1
|
Li T, Dou J, Zhang H, Su X, Liu Y, Gao M, Xiao J, Xu W, Gao J. Optimized gated a SPECT-derived myocardial salvage index: its prognostic significance in predicting major adverse cardiac events following acute myocardial infarction percussion. Ann Nucl Med 2024; 38:219-230. [PMID: 38175381 DOI: 10.1007/s12149-023-01894-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
PURPOSE Estimate myocardial salvage index (MSI) using a single-gated Single-Photon Emission Computed Tomography (SPECT) myocardial perfusion imaging (GSMPI) early after percutaneous coronary intervention (PCI) in patients with acute myocardial infarction (AMI) and compare its predictive value with the traditional method especially for post-PCI left ventricular ejection fraction (LVEF) improvement and major adverse cardiac events (MACEs). METHODS GSMPI was performed in 62 patients with AMI early after PCI (3-10 days). The MSI and the conventional parameters were obtained, including total perfusion deficit, LVEF, peak ejection rate (PER), and peak filling rate (PFR). The new calculation method (scoring evaluation method means the extent of abnormality is the percentage of the total scores of abnormal segments divided by the sum of the maximum scores of all myocardial segments using 4-point and 5-point scale semi-quantitative scoring method) and the reference method (number evaluation method means the extent of abnormality is the percentage of the number of abnormal segments divided by the total number of myocardial segments) were applied to acquire the MSI. We compared the predictive ability of the 2 methods based on the area under the receiver operating characteristic curve for LVEF improvement 6 months after PCI using MSI. The Kaplan-Meier method was used for depicting survival curves for predicting MACEs by the 2 methods. Cox proportional-hazards regression was applied to confirm the independent predictors of MACEs. RESULTS The MSI obtained by the new method indicated stronger prognostic significance in LVEF improvement [area under the curve (AUC): 0.793, 95% confidence interval (CI) 0.620-0.912, P < .001] compared with the reference method (AUC: 0.634, 95%CI 0.452-0.792, P = .187). Delong's test revealed a statistically significant difference in AUCs between the 2 methods (P < .05, 95%CI 0.003-0.316). The diagnostic value of the scoring evaluation method was higher than that of the number evaluation method. The Cox prevalence of MACEs was substantially higher in the < median MSI group than in the ≥ median MSI group (hazard ratio: 0.172; 95% CI 0.041-0.724; P < .05] using the new method, whereas no considerable differences were observed between the 2 groups using the reference method (P = .12). Further, the multivariate Cox regression analysis revealed that MSI was an independent indicator for predicting MACEs (P < .05). CONCLUSION The MSI obtained from a simple GSMPI early after PCI, using the scoring evaluation method, was a reliable prognostic indicator for predicting LVEF improvement and MACEs in AMI. It remarkably improved the prognostic value compared with the previous reference methods.
Collapse
Affiliation(s)
- Ting Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, The Huan-Hu-Xi Road, Ti-Yuan-Bei, HeXi District, Tianjin, 300060, People's Republic of China
- Department of Nuclear Medicine, Tianjin Chest Hospital, No. 261 Tai Er Zhuang Road, Jinnan District, Tianjin, 300222, People's Republic of China
- Graduate School, Tianjin Medical University, No. 22 Qi Xiang Tai Road, Heping District, Tianjin, 300070, People's Republic of China
| | - Jing Dou
- Department of Cardiology, Tianjin Chest Hospital, No. 261 Tai Er Zhuang Road, Jinnan District, Tianjin, 300222, People's Republic of China
| | - Hong Zhang
- Department of Radiology, Tianjin Chest Hospital, No. 261 Tai Er Zhuang Road, Jinnan District, Tianjin, 300222, People's Republic of China
| | - Xuexiao Su
- Department of Nuclear Medicine, Tianjin Chest Hospital, No. 261 Tai Er Zhuang Road, Jinnan District, Tianjin, 300222, People's Republic of China
| | - Yin Liu
- Department of Cardiology, Tianjin Chest Hospital, No. 261 Tai Er Zhuang Road, Jinnan District, Tianjin, 300222, People's Republic of China
| | - Mingdong Gao
- Department of Cardiology, Tianjin Chest Hospital, No. 261 Tai Er Zhuang Road, Jinnan District, Tianjin, 300222, People's Republic of China
| | - Jianyong Xiao
- Department of Cardiology, Tianjin Chest Hospital, No. 261 Tai Er Zhuang Road, Jinnan District, Tianjin, 300222, People's Republic of China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, The Huan-Hu-Xi Road, Ti-Yuan-Bei, HeXi District, Tianjin, 300060, People's Republic of China.
| | - Jing Gao
- Cardiovascular Institute, Tianjin Chest Hospital, No. 261 Tai Er Zhuang Road, Jinnan District, Tianjin, 300222, People's Republic of China.
- Thoracic Clinical College, Tianjin Medical University, No. 22 Qi Xiang Tai Road, Heping District, Tianjin, 300070, People's Republic of China.
| |
Collapse
|
2
|
Zhao K, Chen X, Bian Y, Zhou Z, Wei X, Zhang J. Broadening horizons: The role of ferroptosis in myocardial ischemia-reperfusion injury. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2269-2286. [PMID: 37119287 DOI: 10.1007/s00210-023-02506-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/19/2023] [Indexed: 05/01/2023]
Abstract
Ferroptosis is a novel type of regulated cell death (RCD) discovered in recent years, where abnormal intracellular iron accumulation leads to the onset of lipid peroxidation, which further leads to the disruption of intracellular redox homeostasis and triggers cell death. Iron accumulation with lipid peroxidation is considered a hallmark of ferroptosis that distinguishes it from other RCDs. Myocardial ischemia-reperfusion injury (MIRI) is a process of increased myocardial cell injury that occurs during coronary reperfusion after myocardial ischemia and is associated with high post-infarction mortality. Multiple experiments have shown that ferroptosis plays an important role in MIRI pathophysiology. This review systematically summarized the latest research progress on the mechanisms of ferroptosis. Then we report the possible link between the occurrence of MIRI and ferroptosis in cardiomyocytes. Finally, we discuss and analyze the related drugs that target ferroptosis to attenuate MIRI and its action targets, and point out the shortcomings of the current state of relevant research and possible future research directions. It is hoped to provide a new avenue for improving the prognosis of the acute coronary syndrome.
