1
|
Priya A, Mol N, Singh AK, Aditya AK, Ray AK. "Unveiling the impacts of climatic cold events on the cardiovascular health in animal models". THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 971:179028. [PMID: 40073773 DOI: 10.1016/j.scitotenv.2025.179028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/01/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
Climate change is increasingly driving extreme weather events, leading to drastic temperature fluctuations worldwide. While overall temperatures rise, many regions are simultaneously experiencing severe cold spells that threaten the health of human populations, especially to vulnerable populations including the elderly and those with pre-existing conditions. Exposure to cold stress triggers significant physiological and biochemical disruptions. As cardiovascular diseases (CVDs) rank among the leading causes of global morbidity and mortality, the exacerbation of these conditions by cold exposure underscores critical public health challenges. The complex pathophysiological processes in cold-induced CVDs require careful analysis at an organ-system level, making animal models an ideal tool for replicating human physiological and molecular responses in a controlled environment. However, a detailed mechanism linking cold exposure and cardiovascular dysfunction remains incompletely understood, particularly in the context of animal models. Therefore, this comprehensive review aims to address and analyze from traditional rodent models to less conventional ruminants, broilers, canines, and primate animal models to understand cold stress-induced CVDs, with an extensive account of the potential molecular mechanisms and pathways such as oxidative stress, inflammation, vasomotor dysfunction, and apoptosis, along with emerging roles of cold shock proteins (CSPs), etc. We also delve into various potential therapeutic approaches and preventive measures in cold stress conditions. In conclusion, this review is the first to comprehensively address the underexplored cardiovascular complications arising from cold stress and their underlying mechanisms, particularly using animal models. Furthermore, it provides a foundation for advancing the development of more effective and targeted therapies through translational research.
Collapse
Affiliation(s)
- Anjali Priya
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Nidhi Mol
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Alok Kumar Singh
- Department of Zoology, Ramjas College, University of Delhi, New Delhi, India
| | - Abhishek Kumar Aditya
- Department of Medicine, K.D. Medical College, Hospital and Research Centre, Mathura, India
| | - Ashwini Kumar Ray
- Department of Environmental Studies, University of Delhi, New Delhi, India.
| |
Collapse
|
2
|
Brand T, Baumgarten BT, Denzinger S, Reinders Y, Kleindl M, Schanbacher C, Funk F, Gedik N, Jabbasseh M, Kleinbongard P, Dudek J, Szendroedi J, Tolstik E, Schuh K, Krüger M, Dobrev D, Cuello F, Sickmann A, Schmitt JP, Lorenz K. From Ca 2+ dysregulation to heart failure: β-adrenoceptor activation by RKIP postpones molecular damages and subsequent cardiac dysfunction in mice carrying mutant PLN R9C by correction of aberrant Ca 2+-handling. Pharmacol Res 2025; 211:107558. [PMID: 39742932 DOI: 10.1016/j.phrs.2024.107558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Impaired cardiomyocyte Ca2+ handling is a central hallmark of heart failure (HF), which causes contractile dysfunction and arrhythmias. However, the underlying molecular mechanisms and the precise contribution of defects in Ca2+-cycling regulation in the development of HF are still not completely resolved. Here, we used transgenic mice that express a human mutation in the cardiomyocyte Ca2+-regulator phospholamban (PLNR9C-tg) causing severe HF due to a reduction in Ca2+ reuptake into the sarco(endo)plasmic reticulum (SR). PLNR9C-induced HF is a rapidly progressing condition characterized by prominent Ca2+ cycling and relaxation defects and premature death of mutation carriers. We found that endoplasmic reticulum (ER) and mitochondrial function are affected even before transition to overt HF. Early correction of aberrant Ca2+ cycling by cardiac expression of the Raf kinase inhibitor protein (RKIP), an endogenous activator of β-adrenoceptors (βAR), delayed the cellular alterations, functional failure and prolonged lifespan. Our study highlights the importance of early and persistent correction of Ca2 + dynamics, not only for excitation/contraction coupling, but also for the prevention of rather irreparable events on cardiac energetics and ER stress adaptations. The latter may even impede with later onset of Ca2+-related therapeutic interventions and should gain more focus for HF treatment.
Collapse
Affiliation(s)
- Theresa Brand
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, Würzburg 97078, Germany
| | - Bettina Tanitha Baumgarten
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, Dortmund 44139, Germany
| | - Sabrina Denzinger
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, Würzburg 97078, Germany
| | - Yvonne Reinders
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, Dortmund 44139, Germany
| | - Miriam Kleindl
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, Dortmund 44139, Germany
| | - Constanze Schanbacher
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, Würzburg 97078, Germany
| | - Florian Funk
- Institute of Pharmacology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
| | - Nilgün Gedik
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Hufelandstr. 55, Essen 45122, Germany
| | - Mahmood Jabbasseh
- Institute of Pharmacology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Hufelandstr. 55, Essen 45122, Germany
| | - Jan Dudek
- Comprehensive Heart Failure Center, University Hospital of Würzburg, Am Schwarzenberg 15, Würzburg 97078, Germany
| | - Julia Szendroedi
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Elen Tolstik
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, Dortmund 44139, Germany
| | - Kai Schuh
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Martina Krüger
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany; Institute of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Germany; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, United States; Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Quebec, Canada
| | - Friederike Cuello
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany; Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, Dortmund 44139, Germany
| | - Joachim P Schmitt
- Institute of Pharmacology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany.
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, Würzburg 97078, Germany; Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, Dortmund 44139, Germany; Comprehensive Heart Failure Center, University Hospital of Würzburg, Am Schwarzenberg 15, Würzburg 97078, Germany.
| |
Collapse
|
3
|
Molecular Signaling to Preserve Mitochondrial Integrity against Ischemic Stress in the Heart: Rescue or Remove Mitochondria in Danger. Cells 2021; 10:cells10123330. [PMID: 34943839 PMCID: PMC8699551 DOI: 10.3390/cells10123330] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases are one of the leading causes of death and global health problems worldwide, and ischemic heart disease is the most common cause of heart failure (HF). The heart is a high-energy demanding organ, and myocardial energy reserves are limited. Mitochondria are the powerhouses of the cell, but under stress conditions, they become damaged, release necrotic and apoptotic factors, and contribute to cell death. Loss of cardiomyocytes plays a significant role in ischemic heart disease. In response to stress, protective signaling pathways are activated to limit mitochondrial deterioration and protect the heart. To prevent mitochondrial death pathways, damaged mitochondria are removed by mitochondrial autophagy (mitophagy). Mitochondrial quality control mediated by mitophagy is functionally linked to mitochondrial dynamics. This review provides a current understanding of the signaling mechanisms by which the integrity of mitochondria is preserved in the heart against ischemic stress.
Collapse
|
4
|
Yang X, Wang Y, Wu C, Ling EA. Animal Venom Peptides as a Treasure Trove for New Therapeutics Against Neurodegenerative Disorders. Curr Med Chem 2019; 26:4749-4774. [PMID: 30378475 DOI: 10.2174/0929867325666181031122438] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/08/2018] [Accepted: 10/24/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and cerebral ischemic stroke, impose enormous socio-economic burdens on both patients and health-care systems. However, drugs targeting these diseases remain unsatisfactory, and hence there is an urgent need for the development of novel and potent drug candidates. METHODS Animal toxins exhibit rich diversity in both proteins and peptides, which play vital roles in biomedical drug development. As a molecular tool, animal toxin peptides have not only helped clarify many critical physiological processes but also led to the discovery of novel drugs and clinical therapeutics. RESULTS Recently, toxin peptides identified from venomous animals, e.g. exenatide, ziconotide, Hi1a, and PcTx1 from spider venom, have been shown to block specific ion channels, alleviate inflammation, decrease protein aggregates, regulate glutamate and neurotransmitter levels, and increase neuroprotective factors. CONCLUSION Thus, components of venom hold considerable capacity as drug candidates for the alleviation or reduction of neurodegeneration. This review highlights studies evaluating different animal toxins, especially peptides, as promising therapeutic tools for the treatment of different neurodegenerative diseases and disorders.
Collapse
Affiliation(s)
- Xinwang Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Ying Wang
- Key Laboratory of Chemistry in Ethnic Medicine Resource, State Ethnic Affairs Commission & Ministry of Education, School of Ethnomedicine and Ethnopharmacy, Yunnan Minzu University, Kunming 650500, Yunnan, China
| | - Chunyun Wu
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
5
|
Li S, Chopra A, Keung W, Chan CWY, Costa KD, Kong CW, Hajjar RJ, Chen CS, Li RA. Sarco/endoplasmic reticulum Ca2+-ATPase is a more effective calcium remover than sodium-calcium exchanger in human embryonic stem cell-derived cardiomyocytes. Am J Physiol Heart Circ Physiol 2019; 317:H1105-H1115. [DOI: 10.1152/ajpheart.00540.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Human pluripotent stem cell (hPSCs)-derived ventricular (V) cardiomyocytes (CMs) display immature Ca2+–handing properties with smaller transient amplitudes and slower kinetics due to such differences in crucial Ca2+-handling proteins as the poor sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump but robust Na+-Ca2+ exchanger (NCX) activities in human embryonic stem cell (ESC)-derived VCMs compared with adult. Despite their fundamental importance in excitation-contraction coupling, the relative contribution of SERCA and NCX to Ca2+-handling of hPSC-VCMs remains unexplored. We systematically altered the activities of SERCA and NCX in human embryonic stem cell-derived ventricular cardiomyocytes (hESC-VCMs) and their engineered microtissues, followed by examining the resultant phenotypic consequences. SERCA overexpression in hESC-VCMs shortened the decay of Ca2+ transient at low frequencies (0.5 Hz) without affecting the amplitude, SR Ca2+ content and Ca2+ baseline. Interestingly, short hairpin RNA-based NCX suppression did not prolong the transient decay, indicating a compensatory response for Ca2+ removal. Although hESC-VCMs and their derived microtissues exhibited negative frequency-transient/force responses, SERCA overexpression rendered them less negative at high frequencies (>2 Hz) by accelerating Ca2+ sequestration. We conclude that for hESC-VCMs and their microtissues, SERCA, rather than NCX, is the main Ca2+ remover during diastole; poor SERCA expression is the leading cause for immature negative-frequency/force responses, which can be partially reverted by forced expression. Combinatorial approach to mature calcium handling in hESC-VCMs may help shed further mechanistic insights. NEW & NOTEWORTHY In this study of human pluripotent stem cell-derived cardiomyocytes, we studied the role of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) and Na+-Ca2+ exchanger (NCX) in Ca2+ handling. Our data support the notion that SERCA is more effective in cytosolic calcium removal than the NCX.
Collapse
Affiliation(s)
- Sen Li
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Anant Chopra
- Department of Bioengineering, Boston University, Boston, Massachusetts
- Harvard Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts
| | - Wendy Keung
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Camie W. Y. Chan
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Kevin D. Costa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, New York
| | - Chi-Wing Kong
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Roger J. Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, New York
| | - Christopher S. Chen
- Department of Bioengineering, Boston University, Boston, Massachusetts
- Harvard Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts
| | - Ronald A. Li
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
- Ming-Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong
| |
Collapse
|
6
|
Bae Y, Thuy LT, Lee YH, Ko KS, Han J, Choi JS. Polyplexes of Functional PAMAM Dendrimer/Apoptin Gene Induce Apoptosis of Human Primary Glioma Cells In Vitro. Polymers (Basel) 2019; 11:E296. [PMID: 30960280 PMCID: PMC6419211 DOI: 10.3390/polym11020296] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/01/2019] [Accepted: 02/07/2019] [Indexed: 01/21/2023] Open
Abstract
Highly efficient and safe gene delivery has become an important aspect of neuronal gene therapy. We evaluated the ability of polyamidoamine (PAMAM) dendrimer grafted with phenylalanine, histidine, and arginine (PAMAM-FHR), a nonviral gene delivery vector, to deliver a therapeutic, tumor cell-specific killer gene, apoptin, into the human primary glioma cell line GBL-14 and human dermal fibroblasts. We performed a transfection assay using plasmids of luciferase and enhanced green fluorescent protein (EGFP) and assessed cell viability. Both cell lines were treated with complexes of PAMAM-FHR and apoptin after which their intracellular uptake and localization were examined by fluorescence-activated cell sorting (FACS)analysis and confocal laser scanning microscopy. Confocal microscopy showed that the PAMAM-FHR escaped from the endo-lysosome into the cytosol. Cell cycle phase distribution analysis, annexin V staining, and a tetramethylrhodamine ethyl ester (TMRE) assay established that apoptin triggered apoptosis in the GBL-14 cell line but not in normal fibroblasts. These results indicated that the PAMAM-FHR/apoptin complex is an effective gene vehicle for cancer therapy in vitro.
