00026.2013] [Citation(s) in RCA: 3352] [Impact Index Per Article: 335.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Byproducts of normal mitochondrial metabolism and homeostasis include the buildup of potentially damaging levels of reactive oxygen species (ROS), Ca(2+), etc., which must be normalized. Evidence suggests that brief mitochondrial permeability transition pore (mPTP) openings play an important physiological role maintaining healthy mitochondria homeostasis. Adaptive and maladaptive responses to redox stress may involve mitochondrial channels such as mPTP and inner membrane anion channel (IMAC). Their activation causes intra- and intermitochondrial redox-environment changes leading to ROS release. This regenerative cycle of mitochondrial ROS formation and release was named ROS-induced ROS release (RIRR). Brief, reversible mPTP opening-associated ROS release apparently constitutes an adaptive housekeeping function by the timely release from mitochondria of accumulated potentially toxic levels of ROS (and Ca(2+)). At higher ROS levels, longer mPTP openings may release a ROS burst leading to destruction of mitochondria, and if propagated from mitochondrion to mitochondrion, of the cell itself. The destructive function of RIRR may serve a physiological role by removal of unwanted cells or damaged mitochondria, or cause the pathological elimination of vital and essential mitochondria and cells. The adaptive release of sufficient ROS into the vicinity of mitochondria may also activate local pools of redox-sensitive enzymes involved in protective signaling pathways that limit ischemic damage to mitochondria and cells in that area. Maladaptive mPTP- or IMAC-related RIRR may also be playing a role in aging. Because the mechanism of mitochondrial RIRR highlights the central role of mitochondria-formed ROS, we discuss all of the known ROS-producing sites (shown in vitro) and their relevance to the mitochondrial ROS production in vivo.
Collapse
Affiliation(s)
- Dmitry B Zorov
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; and Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Magdalena Juhaszova
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; and Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Steven J Sollott
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; and Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
177
|
Wang C, Xie N, Zhang H, Li Y, Wang Y. Puerarin protects against β-amyloid-induced microglia apoptosis via a PI3K-dependent signaling pathway. Neurochem Res 2014; 39:2189-96. [PMID: 25173404 DOI: 10.1007/s11064-014-1420-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 07/17/2014] [Accepted: 08/13/2014] [Indexed: 11/26/2022]
Abstract
Puerarin extracted from Radix puerariae is well-known for its anti-oxidative and neuroprotective activities. In this study, we investigated the protective effect of puerarin on amyloid-β protein (Aβ)-induced cytotoxicity and its potential mechanisms in BV-2 and primary microglial cells. We found that pretreatment with puerarin afforded protection against Aβ-induced cytotoxicity through inhibiting apoptosis in BV-2 and primary microglial cells. This result was also confirmed by the activated caspase-3 assay. Phospho-Akt and Bcl-2 expression increased after pretreatment with puerarin in BV-2 and primary microglial cells exposed to Aβ, whereas Bax expression and cytochrome c release decreased. In addition, puerarin treatment prevented the loss of mitochondrial membrane potential and reactive oxygen species production. Interestingly, these effects of puerarin against Aβ insult were abolished by LY294002, an inhibitor of PI3K phosphorylation. Taken together, these findings suggest that puerarin prevents Aβ-induced microglial apoptosis via the activation of PI3K/Akt signaling pathway, and might be a potential preventive or therapeutic agent for Alzheimer's disease.
Collapse
Affiliation(s)
- Cui Wang
- Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | | | | | | | | |
Collapse
|
178
|
Changes in the loading conditions induced by vagal stimulation modify the myocardial infarct size through sympathetic-parasympathetic interactions. Pflugers Arch 2014; 467:1509-1522. [DOI: 10.1007/s00424-014-1591-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 07/10/2014] [Accepted: 07/28/2014] [Indexed: 10/24/2022]
|
179
|
Kim DE, Kim B, Shin HS, Kwon HJ, Park ES. The protective effect of hispidin against hydrogen peroxide-induced apoptosis in H9c2 cardiomyoblast cells through Akt/GSK-3β and ERK1/2 signaling pathway. Exp Cell Res 2014; 327:264-75. [PMID: 25128810 DOI: 10.1016/j.yexcr.2014.07.037] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 07/28/2014] [Accepted: 07/30/2014] [Indexed: 10/24/2022]
Abstract
Hispidin, a phenolic compound from Phellinus linteus (a medicinal mushroom), has been shown to possess strong anti-oxidant, anti-cancer, anti-diabetic, and anti-dementia properties. However, the cardioprotective efficacy of hispidin has not yet been investigated. In the present study, we investigated the protective effect of hispidin against oxidative stress-induced apoptosis in H9c2 cardiomyoblast cells and neonatal rat ventricular myocytes. While the treatment of H9c2 cardiomyoblast cells with hydrogen peroxide caused a loss of cell viability and an increase in the number of apoptotic cells, hispidin significantly protected the cells against hydrogen peroxide-induced cell death without any cytotoxicity as determined by XTT assay, LDH release assay, Hoechst 33342 assay, and Western blotting of apoptosis proteins such as caspase-3, Bax, and Bcl-2. Our data also shows that hispidin significantly scavenged intracellular ROS, and markedly enhanced the expression of antioxidant enzymes such as heme oxygenase-1 and catalase, which was accompanied by the concomitant activation of Akt/GSK-3β and ERK1/2 phosphorylation in H9c2 cardiomyoblast cells. The effects of hispidin on Akt and ERK phosphorylation were abrogated by LY294002 (a PI3K/Akt inhibitor) and U0126 (an ERK1/2 inhibitor). The effect of hispidin on GSK-3b activities was also blocked by LY294002. Furthermore, inhibiting the Akt/GSK-3β and ERK1/2 pathway by these inhibitors significantly reversed the hispidin-induced Bax and Bcl-2 expression, apoptosis induction, and ROS production. These findings indicate that hispidin protects against apoptosis in H9c2 cardiomyoblast cells exposed to hydrogen peroxide through reducing intracellular ROS production, regulating apoptosis-related proteins, and the activation of the Akt/GSK-3β and ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Dae-Eun Kim
- Department of Biomaterials Science and Engineering, Translational Research Center for Protein Function Control, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Department of Biomedical Laboratory Science, Kyungbok University, Sinbuk-myeon, Pochen, Gyeonggi 487-717, Republic of Korea
| | - Bokyung Kim
- Department of Physiology, Institute of Functional Genomics, Konkuk University School of Medicine, Chungju, Chungbuk 380-701, Republic of Korea
| | - Hwa-Sup Shin
- Department of Biomedical Chemistry, Konkuk University, Chungju, Chungbuk 380-701, Republic of Korea
| | - Ho Jeong Kwon
- Department of Biomaterials Science and Engineering, Translational Research Center for Protein Function Control, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Department of Biotechnology, Translational Research Center for Protein Function Control, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Department of Internal Medicine, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea.