Collapse
Affiliation(s)
- Ke Zhao
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Xiaoshu Chen
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yujing Bian
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Zhou Zhou
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Xijin Wei
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, China.
| | - Juan Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, China.
| |
Collapse
|
3
|
Yu S, Wang B, Li G, Guo X, Yang H, Sun Y. Habitual Tea Consumption Increases the Incidence of Metabolic Syndrome in Middle-Aged and Older Individuals. Nutrients 2023; 15:nu15061448. [PMID: 36986178 PMCID: PMC10055940 DOI: 10.3390/nu15061448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
In middle-aged and elderly individuals, the relationship between tea consumption and incident metabolic syndrome (MetS) is still unclear. Therefore, this study intends to figure out the relationship between tea-drinking frequency and MetS in rural middle-aged and older Chinese residents. In the Northeast China Rural Cardiovascular Health Study, 3632 middle-aged or older individuals (mean age 57 ± 8, 55.2% men) without MetS were included at baseline during 2012–2013 and were followed up on between 2015–2017. Participants showing differential tea consumption frequency were divided into the following classes: non-habitual tea drinkers, occasional tea drinkers, 1–2 times/day drinkers, and ≥3 times/day drinkers. Data showed that non-habitual tea drinking was more common among women. The frequency of tea consumption was higher in ethnic groups other than Han and among singles, as well as in concurrent smokers and drinkers and individuals with primary or lower educational status. The increasing tea consumption was in line with baseline elevations in body mass index, systolic and diastolic blood pressure, high-density lipoprotein cholesterol (HDL-C), and AST/ALT ratio. Multivariate logistic regression analysis confirmed that occasional tea drinking increased the incidence of low HDL-C [OR (95% CI): 1.268 (1.015, 1.584)], high waist circumference [OR (95% CI): 1.336 (1.102, 1.621)], and MetS [OR (95% CI): 1.284 (1.050, 1.570)]. In addition, 1–2 times/day tea drinking increased the cumulative incidence of high TG [OR (95% CI): 1.296 (1.040, 1.616)], high waist circumference [OR (95% CI): 1.296 (1.044, 1.609)] and MetS [OR (95% CI): 1.376 (1.030, 1.760)]. We demonstrated that regular tea consumption is correlated with a greater incidence of metabolic disorders and MetS. Our findings may help clarify the contradictory association reported between tea drinking and MetS development in middle-aged and older residents of rural China.
Collapse
Affiliation(s)
- Shasha Yu
- Department of Cardiology, First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang 110001, China
| | - Bo Wang
- Department of Cardiology, First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang 110001, China
| | - Guangxiao Li
- Department of Clinical Epidemiology, Institute of Cardiovascular Diseases, First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaofan Guo
- Department of Cardiology, First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang 110001, China
| | - Hongmei Yang
- Department of Cardiology, First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang 110001, China
| | - Yingxian Sun
- Department of Cardiology, First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang 110001, China
- Correspondence: ; Tel.: +86-024-8328-2888; Fax: +86-24-8328-2346
| |
Collapse
|
4
|
Ayyadurai VAS, Deonikar P, Fields C. Mechanistic Understanding of D-Glucaric Acid to Support Liver Detoxification Essential to Muscle Health Using a Computational Systems Biology Approach. Nutrients 2023; 15:733. [PMID: 36771439 PMCID: PMC9921405 DOI: 10.3390/nu15030733] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/10/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
Liver and muscle health are intimately connected. Nutritional strategies that support liver detoxification are beneficial to muscle recovery. Computational-in silico-molecular systems' biology analysis of supplementation of calcium and potassium glucarate salts and their metabolite D-glucaric acid (GA) reveals their positive effect on mitigation of liver detoxification via four specific molecular pathways: (1) ROS production, (2) deconjugation, (3) apoptosis of hepatocytes, and (4) β-glucuronidase synthesis. GA improves liver detoxification by downregulating hepatocyte apoptosis, reducing glucuronide deconjugates levels, reducing ROS production, and inhibiting β-Glucuronidase enzyme that reduces re-absorption of toxins in hepatocytes. Results from this in silico study provide an integrative molecular mechanistic systems explanation for the mitigation of liver toxicity by GA.