Collapse
Affiliation(s)
- Yoonhee Bae
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea.
| | - Le Thi Thuy
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea.
| | - Young Hwa Lee
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea.
| | - Kyung Soo Ko
- Department of Internal Medicine, Sanggye Paik Hospital, Cardiovascular and Metabolic Disease Center, Inje University, Seoul 100-032, Korea.
| | - Jin Han
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea.
| | - Joon Sig Choi
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea.
| |
Collapse
|
7
|
Li JZ, Zhou LY, Peng YL, Fan J. Pseudomonas bacteriocin syringacin M released upon desiccation suppresses the growth of sensitive bacteria in plant necrotic lesions. Microb Biotechnol 2019; 13:134-147. [PMID: 30672132 PMCID: PMC6922522 DOI: 10.1111/1751-7915.13367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/01/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022] Open
Abstract
Bacteriocins are regarded as important factors mediating microbial interactions, but their exact role in community ecology largely remains to be elucidated. Here, we report the characterization of a mutant strain, derived from Pseudomonas syringae pv. tomato DC3000 (Pst), that was incapable of growing in plant extracts and causing disease. Results showed that deficiency in a previously unannotated gene saxE led to the sensitivity of the mutant to Ca2+ in leaf extracts. Transposon insertions in the bacteriocin gene syrM, adjacent to saxE, fully rescued the bacterial virulence and growth of the ΔsaxE mutant in plant extracts, indicating that syrM‐saxE encode a pair of bacteriocin immunity proteins in Pst. To investigate whether the syrM‐saxE system conferred any advantage to Pst in competition with other SyrM‐sensitive pathovars, we compared the growth of a SyrM‐sensitive strain co‐inoculated with Pst strains with or without the syrM gene and observed a significant syrM‐dependent growth reduction of the sensitive bacteria on plate and in lesion tissues upon desiccation–rehydration treatment. These findings reveal an important biological role of SyrM‐like bacteriocins and help to understand the complex strategies used by P. syringae in adaptation to the phyllosphere niche in the context of plant disease.
Collapse
Affiliation(s)
- Jun-Zhou Li
- Ministry of Agriculture Key Laboratory for Crop Pest Monitoring and Green Control, China Agricultural University, Beijing, 100193, China
| | - Li-Ying Zhou
- Ministry of Agriculture Key Laboratory for Crop Pest Monitoring and Green Control, China Agricultural University, Beijing, 100193, China
| | - You-Liang Peng
- Ministry of Agriculture Key Laboratory for Crop Pest Monitoring and Green Control, China Agricultural University, Beijing, 100193, China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100193, China
| | - Jun Fan
- Ministry of Agriculture Key Laboratory for Crop Pest Monitoring and Green Control, China Agricultural University, Beijing, 100193, China.,Joint Laboratory for International Cooperation in Crop Molecular Breeding, Ministry of Education, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
8
|
Cagle BS, Crawford RA, Doorn JA. Biogenic Aldehyde-Mediated Mechanisms of Toxicity in Neurodegenerative Disease. CURRENT OPINION IN TOXICOLOGY 2018; 13:16-21. [PMID: 31304429 DOI: 10.1016/j.cotox.2018.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Oxidative decomposition of several biomolecules produces reactive aldehydes. Monoamine neurotransmitters are enzymatically converted to aldehydes via monoamine oxidase followed by further metabolism such as carbonyl oxidation/reduction. Elevated levels of aldehyde intermediates are implicated as factors in several pathological conditions, including Parkinson's disease. The biogenic aldehydes produced from dopamine, norepinephrine and serotonin are known to be toxic, generate reactive oxygen species and/or cause aggregation of proteins such as α-synuclein. Polyunsaturated lipids undergo oxidative decomposition to produce biogenic aldehydes, including 4-hydroxy-2-nonenal and malondialdehyde. These lipid aldehydes, some including an α,β-unsaturated carbonyl, target important proteins such as α-synuclein, proteasome degradation and G-protein-coupled signaling. Overproduction of biogenic aldehydes is a hypothesized factor in neurodegeneration; preventing their formation or scavenging may provide means for neuroprotection.
Collapse
Affiliation(s)
- Brianna S Cagle
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, 115 South Grand Ave, Iowa City, IA 52242-1112, USA
| | - Rachel A Crawford
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, 115 South Grand Ave, Iowa City, IA 52242-1112, USA
| | - Jonathan A Doorn
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, 115 South Grand Ave, Iowa City, IA 52242-1112, USA
| |
Collapse
|
9
|
de Souza JM, Goncalves BDC, Gomez MV, Vieira LB, Ribeiro FM. Animal Toxins as Therapeutic Tools to Treat Neurodegenerative Diseases. Front Pharmacol 2018; 9:145. [PMID: 29527170 PMCID: PMC5829052 DOI: 10.3389/fphar.2018.00145] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/09/2018] [Indexed: 12/21/2022] Open
Abstract
Neurodegenerative diseases affect millions of individuals worldwide. So far, no disease-modifying drug is available to treat patients, making the search for effective drugs an urgent need. Neurodegeneration is triggered by the activation of several cellular processes, including oxidative stress, mitochondrial impairment, neuroinflammation, aging, aggregate formation, glutamatergic excitotoxicity, and apoptosis. Therefore, many research groups aim to identify drugs that may inhibit one or more of these events leading to neuronal cell death. Venoms are fruitful natural sources of new molecules, which have been relentlessly enhanced by evolution through natural selection. Several studies indicate that venom components can exhibit selectivity and affinity for a wide variety of targets in mammalian systems. For instance, an expressive number of natural peptides identified in venoms from animals, such as snakes, scorpions, bees, and spiders, were shown to lessen inflammation, regulate glutamate release, modify neurotransmitter levels, block ion channel activation, decrease the number of protein aggregates, and increase the levels of neuroprotective factors. Thus, these venom components hold potential as therapeutic tools to slow or even halt neurodegeneration. However, there are many technological issues to overcome, as venom peptides are hard to obtain and characterize and the amount obtained from natural sources is insufficient to perform all the necessary experiments and tests. Fortunately, technological improvements regarding heterologous protein expression, as well as peptide chemical synthesis will help to provide enough quantities and allow chemical and pharmacological enhancements of these natural occurring compounds. Thus, the main focus of this review is to highlight the most promising studies evaluating animal toxins as therapeutic tools to treat a wide variety of neurodegenerative conditions, including Alzheimer's disease, Parkinson's disease, brain ischemia, glaucoma, amyotrophic lateral sclerosis, and multiple sclerosis.
Collapse
Affiliation(s)
- Jessica M. de Souza
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bruno D. C. Goncalves
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcus V. Gomez
- Department of Neurotransmitters, Instituto de Ensino e Pesquisa Santa Casa, Belo Horizonte, Brazil
| | - Luciene B. Vieira
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabiola M. Ribeiro
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
10
|
Chapa-Dubocq X, Makarov V, Javadov S. Simple kinetic model of mitochondrial swelling in cardiac cells. J Cell Physiol 2018; 233:5310-5321. [PMID: 29215716 DOI: 10.1002/jcp.26335] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/27/2017] [Indexed: 12/12/2022]
Abstract
Mitochondria play an important role in both cell survival and cell death. In response to oxidative stress, they undergo opening of non-selective permeability transition pores (PTP) in the inner mitochondrial membrane. Sustained PTP opening triggers mitochondrial swelling due to increased colloidal osmotic pressure in the matrix accompanied by mitochondrial membrane depolarization and ATP hydrolysis. Mitochondrial swelling is the major factor leading to mitochondria-mediated cell death through both apoptosis and necrosis. Hence, precise estimation of the threshold parameters of the transition of reversible swelling to irreversible swelling is important for understanding the mechanisms of PTP-mediated cell death as well as for the development of new therapeutic approaches targeting the mitochondria under pathological conditions. In this study, we designed a simple kinetic model of the Ca2+ -induced mitochondrial swelling that describes the mechanisms of transition from reversible to irreversible swelling in cardiac mitochondria. Values of kinetic parameters calculated using parameter estimation techniques that fit experimental data of mitochondrial swelling with minimum average differences between the experimental data and model parameters. Overall, this study provides a kinetic model verified by data simulation and model fitting that adequately describes the dynamics of mitochondrial swelling.
Collapse
Affiliation(s)
- Xavier Chapa-Dubocq
- Department of Physiology and Biophysics, Medical Sciences Campus University of Puerto Rico, San Juan, Puerto Rico
| | - Vladimir Makarov
- Department of Physics, University of Puerto Rico Rio Piedras Campus, San Juan, Puerto Rico
| | - Sabzali Javadov
- Department of Physiology and Biophysics, Medical Sciences Campus University of Puerto Rico, San Juan, Puerto Rico
| |
Collapse
|
11
|
Lu J, Lee YK, Ran X, Lai WH, Li RA, Keung W, Tse K, Tse HF, Yao X. An abnormal TRPV4-related cytosolic Ca2+ rise in response to uniaxial stretch in induced pluripotent stem cells-derived cardiomyocytes from dilated cardiomyopathy patients. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2964-2972. [DOI: 10.1016/j.bbadis.2017.07.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/15/2017] [Accepted: 07/24/2017] [Indexed: 01/01/2023]
|
12
|
Shoshan-Barmatz V, De S, Meir A. The Mitochondrial Voltage-Dependent Anion Channel 1, Ca 2+ Transport, Apoptosis, and Their Regulation. Front Oncol 2017; 7:60. [PMID: 28443244 PMCID: PMC5385329 DOI: 10.3389/fonc.2017.00060] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/17/2017] [Indexed: 01/08/2023] Open
Abstract
In the outer mitochondrial membrane, the voltage-dependent anion channel 1 (VDAC1) functions in cellular Ca2+ homeostasis by mediating the transport of Ca2+ in and out of mitochondria. VDAC1 is highly Ca2+-permeable and modulates Ca2+ access to the mitochondrial intermembrane space. Intramitochondrial Ca2+ controls energy metabolism by enhancing the rate of NADH production via modulating critical enzymes in the tricarboxylic acid cycle and fatty acid oxidation. Mitochondrial [Ca2+] is regarded as an important determinant of cell sensitivity to apoptotic stimuli and was proposed to act as a "priming signal," sensitizing the organelle and promoting the release of pro-apoptotic proteins. However, the precise mechanism by which intracellular Ca2+ ([Ca2+]i) mediates apoptosis is not known. Here, we review the roles of VDAC1 in mitochondrial Ca2+ homeostasis and in apoptosis. Accumulated evidence shows that apoptosis-inducing agents act by increasing [Ca2+]i and that this, in turn, augments VDAC1 expression levels. Thus, a new concept of how increased [Ca2+]i activates apoptosis is postulated. Specifically, increased [Ca2+]i enhances VDAC1 expression levels, followed by VDAC1 oligomerization, cytochrome c release, and subsequently apoptosis. Evidence supporting this new model suggesting that upregulation of VDAC1 expression constitutes a major mechanism by which apoptotic stimuli induce apoptosis with VDAC1 oligomerization being a molecular focal point in apoptosis regulation is presented. A new proposed mechanism of pro-apoptotic drug action, namely Ca2+-dependent enhancement of VDAC1 expression, provides a platform for developing a new class of anticancer drugs modulating VDAC1 levels via the promoter and for overcoming the resistance of cancer cells to chemotherapy.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Soumasree De
- Department of Life Sciences, National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Meir
- Department of Life Sciences, National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
13
|
Lv D, Bai Z, Yang L, Li X, Chen X. Lipid emulsion reverses bupivacaine-induced apoptosis of h9c2 cardiomyocytes: PI3K/Akt/GSK-3β signaling pathway. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 42:85-91. [PMID: 26809062 DOI: 10.1016/j.etap.2016.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 01/02/2016] [Accepted: 01/05/2016] [Indexed: 06/05/2023]
Abstract
Some findings have suggested that the rescue of bupivacaine (BPV)-induced cardiotoxicity by lipid emulsion (LE) is associated with inhibition of mitochondrial permeability transition pore (mPTP). However, the mechanism of this rescue action is not clearly known. In this study, the roles of phosphoinositide 3-kinase (PI3K)/Akt and glycogen synthase kinase-3β (GSK-3β) in the molecular mechanism of LE-induced protection and its relationship with mPTP were explored. h9c2 cardiomyocytes were randomly divided into several groups: control, BPV, LE, BPV+LE. To study the effect of LE on mPTP, atractyloside (Atr, 20 μM, mPTP opener) and cyclosporine A (CsA, 10 μM, mPTP blocker) were used. To unravel whether LE protects heart through the PI3K/Akt/GSK-3β signaling pathway, cells were treated with LY294002 (LY, 30 μM, PI3K blocker) or TWS119 (TWS 10 μM, GSK-3β blocker). Later mitochondrial respiratory chain complexes, apoptosis, opening of mPTP and phosphorylation levels of Akt/GSK-3β were measured. LE significantly improved the mitochondrial functions in h9c2 cardiomyocytes. LE reversed the BPV-induced apoptosis and the opening of mPTP. The effect of LE was not only enhanced by CsA and TWS, but also abolished by Atr and LY. LE also increased the phosphorylation levels of Akt and GSK-3β. These results suggested that LE can reverse the apoptosis in cardiomyocytes by BPV and a mechanism of its action is inhibition of mPTP opening through the PI3K/Akt/GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Danni Lv
- Ning Xia Medical University, Yin Chuan, China.