| | - Eun-Seok Park
- Department of Biomedical Laboratory Science, Kyungbok University, Sinbuk-myeon, Pochen, Gyeonggi 487-717, Republic of Korea.
| |
Collapse
|
180
|
Sun L, Chen C, Jiang B, Li Y, Deng Q, Sun M, An X, Yang X, Yang Y, Zhang R, Lu Y, Zhu DS, Huo Y, Feng GS, Zhang Y, Luo J. Grb2-associated binder 1 is essential for cardioprotection against ischemia/reperfusion injury. Basic Res Cardiol 2014; 109:420. [PMID: 24951957 DOI: 10.1007/s00395-014-0420-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 05/30/2014] [Accepted: 05/30/2014] [Indexed: 12/20/2022]
Abstract
We have shown recently that endothelial Grb-2-associated binder 1 (Gab1), an intracellular scaffolding adaptor, has a protective effect against limb ischemia via mediating angiogenic signaling pathways. However, the role of Gab1 in cardiac ischemia/reperfusion (I/R) injury remains unknown. In this study, we show that Gab1 is required for cardioprotection against I/R injury. I/R injury led to remarkable phosphorylation of Gab1 in cardiomyocytes. Compared with controls, the mice with cardiomyocyte-specific deletion of Gab1 gene (CGKO mice) exhibited an increase in infarct size and a decrease in cardiac function after I/R injury. Consistently, in hearts of CGKO mice subjected to I/R, the activation of caspase 3 and myocardial apoptosis was markedly enhanced whereas the activation of protein kinase B (Akt) and mitogen-activated protein kinase (MAPK), which are critical for cardiomyocyte survival, was attenuated. Oxidative stress is regarded as a major contributor to myocardial I/R injury. To examine the role of Gab1 in oxidative stress directly, isolated adult cardiomyocytes were subject to oxidant hydrogen peroxide and the cardioprotective effects of Gab1 were confirmed. Furthermore, we found that the phosphorylation of Gab1 and Gab1-mediated activation of Akt and MAPK by oxidative stress was suppressed by ErbB receptor and Src kinase inhibitors, accompanied by an increase in apoptotic cell death. In conclusion, our results suggest that Gab1 is essential for cardioprotection against I/R oxidative injury via mediating survival signaling.
Collapse
Affiliation(s)
- Lulu Sun
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Apelin-APJ effects of ginsenoside-Rb1 depending on hypoxia-induced factor 1α in hypoxia neonatal cardiomyocytes. Chin J Integr Med 2014; 21:139-46. [PMID: 24893658 DOI: 10.1007/s11655-014-1774-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate whether ginsenoside-Rb1 (Gs-Rb1) inhibits the apoptosis of hypoxia cardiomyocytes by up-regulating apelin-APJ system and whether the system is affected by hypoxia-induced factor 1α (Hif-1α). METHODS Neonatal rat cardiomyocytes were randomly divided into 6 groups: a control group, a simple CoCl group, a simple Gs-Rb1 group, a CoCl and Gs-Rb1 hypoxia group, a CoCl and 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole (YC-1) group, a CoCl and YC-1 group and a Gs-Rb1 group, in which YC-1 inhibits the synthesis and accelerates the degradation of Hif-1a. The concentration of CoCl, Gs-Rb1 and YC-1 was 500 μmol/L, 200 μmol/L and 5 μmol/L, respectively; the apoptosis ratio was analyzed with a flow cytometer; and apelin, APJ and Hif-1α were assayed with immunocytochemistry, Western blot assays and reverse transcription polymerase chain reaction (RT-PCR). RESULTS (1) The anti-apoptosis effect of Gs-Rb1 on hypoxia cardiomyocytes was significantly inhibited by YC-1; (2) Hypoxia significantly up-graded the expression of mRNA and protein of apelin; this effect was further reinforced by Gs-Rb1 and significantly inhibited by YC-1; (3) Gs-Rb1 further strengthened the expression of APJ mRNA and APJ proteins once hypoxia occurred, which was significantly inhibited by YC-1; (4) Gs-Rb1 significantly increased the expression of Hif-1α, which was completely abolished by YC-1; (5) There was a negative relationship between AR and apelin (or APJ, including mRNA and protein), a positive correlation between apelin (or APJ) protein and Hif-1a protein, in hypoxia cardiomyocytes. CONCLUSION The apelin-APJ system plays an important role in the anti-apoptosis effect of Gs-Rb1 on hypoxia neonatal cardiomyocytes, which was partly adjusted by Hif-1α.