Collapse
Affiliation(s)
- V. A. Shiva Ayyadurai
- Systems Biology Group, CytoSolve Research Division, CytoSolve, Inc., Cambridge, MA 02138, USA
| | - Prabhakar Deonikar
- Systems Biology Group, CytoSolve Research Division, CytoSolve, Inc., Cambridge, MA 02138, USA
| | - Christine Fields
- Applied Food Sciences Inc., 8708 South Congress Suite 290, Austin, TX 78745, USA
| |
Collapse
|
5
|
San-Martín-Martínez D, Serrano-Lemus D, Cornejo V, Gajardo AIJ, Rodrigo R. Pharmacological Basis for Abrogating Myocardial Reperfusion Injury Through a Multi-Target Combined Antioxidant Therapy. Clin Pharmacokinet 2022; 61:1203-1218. [DOI: 10.1007/s40262-022-01151-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2022] [Indexed: 11/29/2022]
|
6
|
Wischmann P, Chennupati R, Solga I, Funk F, Becher S, Gerdes N, Anker S, Kelm M, Jung C. Safety and efficacy of iron supplementation after myocardial infarction in mice with moderate blood loss anaemia. ESC Heart Fail 2021; 8:5445-5455. [PMID: 34636175 PMCID: PMC8712778 DOI: 10.1002/ehf2.13639] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 09/12/2021] [Accepted: 09/19/2021] [Indexed: 11/28/2022] Open
Abstract
Aims Iron deficiency is frequently observed in patients with acute coronary syndrome and associates with poor prognosis after acute myocardial infarction (AMI). Anaemia is linked to dysregulation of iron metabolism, red blood cell dysfunction, and increased reactive oxygen species generation. Iron supplementation in chronic heart failure is safe and improves cardiac exercise capacity. Increases in iron during ischaemia or immediately after reperfusion are associated with detrimental effects on left ventricular (LV) function. The safety and applicability of iron during or immediately after reperfusion of AMI in anaemia are not known. We aimed to study the safety and efficacy of iron supplementation within 1 h or deferred to 24 h after reperfusion of AMI by analysing LV function and infarct size. Methods and results In a mouse model of moderate blood loss anaemia (n = 6–8 mice/group), the effects of iron supplementation (20 mg iron as ferric carboxymaltose per kg body weight) within 1 h and deferred to 24 h after ischaemia/reperfusion were assessed. Cardiac function was analysed in vivo by echocardiography at baseline (Day 3) with and without anaemia, after AMI (24 h), and after administration of intravenous iron. Anaemia was characterized by iron deficiency and a trend towards increased haemolysis, which was supported by increased plasma free‐haemoglobin [sham vs. anaemia (n = 8/group): P < 0.05]. Anaemia increased heart rate, LV end‐diastolic volume, stroke volume, and cardiac output, while LV end‐systolic volume remained unchanged at baseline. Superimposition of AMI deteriorated global LV function, whereas infarct sizes remained unaffected [sham vs. anaemia (n = 6/group): P = 0.9]. Deferred iron supplementation 24 h after ischaemia/reperfusion resulted in reversal of end‐systolic volume increase and reduced infarct size [% of area at risk: sham vs. anaemia + iron after 24 h; (n = 6/group); 48 ± 7 vs. 38 ± 7; P < 0.05], whereas administration within 1 h after reperfusion was neutral [sham vs. anaemia + iron; (n = 6/group); 48 ± 7 vs. 42 ± 8; P = 0.56]. Moreover, iron application after reperfused AMI showed unaltered mortality compared with sham. Conclusions Iron supplementation 24 h after reperfusion of AMI is safe and reversed enlargement of end‐systolic volume after AMI resulting in increased stroke volume and cardiac output. This highlights its potential as adjunctive treatment in anaemia with ID after reperfused AMI. Time point of iron application after reperfusion appears critical.
Collapse
Affiliation(s)
- Patricia Wischmann
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstr. 5, Düsseldorf, 40225, Germany
| | - Ramesh Chennupati
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstr. 5, Düsseldorf, 40225, Germany
| | - Isabella Solga
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstr. 5, Düsseldorf, 40225, Germany
| | - Felix Funk
- Department of Nanomedicines, Vifor Pharma Management Ltd, Glattbrugg, Switzerland
| | - Stefanie Becher
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstr. 5, Düsseldorf, 40225, Germany
| | - Norbert Gerdes
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstr. 5, Düsseldorf, 40225, Germany
| | - Stefan Anker
- Department of Cardiology, Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstr. 5, Düsseldorf, 40225, Germany
| | - Christian Jung
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstr. 5, Düsseldorf, 40225, Germany
| |
Collapse
|
7
|
Reinhold J, Papadopoulou C, Baral R, Vassiliou VS. Iron deficiency for prognosis in acute coronary syndrome - A systematic review and meta-analysis. Int J Cardiol 2020; 328:46-54. [PMID: 33326805 DOI: 10.1016/j.ijcard.2020.12.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/23/2020] [Accepted: 12/05/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Iron deficiency (ID) is an important predictor of adverse outcomes in patients with heart failure, however it is unclear whether ID also affects prognosis in patients with acute coronary syndrome (ACS). The aim of this systematic review and meta-analysis was to assess the prognostic value of iron deficiency in patients with ACS. METHODS We searched PubMed, Web of Science, and the Cochrane library and included cohort studies of patients with ACS that were stratified by ID status. There were no restrictions on definition of ACS or ID. Studies were systematically appraised and data extracted by two independent reviewers. Meta-analysis was performed where two or more studies reported on the same pre-determined outcome measure. RESULTS Seven studies with 2821 participants were identified, reporting a high prevalence of ID in the ACS population. Three studies reported worse long-term outcomes in the ID population, whereas short-term outcomes were heterogeneous across studies. CONCLUSIONS Patients with ID presenting with ACS may have a worse long-term prognosis but more studies are required for confirmation. A role for ID in prognosis of patients with ACS and as a potentially treatable condition may have implication for the current management of this patient population.
Collapse
Affiliation(s)
- Johannes Reinhold
- University of East Anglia (UEA), Norwich Research Park, Norwich NR4 7TJ, United Kingdom; Norfolk and Norwich University Hospital, Colney Lane, Norwich NR4 7UY, United Kingdom.