| | - Zhixia Bai
- Department of Anesthesiology, Tumor Hospital, General Hospital of Ning Xia Medical University, Yin Chuan, China.
| | - Libin Yang
- Department of Anesthesiology, First People's Hospital, Shi Zui Shan, China.
| | - Xiaohui Li
- Ning Xia Medical University, Yin Chuan, China.
| | - Xuexin Chen
- Department of Anesthesiology, Tumor Hospital, General Hospital of Ning Xia Medical University, Yin Chuan, China.
| |
Collapse
|
14
|
Sharma V, O'Halloran DM. Nematode Sodium Calcium Exchangers: A Surprising Lack of Transport. Bioinform Biol Insights 2016; 10:1-4. [PMID: 26848260 PMCID: PMC4737524 DOI: 10.4137/bbi.s37130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 12/28/2015] [Accepted: 01/02/2016] [Indexed: 12/14/2022] Open
Abstract
Na+/Ca2+ exchangers are low-affinity, high-capacity transporters that rapidly transport calcium against a gradient of Na+ ions. Na+/Ca2+ exchangers are divided into three groups based upon substrate specificity: Na+/Ca2+ exchangers (NCX), Na+/Ca2+/K+ exchangers (NCKX), and Ca2+/cation exchangers (NCLX). In mammals, there are three NCX genes, five NCKX genes, and a single NCLX gene. The genome of the nematode Caenorhabditis elegans contains 10 Na+/Ca2+ exchanger genes: three NCX, five NCLX, and two NCKX genes. In a previous study, we characterized the structural and taxonomic specializations within the family of Na+/Ca2+ exchangers across the phylum Nematoda and observed a complex picture of Na+/Ca2+ exchanger evolution across diverse nematode species. We noted multiple cases of putative gene gain and loss and, most surprisingly, did not detect members of the NCLX type of exchangers within subsets of nematode species. In this commentary, we discuss these findings and speculate on the functional outcomes and physiology of these observations. Our data highlight the importance of studying diverse systems in order to get a deeper understanding of the evolution and regulation of Ca2+ signaling critical for animal function.
Collapse
Affiliation(s)
- Vishal Sharma
- Department of Biological Sciences, The George Washington University, Washington, DC, USA
| | - Damien M O'Halloran
- Department of Biological Sciences, The George Washington University, Washington, DC, USA.; Institute for Neuroscience, The George Washington University, Washington, DC, USA
| |
Collapse
|
15
|
Mabandla MV, Nyoka M, Daniels WMU. Early use of oleanolic acid provides protection against 6-hydroxydopamine induced dopamine neurodegeneration. Brain Res 2015; 1622:64-71. [PMID: 26111646 DOI: 10.1016/j.brainres.2015.06.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 06/11/2015] [Accepted: 06/13/2015] [Indexed: 12/13/2022]
Abstract
Oleanolic acid is a triterpenoid that has been shown to possess antioxidant properties. In this study we investigated the effects of oleanolic acid in a parkinsonian rat model. Unilateral 6-hydroxydopamine (6-OHDA) lesions were carried out on postnatal day (PND) 60 in 4 groups viz. (1) Rats that started oleanolic acid treatment 7 days prior to lesion. (2) Rats not treated with oleanolic acid. (3) Rats that started oleanolic acid treatment 1 day post-lesion. (4) Rats treated with oleanolic acid 7 days post-lesion. The degree of forelimb impairment was assessed using limb use asymmetry and forelimb akinesia tests. Neurochemical changes were assessed using a Dopamine ELISA kit and mitochondrial apoptosis was measured using a mitochondrial apoptosis detection kit. In this study, animals injected with 6-OHDA displayed forelimb use asymmetry that was ameliorated by treatment with oleanolic acid 7 days pre- and 1 day post-lesion. In the cylinder test, rats injected with 6-OHDA favored using the forelimb ipsilateral (unimpaired) to the lesioned hemisphere while rats treated with oleanolic acid used the forelimb contralateral (impaired) to the lesioned hemisphere significantly more. Rats treated with oleanolic acid 7 days pre- and 1 day post-lesion had more dopamine in the striatum than the non-treated or the 7 days after lesion rats. Similarly, 6-OHDA-induced membrane depolarization was decreased in rats that received oleanolic acid treatment pre- or immediately post-lesion. This suggests that early treatment with oleanolic acid protects dopamine neurons from the toxic effects of 6-OHDA.
Collapse
Affiliation(s)
- Musa V Mabandla
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa.
| | - Mpumelelo Nyoka
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Willie M U Daniels
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| |
Collapse
|
16
|
Rutkovskiy A, Mariero LH, Vaage J. Deletion of the aquaporin-4 gene alters expression and phosphorylation of protective kinases in the mouse heart. Scandinavian Journal of Clinical and Laboratory Investigation 2014; 74:500-5. [PMID: 24792367 DOI: 10.3109/00365513.2014.905698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIM Aquaporins are channel-forming proteins highly permeable to water and some small molecular solutes. We have previously shown that aquaporin-4 knockout mice have increased tolerance to ischemia. However, the mechanism of cardioprotection was unclear. The aim of the current study was to investigate the effects of aquaporin-4 deletion on baseline expression and phosphorylation of some cardioprotective protein kinases. METHODS Proteins were extracted from hearts of aquaporin-4 knockout mice and littermate wild-type controls and analyzed with Western blot. Samples were taken from young (≤ 6 months of age), and old (≥ 1 year) mice. RESULTS Western blots showed three different isoforms of aquaporin-4 in wild types, likely representing M1, M23, and Mz. Total AMP-dependent kinase expression was decreased in aquaporin-4 knockout hearts by 18 ± 13% (p = 0.02), while the expression of Akt kinase, extracellular signal regulated kinase 1/2, protein kinase C-epsilon, mitogen-associated kinase P38 and C-Jun N-terminal kinase was unchanged. The phosphorylation of Akt kinase was reduced in hearts from knockout mice by 41 ± 16% (p = 0.01). No other alterations in phosphorylation were found. These effects were only detected in young mice. CONCLUSION Deletion of the aquaporin-4 gene decreased AMP-dependent kinase expression and Akt kinase phosphorylation in the heart. These changes may influence energy metabolism and endogenous cardioprotection.
Collapse
Affiliation(s)
- Arkady Rutkovskiy
- Department of Emergency and Intensive Care at the Institute of Clinical Medicine , Norway
| | | | | |
Collapse
|
17
|
Weisthal S, Keinan N, Ben-Hail D, Arif T, Shoshan-Barmatz V. Ca(2+)-mediated regulation of VDAC1 expression levels is associated with cell death induction. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2270-81. [PMID: 24704533 DOI: 10.1016/j.bbamcr.2014.03.021] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 03/07/2014] [Accepted: 03/25/2014] [Indexed: 12/16/2022]
Abstract
VDAC1, an outer mitochondrial membrane (OMM) protein, is crucial for regulating mitochondrial metabolic and energetic functions and acts as a convergence point for various cell survival and death signals. VDAC1 is also a key player in apoptosis, involved in cytochrome c (Cyto c) release and interactions with anti-apoptotic proteins. Recently, we demonstrated that various pro-apoptotic agents induce VDAC1 oligomerization and proposed that a channel formed by VDAC1 oligomers mediates cytochrome c release. As VDAC1 transports Ca(2+) across the OMM and because Ca(2+) has been implicated in apoptosis induction, we addressed the relationship between cytosolic Ca(2+) levels ([Ca(2)(+)]i), VDAC1 oligomerization and apoptosis induction. We demonstrate that different apoptosis inducers elevate cytosolic Ca(2+) and induce VDAC1 over-expression. Direct elevation of [Ca(2+)]i by the Ca(2+)-mobilizing agents A23187, ionomycin and thapsigargin also resulted in VDAC1 over-expression, VDAC1 oligomerization and apoptosis. In contrast, decreasing [Ca(2+)]i using the cell-permeable Ca(2+)-chelating reagent BAPTA-AM inhibited VDAC1 over-expression, VDAC1 oligomerization and apoptosis. Correlation between the increase in VDAC1 levels and oligomerization, [Ca(2+)]i levels and apoptosis induction, as induced by H2O2 or As2O3, was also obtained. On the other hand, cells transfected to overexpress VDAC1 presented Ca(2+)-independent VDAC1 oligomerization, cytochrome c release and apoptosis, suggesting that [Ca(2+)]i elevation is not a pre-requisite for apoptosis induction when VDAC1 is over-expressed. The results suggest that Ca(2+) promotes VDAC1 over-expression by an as yet unknown signaling pathway, leading to VDAC1 oligomerization, ultimately resulting in apoptosis. These findings provide a new insight into the mechanism of action of existing anti-cancer drugs involving induction of VDAC1 over-expression as a mechanism for inducing apoptosis. This article is part of a Special Issue entitled: Calcium Signaling in Health and Disease. Guest Editors: Geert Bultynck, Jacques Haiech, Claus W. Heizmann, Joachim Krebs, and Marc Moreau.
Collapse
Affiliation(s)
- Shira Weisthal
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Nurit Keinan
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Danya Ben-Hail
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Tasleem Arif
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Varda Shoshan-Barmatz
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| |
Collapse
|
18
|
Simard JM, Woo SK, Gerzanich V. Transient receptor potential melastatin 4 and cell death. Pflugers Arch 2012; 464:573-82. [PMID: 23065026 PMCID: PMC3513597 DOI: 10.1007/s00424-012-1166-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 09/25/2012] [Accepted: 09/25/2012] [Indexed: 12/12/2022]
Abstract
Cell death proceeds by way of a variety of “cell death subroutines,” including several types of “apoptosis,” “regulated necrosis,” and others. “Accidental necrosis” due to profound adenosine triphosphate (ATP) depletion or oxidative stress is distinguished from regulated necrosis by the absence of death receptor signaling. However, both accidental and regulated necrosis have in common the process of “oncosis,” a physiological process characterized by Na+ influx and cell volume increase that, in necrotic cell death, is required to produce the characteristic features of membrane blebbing and membrane rupture. Here, we review emerging evidence that the monovalent cation channel, transient receptor potential melastatin 4 (TRPM4), is involved in the cell death process of oncosis. Potential involvement of TRPM4 in oncosis is suggested by the fact that the two principal regulators of TRPM4, intracellular ATP and Ca2+, are both altered during necrosis in the direction that causes TRPM4 channel opening. Under physiological conditions, activation of TRPM4 promotes Na+ influx and cell depolarization. Under pathological conditions, unchecked activation of TRPM4 leads to Na+ overload, cell volume increase, blebbing and cell membrane rupture, the latter constituting the irreversible end stage of necrosis. Emerging data indicate that TRPM4 plays a crucial role as end executioner in the accidental necrotic death of ATP-depleted or redox-challenged endothelial and epithelial cells, both in vitro and in vivo. Future studies will be needed to determine whether TRPM4 also plays a role in regulated necrosis and apoptosis.
Collapse
Affiliation(s)
- J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201-1595, USA.
| | | | | |
Collapse
|
19
|
Li P, Jiao J, Gao G, Prabhakar BS. Control of mitochondrial activity by miRNAs. J Cell Biochem 2012; 113:1104-10. [PMID: 22135235 DOI: 10.1002/jcb.24004] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitochondria supply energy for physiological function and they participate in the regulation of other cellular events including apoptosis, calcium homeostasis, and production of reactive oxygen species. Thus, mitochondria play a critical role in the cells. However, dysfunction of mitochondria is related to a variety of pathological processes and diseases. MicroRNAs (miRNAs) are a class of small noncoding RNAs about 22 nucleotides long, and they can bind to the 3'-untranslated region (3'-UTR) of mRNAs, thereby inhibiting mRNA translation or promoting mRNA degradation. We summarize the molecular regulation of mitochondrial metabolism, structure, and function by miRNAs. Modulation of miRNAs levels may provide a new therapeutic approach for the treatment of mitochondria-related diseases.