Collapse
|
182
|
SGK1 Is Involved in Cardioprotection of Urocortin-1 Against Hypoxia/Reoxygenation in Cardiomyocytes. Can J Cardiol 2014; 30:687-95. [DOI: 10.1016/j.cjca.2014.03.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/17/2014] [Accepted: 03/10/2014] [Indexed: 12/26/2022] Open
|
183
|
Ischemic Postconditioning Reduces Infarct Size Through the α1-Adrenergic Receptor Pathway. J Cardiovasc Pharmacol 2014; 63:504-11. [DOI: 10.1097/fjc.0000000000000074] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
184
|
Park ES, Kang JC, Jang YC, Park JS, Jang SY, Kim DE, Kim B, Shin HS. Cardioprotective effects of rhamnetin in H9c2 cardiomyoblast cells under H₂O₂-induced apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2014; 153:552-560. [PMID: 24607510 DOI: 10.1016/j.jep.2014.02.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 01/21/2014] [Accepted: 02/11/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Many studies have emphasized that flavonoids, found in various fruits, vegetables, and seeds, as well as tea and red wine, have potential health-promoting and disease-preventing effects. Rhamnetin is a flavonoid that exhibits antioxidant capabilities. However, little is known about its effect on cardiac myocytes under oxidative stress and the underlying mechanisms. MATERIALS AND METHODS H9c2 cardiomyoblast cells were subjected to H2O2, to study the protective effect of rhamnetin on cell viability, apoptosis, and ROS production. Signaling proteins related to apoptosis, survival, and redox were analyzed by Western blot. Furthermore, the mRNA expressions of SIRTs were tested by real time-polymerase chain reaction (PCR). RESULTS We investigated the protective effects of rhamnetin against H₂O₂-induced apoptosis in H9c2 cardiomyoblasts. Rhamnetin protected cells against H₂O₂-induced cell death without any cytotoxicity, as determined by the XTT assay, LDH assay, TUNEL assay, Hoechst 33342 assay, and Western blot analysis of apoptosis-related proteins. Rhamnetin also enhanced the expression of catalase and Mn-SOD, thereby inhibiting production of intracellular ROS. Furthermore, rhamnetin recovered the H₂O₂-induced decrease in phosphorylation of Akt/GSK-3β and MAPKs (ERK1/2, p38 MAPK, and JNK) and pretreatment with their inhibitors, attenuating the rhamnetin-induced cytoprotective effect. Further studies with real time-PCR and a sirtuin inhibitor showed that cardioprotection by rhamnetin occurred through induction of SIRT3 and SIRT4. CONCLUSIONS Taken together, these results suggest that rhamnetin may have novel therapeutic potential to protect the heart from ischemia-related injury.
Collapse
Affiliation(s)
- Eun-Seok Park
- Department of Biomedical Chemistry, College of Biomedical and Health science, Konkuk University, 322 Danwol-Dong, Chungju 380-701, Chungbuk, Republic of Korea
| | - Jun Chul Kang
- Department of Biomedical Chemistry, College of Biomedical and Health science, Konkuk University, 322 Danwol-Dong, Chungju 380-701, Chungbuk, Republic of Korea
| | - Yong Chang Jang
- Department of Biomedical Chemistry, College of Biomedical and Health science, Konkuk University, 322 Danwol-Dong, Chungju 380-701, Chungbuk, Republic of Korea
| | - Jong Seok Park
- Department of Biomedical Laboratory Science, Taegu Health College, Taegu 702-722, Republic of Korea
| | - Shin Yi Jang
- Cardiovascular Imaging Center, Samsung Medical Center, Seoul 135-710, Republic of Korea
| | - Dae-Eun Kim
- Department of Biomedical Laboratory Science, Kyungbok University, Pochen 487-717, Republic of Korea
| | - Bokyung Kim
- Department of Physiology, Institute of Functional Genomics, Konkuk University School of Medicine, Chungju 380-701, Chungbuk, Republic of Korea
| | - Hwa-Sup Shin
- Department of Biomedical Chemistry, College of Biomedical and Health science, Konkuk University, 322 Danwol-Dong, Chungju 380-701, Chungbuk, Republic of Korea.
| |
Collapse
|
185
|
Ma LL, Ge HW, Kong FJ, Qian LB, Hu BC, Li Q, Xu L, Liu JQ, Xu YX, Sun RH. Ventricular hypertrophy abrogates intralipid-induced cardioprotection by alteration of reperfusion injury salvage kinase/glycogen synthase kinase 3β signal. Shock 2014; 41:435-442. [PMID: 24430545 DOI: 10.1097/shk.0000000000000130] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Recent studies have demonstrated that intralipid (ILP) conferred myocardial protection against ischemia-reperfusion (IR) injury through activation of reperfusion injury salvage kinase (RISK) pathway. As RISK signal has been shown to be impaired in hypertrophied myocardium, we investigated whether ILP-induced cardiac protection was maintained in hypertrophied rat hearts. Transverse aortic constriction was performed on male Sprague-Dawley rats to induce left ventricular hypertrophy, then sham-operated or hypertrophied rat hearts were isolated and perfused retrogradely by the Langendorff for 30 min (equilibration) followed by 40 min of ischemia and then 120 min of reperfusion. The isolated hearts received 15-min episode of 1% ILP separated by 15 min of washout or three episodes of 5-min ischemia followed by 5-min reperfusion before ischemia. The hemodynamics, infarct size, apoptosis, phosphorylated protein kinase B (p-Akt), phosphorylated extracellular regulated protein kinase 1/2 (ERK1/2), phosphorylated glycogen synthase kinase 3β (GSK3β), Bcl-2, phosphorylated Bad, and Bax were determined. We found that ILP significantly improved left ventricular hemodynamics and reduced infarct size and the number of TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling)-positive cells in the sham-operated rat hearts exposed to IR. However, such myocardial infarct-sparing effect of ILP was completely blocked by phosphatidylinositol-3-kinase inhibitor wortmannin, but only partially by mitogen-activated protein kinase kinase inhibitor PD98059 in sham-operated hearts. Intralipd upregulated the phosphorylation of Akt, extracellular regulated protein kinase 1/2 (ERK1/2), and their downstream target of GSK3β and antiapoptotic Bcl-2 expression in healthy rat hearts. Nonetheless, ILP failed to improve left ventricular hemodynamics and reduced infarct size and apoptosis and increase the phosphorylated Akt, ERK1/2, GSK3β, and antiapoptotic Bcl-2 in hypertrophied myocardium. In contrast, ischemic preconditioning increased the phosphorylation of Akt, ERK1/2 and GSK3β, improved heart pump function, and reduced myocardial necrosis in sham-operated hearts, a phenomenon partially attenuated by ventricular hypertrophy. Interestingly, GSK inhibitor SB216763 conferred cardioprotection against IR injury in sham-operated hearts, but failed to exert cardioprotection in hypertrophied myocardium. Our results indicated that ventricular hypertrophy abrogated ILP-induced cardioprotection against IR injury by alteration of RISK/GSK3β signal.