| | - Charikleia Papadopoulou
- University of Cambridge, The Old Schools, Trinity Ln, Cambridge CB2 1TN, United Kingdom; Royal Papworth Hospital, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0AY, United Kingdom
| | - Ranu Baral
- University of East Anglia (UEA), Norwich Research Park, Norwich NR4 7TJ, United Kingdom; Norfolk and Norwich University Hospital, Colney Lane, Norwich NR4 7UY, United Kingdom
| | - Vassilios S Vassiliou
- University of East Anglia (UEA), Norwich Research Park, Norwich NR4 7TJ, United Kingdom; Norfolk and Norwich University Hospital, Colney Lane, Norwich NR4 7UY, United Kingdom
| |
Collapse
|
8
|
Behrouzi B, Weyers JJ, Qi X, Barry J, Rabadia V, Manca D, Connelly J, Spino M, Wood JC, Strauss BH, Wright GA, Ghugre NR. Action of iron chelator on intramyocardial hemorrhage and cardiac remodeling following acute myocardial infarction. Basic Res Cardiol 2020; 115:24. [PMID: 32140789 DOI: 10.1007/s00395-020-0782-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/17/2020] [Indexed: 12/22/2022]
Abstract
Intramyocardial hemorrhage is an independent predictor of adverse outcomes in ST-segment elevation myocardial infarction (STEMI). Iron deposition resulting from ischemia-reperfusion injury (I/R) is pro-inflammatory and has been associated with adverse remodeling. The role of iron chelation in hemorrhagic acute myocardial infarction (AMI) has never been explored. The purpose of this study was to investigate the cardioprotection offered by the iron-chelating agent deferiprone (DFP) in a porcine AMI model by evaluating hemorrhage neutralization and subsequent cardiac remodeling. Two groups of animals underwent a reperfused AMI procedure: control and DFP treated (N = 7 each). A comprehensive MRI examination was performed in healthy state and up to week 4 post-AMI, followed by histological assessment. Infarct size was not significantly different between the two groups; however, the DFP group demonstrated earlier resolution of hemorrhage (by T2* imaging) and edema (by T2 imaging). Additionally, ventricular enlargement and myocardial hypertrophy (wall thickness and mass) were significantly smaller with DFP, suggesting reduced adverse remodeling, compared to control. The histologic results were consistent with the MRI findings. To date, there is no effective targeted therapy for reperfusion hemorrhage. Our proof-of-concept study is the first to identify hemorrhage-derived iron as a therapeutic target in I/R and exploit the cardioprotective properties of an iron-chelating drug candidate in the setting of AMI. Iron chelation could potentially serve as an adjunctive therapy in hemorrhagic AMI.
Collapse
Affiliation(s)
- Bita Behrouzi
- Department of Physics and Physiology, University of Toronto, Toronto, ON, Canada
| | - Jill J Weyers
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Xiuling Qi
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Jennifer Barry
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | | | | | | | - Michael Spino
- ApoPharma Inc, Toronto, ON, Canada.,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - John C Wood
- Childrens Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Bradley H Strauss
- Schulich Heart Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Graham A Wright
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada.,Schulich Heart Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Nilesh R Ghugre
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada. .,Schulich Heart Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Affiliation(s)
- Robert A. Kloner
- Huntington Medical Research InstitutesPasadenaCA
- Division of Cardiovascular MedicineDepartment of MedicineKeck School of Medicine at University of Southern CaliforniaLos AngelesCA
| |
Collapse
|
10
|
Iron deficiency in patients with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention. Int J Cardiol 2020; 300:14-19. [PMID: 31399299 DOI: 10.1016/j.ijcard.2019.07.083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/18/2019] [Accepted: 07/28/2019] [Indexed: 12/11/2022]
|
11
|
Sharma S, Leaf DE. Iron Chelation as a Potential Therapeutic Strategy for AKI Prevention. J Am Soc Nephrol 2019; 30:2060-2071. [PMID: 31554656 DOI: 10.1681/asn.2019060595] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AKI remains a major public health concern. Despite years of investigation, no intervention has been demonstrated to reliably prevent AKI in humans. Thus, development of novel therapeutic targets is urgently needed. An important role of iron in the pathophysiology of AKI has been recognized for over three decades. When present in excess and in nonphysiologic labile forms, iron is toxic to the kidneys and multiple other organs, whereas iron chelation is protective across a broad spectrum of insults. In humans, small studies have investigated iron chelation as a novel therapeutic strategy for prevention of AKI and extrarenal acute organ injury, and have demonstrated encouraging initial results. In this review, we examine the existing data on iron chelation for AKI prevention in both animal models and human studies. We discuss practical considerations for future clinical trials of AKI prevention using iron chelators, including selection of the ideal clinical setting, patient population, iron chelating agent, and dosing regimen. Finally, we compare the key differences among the currently available iron chelators, including pharmacokinetics, routes of administration, and adverse effects.
Collapse
Affiliation(s)
- Shreyak Sharma
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
12
|
Chawla LS, Beers-Mulroy B, Tidmarsh GF. Therapeutic Opportunities for Hepcidin in Acute Care Medicine. Crit Care Clin 2019; 35:357-374. [DOI: 10.1016/j.ccc.2018.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
13
|
Leaf DE, Rajapurkar M, Lele SS, Mukhopadhyay B, Boerger EAS, Mc Causland FR, Eisenga MF, Singh K, Babitt JL, Kellum JA, Palevsky PM, Christov M, Waikar SS. Iron, Hepcidin, and Death in Human AKI. J Am Soc Nephrol 2019; 30:493-504. [PMID: 30737269 PMCID: PMC6405140 DOI: 10.1681/asn.2018100979] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/30/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Iron is a key mediator of AKI in animal models, but data on circulating iron parameters in human AKI are limited. METHODS We examined results from the ARF Trial Network study to assess the association of plasma catalytic iron, total iron, transferrin, ferritin, free hemoglobin, and hepcidin with 60-day mortality. Participants included critically ill patients with AKI requiring RRT who were enrolled in the study. RESULTS Of the 807 study participants, 409 (51%) died by day 60. In both unadjusted and multivariable adjusted models, higher plasma concentrations of catalytic iron were associated with a significantly greater risk of death, as were lower concentrations of hepcidin. After adjusting for other factors, patients with catalytic iron levels in the highest quintile versus the lowest quintile had a 4.06-fold increased risk of death, and patients with hepcidin levels in the lowest quintile versus the highest quintile of hepcidin had a 3.87-fold increased risk of death. These findings were consistent across multiple subgroups. Other iron markers were also associated with death, but the magnitude of the association was greatest for catalytic iron and hepcidin. Higher plasma concentrations of catalytic iron and lower concentrations of hepcidin are each independently associated with mortality in critically ill patients with AKI requiring RRT. CONCLUSIONS These findings suggest that plasma concentrations of catalytic iron and hepcidin may be useful prognostic markers in patients with AKI. Studies are needed to determine whether strategies to reduce catalytic iron or increase hepcidin might be beneficial in this patient population.