Collapse
Affiliation(s)
- Peifeng Li
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | |
Collapse
|
20
|
Clementi EA, Marks LR, Duffey ME, Hakansson AP. A novel initiation mechanism of death in Streptococcus pneumoniae induced by the human milk protein-lipid complex HAMLET and activated during physiological death. J Biol Chem 2012; 287:27168-82. [PMID: 22700972 DOI: 10.1074/jbc.m112.371070] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
To cause colonization or infection, most bacteria grow in biofilms where differentiation and death of subpopulations is critical for optimal survival of the whole population. However, little is known about initiation of bacterial death under physiological conditions. Membrane depolarization has been suggested, but never shown to be involved, due to the difficulty of performing such studies in bacteria and the paucity of information that exists regarding ion transport mechanisms in prokaryotes. In this study, we performed the first extensive investigation of ion transport and membrane depolarization in a bacterial system. We found that HAMLET, a human milk protein-lipid complex, kills Streptococcus pneumoniae (the pneumococcus) in a manner that shares features with activation of physiological death from starvation. Addition of HAMLET to pneumococci dissipated membrane polarity, but depolarization per se was not enough to trigger death. Rather, both HAMLET- and starvation-induced death of pneumococci specifically required a sodium-dependent calcium influx, as shown using calcium and sodium transport inhibitors. This mechanism was verified under low sodium conditions, and in the presence of ionomycin or monensin, which enhanced pneumococcal sensitivity to HAMLET- and starvation-induced death. Pneumococcal death was also inhibited by kinase inhibitors, and indicated the involvement of Ser/Thr kinases in these processes. The importance of this activation mechanism was made evident, as dysregulation and manipulation of physiological death was detrimental to biofilm formation, a hallmark of bacterial colonization. Overall, our findings provide novel information on the role of ion transport during bacterial death, with the potential to uncover future antimicrobial targets.
Collapse
Affiliation(s)
- Emily A Clementi
- Department of Microbiology and Immunology, University at Buffalo, State University of New York, Buffalo, New York 14214, USA
| | | | | | | |
Collapse
|
21
|
Excitation-contraction coupling in ventricular myocytes is enhanced by paracrine signaling from mesenchymal stem cells. J Mol Cell Cardiol 2012; 52:1249-56. [PMID: 22465692 DOI: 10.1016/j.yjmcc.2012.03.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 02/14/2012] [Accepted: 03/12/2012] [Indexed: 12/17/2022]
Abstract
In clinical trials mesenchymal stem cells (MSCs) are transplanted into cardiac ischemic regions to decrease infarct size and improve contractility. However, the mechanism and time course of MSC-mediated cardioprotection are incompletely understood. We tested the hypothesis that paracrine signaling by MSCs promotes changes in cardiac excitation-contraction (EC) coupling that protects myocytes from cell death and enhances contractility. Isolated mouse ventricular myocytes (VMs) were treated with control tyrode, MSC conditioned-tyrode (ConT) or co-cultured with MSCs. The Ca handling properties of VMs were monitored by laser scanning confocal microscopy and whole cell voltage clamp. ConT superfusion of VMs resulted in a time dependent increase of the Ca transient amplitude (ConT(15min): ΔF/F(0)=3.52±0.38, n=14; Ctrl(15min): ΔF/F(0)=2.41±0.35, n=14) and acceleration of the Ca transient decay (τ: ConT: 269±18ms n=14; vs. Ctrl: 315±57ms, n=14). Voltage clamp recordings confirmed a ConT induced increase in I(Ca,L) (ConT: -5.9±0.5 pA/pF n=11; vs. Ctrl: -4.04±0.3 pA/pF, n=12). The change of τ resulted from increased SERCA activity. Changes in the Ca transient amplitude and τ were prevented by the PI3K inhibitors Wortmannin (100nmol/L) and LY294002 (10μmol/L) and the Akt inhibitor V (20μmol/L) indicating regulation through PI3K signal transduction and Akt activation which was confirmed by western blotting. A change in τ was also prevented in eNOS(-/-) myocytes or by inhibition of eNOS suggesting an NO mediated regulation of SERCA activity. Since paracrine signaling further resulted in increased survival of VMs we propose that the Akt induced change in Ca signaling is also a mechanism by which MSCs mediate an anti-apoptotic effect.
Collapse
|
22
|
O'Sullivan JF, Leblond AL, Kelly G, Kumar AH, Metharom P, Büneker CK, Alizadeh-Vikali N, Hristova I, Hynes BG, O'Connor R, Caplice NM. Potent Long-Term Cardioprotective Effects of Single Low-Dose Insulin-Like Growth Factor-1 Treatment Postmyocardial Infarction. Circ Cardiovasc Interv 2011; 4:327-35. [DOI: 10.1161/circinterventions.110.960765] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background—
Insulin-like growth factor-1 (IGF-1) is recognized as an important regulator of cardiac structure and cardiomyocyte homeostasis. The prosurvival and antiapoptotic effects of IGF-1 have been investigated in vitro and in rodent models of myocardial infarction (MI). However, the clinical application of IGF-1 has been hampered by dose-dependent side effects both acutely and during chronic administration. We hypothesized that single, low-dose IGF-1 (LD-IGF-1) administered locally and early in the reperfusion phase after acute MI in a large animal model would avoid significant side effects but would have prosurvival effects that would manifest in long-term structural and functional improvement after MI treatment.
Methods and Results—
Forty-four female Landrace pigs underwent intracoronary administration of LD-IGF-1 or saline 2 hours into the reperfusion phase of acute left anterior descending artery occlusion MI. In the area of infarction, IGF-1 receptor and signaling responses were activated at 30 minutes and cardiomyocyte cell death attenuated at 24 hours after LD-IGF-1 but not saline treatment. Hemodynamic and structural studies using pressure-volume loop, CT, and triphenyltetrazolium chloride analysis 2 months post-MI confirmed a marked reduction in infarct size, attenuation of wall thinning, and augmentation of wall motion in the LD-IGF-1-treated but not in the saline-treated animals. These regional structural benefits were associated with global reductions in left ventricular volumes and significant improvement in left ventricular systolic and diastolic function.
Conclusions—
One-time LD-IGF-1 effects potent acute myocardial salvage in a preclinical model of left anterior descending artery occlusive MI, extending to long-term benefits in MI size, wall structure, and function and underscoring its potential as an adjunctive therapeutic agent.
Collapse
Affiliation(s)
- John F. O'Sullivan
- From the Centre for Research in Vascular Biology (J.F.O., A.-L.L., A.H.S.K., P.M., C.K.B., N.A.-V., I.H., B.G.H., N.M.C.) and Cell Biology Laboratory (G.K., R.O.), Department of Biochemistry, Biosciences Institute, University College Cork, Cork, Ireland
| | - Anne-Laure Leblond
- From the Centre for Research in Vascular Biology (J.F.O., A.-L.L., A.H.S.K., P.M., C.K.B., N.A.-V., I.H., B.G.H., N.M.C.) and Cell Biology Laboratory (G.K., R.O.), Department of Biochemistry, Biosciences Institute, University College Cork, Cork, Ireland
| | - Geraldine Kelly
- From the Centre for Research in Vascular Biology (J.F.O., A.-L.L., A.H.S.K., P.M., C.K.B., N.A.-V., I.H., B.G.H., N.M.C.) and Cell Biology Laboratory (G.K., R.O.), Department of Biochemistry, Biosciences Institute, University College Cork, Cork, Ireland
| | - Arun H.S. Kumar
- From the Centre for Research in Vascular Biology (J.F.O., A.-L.L., A.H.S.K., P.M., C.K.B., N.A.-V., I.H., B.G.H., N.M.C.) and Cell Biology Laboratory (G.K., R.O.), Department of Biochemistry, Biosciences Institute, University College Cork, Cork, Ireland
| | - Pat Metharom
- From the Centre for Research in Vascular Biology (J.F.O., A.-L.L., A.H.S.K., P.M., C.K.B., N.A.-V., I.H., B.G.H., N.M.C.) and Cell Biology Laboratory (G.K., R.O.), Department of Biochemistry, Biosciences Institute, University College Cork, Cork, Ireland
| | - Chirlei K. Büneker
- From the Centre for Research in Vascular Biology (J.F.O., A.-L.L., A.H.S.K., P.M., C.K.B., N.A.-V., I.H., B.G.H., N.M.C.) and Cell Biology Laboratory (G.K., R.O.), Department of Biochemistry, Biosciences Institute, University College Cork, Cork, Ireland
| | - Niki Alizadeh-Vikali
- From the Centre for Research in Vascular Biology (J.F.O., A.-L.L., A.H.S.K., P.M., C.K.B., N.A.-V., I.H., B.G.H., N.M.C.) and Cell Biology Laboratory (G.K., R.O.), Department of Biochemistry, Biosciences Institute, University College Cork, Cork, Ireland
| | - Ivalina Hristova
- From the Centre for Research in Vascular Biology (J.F.O., A.-L.L., A.H.S.K., P.M., C.K.B., N.A.-V., I.H., B.G.H., N.M.C.) and Cell Biology Laboratory (G.K., R.O.), Department of Biochemistry, Biosciences Institute, University College Cork, Cork, Ireland
| | - Brian G. Hynes
- From the Centre for Research in Vascular Biology (J.F.O., A.-L.L., A.H.S.K., P.M., C.K.B., N.A.-V., I.H., B.G.H., N.M.C.) and Cell Biology Laboratory (G.K., R.O.), Department of Biochemistry, Biosciences Institute, University College Cork, Cork, Ireland
| | - Rosemary O'Connor
- From the Centre for Research in Vascular Biology (J.F.O., A.-L.L., A.H.S.K., P.M., C.K.B., N.A.-V., I.H., B.G.H., N.M.C.) and Cell Biology Laboratory (G.K., R.O.), Department of Biochemistry, Biosciences Institute, University College Cork, Cork, Ireland
| | - Noel M. Caplice
- From the Centre for Research in Vascular Biology (J.F.O., A.-L.L., A.H.S.K., P.M., C.K.B., N.A.-V., I.H., B.G.H., N.M.C.) and Cell Biology Laboratory (G.K., R.O.), Department of Biochemistry, Biosciences Institute, University College Cork, Cork, Ireland
| |
Collapse
|
23
|
Ben-Ami I, Yao Z, Naor Z, Seger R. Gq protein-induced apoptosis is mediated by AKT kinase inhibition that leads to protein kinase C-induced c-Jun N-terminal kinase activation. J Biol Chem 2011; 286:31022-31031a. [PMID: 21757743 DOI: 10.1074/jbc.m111.247726] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
G(q) protein-coupled receptors (G(q)PCRs) regulate various cellular processes, including mainly proliferation and differentiation. In a previous study we found that in prostate cancer cells, the G(q)PCR of gonadotropin-releasing hormone (GnRH) induces apoptosis by reducing the PKC-dependent AKT activity and elevating JNK phosphorylation. Because it was thought that G(q)PCRs mainly induce activation of AKT, we first undertook to examine how general this phenomenon is. In a screen of 21 cell lines we found that PKC activation results in the reduction of AKT activity, which correlates nicely with JNK activation and in some cases with apoptosis. To understand further the signaling pathways involved in this stimulation, we studied in detail SVOG-4O and αT3-1 cells. We found that prostaglandin F2α and GnRH agonist (GnRH-a) indeed induce significant Gα(q)- and PKC-dependent apoptosis in these cells. This is mediated by two signaling branches downstream of PKC, which converge at the level of MLK3 upstream of JNK. One branch consists of c-Src activation of the JNK cascade, and the second involves reduction of AKT activity that alleviates its inhibitory effect on MLK3 to allow the flow of the c-Src signal to JNK. At the MAPKK level, we found that the signal is transmitted by MKK7 and not MKK4. Our results present a general mechanism that mediates a G(q)PCR-induced, death receptor-independent, apoptosis in physiological, as well as cancer-related systems.
Collapse
Affiliation(s)
- Ido Ben-Ami
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Obstetrics and Gynecology, Assaf Harofeh Medical Center, Zerifin, Sackler School of Medicine, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Zhong Yao
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Zvi Naor
- Department Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Rony Seger
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
24
|
Usta E, Mustafi M, Artunc F, Walker T, Voth V, Aebert H, Ziemer G. The challenge to verify ceramide's role of apoptosis induction in human cardiomyocytes--a pilot study. J Cardiothorac Surg 2011; 6:38. [PMID: 21443760 PMCID: PMC3079610 DOI: 10.1186/1749-8090-6-38] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 03/28/2011] [Indexed: 12/19/2022] Open
Abstract
Background Cardioplegia and reperfusion of the myocardium may be associated with cardiomyocyte apoptosis and subsequent myocardial injury. In order to establish a pharmacological strategy for the prevention of these events, this study aimed to verify the reliability of our human cardiac model and to evaluate the pro-apoptotic properties of the sphingolipid second messenger ceramide and the anti-apoptotic properties of the acid sphingomyelinase inhibitor amitryptiline during simulated cardioplegia and reperfusion ex vivo. Methods Cardiac biopsies were retrieved from the right auricle of patients undergoing elective CABG before induction of cardiopulmonary bypass. Biopsies were exposed to ex vivo conditions of varying periods of cp/rep (30/10, 60/20, 120/40 min). Groups: I (untreated control, n = 10), II (treated control cp/rep, n = 10), III (cp/rep + ceramide, n = 10), IV (cp/rep + amitryptiline, n = 10) and V (cp/rep + ceramide + amitryptiline, n = 10). For detection of apoptosis anti-activated-caspase-3 and PARP-1 cleavage immunostaining were employed. Results In group I the percentage of apoptotic cardiomyocytes was significantly (p < 0.05) low if compared to group II revealing a time-dependent increase. In group III ceramid increased and in group IV amitryptiline inhibited apoptosis significantly (p < 0.05). In contrast in group V, under the influence of ceramide and amitryptiline the induction of apoptosis was partially suppressed. Conclusion Ceramid induces and amitryptiline suppresses apoptosis significantly in our ex vivo setting. This finding warrants further studies aiming to evaluate potential beneficial effects of selective inhibition of apoptosis inducing mediators on the suppression of ischemia/reperfusion injury in clinical settings.