Collapse
Affiliation(s)
- Lei-Lei Ma
- *Department of Critical Care Medicine, Zhejiang Provincial People's Hospital; and †Department of Anesthesiology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou; ‡Department of Urology, Peking University Shougang Hospital, Beijing; and §Department of Physiology, Zhejiang Medical College, Hangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
MicroRNA-26 was decreased in rat cardiac hypertrophy model and may be a promising therapeutic target. J Cardiovasc Pharmacol 2014; 62:312-9. [PMID: 23719092 DOI: 10.1097/fjc.0b013e31829b82e6] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
MicroRNA (miR)-26 was found to be downregulated in cardiac diseases. In this study, the critical role of miR-26 in myocardial hypertrophy in both in vivo and in vitro was investigated. Sixteen male Wistar rats that underwent sham or transverse abdominal aortic constriction (TAAC) surgery were divided into control or TAAC group. Cardiomyocytes were isolated from neonatal Sprague-Dawley rats. Our study demonstrated that miR-26a/b was downregulated in both TAAC rat model and cardiomyocytes. The results of luciferase assays also suggested that glycogen synthase kinase 3β (GSK3β) may be a direct target of miR-26. The overexpression of miR-26 attenuated GSK3β expression and inhibited myocardial hypertrophy. The downregulation of miR-26 reversed these effects. Furthermore, silence of GSK3β gene phenocopied the anti-hypertrophy effects of miR-26, whereas overexpression of this protein attenuated the effects of miR-26. Taken together, these data suggest that miR-26 regulates pathological structural changes in the rat heart, which may be associated with suppression of the GSK3β signaling pathway, and implicate the potential application of miR-26 in diagnosis and therapy of cardiac hypertrophy.
Collapse
|
187
|
Issan Y, Kornowski R, Aravot D, Shainberg A, Laniado-Schwartzman M, Sodhi K, Abraham NG, Hochhauser E. Heme oxygenase-1 induction improves cardiac function following myocardial ischemia by reducing oxidative stress. PLoS One 2014; 9:e92246. [PMID: 24658657 PMCID: PMC3962395 DOI: 10.1371/journal.pone.0092246] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 02/20/2014] [Indexed: 01/23/2023] Open
Abstract
Background Oxidative stress plays a key role in exacerbating diabetes and cardiovascular disease. Heme oxygenase-1 (HO-1), a stress response protein, is cytoprotective, but its role in post myocardial infarction (MI) and diabetes is not fully characterized. We aimed to investigate the protection and the mechanisms of HO-1 induction in cardiomyocytes subjected to hypoxia and in diabetic mice subjected to LAD ligation. Methods In vitro: cultured cardiomyocytes were treated with cobalt-protoporphyrin (CoPP) and tin protoporphyrin (SnPP) prior to hypoxic stress. In vivo: CoPP treated streptozotocin-induced diabetic mice were subjected to LAD ligation for 2/24 h. Cardiac function, histology, biochemical damage markers and signaling pathways were measured. Results HO-1 induction lowered release of lactate dehydrogenase (LDH) and creatine phospho kinase (CK), decreased propidium iodide staining, improved cell morphology and preserved mitochondrial membrane potential in cardiomyocytes. In diabetic mice, Fractional Shortening (FS) was lower than non-diabetic mice (35±1%vs.41±2, respectively p<0.05). CoPP-treated diabetic animals improved cardiac function (43±2% p<0.01), reduced CK, Troponin T levels and infarct size compared to non-treated diabetic mice (P<0.01, P<0.001, P<0.01 respectively). CoPP-enhanced HO-1 protein levels and reduced oxidative stress in diabetic animals, as indicated by the decrease in superoxide levels in cardiac tissues and plasma TNFα levels (p<0.05). The increased levels of HO-1 by CoPP treatment after LAD ligation led to a shift of the Bcl-2/bax ratio towards the antiapoptotic process (p<0.05). CoPP significantly increased the expression levels of pAKT and pGSK3β (p<0.05) in cardiomyocytes and in diabetic mice with MI. SnPP abolished CoPP's cardioprotective effects. Conclusions HO-1 induction plays a role in cardioprotection against hypoxic damage in cardiomyocytes and in reducing post ischemic cardiac damage in the diabetic heart as proved by the increased levels of pAKT with a concomitant inhibition of pGSK3β leading to preserved mitochondrial membrane potential.
Collapse
Affiliation(s)
- Yossi Issan
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel-Aviv University, Petah-Tikva, Israel
| | - Ran Kornowski
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel-Aviv University, Petah-Tikva, Israel
- Cardiology Department, Rabin Medical Center, Tel-Aviv University, Petah-Tikva, Israel
| | - Dan Aravot
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel-Aviv University, Petah-Tikva, Israel
- Cardiac Surgery department, Rabin Medical Center, Tel-Aviv University, Petah-Tikva, Israel
| | - Asher Shainberg
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Komal Sodhi
- Department of Internal Medicine, Marshall University Joan C. Edwards School of Medicine, Huntington, West Virginia, United States of America
| | - Nader G. Abraham
- Department of Internal Medicine, Marshall University Joan C. Edwards School of Medicine, Huntington, West Virginia, United States of America
| | - Edith Hochhauser
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel-Aviv University, Petah-Tikva, Israel
- Cardiac Surgery department, Rabin Medical Center, Tel-Aviv University, Petah-Tikva, Israel
- * E-mail:
| |
Collapse
|
188
|
Brooks MJ, Andrews DT. Molecular mechanisms of ischemic conditioning: translation into patient outcomes. Future Cardiol 2014; 9:549-68. [PMID: 23834695 DOI: 10.2217/fca.13.30] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Following the initiation of an ischemic insult, reperfusion injury (RI) can result in numerous deleterious cardiac effects, including cardiomyocyte death. Experimental data have suggested that ischemic conditioning, when delivered either before or after the ischemic event, can provide considerable cardioprotection against RI. Ischemic conditioning involves delivering brief repetitive cycles of ischemia to the myocardium (local) or to another distal organ or structure (remote). This review will discuss recent advances in the molecular mechanisms involved in RI, the signaling pathways recruited by ischemic conditioning and conclude with an appraisal of the evidence for the use of ischemic conditioning in current clinical practice.