Collapse
Affiliation(s)
- David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts;
| | | | | | | | - Emily A S Boerger
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Michele F Eisenga
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Departments of
| | - Karandeep Singh
- Learning Health Sciences and
- Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jodie L Babitt
- Nephrology Division, Program in Membrane Biology, Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Paul M Palevsky
- Renal Section, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Marta Christov
- Department of Medicine, New York Medical College, Valhalla, New York
| | - Sushrut S Waikar
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
14
|
Catapano MC, Karlíčková J, Tvrdý V, Sharma S, Prasad AK, Saso L, Chhillar AK, Kuneš J, Pour M, Parmar VS, Mladěnka P. Mono and dihydroxy coumarin derivatives: Copper chelation and reduction ability. J Trace Elem Med Biol 2018; 46:88-95. [PMID: 29413115 DOI: 10.1016/j.jtemb.2017.11.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/07/2017] [Accepted: 11/23/2017] [Indexed: 01/07/2023]
Abstract
UNLABELLED Due to the limited array of the currently available copper chelators, research of such compounds continues to be of clinical interest. Notably, o-dihydroxycoumarins have been previously shown to be potent iron chelators under neutral conditions. Within this study, the interaction of a series of natural coumarins and their synthetic analogs with copper has been evaluated in order to obtain structure-activity relationships under different pathophysiological pH conditions. Both competitive and non-competitive methods have been employed. Analysis of cupric ion reduction has also been performed. Under mildly competitive conditions, cupric chelation was observed for o-dihydroxycoumarins, and partially for o-diacetoxycoumarin. Non-competitive studies showed that cuprous ions are not chelated at all and that the stoichiometries of the most active 6,7- and 7,8-dihydroxycoumarins to cupric ions ranged from 1:1 to 2:1 depending on pH and concentration. Interestingly, under highly competitive conditions, coumarins were not capable of chelating cupric ions, either. Reduction experiments have shown that 13 out of the 15 coumarins included in this study reduced cupric ions. However, significant differences depending on their structures were apparent in their potencies. O-dihydroxycoumarins were the most potent ones again. CONCLUSION O-dihydroxycoumarins are moderately active cupric ion chelators with potent copper reducing properties.
Collapse
Affiliation(s)
- Maria Carmen Catapano
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Králové, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Jana Karlíčková
- Department of Pharmaceutical Botany and Ecology, Charles University, Faculty of Pharmacy in Hradec Králové, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Václav Tvrdý
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Králové, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Sweta Sharma
- Bioorganic Laboratory, Department of Chemistry, University of Delhi, Delhi 110 007, India
| | - Ashok K Prasad
- Bioorganic Laboratory, Department of Chemistry, University of Delhi, Delhi 110 007, India
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, Italy
| | - Anil K Chhillar
- Centre of Biotechnology, Maharshi Dayanand University, Rohtak 124 001, Haryana, India
| | - Jiří Kuneš
- Department of Inorganic and Organic Chemistry, Charles University, Faculty of Pharmacy in Hradec Králové, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Milan Pour
- Department of Inorganic and Organic Chemistry, Charles University, Faculty of Pharmacy in Hradec Králové, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Virinder S Parmar
- Bioorganic Laboratory, Department of Chemistry, University of Delhi, Delhi 110 007, India; Institute of Advanced Sciences, 86-410 Faunce Corner Mall Road, Dartmouth, MA 02747, USA
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Králové, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic.
| |
Collapse
|
15
|
Sinning C, Westermann D, Clemmensen P. Oxidative stress in ischemia and reperfusion: current concepts, novel ideas and future perspectives. Biomark Med 2017; 11:11031-1040. [DOI: 10.2217/bmm-2017-0110] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Oxidative stress remains a major contributor to myocardial injury after ischemia followed by reperfusion (I/R) as the reperfusion of the myocardial infarction (MI) area inevitably leads to a cascade of I/R injury. This review focused on concepts of the antioxidative defense system and elucidates recent research using antioxidants like vitamin C, E and β-carotene or essential trace elements to activate compounds of antioxidative pathways in the circulation. In this context, important defense mechanisms like superoxide dismutase and glutathione peroxidase will be described. Furthermore, the different mechanisms through which myocardial protection can be addressed, like ischemic postconditioning in myocardial infarction or adjunctive measures like targeted temperature management as well as new theories, including the role of iron in I/R injury, will be discussed.
Collapse
Affiliation(s)
- Christoph Sinning
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Dirk Westermann
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
- German Center of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Peter Clemmensen
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
- Department of Regional Health Research, Faculty of Health Sciences, Nykøbing Falster & Odense, Denmark
| |
Collapse
|
16
|
Kim J, Kim J, Kook H, Park WJ. PICOT alleviates myocardial ischemia-reperfusion injury by reducing intracellular levels of reactive oxygen species. Biochem Biophys Res Commun 2017; 485:807-813. [PMID: 28257842 DOI: 10.1016/j.bbrc.2017.02.136] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 12/31/2022]
Abstract
Excessive generation of reactive oxygen species (ROS) is one of the main causes of myocardial ischemia-reperfusion (I/R) injury. In this study, we investigated the role of protein kinase C-interacting cousin of thioredoxin (PICOT; Grx3) during myocardial I/R using PICOT transgenic (TG) and knockdown (KD) mice. Infarction and apoptosis were attenuated in PICOT TG mice but exacerbated in PICOT KD mice upon I/R. In parallel, I/R-induced generation of ROS was attenuated in PICOT TG mice but exacerbated in PICOT KD mice. Angiotensin II (AngII)-mediated increases in ROS and free iron levels were also attenuated in cardiomyocytes isolated from PICOT TG mice but exacerbated in cardiomyocytes from PICOT KD mice. Accordingly, H2O2-mediated cell death was attenuated in cardiomyocytes isolated from PICOT TG mice but exacerbated in cardiomyocytes from PICOT KD mice. Taken together, these data show that PICOT alleviates myocardial I/R injury by regulating intracellular ROS and free iron levels. We suggest that PICOT presents a novel therapeutic strategy for myocardial I/R injury.