Collapse
Affiliation(s)
- Engin Usta
- Children's University Hospital, Div, Congenital & Pediatric Cardiac Surgery; University Hospital Tübingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
25
|
Usta E, Mustafi M, Straub A, Ziemer G. The nonselective beta-blocker carvedilol suppresses apoptosis in human cardiac tissue: a pilot study. Heart Surg Forum 2011; 13:E218-22. [PMID: 20719722 DOI: 10.1532/hsf98.20091179] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Cardioplegia and reperfusion of the myocardium may be associated with cardiomyocyte apoptosis and subsequent myocardial injury. To establish a pharmacologic strategy for the prevention of these events, this study aimed to verify the reliability of our human cardiac model and to evaluate the antiapoptotic properties of the nonselective beta-blocker carvedilol during simulated cardioplegia and reperfusion ex vivo. METHODS Cardiac biopsies were retrieved before induction of cardiopulmonary bypass from the auricle of the right atrium of patients undergoing elective coronary artery bypass grafting. Biopsies were exposed to ex vivo conditions of varying periods of cardioplegia/reperfusion (30/10 minutes, 60/20 minutes, 120/40 minutes). Group I was the untreated control (n = 15), group II was the treated control (cardioplegia/reperfusion, n = 15), and group III was the experimental group (cardioplegia/reperfusion plus carvedilol, n = 15). Immunostaining for antibodies to activated caspase 3 and poly(ADP-ribose) polymerase 1 (PARP-1) cleavage was used to detect apoptosis. RESULTS The percentage of apoptotic cardiomyocytes was significantly lower (P < .05) in group I than in group II, revealing a time-dependent increase. In group III, carvedilol treatment suppressed apoptosis significantly (P < .05). CONCLUSION Carvedilol significantly suppresses apoptosis in our ex vivo setting. This finding warrants further studies to evaluate the potential beneficial effects of carvedilol in suppressing ischemia/reperfusion injury in clinical settings.
Collapse
Affiliation(s)
- Engin Usta
- Department of Thoracic, Cardiac and Vascular Surgery, Tübingen University Hospital, Tübingen, Germany.
| | | | | | | |
Collapse
|
26
|
Si KW, Liu JT, He LC, Li XK, Gou W, Liu CH, Li XQ. Effects of Caulophine on Caffeine-induced Cellular Injury and Calcium Homeostasis in Rat Cardiomyocytes. Basic Clin Pharmacol Toxicol 2010; 107:976-81. [DOI: 10.1111/j.1742-7843.2010.00618.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Dorn GW, Scorrano L. Two close, too close: sarcoplasmic reticulum-mitochondrial crosstalk and cardiomyocyte fate. Circ Res 2010; 107:689-99. [PMID: 20847324 DOI: 10.1161/circresaha.110.225714] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitochondria are key organelles in cell life whose dysfunction is associated with a variety of diseases. Their crucial role in intermediary metabolism and energy conversion makes them a preferred target in tissues, such as the heart, where the energetic demands are very high. In the cardiomyocyte, the spatial organization of mitochondria favors their interaction with the sarcoplasmic reticulum, thereby offering a mechanism for Ca(2+)-mediated crosstalk between these 2 organelles. Recently, the molecular basis for this interaction has begun to be unraveled, and we are learning how endoplasmic reticulum-mitochondrial interactions are often exploited by death signals, such as proapoptotic Bcl-2 family members, to amplify the cell death cascade. Here, we review our present understanding of the structural basis and the functional consequences of the close interaction between sarcoplasmic reticulum and mitochondria on cardiomyocyte function and death.
Collapse
Affiliation(s)
- Gerald W Dorn
- Washington University Center for Pharmacogenomics, Campus Box 8220, 660 S Euclid Ave, St Louis, MO 63110, USA.
| | | |
Collapse
|
28
|
Sapia L, Palomeque J, Mattiazzi A, Petroff MV. Na+/K+-ATPase inhibition by ouabain induces CaMKII-dependent apoptosis in adult rat cardiac myocytes. J Mol Cell Cardiol 2010; 49:459-68. [PMID: 20435043 DOI: 10.1016/j.yjmcc.2010.04.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 04/12/2010] [Accepted: 04/21/2010] [Indexed: 02/03/2023]
Abstract
The positive inotropic effect produced by Na(+)/K(+)-ATPase inhibition has been used for the treatment of heart failure for over 200 years. Recently, administration of toxic doses of ouabain has been shown to induce cardiac myocyte apoptosis. However, whether prolonged administration of non-toxic doses of ouabain can also promote cardiac myocyte cell death has never been explored. The aim of this study was to assess whether non-toxic doses of ouabain can induce myocyte apoptosis and if so, to examine the underlying mechanisms. For this purpose, cardiac myocytes from rat and cat, two species with different sensitivity to digitalis, were cultured for 24h in the presence or absence of 2 microM (rat) and 25 nm-2 microM ouabain (cat). Cell viability and apoptosis assays showed that ouabain produced, in the rat, a 43+/-5% decrease in cell viability due to apoptosis (enhanced caspase-3 activity, increased Bax/Bcl-2 and TUNEL-positive nuclei) and necrosis (LDH release and trypan blue staining). Similar results were obtained with 25 nM ouabain in the cat. Ouabain-induced reduction in cell viability was prevented by the NCX inhibitor KB-R7943 and by the CaMKII inhibitors, KN93 and AIP. Furthermore, CaMKII overexpression exacerbated ouabain-induced cell mortality which in contrast was reduced in transgenic mice with chronic CaMKII inhibition. However, KN93 failed to affect ouabain-induced inotropy. In addition, whereas ERK(1/2) inhibition with PD-98059 had no effect on cell mortality, PI3K inhibition with wortmannin, exacerbated myocyte death. We conclude that ouabain triggers an apoptotic cascade that involves NCX and CaMKII as a downstream effector. Ouabain simultaneously activates an antiapoptotic cascade involving PI3K/AKT which is however, insufficient to completely repress apoptosis. The finding that KN93 prevents ouabain-induced apoptosis without affecting inotropy suggests the potential use of CaMKII inhibitors as an adjunct to digitalis treatment for cardiovascular disease.
Collapse
Affiliation(s)
- Luciana Sapia
- Centro de Investigaciones Cardiovasculares, Conicet La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata 1900, Argentina
| | | | | | | |
Collapse
|
29
|
Usta E, Renovanz M, Mustafi M, Ziemer G, Aebert H. Human cardiac tissue in a microperfusion chamber simulating extracorporeal circulation--ischemia and apoptosis studies. J Cardiothorac Surg 2010; 5:3. [PMID: 20082695 PMCID: PMC2821307 DOI: 10.1186/1749-8090-5-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 01/18/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND After coronary artery bypass grafting ischemia/reperfusion injury inducing cardiomyocyte apoptosis may occur. This surgery-related inflammatory reaction appears to be of extreme complexity with regard to its molecular, cellular and tissue mechanisms and many studies have been performed on animal models. However, finding retrieved from animal studies were only partially confirmed in humans. To investigate this phenomenon and to evaluate possible therapies in vitro, adequate human cardiomyocyte models are required. We established a tissue model of human cardiomyocytes preserving the complex tissue environment. To our knowledge human cardiac tissue has not been investigated in an experimental setup mimicking extracorporeal circulation just in accordance to clinical routine, yet. METHODS Cardiac biopsies were retrieved from the right auricle of patients undergoing elective coronary artery bypass grafting before cardiopulmonary bypass. The extracorporeal circulation was simulated by submitting the biopsies to varied conditions simulating cardioplegia (cp) and reperfusion (rep) in a microperfusion chamber. Cp/rep time sets were 20/7, 40/13 and 60/20 min. For analyses of the calcium homoeostasis the fluorescent calcium ion indicator FURA-2 and for apoptosis detection PARP-1 cleavage immunostaining were employed. Further the anti-apoptotic effect of carvedilol [10 microM] was investigated by adding into the perfusate. RESULTS Viable cardiomyocytes presented an intact calcium homoeostasis under physiologic conditions. Following cardioplegia and reperfusion a time-dependent elevation of cytosolic calcium as a sign of disarrangement of the calcium homoeostasis occurred. PARP-1 cleavage also showed a time-dependence whereas reperfusion had the highest impact on apoptosis. Cardioplegia and carvedilol could reduce apoptosis significantly, lowering it between 60-70% (p < 0.05). CONCLUSIONS Our human cardiac preparation served as a reliable cellular model tool to study apoptosis in vitro. Decisively cardiac tissue from the right auricle can be easily obtained at nearly every cardiac operation avoiding biopsying of the myocardium or even experiments on animals.The apoptotic damage induced by the ischemia/reperfusion stimulus could be significantly reduced by the cold crystalloid cardioplegia. The additional treatment of cardiomyocytes with a non-selective beta-blocker, carvedilol had even a significantly higher reduction of apoptotis.
Collapse
Affiliation(s)
- Engin Usta
- Department of Thoracic-, Cardiac- and Vascular Surgery, Tübingen University Hospital, Germany.
| | | | | | | | | |
Collapse
|
30
|
Highly pathogenic H5N1 avian influenza virus induces extracellular Ca2+ influx, leading to apoptosis in avian cells. J Virol 2010; 84:3068-78. [PMID: 20053741 DOI: 10.1128/jvi.01923-09] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, we show that the highly pathogenic H5N1 avian influenza virus (AIV) (A/crow/Kyoto/53/04 and A/chicken/Egypt/CL6/07) induced apoptosis in duck embryonic fibroblasts (DEF). In contrast, apoptosis was reduced among cells infected with low-pathogenic AIVs (A/duck/HK/342/78 [H5N2], A/duck/HK/820/80 [H5N3], A/wigeon/Osaka/1/01 [H7N7], and A/turkey/Wisconsin/1/66 [H9N2]). Thus, we investigated the molecular mechanisms of apoptosis induced by H5N1-AIV infection. Caspase-dependent and -independent pathways contributed to the cytopathic effects. We further showed that, in the induction of apoptosis, the hemagglutinin of H5N1-AIV played a major role and its cleavage sequence was not critical. We also observed outer membrane permeabilization and loss of the transmembrane potential of the mitochondria of infected DEF, indicating that mitochondrial dysfunction was caused by the H5N1-AIV infection. We then analyzed Ca(2+) dynamics in the infected cells and demonstrated an increase in the concentration of Ca(2+) in the cytosol ([Ca(2+)](i)) and mitochondria ([Ca(2+)](m)) after H5N1-AIV infection. Regardless, gene expression important for regulating Ca(2+) efflux from the endoplasmic reticulum did not significantly change after H5N1-AIV infection. These results suggest that extracellular Ca(2+) may enter H5N1-AIV-infected cells. Indeed, EGTA, which chelates extracellular free Ca(2+), significantly reduced the [Ca(2+)](i), [Ca(2+)](m), and apoptosis induced by H5N1-AIV infection. In conclusion, we identified a novel mechanism for influenza A virus-mediated cell death, which involved the acceleration of extracellular Ca(2+) influx, leading to mitochondrial dysfunction and apoptosis. These findings may be useful for understanding the pathogenesis of H5N1-AIV in avian species as well as the impact of Ca(2+) homeostasis on influenza A virus infection.
Collapse
|
31
|
Abstract
Biological actions resulting from phosphoinositide synthesis trigger multiple downstream signalling cascades by recruiting proteins with pleckstrin homology domains, including phosphoinositide-dependent kinase-1 and protein kinase B (also known as Akt). Retrospectively, more attention has been focused on the plasma membrane-associated interactions of these molecules and resulting cytoplasmic target activation. The complex biological activities exerted by Akt activation suggest, however, that more subtle and complex subcellular control mechanisms are involved. This review examines the regulation of Akt activity from the perspective of subcellular compartmentalization and focuses specifically upon the actions of Akt activation downstream from phosphoinositide synthesis that influence cell biology by altering nuclear signalling leading to Pim-1 kinase induction as well as hexokinase phosphorylation that, together with Akt, serves to preserve mitochondrial integrity.