Collapse
Affiliation(s)
- Matthew J Brooks
- Department of Cardiology, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | | |
Collapse
|
189
|
Földes G, Mioulane M, Kodagoda T, Lendvai Z, Iqbal A, Ali NN, Schneider MD, Harding SE. Immunosuppressive Agents Modulate Function, Growth, and Survival of Cardiomyocytes and Endothelial Cells Derived from Human Embryonic Stem Cells. Stem Cells Dev 2014; 23:467-76. [DOI: 10.1089/scd.2013.0229] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Gábor Földes
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Maxime Mioulane
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| | - Thusharika Kodagoda
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| | | | - Adeel Iqbal
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| | - Nadire N. Ali
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| | - Michael D. Schneider
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| | - Sian E. Harding
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| |
Collapse
|
190
|
Peart JN, Pepe S, Reichelt ME, Beckett N, See Hoe L, Ozberk V, Niesman IR, Patel HH, Headrick JP. Dysfunctional survival-signaling and stress-intolerance in aged murine and human myocardium. Exp Gerontol 2014. [PMID: 24316036 DOI: 10.1016/j.exger.2013.11.015.pubmed:24316036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Changes in cytoprotective signaling may influence cardiac aging, and underpin sensitization to ischemic insult and desensitization to 'anti-ischemic' therapies. We tested whether age-dependent shifts in ischemia-reperfusion (I-R) tolerance in murine and human myocardium are associated with reduced efficacies and coupling of membrane, cytoplasmic and mitochondrial survival-signaling. Hormesis (exemplified in ischemic preconditioning; IPC) and expression of proteins influencing signaling/stress-resistance were also assessed in mice. Mouse hearts (18 vs. 2-4 mo) and human atrial tissue (75±2 vs. 55±2 yrs) exhibited profound age-dependent reductions in I-R tolerance. In mice aging negated cardioprotection via IPC, G-protein coupled receptor (GPCR) agonism (opioid, A1 and A3 adenosine receptors) and distal protein kinase c (PKC) activation (4 nM phorbol 12-myristate 13-acetate; PMA). In contrast, p38-mitogen activated protein kinase (p38-MAPK) activation (1 μM anisomycin), mitochondrial ATP-sensitive K(+) channel (mKATP) opening (50 μM diazoxide) and permeability transition pore (mPTP) inhibition (0.2 μM cyclosporin A) retained protective efficacies in older hearts (though failed to eliminate I-R tolerance differences). A similar pattern of change in protective efficacies was observed in human tissue. Murine hearts exhibited molecular changes consistent with altered membrane control (reduced caveolin-3, cholesterol and caveolae), kinase signaling (reduced p70 ribosomal s6 kinase; p70s6K) and stress-resistance (increased G-protein receptor kinase 2, GRK2; glycogen synthase kinase 3β, GSK3β; and cytosolic cytochrome c). In summary, myocardial I-R tolerance declines with age in association with dysfunctional hormesis and transduction of survival signals from GPCRs/PKC to mitochondrial effectors. Differential changes in proteins governing caveolar and mitochondrial function may contribute to signal dysfunction and stress-intolerance.
Collapse
Affiliation(s)
- Jason N Peart
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Australia
| | - Salvatore Pepe
- Heart Research, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Melissa E Reichelt
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Nikkie Beckett
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Australia
| | - Louise See Hoe
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Australia
| | - Victoria Ozberk
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Australia
| | | | - Hemal H Patel
- VA San Diego Healthcare System, San Diego, USA; Department of Anesthesiology, University of California San Diego, USA
| | - John P Headrick
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Australia.
| |
Collapse
|
191
|
Miura T. HASF, a PKC-ε activator with novel features for cardiomyocyte protection. J Mol Cell Cardiol 2014; 69:1-3. [PMID: 24486196 DOI: 10.1016/j.yjmcc.2014.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 01/20/2014] [Accepted: 01/22/2014] [Indexed: 12/26/2022]
Affiliation(s)
- Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1 W-16, Chuo-ku Sapporo 060-8543, Japan.
| |
Collapse
|
192
|
Peart JN, Pepe S, Reichelt ME, Beckett N, See Hoe L, Ozberk V, Niesman IR, Patel HH, Headrick JP. Dysfunctional survival-signaling and stress-intolerance in aged murine and human myocardium. Exp Gerontol 2013; 50:72-81. [PMID: 24316036 DOI: 10.1016/j.exger.2013.11.015] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 11/03/2013] [Accepted: 11/26/2013] [Indexed: 11/26/2022]
Abstract
Changes in cytoprotective signaling may influence cardiac aging, and underpin sensitization to ischemic insult and desensitization to 'anti-ischemic' therapies. We tested whether age-dependent shifts in ischemia-reperfusion (I-R) tolerance in murine and human myocardium are associated with reduced efficacies and coupling of membrane, cytoplasmic and mitochondrial survival-signaling. Hormesis (exemplified in ischemic preconditioning; IPC) and expression of proteins influencing signaling/stress-resistance were also assessed in mice. Mouse hearts (18 vs. 2-4 mo) and human atrial tissue (75±2 vs. 55±2 yrs) exhibited profound age-dependent reductions in I-R tolerance. In mice aging negated cardioprotection via IPC, G-protein coupled receptor (GPCR) agonism (opioid, A1 and A3 adenosine receptors) and distal protein kinase c (PKC) activation (4 nM phorbol 12-myristate 13-acetate; PMA). In contrast, p38-mitogen activated protein kinase (p38-MAPK) activation (1 μM anisomycin), mitochondrial ATP-sensitive K(+) channel (mKATP) opening (50 μM diazoxide) and permeability transition pore (mPTP) inhibition (0.2 μM cyclosporin A) retained protective efficacies in older hearts (though failed to eliminate I-R tolerance differences). A similar pattern of change in protective efficacies was observed in human tissue. Murine hearts exhibited molecular changes consistent with altered membrane control (reduced caveolin-3, cholesterol and caveolae), kinase signaling (reduced p70 ribosomal s6 kinase; p70s6K) and stress-resistance (increased G-protein receptor kinase 2, GRK2; glycogen synthase kinase 3β, GSK3β; and cytosolic cytochrome c). In summary, myocardial I-R tolerance declines with age in association with dysfunctional hormesis and transduction of survival signals from GPCRs/PKC to mitochondrial effectors. Differential changes in proteins governing caveolar and mitochondrial function may contribute to signal dysfunction and stress-intolerance.