Collapse
Affiliation(s)
- Jihwa Kim
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Jooyeon Kim
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology and Medical Research Center for Gene Regulation, Chonnam National University Medical School, 160 Baekseo-ro, Dong-ku, Gwangju 61469, Republic of Korea
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea.
| |
Collapse
|
17
|
Uberti F, Morsanuto V, Lattuada D, Colciaghi B, Cochis A, Bulfoni A, Colombo P, Bolis G, Molinari C. Protective effects of vitamin D3 on fimbrial cells exposed to catalytic iron damage. J Ovarian Res 2016; 9:34. [PMID: 27317433 PMCID: PMC4912710 DOI: 10.1186/s13048-016-0243-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/31/2016] [Indexed: 12/13/2022] Open
Abstract
Background Recently, vitamin D3 (1alpha, 25-dihydroxyvitamin D) has shown its capability to take part in many extraskeletal functions and its serum levels have been related to patient survival rate and malignancy of many types of neoplasms, including ovarian cancers. Catalytic iron is a free circulating form of iron that is able to generate reactive oxygen species and consequently to promote a number of cellular and tissutal dysfunctions including tumorigenesis. In fertile women an important source of catalytic iron is derived from retrograde menstruation. Epithelial secretory cells from fimbriae of fallopian tubes are greatly exposed to catalytic iron derived from menstrual reflux and so represent the site of origin for most serous ovarian cancers. The aim of this study was to assess whether vitamin D3 can play a role in counteracting catalytic iron-induced oxidative stress in cells from fimbriae of fallopian tubes. Methods The cells, isolated from women undergoing isteroannessiectomy, were treated with catalytic iron 50-75-100 mM and vitamin D3 at a concentration ranging from 0.01 to 10 nM to study cell viability, radical oxygen species production, p53, pan-Ras, Ki67 and c-Myc protein expressions through Western Blot, and immunocytochemistry or immunofluorescence analysis. Results The pre-treatment with vitamin D3 1 nM showed its beneficial effects that consists in a significant decrease in ROS production. In addition a novel finding is represented by the demonstration that pre-treatment with vitamin D3 is also able to significantly counteract tumoral biomarkers activation, such as p53, pan-Ras, Ki67 and c-Myc, and consequently the catalytic iron-induced cellular injury. Conclusions This study demonstrates for the first time that vitamin D3 plays an important role in preventing catalytic iron-dependent oxidative stress in cultured fimbrial cells. These results support the hypothesis that vitamin D3 could counteract carcinogenic changes induced by catalytic iron.
Collapse
Affiliation(s)
- Francesca Uberti
- Physiology Laboratory, Department of Translational Medicine, UPO - University of Eastern Piedmont, Via Solaroli 17, Novara, 28100, Italy.
| | - Vera Morsanuto
- Physiology Laboratory, Department of Translational Medicine, UPO - University of Eastern Piedmont, Via Solaroli 17, Novara, 28100, Italy
| | - Debora Lattuada
- Department of Obstetrics and Gynecology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Barbara Colciaghi
- Department of Obstetrics and Gynecology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Andrea Cochis
- Department of Biomedical, Surgical and Dental Sciences, Milan State University, via Beldiletto 1, Milan, 20142, Italy
| | - Alessandro Bulfoni
- Department of Obstetrics and Gynecology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Paola Colombo
- Department of Obstetrics and Gynecology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Giorgio Bolis
- Department of Obstetrics and Gynecology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Claudio Molinari
- Physiology Laboratory, Department of Translational Medicine, UPO - University of Eastern Piedmont, Via Solaroli 17, Novara, 28100, Italy
| |
Collapse
|
18
|
Filipský T, Říha M, Hašková P, Pilařová V, Nováková L, Semecký V, Vávrová J, Holečková M, Palicka V, Šimůnek T, Hrdina R, Mladěnka P. Intravenous rutin in rat exacerbates isoprenaline-induced cardiotoxicity likely due to intracellular oxidative stress. Redox Rep 2016; 22:78-90. [PMID: 27077454 DOI: 10.1080/13510002.2016.1159817] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVES Rutin, quercetin-3-O-rutinoside, a natural flavonol glycoside, has shown various in vitro benefits with potential use treating human diseases, especially cardiovascular system disorders. Antioxidant properties are assumed to underlie the majority of these benefits. Yet rutin pro-oxidant properties have been reported as well. Our research group has recently shown aggravating effects on isoprenaline (ISO)-induced cardiotoxicity in Wistar:Han rats after 24 hours. METHODS This study was designed to examine in more detail the reasons for the negative effects of rutin (11.5 and 46 mg/kg, i.v.) after administration of ISO (100 mg/kg, s.c.) in rats within 2 hours of continuous experiment and in the H9c2 cardiomyoblast-derived cell line. RESULTS Like our previous findings, rutin did not (11.5 or 46 mg/kg, i.v.) reduce the ISO-induced mortality within 2 hours although the lower dose significantly reduced cardiac troponin T (cTnT) and partly improved the histological findings. In contrast, the higher dose increased the mortality in comparison with solvent (1.26% w/v sodium bicarbonate). This was not caused by any specific haemodynamic disturbances. It appears to be associated with oxidative stress as rutin enhanced intracellular reactive oxygen species formation in vitro and had the tendency to increase it in vivo. CONCLUSIONS Rutin, likely due to its pro-oxidative effects, can exacerbate catecholamine cardiotoxicity depending on the dose used.