Collapse
Affiliation(s)
- Shigeki Miyamoto
- Department of Pharmacology, University of California, La Jolla, San Diego, CA 92093-0636, USA
| | - Marta Rubio
- Department of Biology, SDSU Heart Institute, San Diego State University, NLS 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Mark A. Sussman
- Department of Biology, SDSU Heart Institute, San Diego State University, NLS 426, 5500 Campanile Drive, San Diego, CA 92182, USA
- Corresponding author. Tel: +1 619 594 2983; +1 619 594 2610. E-mail address:
| |
Collapse
|
32
|
Miyamoto S, Murphy AN, Brown JH. Akt mediated mitochondrial protection in the heart: metabolic and survival pathways to the rescue. J Bioenerg Biomembr 2009; 41:169-80. [PMID: 19377835 PMCID: PMC2732429 DOI: 10.1007/s10863-009-9205-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiomyocyte death is now recognized as a critical factor in the development of heart disease. Mitochondria are not only responsible for energy production to ensure that cardiac output meets the body's energy demands, but they serve as critical integrators of cell survival signals. Numerous stressors are known to induce cell death by necrosis and/or apoptosis mediated through mitochondrial dysregulation. Anti- and pro-apoptotic Bcl-2 family proteins regulate apoptosis by controlling mitochondrial outer membrane permeability, whereas opening of the mitochondrial permeability transition pore (PT-pore) induces large amplitude permeability of the inner membrane and consequent rupture of the outer membrane. Akt is one of the best described survival kinases activated by receptor ligands and its activation preserves mitochondrial integrity and protects cardiomyocytes against necrotic and apoptotic death. The mechanisms responsible for Akt-mediated mitochondrial protection have not been fully elucidated. There is, however, accumulating evidence that multiple Akt target molecules, recruited through both transcriptional and post-transcriptional mechanisms, directly impinge upon and protect mitochondria. In this review we discuss mechanisms by which Akt activation can effect changes at the mitochondria that protect cardiomyocytes and attenuate pathophysiological responses of the heart.
Collapse
Affiliation(s)
- Shigeki Miyamoto
- Department of Pharmacology, University of California, 9500 Gilman Dr., La Jolla, San Diego, CA 92093-0636, USA.
| | | | | |
Collapse
|
33
|
Hausenloy DJ, Ong SB, Yellon DM. The mitochondrial permeability transition pore as a target for preconditioning and postconditioning. Basic Res Cardiol 2009; 104:189-202. [PMID: 19242644 DOI: 10.1007/s00395-009-0010-x] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 01/25/2009] [Accepted: 01/27/2009] [Indexed: 12/19/2022]
Abstract
The experimental evidence supporting the mitochondrial permeability transition pore (mPTP) as a major mediator of lethal myocardial reperfusion injury and therefore a critical target for cardioprotection is persuasive. Although, its molecular identity eludes investigators, it is generally accepted that mitochondrial cyclophilin-D, the target for the inhibitory effects of cyclosporine-A on the mPTP, is a regulatory component of the mPTP. Animal myocardial infarction studies and a recent clinical proof-of-concept study have demonstrated that pharmacologically inhibiting its opening at the onset of myocardial reperfusion reduces myocardial infarct size in the region of 30-50%. Interestingly, the inhibition of mPTP opening at this time appears to underpin the infarct-limiting effects of the endogenous cardioprotective strategies of ischemic preconditioning (IPC) and postconditioning (IPost). However, the mechanism underlying this inhibitory action of IPC and IPost on mPTP opening is unclear. The objectve of this review article will be to explore the potential mechanisms which link IPC and IPost to mPTP inhibition in the reperfused heart.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Institute and Center for Cardiology, University College London Hospitals and Medical School, Grafton Way, London, UK.
| | | | | |
Collapse
|
34
|
Dorn GW. Apoptotic and non-apoptotic programmed cardiomyocyte death in ventricular remodelling. Cardiovasc Res 2009; 81:465-73. [PMID: 18779231 PMCID: PMC2721651 DOI: 10.1093/cvr/cvn243] [Citation(s) in RCA: 219] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 08/26/2008] [Accepted: 08/28/2008] [Indexed: 12/25/2022] Open
Abstract
A defining cellular event in the transition from compensated hypertrophy to dilated cardiomyopathy is cardiomyocyte drop-out due to apoptosis, programmed necrosis, and autophagy. The importance of apoptosis in heart failure has been recognized for over a decade, while other forms of programmed cell death have more recently been appreciated, and their pathophysiological roles continue to be defined in experimental and clinical heart failure. The major focus of this review is on apoptosis in heart failure, with a discussion of molecular cross-talk between apoptosis, autophagy, and programmed necrosis.
Collapse
Affiliation(s)
- Gerald W Dorn
- Center for Pharmacogenomics and Cardiovascular Division, Department of Internal Medicine, Washington University, 660 S. Euclid Ave., Campus Box 8086, St Louis, MO 63110, USA.
| |
Collapse
|
35
|
Diwan A, Matkovich SJ, Yuan Q, Zhao W, Yatani A, Brown JH, Molkentin JD, Kranias EG, Dorn GW. Endoplasmic reticulum-mitochondria crosstalk in NIX-mediated murine cell death. J Clin Invest 2008; 119:203-12. [PMID: 19065046 DOI: 10.1172/jci36445] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Accepted: 10/15/2008] [Indexed: 01/09/2023] Open
Abstract
Transcriptional upregulation of the proapoptotic BCL2 family protein NIX limits red blood cell formation and can cause heart failure by inducing cell death, but the requisite molecular events are poorly defined. Here, we show complementary mechanisms for NIX-mediated cell death involving direct and ER/sarcoplasmic reticulum-mediated (ER/SR-mediated) mitochondria disruption. Endogenous cardiac NIX and recombinant NIX localize both to the mitochondria and to the ER/SR. In genetic mouse models, cardiomyocyte ER/SR calcium stores are proportional to the level of expressed NIX. Whereas Nix ablation was protective in a mouse model of apoptotic cardiomyopathy, genetic correction of the decreased SR calcium content of Nix-null mice restored sensitivity to cell death and reestablished cardiomyopathy. Nix mutants specific to ER/SR or mitochondria activated caspases and were equally lethal, but only ER/SR-Nix caused loss of the mitochondrial membrane potential. These results establish a new function for NIX as an integrator of transcriptional and calcium-mediated signals for programmed cell death.
Collapse
Affiliation(s)
- Abhinav Diwan
- Center for Pharmacogenomics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hensler ME, Miyamoto S, Nizet V. Group B streptococcal beta-hemolysin/cytolysin directly impairs cardiomyocyte viability and function. PLoS One 2008; 3:e2446. [PMID: 18560574 PMCID: PMC2409074 DOI: 10.1371/journal.pone.0002446] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2008] [Accepted: 05/09/2008] [Indexed: 12/03/2022] Open
Abstract
Background Group B Streptococcus (GBS) is a leading cause of neonatal sepsis where myocardial dysfunction is an important contributor to poor outcome. Here we study the effects of the GBS pore-forming β-hemolysin/cytolysin (Bh/c) exotoxin on cardiomyocyte viability, contractility, and calcium transients. Methodology/Principal Findings HL-1 cardiomyocytes exposed to intact wild-type (WT) or isogenic Δβh/c mutant GBS, or to cell-free extracts from either strain, were assessed for viability by trypan blue exclusion and for apoptosis by TUNEL staining. Functionality of exposed cardiomyocytes was analyzed by visual quantitation of the rate and extent of contractility. Mitochondrial membrane polarization was measured in TMRE-loaded cells exposed to GBS βh/c. Effects of GBS βh/c on calcium transients were studied in fura-2AM-loaded primary rat ventricular cardiomyocytes. Exposure of HL-1 cardiomyocytes to either WT GBS or βh/c extracts significantly reduced both rate and extent of contractility and later induced necrotic and apoptotic cell death. No effects on cardiomyocyte viability or function were observed after treatment with Δβh/c mutant bacteria or extracts. The βh/c toxin was associated with complete and rapid loss of detectable calcium transients in primary neonatal rat ventricular cardiomyocytes and induced a loss of mitochondrial membrane polarization. These effects on viability and function were abrogated by the βh/c inhibitor, dipalmitoyl phosphatidylcholine (DPPC). Conclusions/Significance Our data show a rapid loss of cardiomyocyte viability and function induced by GBS βh/c, and these deleterious effects are inhibited by DPPC, a normal constituent of human pulmonary surfactant.. These findings have clinical implications for the cardiac dysfunction observed in neonatal GBS infections.
Collapse
Affiliation(s)
- Mary E. Hensler
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
| | - Shigeki Miyamoto
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
37
|
Roderick HL, Cook SJ. Ca2+ signalling checkpoints in cancer: remodelling Ca2+ for cancer cell proliferation and survival. Nat Rev Cancer 2008; 8:361-75. [PMID: 18432251 DOI: 10.1038/nrc2374] [Citation(s) in RCA: 559] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increases in cytosolic free Ca2+ ([Ca2+]i) represent a ubiquitous signalling mechanism that controls a variety of cellular processes, including proliferation, metabolism and gene transcription, yet under certain conditions increases in intracellular Ca2+ are cytotoxic. Thus, in using Ca2+ as a messenger, cells walk a tightrope in which [Ca2+]i is strictly maintained within defined boundaries. To adhere to these boundaries and to sustain their modified phenotype, many cancer cells remodel the expression or activity of their Ca2+ signalling apparatus. Here, we review the role of Ca2+ in promoting cell proliferation and cell death, how these processes are remodelled in cancer and the opportunities this might provide for therapeutic intervention.
Collapse
Affiliation(s)
- H Llewelyn Roderick
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK.
| | | |
Collapse
|
38
|
DeGeorge BR, Gao E, Boucher M, Vinge LE, Martini JS, Raake PW, Chuprun JK, Harris DM, Kim GW, Soltys S, Eckhart AD, Koch WJ. Targeted Inhibition of Cardiomyocyte Gi Signaling Enhances Susceptibility to Apoptotic Cell Death in Response to Ischemic Stress. Circulation 2008; 117:1378-87. [DOI: 10.1161/circulationaha.107.752618] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background—
A salient characteristic of dysfunctional myocardium progressing to heart failure is an upregulation of the adenylyl cyclase inhibitory guanine nucleotide (G) protein α subunit, Gα
i2
. It has not been determined conclusively whether increased Gi activity in the heart is beneficial or deleterious in vivo. Gi signaling has been implicated in the mechanism of cardioprotective agents; however, no in vivo evidence exists that any of the Gα subunits are cardioprotective. We have created a novel molecular tool to specifically address the role of Gi proteins in normal and dysfunctional myocardium.
Methods and Results—
We have developed a class-specific Gi inhibitor peptide, GiCT, composed of the region of Gα
i2
that interacts specifically with G protein–coupled receptors. GiCT inhibits Gi signals specifically in vitro and in vivo, whereas Gs and Gq signals are not affected. In vivo expression of GiCT in transgenic mice effectively causes a “functional knockout” of cardiac Gα
i2
signaling. Inducible, cardiac-specific GiCT transgenic mice display a baseline phenotype consistent with nontransgenic mice. However, when subjected to ischemia/reperfusion injury, GiCT transgenic mice demonstrate a significant increase in infarct size compared with nontransgenic mice (from 36.9±2.5% to 50.9±4.3%). Mechanistically, this post-ischemia/reperfusion phenotype includes increased myocardial apoptosis and resultant decreased contractile performance.
Conclusions—
Overall, our results demonstrate the in vivo utility of GiCT to dissect specific mechanisms attributed to Gi signaling in stressed myocardium. Our results with GiCT indicate that upregulation of Gα
i2
is an adaptive protective response after ischemia to shield myocytes from apoptosis.