Collapse
Affiliation(s)
- Jason N Peart
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Australia
| | - Salvatore Pepe
- Heart Research, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Melissa E Reichelt
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Nikkie Beckett
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Australia
| | - Louise See Hoe
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Australia
| | - Victoria Ozberk
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Australia
| | | | - Hemal H Patel
- VA San Diego Healthcare System, San Diego, USA; Department of Anesthesiology, University of California San Diego, USA
| | - John P Headrick
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Australia.
| |
Collapse
|
193
|
Wang Z, Ge Y, Bao H, Dworkin L, Peng A, Gong R. Redox-sensitive glycogen synthase kinase 3β-directed control of mitochondrial permeability transition: rheostatic regulation of acute kidney injury. Free Radic Biol Med 2013; 65:849-858. [PMID: 23973862 PMCID: PMC3859848 DOI: 10.1016/j.freeradbiomed.2013.08.169] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 07/31/2013] [Accepted: 08/16/2013] [Indexed: 01/29/2023]
Abstract
Mitochondrial dysfunction plays a pivotal role in necroapoptotic cell death and in the development of acute kidney injury (AKI). Evidence suggests that glycogen synthase kinase (GSK) 3β resides at the nexus of multiple signaling pathways implicated in the regulation of mitochondrial permeability transition (MPT). In cultured renal tubular epithelial cells, a discrete pool of GSK3β was detected in mitochondria. Coimmunoprecipitation assay confirmed that GSK3β physically interacts with cyclophilin F and voltage-dependent anion channel (VDAC), key MPT regulators that possess multiple GSK3β phosphorylation consensus motifs, suggesting that GSK3β has a direct control of MPT. Upon a strong burst of reactive oxygen species elicited by the pro-oxidant herbicide paraquat, the activity of the redox-sensitive GSK3β was drastically enhanced. This was accompanied by augmented phosphorylation of cyclophilin F and VDAC, associated with MPT and cell death. Inhibition of GSK3β by either the selective inhibitor 4-Benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8) or forced expression of a kinase-dead mutant obliterated paraquat-induced phosphorylation of cyclophilin F and VDAC, prevented MPT, and improved cellular viability. Conversely, ectopic expression of a constitutively active GSK3β amplified the effect of paraquat on cyclophilin F and VDAC phosphorylation and sensitized cells to paraquat-induced MPT and death. In vivo, paraquat injection elicited marked oxidant stress in the kidney and resulted in acute kidney dysfunction and massive tubular apoptosis and necrosis. Consistent with in vitro findings, the activity of GSK3β was augmented in the kidney after paraquat injury, associated with increased phosphorylation of cyclophilin F and VDAC and sensitized MPT. TDZD-8 blocked GSK3β activity in the kidney, intercepted cyclophilin F and VDAC phosphorylation, prevented MPT, attenuated tubular cell death, and ameliorated paraquat-induced AKI. Our data suggest that the redox-sensitive GSK3β regulates renal tubular injury in AKI by controlling the activity of MPT regulators.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI 02903, USA
| | - Yan Ge
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI 02903, USA
| | - Hui Bao
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI 02903, USA
| | - Lance Dworkin
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI 02903, USA
| | - Ai Peng
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Rujun Gong
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI 02903, USA.
| |
Collapse
|
194
|
Brooks MM, Neelam S, Cammarata PR. Lenticular mitoprotection. Part B: GSK-3β and regulation of mitochondrial permeability transition for lens epithelial cells in atmospheric oxygen. Mol Vis 2013; 19:2451-67. [PMID: 24319338 PMCID: PMC3850969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 11/26/2013] [Indexed: 12/02/2022] Open
Abstract
PURPOSE Loss of integrity of either the inner or outer mitochondrial membrane results in the dissipation of the mitochondrial electrochemical gradient that leads to mitochondrial membrane permeability transition (mMPT). This study emphasizes the role of glycogen synthase kinase 3beta (GSK-3β) in maintaining mitochondrial membrane potential, thus preventing mitochondrial depolarization (hereafter termed mitoprotection). Using 3-(2,4-dichlorophenyl)-4-(1-methyl-1H-indol-3-yl)-1H-pyrrole-2,5-dione (SB216763), an inhibitor of GSK-3β, and drawing a distinction between it and 1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio] butadiene (UO126), an inhibitor of extracellular-signal-regulated kinase (ERK) phosphorylation, the means by which GSK-3β influences mitoprotection in cultured human lens epithelial (HLE-B3) cells and normal, secondary cultures of bovine lens epithelial cells, maintained in atmospheric oxygen, was investigated. METHODS Virally transfected human lens epithelial cells (HLE-B3) and normal cultures of bovine lens epithelial cells were exposed to acute hypoxic conditions (about 1% O2) followed by exposure to atmospheric oxygen (about 21% O2). Specific antisera and western blot analysis was used to examine the state of phosphorylation of ERK and GSK-3β, as well as the phosphorylation of a downstream substrate of GSK-3β, glycogen synthase (GS, useful in monitoring GSK-3β activity). The potentiometric dye, 1H-benzimidazolium-5,6-dichloro-2-[3-(5,6-dichloro-1,3-diethyl-1,3-dihydro-2H-benzimidazol-2-ylidene)-1-propenyl]-1,3-diethyl-iodide (JC-1), was used to monitor mitochondrial depolarization upon exposure of inhibitor treatment relative to the control cells (mock inhibition) in atmospheric oxygen. Caspase-3 activation was scrutinized to determine whether mitochondrial depolarization inevitably leads to apoptosis. RESULTS Treatment of HLE-B3 cells with SB216763 (12 µM) inactivated GSK-3β activity as verified by the enzyme's inability to phosphorylate its substrate, GS. SB216763-treated cells were not depolarized relative to the control cells as demonstrated with JC-1 fluorescent dye analysis. The HLE-B3 cells treated with UO126, which similarly blocked phosphorylation of GS, were nevertheless prone to mMPT relative to the control cells. Western blot analysis determined that Bcl-2-associated X (BAX) levels were unchanged for SB216763-treated or UO126-treated HLE-B3 cells when compared to their respective control cells. However, unlike the SB216763-treated cells, the UO126-treated cells showed a marked absence of Bcl-2, as well as phosphorylated Bcl-2 relative to the controls. UO126 treatment of bovine lens epithelial cells showed similar results with pBcl-2 levels, while the Bcl-2 content appeared unchanged relative to the control cells. HLE-B3 and normal bovine lens cell cultures showed susceptibility to mMPT associated with the loss of pBcl-2 by UO126 treatment. CONCLUSIONS MITOCHONDRIAL DEPOLARIZATION MAY OCCUR BY ONE OF TWO KEY OCCURRENCES: interruption of the electrochemical gradient across the inner mitochondrial membrane resulting in mMPT or by disruption of the integrity of the inner or outer mitochondrial membrane. The latter scenario is generally tightly regulated by members of the Bcl-2 family of proteins. Inhibition of GSK-3β activity by SB216763 blocks mMPT by preventing the opening of the mitochondrial permeability transition pore. UO126, likewise, inhibits GSK-3β activity, but unlike SB216763, inhibition of ERK phosphorylation induces the loss of intracellular pBcl-2 levels under conditions where intracellular BAX levels remain constant. These results suggest that the lenticular mitoprotection normally afforded by the inactivation of GSK-3β activity may, however, be bypassed by a loss of pBcl-2, an anti-apoptotic member of the Bcl-2 family. Bcl-2 prevents the translocation of BAX to the mitochondrial outer membrane inhibiting depolarization by disrupting the normal electrochemical gradient leading to mMPT.