Collapse
Affiliation(s)
- Tomáš Filipský
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové , Charles University in Prague , Heyrovského 1203, 500 05 Czech Republic
| | - Michal Říha
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové , Charles University in Prague , Heyrovského 1203, 500 05 Czech Republic
| | - Pavlína Hašková
- b Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové , Charles University in Prague , Heyrovského 1203, 500 05 Czech Republic
| | - Veronika Pilařová
- c Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové , Charles University in Prague , Heyrovského 1203, 500 05 Czech Republic
| | - Lucie Nováková
- c Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové , Charles University in Prague , Heyrovského 1203, 500 05 Czech Republic
| | - Vladimír Semecký
- d Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové , Charles University in Prague , Heyrovského 1203, 500 05 Czech Republic
| | - Jaroslava Vávrová
- e Faculty of Medicine in Hradec Králové , Charles University in Prague , Šimkova 870, 500 38 Czech Republic.,f University Hospital Hradec Králové , Sokolská 581, 500 05 Czech Republic
| | - Magdaléna Holečková
- e Faculty of Medicine in Hradec Králové , Charles University in Prague , Šimkova 870, 500 38 Czech Republic.,f University Hospital Hradec Králové , Sokolská 581, 500 05 Czech Republic
| | - Vladimir Palicka
- e Faculty of Medicine in Hradec Králové , Charles University in Prague , Šimkova 870, 500 38 Czech Republic.,f University Hospital Hradec Králové , Sokolská 581, 500 05 Czech Republic
| | - Tomáš Šimůnek
- b Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové , Charles University in Prague , Heyrovského 1203, 500 05 Czech Republic
| | - Radomír Hrdina
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové , Charles University in Prague , Heyrovského 1203, 500 05 Czech Republic
| | - Přemysl Mladěnka
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové , Charles University in Prague , Heyrovského 1203, 500 05 Czech Republic
| |
Collapse
|
19
|
Protective Effects of D-Penicillamine on Catecholamine-Induced Myocardial Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:5213532. [PMID: 26788248 PMCID: PMC4691625 DOI: 10.1155/2016/5213532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/15/2015] [Indexed: 01/07/2023]
Abstract
Iron and copper release participates in the myocardial injury under ischemic conditions and hence protection might be achieved by iron chelators. Data on copper chelation are, however, sparse. The effect of the clinically used copper chelator D-penicillamine in the catecholamine model of acute myocardial injury was tested in cardiomyoblast cell line H9c2 and in Wistar Han rats. D-Penicillamine had a protective effect against catecholamine-induced injury both in vitro and in vivo. It protected H9c2 cells against the catecholamine-induced viability loss in a dose-dependent manner. In animals, both intravenous D-penicillamine doses of 11 (low) and 44 mg/kg (high) decreased the mortality caused by s.c. isoprenaline (100 mg/kg) from 36% to 14% and 22%, respectively. However, whereas the low D-penicillamine dose decreased the release of cardiac troponin T (specific marker of myocardial injury), the high dose resulted in an increase. Interestingly, the high dose led to a marked elevation in plasma vitamin C. This might be related to potentiation of oxidative stress, as suggested by additional in vitro experiments with D-penicillamine (iron reduction and the Fenton reaction). In conclusion, D-penicillamine has protective potential against catecholamine-induced cardiotoxicity; however the optimal dose selection seems to be crucial for further application.
Collapse
|
20
|
Roghi A, Poggiali E, Duca L, Mafrici A, Pedrotti P, Paccagnini S, Brenna S, Galli A, Consonni D, Cappellini MD. Role of Non-Transferrin-Bound Iron in the pathogenesis of cardiotoxicity in patients with ST-elevation myocardial infarction assessed by Cardiac Magnetic Resonance Imaging. Int J Cardiol 2015; 199:326-32. [DOI: 10.1016/j.ijcard.2015.07.056] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 07/18/2015] [Indexed: 12/15/2022]
|
21
|
Eguchi A, Naito Y, Iwasaku T, Okuhara Y, Morisawa D, Sawada H, Nishimura K, Oboshi M, Fujii K, Mano T, Masuyama T, Hirotani S. Association of dietary iron restriction with left ventricular remodeling after myocardial infarction in mice. Heart Vessels 2015; 31:222-9. [PMID: 25573257 DOI: 10.1007/s00380-014-0621-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 12/26/2014] [Indexed: 11/24/2022]
Abstract
Several epidemiologic studies have reported that body iron status and dietary iron intake are related to an increased risk of acute myocardial infarction (MI). However, it is completely unknown whether dietary iron reduction impacts the development of left ventricular (LV) remodeling after MI. Here, we investigate the effect of dietary iron restriction on the development of LV remodeling after MI in an experimental model. MI was induced in C57BL/6 J mice (9-11 weeks of age) by the permanent ligation of the left anterior descending coronary artery (LAD). At 2 weeks after LAD ligation, mice were randomly divided into two groups and were given a normal diet or an iron-restricted diet for 4 weeks. Sham operation without LAD ligation was also performed as controls. MI mice exhibited increased LV dilatation and impaired LV systolic function that was associated with cardiomyocyte hypertrophy and interstitial fibrosis in the remote area, as compared with the controls at 6 weeks after MI. In contrast, dietary iron restriction attenuated LV dilatation and impaired LV systolic function coupled to cardiomyocyte hypertrophy and interstitial fibrosis in the remote area. Importantly, cardiac expression of cellular iron transport proteins, transferrin receptor 1 and divalent metal transporter 1 was increased in the remote area of MI mice compared with the controls. Dietary iron restriction attenuated the development of LV remodeling after MI in mice. Cellular iron transport might play a role in the pathophysiological mechanism of LV remodeling after MI.