Collapse
Affiliation(s)
- Brent R. DeGeorge
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| | - Erhe Gao
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| | - Matthieu Boucher
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| | - Leif E. Vinge
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| | - Jeffrey S. Martini
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| | - Philip W. Raake
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| | - J. Kurt Chuprun
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| | - David M. Harris
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| | - Gilbert W. Kim
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| | - Stephen Soltys
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| | - Andrea D. Eckhart
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| | - Walter J. Koch
- From the Center for Translational Medicine (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., D.M.H., G.W.K., S.S., A.D.E., W.J.K.), George Zallie and Family Laboratory of Cardiovascular Gene Therapy (B.R.D., E.G., M.B., L.E.V., J.S.M., P.W.R., J.K.C., G.W.K., S.S., W.J.K.), and Eugene Feiner Laboratory of Vascular Biology and Thrombosis (D.M.H., A.D.E.), Thomas Jefferson University, Philadelphia, Pa, and Institute for Surgical Research, University of Oslo, Oslo, Norway (L.E.V.)
| |
Collapse
|
39
|
Yin Z, Jones GN, Towns WH, Zhang X, Abel ED, Binkley PF, Jarjoura D, Kirschner LS. Heart-specific ablation of Prkar1a causes failure of heart development and myxomagenesis. Circulation 2008; 117:1414-22. [PMID: 18316483 DOI: 10.1161/circulationaha.107.759233] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Protein kinase A signaling has long been known to play an important role in cardiac function. Dysregulation of the protein kinase A system, caused by mutation of the protein kinase A regulatory subunit gene PRKAR1A, causes the inherited tumor syndrome Carney complex, which includes cardiac myxomas as one of its cardinal features. Mouse models of this genetic defect have been unsatisfactory because homozygote null animals die early in development and heterozygotes do not exhibit a cardiac phenotype. METHODS AND RESULTS To study the cardiac-specific effects resulting from complete loss of Prkar1a, we used cre-lox technology to generate mice lacking this protein specifically in cardiomyocytes. Conditional knockout mice died at day 11.5 to 12.5 of embryogenesis with thin-walled, dilated hearts. These hearts showed elevated protein kinase A activity and decreased cardiomyocyte proliferation before demise. Analysis of the expression of transcription factors required for cardiogenesis revealed downregulation of key cardiac transcription factors such as the serum response factor, Gata4, and Nkx2-5. Although heart wall thickness was reduced overall, specific areas exhibited morphological changes consistent with myxomatous degeneration in the walls of knockout hearts. CONCLUSIONS Loss of Prkar1a from the heart causes a failure of proper myocardial development with subsequent cardiac failure and embryonic demise. These changes appear to be due to suppression of cardiac-specific transcription by increased protein kinase A activity. These biochemical changes lead to myxoma-like changes, indicating that these mice may be a good model with which to study the formation of these tumors.
Collapse
Affiliation(s)
- Zhirong Yin
- Department of Molecular Virology, Immunology, and Molecular Genetics, Ohio State University, Columbus, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Formisano L, Saggese M, Secondo A, Sirabella R, Vito P, Valsecchi V, Molinaro P, Di Renzo G, Annunziato L. The two isoforms of the Na+/Ca2+ exchanger, NCX1 and NCX3, constitute novel additional targets for the prosurvival action of Akt/protein kinase B pathway. Mol Pharmacol 2008; 73:727-37. [PMID: 18079274 DOI: 10.1124/mol.107.042549] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
The proteins NCX1, NCX2, and NCX3 expressed on the plasma membrane of neurons play a crucial role in ionic regulation because they are the major bidirectional system promoting the efflux and influx of Na(+) and Ca(2+) ions. Here, we demonstrate that NCX1 and NCX3 proteins are novel additional targets for the survival action of the phosphatidylinositol 3-kinase (PI3-K)/Akt pathway. Indeed, the doxycycline-dependent overexpression of constitutively active Akt1 in tetracycline (Tet)-Off PC-12 positive mutants and the exposure of Tet-Off PC-12 wild type to nerve growth factor induced an up-regulation of NCX1 and NCX3 proteins. NCX1 up-regulation induced by Akt1 activation occurred at the transcriptional level because NCX1 mRNA increased, and it was counteracted by cAMP response element-binding protein 1 inhibition through small interfering RNA strategy. In contrast, Akt1-induced NCX3 up-regulation recognized a post-transcriptional mechanism occurring at the proteasome level because 1) NCX3 transcript did not increase and 2) the proteasome inhibitor N-benzyloxycarbonyl (Z)-Leu-Leu-leucinal (MG-132) did not further enhance NCX3 protein levels in Akt1 active mutants as it would be expected if the ubiquitin-proteasome complex was not already blocked by Akt1 pathway. As expected, in PC-12 Tet-Off wild-type cells MG-132 enhanced NCX3 protein levels. This up-regulation produced an increased activity of NCX function. Furthermore, NCX1 and NCX3 up-regulation contributed to the survival action of Akt1 during chemical hypoxia because both the silencing of NCX1 or NCX3 and the pharmacological paninhibition of NCX isoforms reduced the prosurvival property of Akt1. Together, these results indicated that NCX1 and NCX3 represent novel additional molecular targets for the prosurvival action of PI3-K/Akt pathway.
Collapse
Affiliation(s)
- Luigi Formisano
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Diwan A, Wansapura J, Syed FM, Matkovich SJ, Lorenz JN, Dorn GW. Nix-mediated apoptosis links myocardial fibrosis, cardiac remodeling, and hypertrophy decompensation. Circulation 2008; 117:396-404. [PMID: 18178777 PMCID: PMC2538800 DOI: 10.1161/circulationaha.107.727073] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Pathological cardiac hypertrophy inevitably remodels, leading to functional decompensation. Although modulation of apoptosis-regulating genes occurs in cardiac hypertrophy, a causal role for programmed cardiomyocyte death in left ventricular (LV) remodeling has not been established. METHODS AND RESULTS We targeted the gene for proapoptotic Nix, which is transcriptionally upregulated in pressure overload and Gq-dependent hypertrophies, in the mouse germ line or specifically in cardiomyocytes (knockout [KO]) and conditionally overexpressed it in the heart (transgenic [TG]). Conditional forced Nix expression acted synergistically with the prohypertrophic Gq transgene to increase cardiomyocyte apoptosis (0.8+/-0.1% in GqTG versus 7.8+/-0.6% in GqTG+NixTG; P<0.001), causing lethal cardiomyopathy with LV dilation and depressed systolic function (percent fractional shortening, 39+/-4 versus 23+/-4; P=0.042). In the reciprocal experiment, germ-line Nix ablation significantly reduced cardiomyocyte apoptosis (4.8+/-0.2% in GqTG+NixKO versus 8.4+/-0.5% in GqTG; P=0.001), which improved percent fractional shortening (43+/-3% versus 27+/-3%; P=0.017), attenuated LV remodeling, and largely prevented lethality in the Gq peripartum model of apoptotic cardiomyopathy. Cardiac-specific (Nkx2.5-Cre) Nix KO mice subjected to transverse aortic constriction developed significantly less LV dilation by echocardiography and magnetic resonance imaging, maintained concentric remodeling, and exhibited preserved LV ejection fraction (61+/-2% in transverse aortic constriction cardiac Nix KO versus 36+/-6% in transverse aortic constriction wild-type mice; P=0.003) at 9 weeks, with reduced cardiomyocyte apoptosis at day 4 (1.70+/-0.21% versus 2.73+/-0.35%; P=0.032). CONCLUSIONS Nix-induced cardiomyocyte apoptosis is a major determinant of adverse remodeling in pathological hypertrophies, a finding that suggests therapeutic value for apoptosis inhibition to prevent cardiomyopathic decompensation.
Collapse
Affiliation(s)
- Abhinav Diwan
- Center for Molecular Cardiovascular Research, University of Cincinnati, Cincinnati, OH
| | - Janaka Wansapura
- Imaging Research Center, Department of Radiology, Children's Hospital Medical Center, Cincinnati, OH
| | - Faisal M. Syed
- Center for Molecular Cardiovascular Research, University of Cincinnati, Cincinnati, OH
| | - Scot J. Matkovich
- Center for Molecular Cardiovascular Research, University of Cincinnati, Cincinnati, OH
| | - John N. Lorenz
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, OH
| | - Gerald W. Dorn
- Center for Molecular Cardiovascular Research, University of Cincinnati, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
42
|
Akt mediates mitochondrial protection in cardiomyocytes through phosphorylation of mitochondrial hexokinase-II. Cell Death Differ 2007; 15:521-9. [PMID: 18064042 DOI: 10.1038/sj.cdd.4402285] [Citation(s) in RCA: 283] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Akt activation supports survival of cardiomyocytes against ischemia/reperfusion, which induces cell death through opening of the mitochondrial permeability transition pore (PT-pore). Mitochondrial depolarization induced by treatment of cardiomyocytes with H(2)O(2) is prevented by activation of Akt with leukemia inhibitory factor (LIF). This protective effect is observed even when cardiomyocytes treated with LIF are permeabilized and mitochondrial depolarization is elicited by elevating Ca(2+). Cell fractionation studies demonstrate that LIF treatment increases both total and phosphorylated Akt in the mitochondrial fraction. Furthermore, the association of Akt with HK-II is increased by LIF. HK-II contains consensus sequences for phosphorylation by Akt and LIF treatment induces PI3K- and Akt-dependent HK-II phosphorylation. Addition of recombinant kinase-active Akt to isolated adult mouse heart mitochondria stimulates phosphorylation of HK-II and concomitantly inhibits the ability of Ca(2+) to induce cytochrome c release. This protection is prevented when HK-II is dissociated from mitochondria by incubation with glucose 6-phosphate or HK-II-dissociating peptide. Finally LIF increases HK-II association with mitochondria and dissociation of HK-II from mitochondria attenuates the protective effect of LIF on H(2)O(2)-induced mitochondrial depolarization in cardiomyocytes. We conclude that Akt has a direct effect at the level of the mitochondrion, which is mediated via phosphorylation of HK-II and results in protection of mitochondria against oxidant or Ca(2+)-stimulated PT-pore opening.
Collapse
|
43
|
Robinson P, Griffiths PJ, Watkins H, Redwood CS. Dilated and hypertrophic cardiomyopathy mutations in troponin and alpha-tropomyosin have opposing effects on the calcium affinity of cardiac thin filaments. Circ Res 2007; 101:1266-73. [PMID: 17932326 DOI: 10.1161/circresaha.107.156380] [Citation(s) in RCA: 174] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Dilated cardiomyopathy and hypertrophic cardiomyopathy (HCM) can be caused by mutations in thin filament regulatory proteins of the contractile apparatus. In vitro functional assays show that, in general, the presence of dilated cardiomyopathy mutations decreases the Ca(2+) sensitivity of contractility, whereas HCM mutations increase it. To assess whether this functional phenomenon was a direct result of altered Ca(2+) affinity or was caused by altered troponin-tropomyosin switching, we assessed Ca(2+) binding of the regulatory site of cardiac troponin C in wild-type or mutant troponin complex and thin filaments using a fluorescent probe (2-[4'-{iodoacetamido}aniline]-naphthalene-6-sulfonate) attached to Cys35 of cardiac troponin C. The Ca(2+)-binding affinity (pCa(50)=6.57+/-0.03) of reconstituted troponin complex was unaffected by all of the HCM and dilated cardiomyopathy troponin mutants tested, with the exception of the troponin I Arg145Gly HCM mutation, which caused an increase (DeltapCa(50)=+0.31+/-0.05). However, when incorporated into regulated thin filaments, all but 1 of the 10 troponin and alpha-tropomyosin mutants altered Ca(2+)-binding affinity. Both HCM mutations increased Ca(2+) affinity (DeltapCa(50)=+0.41+/-0.02 and +0.51+/-0.01), whereas the dilated cardiomyopathy mutations decreased affinity (DeltapCa(50)=-0.12+/-0.04 to -0.54+/-0.04), which correlates with our previous functional in vitro assays. The exception was the troponin T Asp270Asn mutant, which caused a significant decrease in cooperativity. Because troponin is the major Ca(2+) buffer in the cardiomyocyte sarcoplasm, we suggest that Ca(2+) affinity changes caused by cardiomyopathy mutant proteins may directly affect the Ca(2+) transient and hence Ca(2+)-sensitive disease state remodeling pathways in vivo. This represents a novel mechanism for this class of mutation.
Collapse
Affiliation(s)
- Paul Robinson
- Department of Cardiovascular Medicine, University of Oxford, United Kingdom
| | | | | | | |
Collapse
|
44
|
Fan Y, Shi L, Gu Y, Zhao Y, Xie J, Qiao J, Yang GY, Wang Y, Lu CZ. Pretreatment with PTD-calbindin D 28k alleviates rat brain injury induced by ischemia and reperfusion. J Cereb Blood Flow Metab 2007; 27:719-28. [PMID: 16868556 DOI: 10.1038/sj.jcbfm.9600373] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcium toxicity remains the central focus of ischemic brain injury. Calcium channel antagonists have been reported to be neuroprotective in ischemic animal models but have failed in clinical trials. Rather than block the calcium channels, calbindin proteins can buffer excessive intracellular Ca2+, and as a result, maintain the calcium homeostasis. In the present study, we investigated the effect of calbindin D 28k (CaBD) in ischemic brain using the novel technique protein transduction domain (PTD)-mediated protein transduction. We generated PTD-CaBD in Escherichia coli, tested its biologic activity in N-methyl-D-aspartate (NMDA)- and oxygen-glucose deprivation (OGD)-induced hippocampal injury models, and examined the protection of the fusion protein using a rat brain focal ischemia model. Infarct volume was determined using 2,3,5-triphenyl-tetrazolium chloride staining; neuronal injury was examined using terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate-biotin nick end labeling (TUNEL) staining and cleaved caspase-3 assay. The results showed that the PTD-CaBD was efficiently delivered into Cos7 cells, hippocampal slice cells, and brain tissue. Pretreatment with PTD-CaBD decreased intracellular free calcium concentration and reduced cell death in NMDA- or OGD-exposed hippocampal slices (P<0.05). Intraperitoneal administration of PTD-CaBD before transient middle cerebral artery occlusion decreased brain infarct volume (280+/-47 versus 166+/-70 mm3, P<0.05), and improved neurologic outcomes compared with the control. Further studies showed that, compared with the control animals, PTD-CaBD decreased TUNEL (58%+/-7% versus 29%+/-3%, P<0.05)- and cleaved caspase-3 (62+/-4/field versus 31+/-6/field, P<0.05)-positive cells in the ischemic boundary zone. These results indicate that systemic administration of PTD-CaBD could attenuate ischemic brain injury, suggesting that PTD-mediated protein transduction might provide a promising and effective approach for the therapies of brain diseases, including cerebral ischemia.