Collapse
|
195
|
Hsieh SR, Hsu CS, Lu CH, Chen WC, Chiu CH, Liou YM. Epigallocatechin-3-gallate-mediated cardioprotection by Akt/GSK-3β/caveolin signalling in H9c2 rat cardiomyoblasts. J Biomed Sci 2013; 20:86. [PMID: 24251870 PMCID: PMC3871020 DOI: 10.1186/1423-0127-20-86] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 11/18/2013] [Indexed: 12/30/2022] Open
Abstract
Background Epigallocatechin-3-gallate (EGCg) with its potent anti-oxidative capabilities is known for its beneficial effects ameliorating oxidative injury to cardiac cells. Although studies have provided convincing evidence to support the cardioprotective effects of EGCg, it remains unclear whether EGCg affect trans-membrane signalling in cardiac cells. Here, we have demonstrated the potential mechanism for cardioprotection of EGCg against H2O2-induced oxidative stress in H9c2 cardiomyoblasts. Results Exposing H9c2 cells to H2O2 suppressed cell viability and altered the expression of adherens and gap junction proteins with increased levels of intracellular reactive oxygen species and cytosolic Ca2+. These detrimental effects were attenuated by pre-treating cells with EGCg for 30 min. EGCg also attenuated H2O2-mediated cell cycle arrest at the G1-S phase through the glycogen synthase kinase-3β (GSK-3β)/β-catenin/cyclin D1 signalling pathway. To determine how EGCg targets H9c2 cells, enhanced green fluorescence protein (EGFP) was ectopically expressed in these cells. EGFP-emission fluorescence spectroscopy revealed that EGCg induced dose-dependent fluorescence changes in EGFP expressing cells, suggesting that EGCg signalling events might trigger proximity changes of EGFP expressed in these cells. Proteomics studies showed that EGFP formed complexes with the 67 kD laminin receptor, caveolin-1 and -3, β-actin, myosin 9, vimentin in EGFP expressing cells. Using in vitro oxidative stress and in vivo myocardial ischemia models, we also demonstrated the involvement of caveolin in EGCg-mediated cardioprotection. In addition, EGCg-mediated caveolin-1 activation was found to be modulated by Akt/GSK-3β signalling in H2O2-induced H9c2 cell injury. Conclusions Our data suggest that caveolin serves as a membrane raft that may help mediate cardioprotective EGCg transmembrane signalling.
Collapse
Affiliation(s)
| | | | | | | | | | - Ying-Ming Liou
- Department of Life Sciences, National Chung-Hsing University, Taichung 402, Taiwan.
| |
Collapse
|
196
|
Gsk-3β inhibitors mimic the cardioprotection mediated by ischemic pre- and postconditioning in hypertensive rats. BIOMED RESEARCH INTERNATIONAL 2013; 2013:317456. [PMID: 24288674 PMCID: PMC3830772 DOI: 10.1155/2013/317456] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 06/17/2013] [Accepted: 09/16/2013] [Indexed: 11/18/2022]
Abstract
The aim of this study was to examine the effects of GSK-3β inhibitors compared with PRE and POS in spontaneously hypertensive rats (SHR). Isolated hearts were submitted to the following protocols: IC: 45 min global ischemia (GI) and 1-hour reperfusion (R); PRE: a cycle of 5 min GI and 10 minutes of R prior to 45 min GI; POS: three cycles of 30 sec GI/30 sec R at the start of R. Other hearts received lithium chloride (LiCl) or indirubin-3′-monoxime,5-iodo-(IMI) as GSK-3β inhibitors. All interventions reduced the infarct size observed in IC group. The expressions of P-GSK-3β and P-Akt decreased in IC and were restored after PRE, POS, and GSK-3β inhibitors treatments. An increase of cytosolic MnSOD activity and lipid peroxidation and a decrease of GSH content observed in IC hearts were attenuated in PRE, POS, and LiCl or IMI treatments. An increase of P-GSK-3β/VDAC physical association and a partial recovery of mitochondrial permeability were also detected after interventions. These data show that, in SHR hearts, GSK-3β inhibitors mimic the cardioprotection afforded by PRE and POS and suggest that a decrease in mitochondrial permeability mediated by P-GSK-3β/VDAC interaction is a crucial event.