Collapse
Affiliation(s)
- Yongfeng Fan
- Department of Neurology, Institute of Neurology, Hua-Shan Hospital, Fudan University, Shanghai, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Del Re DP, Miyamoto S, Brown JH. RhoA/Rho kinase up-regulate Bax to activate a mitochondrial death pathway and induce cardiomyocyte apoptosis. J Biol Chem 2007; 282:8069-78. [PMID: 17234627 DOI: 10.1074/jbc.m604298200] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The small G-protein RhoA regulates the actin cytoskeleton, and its involvement in cell proliferation has also been established. In contrast, little is known about whether RhoA participates in cell survival or apoptosis. In cardiomyocytes in vitro, RhoA induces hypertrophic cell growth and gene expression. In vivo, however, RhoA expression leads to development of heart failure (Sah, V. P., Minamisawa, S., Tam, S. P., Wu, T. H., Dorn, G. W., Ross, J. Jr., Chien, K. R., and Brown, J. H. (1999) J. Clin. Investig. 103, 1627-1634), a condition widely associated with cardiomyocyte apoptosis. We demonstrate here that adenoviral overexpression of activated RhoA in cardiomyocytes induces hypertrophy, which transitions over time to apoptosis, as evidenced by caspase activation and nucleosomal DNA fragmentation. The Rho kinase inhibitors Y-27632 and HA-1077 and expression of a dominant negative Rho kinase block these responses. Caspase-9, but not caspase-8, is activated, and its inhibition prevents DNA fragmentation, consistent with involvement of a mitochondrial death pathway. Interestingly, RhoA expression induces a 3-4-fold up-regulation of the proapoptotic Bcl-2 family protein Bax. RhoA also increases levels of activated Bax and the amount of Bax protein localized at mitochondria. Bax mRNA is increased by RhoA, indicating transcriptional regulation, and the ability of a dominant negative p53 mutant to block Bax up-regulation implicates p53 in this response. The involvement of Bax in RhoA-induced apoptosis was examined by treatment with a Bax-inhibitory peptide, which was found to significantly attenuate DNA fragmentation and caspase-9 and -3 activation. The dominant negative p53 also prevents RhoA-induced apoptosis. We conclude that RhoA/Rho kinase activation up-regulates Bax through p53 to induce a mitochondrial death pathway and cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Dominic P Del Re
- Department of Pharmacology and Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
46
|
Venè R, Arena G, Poggi A, D'Arrigo C, Mormino M, Noonan DM, Albini A, Tosetti F. Novel cell death pathways induced by N-(4-hydroxyphenyl)retinamide: therapeutic implications. Mol Cancer Ther 2007; 6:286-298. [PMID: 17237288 DOI: 10.1158/1535-7163.mct-06-0346] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We previously reported that N-(4-hydroxyphenyl)retinamide (4HPR) inhibits retinoblastoma tumor growth in a murine model in vivo and kills Y79 retinoblastoma cells in vitro. In this work, we assayed different cell death-related parameters, including mitochondrial damage and caspase activation, in Y79 cells exposed to 4HPR. 4HPR induced cytochrome c release from mitochondria, caspase-3 activation, and oligonucleosomal DNA fragmentation. However, pharmacologic inactivation of caspases by the pan-caspase inhibitor BOC-D-fmk, or specific caspase-3 inhibition by Z-DEVD-fmk, was not sufficient to prevent cell death, as assessed by loss of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction, lactate dehydrogenase release, disruption of mitochondrial transmembrane potential (Deltapsi(m)), and ATP depletion. We found that 4HPR causes lysosomal membrane permeabilization and cytosolic relocation of cathepsin D. Pepstatin A partially rescued cell viability and reduced DNA fragmentation and cytosolic cytochrome c. The antioxidant N-acetylcysteine attenuated cathepsin D relocation into the cytosol, suggesting that lysosomal destabilization is dependent on elevation of reactive oxygen species and precedes mitochondrial dysfunction. Activation of AKT, which regulates energy level in the cell, by the retinal survival facto]r insulin-like growth factor I was impaired and insulin-like growth factor I was ineffective against ATP and Deltapsi(m) loss in the presence of 4HPR. Lysosomal destabilization, associated with mitochondrial dysfunction, was induced by 4HPR also in other cancer cell lines, including PC3 prostate adenocarcinoma and the vascular tumor Kaposi sarcoma KS-Imm cells. The novel finding of a lysosome-mediated cell death pathway activated by 4HPR could have implications at clinical level for the development of combination chemoprevention and therapy of cancer.
Collapse
Affiliation(s)
- Roberta Venè
- IRCCS MultiMedica, Polo Scientifico e Tecnologico, Settore Ricerca Oncologica, Via Fantoli 15/16, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Vila-Petroff M, Salas MA, Said M, Valverde CA, Sapia L, Portiansky E, Hajjar RJ, Kranias EG, Mundiña-Weilenmann C, Mattiazzi A. CaMKII inhibition protects against necrosis and apoptosis in irreversible ischemia-reperfusion injury. Cardiovasc Res 2006; 73:689-98. [PMID: 17217936 DOI: 10.1016/j.cardiores.2006.12.003] [Citation(s) in RCA: 185] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2006] [Revised: 11/14/2006] [Accepted: 12/04/2006] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) has been implicated in the regulation of cardiac excitation-contraction coupling (ECC) as well as in apoptotic signaling and adverse remodeling. The goal of the present study is to investigate the role of CaMKII in irreversible ischemia and reperfusion (I/R) injury. METHODS Isovolumic Langendorff perfused rat hearts were subjected to global no-flow I/R (45 min/120 min), and isolated myocytes were subjected to a protocol of simulated I/R (45 min simulated ischemia/60 min reoxygenation) either in the absence or presence of CaMKII inhibition [KN-93 (KN) or the CaMKII inhibitory peptide (AIP)]. RESULTS In I/R hearts, an increase in CaMKII activity at the beginning of reperfusion was confirmed by the significantly increased phosphorylation of the Thr(17) site of phospholamban. In the presence of KN, contractile recovery at the end of reperfusion was almost double that of I/R hearts. This recovery was associated with a significant decrease in the extent of infarction, lactate dehydrogenase release (necrosis), TUNEL-positive cells, caspase-3 activity, and an increase in the Bcl-2/Bax ratio (apoptosis). In isolated myocytes, both KN and AIP prevented simulated I/R-induced spontaneous contractile activity and cell mortality. Similar results were obtained when inhibiting the reverse mode Na(+)/Ca(2+) exchanger (NCX) with KB-R7943, sarcoplasmic reticulum (SR) function with ryanodine and thapsigargin, or SR Ca(2+) release with tetracaine. In contrast, overexpression of CaMKII decreased cell viability from 52+/-3% to 26+/-2%. CONCLUSIONS Taken together, the present findings are the first to establish CaMKII as a fundamental component of a cascade of events integrating the NCX, the SR, and mitochondria that promote cellular apoptosis and necrosis in irreversible I/R injury.
Collapse
Affiliation(s)
- Martin Vila-Petroff
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, 60 y 120, (1900) La Plata, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Li CR, Zhou Z, Zhu D, Sun YN, Dai JM, Wang SQ. Protective effect of paeoniflorin on irradiation-induced cell damage involved in modulation of reactive oxygen species and the mitogen-activated protein kinases. Int J Biochem Cell Biol 2006; 39:426-38. [PMID: 17097910 DOI: 10.1016/j.biocel.2006.09.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2006] [Revised: 09/01/2006] [Accepted: 09/20/2006] [Indexed: 12/26/2022]
Abstract
Ionizing radiation can induce DNA damage and cell death by generating reactive oxygen species (ROS). The objective of this study was to investigate the radioprotective effect of paeoniflorin (PF, a main bioactive component in the traditional Chinese herb peony) on irradiated thymocytes and discover the possible mechanisms of protection. We found 60Co gamma-ray irradiation increased cell death and DNA fragmentation in a dose-dependent manner while increasing intracellular ROS. Pretreatment of thymocytes with PF (50-200 microg/ml) reversed this tendency and attenuated irradiation-induced ROS generation. Hydroxyl-scavenging action of PF in vitro was detected through electron spin resonance assay. Several anti-apoptotic characteristics of PF, including the ability to diminish cytosolic Ca2+ concentration, inhibit caspase-3 activation, and upregulate Bcl-2 and downregulate Bax in 4Gy-irradiated thymocytes were determined. Extracellular regulated kinase (ERK), c-Jun NH2-terminal kinase (JNK), and p38 kinase were activated by 4Gy irradiation, whereas its activations were partly blocked by pretreatment of cells with PF. The presence of ERK inhibitor PD98059, JNK inhibitor SP600125 and p38 inhibitor SB203580 decreased cell death in 4Gy-irradiated thymocytes. These results suggest PF protects thymocytes against irradiation-induced cell damage by scavenging ROS and attenuating the activation of the mitogen-activated protein kinases.
Collapse
Affiliation(s)
- Chun Rong Li
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Taiping Road 27#, Beijing 100850, People's Republic of China
| | | | | | | | | | | |
Collapse
|
49
|
Deshpande A, Mina E, Glabe C, Busciglio J. Different conformations of amyloid beta induce neurotoxicity by distinct mechanisms in human cortical neurons. J Neurosci 2006; 26:6011-8. [PMID: 16738244 PMCID: PMC6675207 DOI: 10.1523/jneurosci.1189-06.2006] [Citation(s) in RCA: 381] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Characterization of soluble oligomeric amyloid beta (Abeta) species in the brains of Alzheimer's disease (AD) patients and transgenic models has raised the possibility that different conformations of Abeta may contribute to AD pathology via different mechanisms. To characterize the toxic effect of different Abeta conformations, we tested side by side the effect of well characterized Abeta oligomers (AbetaOs), Abeta-derived diffusible ligands (ADDLs), and fibrillar Abeta (Abetaf) preparations in human cortical neurons (HCNs). Both AbetaOs and ADDLs bind rapidly and with high affinity to synaptic contacts and cellular membranes. AbetaOs (5 microm) induced rapid and massive neuronal death. Calcium influx accelerated, but was not required for, AbetaO toxicity. AbetaOs elicited a stereotyped succession of cellular changes consistent with the activation of a mitochondrial death apoptotic pathway. At low concentrations AbetaOs caused chronic and subtler mitochondrial alterations but minimal cell death. ADDLs induced similar toxic changes as AbetaOs but on a fivefold longer time scale. Higher concentrations of Abetaf and longer incubation times were required to produce widespread neuritic dystrophy but modest HCN cell death. Thus various Abeta species may play relevant roles in AD, causing neurotoxicity by distinct non-overlapping mechanisms affecting neuronal function and viability over multiple time courses.
Collapse
|
50
|
Abstract
Studies have shown that hypertrophied hearts are unusually vulnerable to ischemia. Compromised O2supply has been postulated as a possible explanation for this phenomenon on the basis of elongated O2diffusion distance and altered coronary vasculature found in hypertrophied myocardium. To examine the postulate, perfused heart experiments followed the metabolic and functional responses of hypertrophic myocardium to ischemia.1H/31P NMR was used to measure cellular oxygenation and energy level during ischemia-reperfusion. The left ventricles from spontaneously hypertensive rats (SHR) were enlarged by 48%. With this moderate degree of hypertrophy, cellular O2and energy levels were normal during baseline perfusion. After an ischemic episode, however, cellular O2was severely deprived in the SHR hearts compared with the normal hearts. Depressed postischemic O2reperfusion correlated well with depressed energetic and functional recovery. The results from the current study thus demonstrate a critical relationship between reperfused O2level and functional recovery in hypertrophic myocardium. The role of reperfused O2, however, is time dependent. During early reperfusion, factor(s) other than O2appear to limit functional recovery. It is when the mechanical function of the heart approaches a new steady state that O2becomes a dominant factor. Meanwhile, the finding of a normal O2level in preischemic SHR hearts defies the notion of preexisting hypoxia as a primer of ischemic damage.
Collapse
Affiliation(s)
- Youngran Chung
- Biochemistry and Molecular Medicine, University of California, Davis, CA 95616-8635, USA.
| |
Collapse
|