Collapse
|
197
|
Xu Y, Ma LL, Zhou C, Zhang FJ, Kong FJ, Wang WN, Qian LB, Wang CC, Liu XB, Yan M, Wang JA. Hypercholesterolemic myocardium is vulnerable to ischemia-reperfusion injury and refractory to sevoflurane-induced protection. PLoS One 2013; 8:e76652. [PMID: 24124583 PMCID: PMC3790738 DOI: 10.1371/journal.pone.0076652] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 08/27/2013] [Indexed: 11/18/2022] Open
Abstract
Recent studies have demonstrated that volatile anesthetic postconditioning confers myocardial protection against ischemia-reperfusion (IR) injury through activation of the reperfusion injury salvage kinase (RISK) pathway. As RISK has been shown to be impaired in hypercholesterolemia. Therefore, we investigate whether anesthetic-induced cardiac protection was maintained in hypercholesterolemic rats. In the present study, normocholesteolemic or hypercholesterolemic rat hearts were subjected to 30 min of ischemia and 2 h of reperfusion. Animals received 2.4% sevoflurane for 5 min or 3 cycles of 10-s ischemia/10-s reperfusion. The hemodynamic parameters, including left ventricular developed pressure, left ventricular end-diastolic pressure and heart rate, were continuously monitored. The infarct size, apoptosis, p-Akt, p-ERK1/2, p-GSK3β were determined. We found that both sevoflurane and ischemic postconditioning significantly improved heart pump function, reduced infarct size and increased the phosphorylation of Akt, ERK1/2 and their downstream target of GSK3β in the healthy rats. In the hypercholesterolemic rats, neither sevoflurane nor ischemic postconditioning improved left ventricular hemodynamics, reduced infarct size and increased the phosphorylated Akt, ERK1/2 and GSK3β. In contrast, GSK inhibitor SB216763 conferred cardioprotection against IR injury in healthy and hypercholesterolemic hearts. In conclusions, hyperchoesterolemia abrogated sevoflurane-induced cardioprotection against IR injury by alteration of upstream signaling of GSK3β and acute GSK inhibition may provide a novel therapeutic strategy to protect hypercholesterolemic hearts against IR injury.
Collapse
Affiliation(s)
- Yong Xu
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejaing, China
| | - Lei-Lei Ma
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejaing, China
| | - Chen Zhou
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejaing, China
| | - Fei-Jiang Zhang
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejaing, China
| | - Fei-Juan Kong
- Department of Anesthesiology, Hangzhou First People’s Hospital, Nanjing Medical University, Hangzhou, Zhejaing, China
| | - Wen-Na Wang
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejaing, China
| | - Ling-Bo Qian
- Department of Physiology, Zhejiang Medical College, Hangzhou, Zhejaing, China
| | - Can-Can Wang
- Department of Physiology, Zhejiang Medical College, Hangzhou, Zhejaing, China
| | - Xian-Bao Liu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejaing, China
| | - Min Yan
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejaing, China
- * E-mail: (MY); (JAW)
| | - Jian-An Wang
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejaing, China
- * E-mail: (MY); (JAW)
| |
Collapse
|
198
|
Yaniv Y, Juhaszova M, Sollott SJ. Age-related changes of myocardial ATP supply and demand mechanisms. Trends Endocrinol Metab 2013; 24:495-505. [PMID: 23845538 PMCID: PMC3783621 DOI: 10.1016/j.tem.2013.06.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 05/30/2013] [Accepted: 06/03/2013] [Indexed: 01/03/2023]
Abstract
In advanced age, the resting myocardial oxygen consumption rate (MVO2) and cardiac work (CW) in the rat remain intact. However, MVO2, CW and cardiac efficiency achieved at high demand are decreased with age, compared to maximal values in the young. Whether this deterioration is due to decrease in myocardial ATP demand, ATP supply, or the control mechanisms that match them remains controversial. Here we discuss evolving perspectives of age-related changes of myocardial ATP supply and demand mechanisms, and critique experimental models used to investigate aging. Specifically, we evaluate experimental data collected at the level of isolated mitochondria, tissue, or organism, and discuss how mitochondrial energetic mechanisms change in advanced age, both at basal and high energy-demand levels.
Collapse
Affiliation(s)
- Yael Yaniv
- Laboratory of Cardiovascular Science, Biomedical Research Center, Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | | | | |
Collapse
|
199
|
Krenz M, Baines C, Kalogeris T, Korthuis R. Cell Survival Programs and Ischemia/Reperfusion: Hormesis, Preconditioning, and Cardioprotection. ACTA ACUST UNITED AC 2013. [DOI: 10.4199/c00090ed1v01y201309isp044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
200
|
Ge XH, Zhu GJ, Geng DQ, Zhang ZJ, Liu CF. Erythropoietin attenuates 6-hydroxydopamine-induced apoptosis via glycogen synthase kinase 3β-mediated mitochondrial translocation of Bax in PC12 cells. Neurol Sci 2013; 33:1249-56. [PMID: 22294054 DOI: 10.1007/s10072-012-0959-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 01/19/2012] [Indexed: 01/25/2023]
Abstract
The aim of this study was to determine the mechanism by which erythropoietin (EPO) suppressed 6-hydroxydopamine (6-OHDA)-induced apoptosis. Our results showed that 6-OHDA remarkably decreased phosphorylation of glycogen synthase kinase 3β (GSK3β) as well as enhanced the level of Bax in the mitochondria. Besides, 6-OHDA decreased the mitochondrial expression of Bcl-2 without altering the cytoplasmic expression of Bcl-2. In line with these results, 6-OHDA treatment enhanced the apoptosis and caspase 3 activity in PC12 cells. These findings indicated that mitochondrial dysfunction was involved in the neurotoxicity of 6-OHDA and GSK3β might act upstream of Bax/Bcl-2 and the caspase 3 pathways in 6-OHDA-treated PC12 cells. Furthermore, EPO reduced 6-OHDA-induced growth inhibition. Western blot exhibited that GSK3β inhibitor 4-benzyl-2-methyl-1, 2,4-thiadiazolidine-3, 5-dione (TDZD8) and EPO not only increased the phosphorylation of GSK3β but also inhibited the mitochondrial translocation of Bax. In agreement with these results, EPO and TDZD8 obviously increased the mitochondrial expression of Bcl-2. Finally, TDZD-8 and EPO significantly suppressed the enhanced apoptosis and activity of caspase 3 induced by 6-OHDA. Taken together, GSK3β-mediated mitochondrial cell death pathway is involved in the neuroprotective effect of EPO against 6-OHDA-induced apoptosis.
Collapse
Affiliation(s)
- Xu-Hua Ge
- Department of Neurology, Second Affiliated Hospital of Soochow University, and Institute of Neuroscience, Soochow University, Jiangsu Province, China
| | | | | | | | | |
Collapse
|