Collapse
Affiliation(s)
- Akiyo Eguchi
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Yoshiro Naito
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan.
| | - Toshihiro Iwasaku
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Yoshitaka Okuhara
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Daisuke Morisawa
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Hisashi Sawada
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Koichi Nishimura
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Makiko Oboshi
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Kenichi Fujii
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Toshiaki Mano
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Tohru Masuyama
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Shinichi Hirotani
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| |
Collapse
|
22
|
Chung JH, Kim YS, Noh K, Lee YM, Chang SW, Kim EC. Deferoxamine promotes osteoblastic differentiation in human periodontal ligament cells via the nuclear factor erythroid 2-related factor-mediated antioxidant signaling pathway. J Periodontal Res 2013; 49:563-73. [PMID: 24111577 DOI: 10.1111/jre.12136] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2013] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND OBJECTIVE Recently it was reported that deferoxamine (DFO), an iron chelator, stimulates bone formation from MG63 and mesenchymal stem cells, but inhibits differentiation in rat calvarial cells; however, the effect of DFO on osteoblastic differentiation in human periodontal ligament cells (hPDLCs) has not been reported. The aim of this study was to investigate the effects and the possible underlying mechanism of DFO on osteoblastic differentiation of hPDLCs. MATERIAL AND METHODS The effect of DFO on osteoblast differentiation was determined by the staining intensity of calcium deposits with Alizarin red and by RT-PCR analysis of the expression of osteoblastic markers. Signal transduction pathways were analyzed by western blotting. RESULTS DFO increased osteogenic differentiation in a concentration-dependent manner by expression of the mRNA for differentiation markers and calcium nodule formation. Exposure of hPDLCs to DFO resulted in increases in the production of reactive oxygen species and in the levels of nuclear factor erythroid 2-related factor (Nrf2) protein in nuclear extractions, as well as a dose-dependent increase in the expression of Nrf2 target genes, including glutathione (GSH), glutathione S-transferase, γ-glutamylcysteine lygase, glutathione reductase and glutathione peroxidase. Pretreatment with Nrf2 small interfering RNA, GSH depletion by buthionine sulfoximine and diethyl maleate, and with antioxidants by N-acetylcysteine and vitamin E, blocked DFO-stimulated osteoblastic differentiation. Furthermore, pretreatment with GSH depletion and antioxidants blocked DFO-induced p38 MAPK, ERK, JNK and nuclear factor-kappaB pathways. CONCLUSION These data indicate, for the first time, that nontoxic DFO promotes osteoblastic differentiation of hPDLCs via modulation of the Nrf2-mediated antioxidant pathway.
Collapse
Affiliation(s)
- J H Chung
- Department of Periodontology, School of Dentistry, Kyung Hee University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
23
|
Rajapurkar MM, Lele SS, Malavade TS, Kansara MR, Hegde UN, Gohel KD, Gang SD, Shah SV, Mukhopadhyay BN. Serum catalytic Iron: A novel biomarker for coronary artery disease in patients on maintenance hemodialysis. Indian J Nephrol 2013; 23:332-7. [PMID: 24049267 PMCID: PMC3764705 DOI: 10.4103/0971-4065.116293] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in maintenance hemodialysis (MHD) patients. We evaluated the role of serum catalytic iron (SCI) as a biomarker for coronary artery disease (CAD) in patients on MHD. SCI was measured in 59 stable MHD patients. All patients underwent coronary angiography. Significant CAD was defined as a > 70% narrowing in at least one epicardial coronary artery. Levels of SCI were compared with a group of healthy controls. Significant CAD was detected in 22 (37.3%) patients, with one vessel disease in 14 (63.63%) and multi-vessel disease in eight (36.36%) patients. The MHD patients had elevated levels of SCI (4.70 ± 1.79 μmol/L) compared with normal health survey participants (0.11 ± 0.01 μmol/L) (P < 0.0001). MHD patients who had no CAD had SCI levels of 1.36 ± 0.34 μmol/L compared with those having significant CAD (8.92 ± 4.12 μmol/L) (P < 0.0001). Patients on MHD and diabetes had stronger correlation between SCI and prevalence of CAD compared with non-diabetics. Patients having one vessel disease had SCI of 8.85 ± 4.67 μmol/L versus multi-vessel disease with SCI of 9.05 ± 8.34 μmol/L, P = 0.48. In multivariate analysis, SCI and diabetes mellitus were independently associated with significant CAD. We confirm the high prevalence of significant CAD in MHD patients. Elevated SCI levels are associated with presence of significant coronary disease in such patients. The association of SCI is higher in diabetic versus the non-diabetic subgroup. This is an important potentially modifiable biomarker of CAD in MHD patients.
Collapse
Affiliation(s)
- M M Rajapurkar
- Department of Nephrology, Muljibhai Patel Urological Hospital, Nadiad, India
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Filipský T, Říha M, Hrdina R, Vávrová K, Mladěnka P. Mathematical calculations of iron complex stoichiometry by direct UV-Vis spectrophotometry. Bioorg Chem 2013; 49:1-8. [PMID: 23832103 DOI: 10.1016/j.bioorg.2013.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 04/15/2013] [Accepted: 06/03/2013] [Indexed: 01/19/2023]
Abstract
The effects of iron-chelating agents on miscellaneous pathologies are currently largely tested. Due to various indications, different properties for chelators are required. A stoichiometry of the complex in relation to pH is one of the crucial factors. Moreover, the published data on the stoichiometry, especially concerning flavonoids, are equivocal. In this study, a new complementary approach was employed for the determination of stoichiometry in 10 iron-chelating agents, including clinically used drugs, by UV-Vis spectrophotometry at relevant pH conditions and compared with the standard Job's method. This study showed that the simple approach based on absorbance at the wavelength of complex absorption maximum was sufficient when the difference between absorption maximum of substance and complex was high. However, in majority of substances this difference was much lower (9-73 nm). The novel complementary approach was able to determine the stoichiometry in all tested cases. The major benefit of this method compared to the standard Job's approach seems to be its capability to reveal a reaction stoichiometry in chelators with moderate affinity to iron. In conclusion, using this complementary method may explain several previous contradictory data and lead to a better understanding of the underlying mechanisms of chelator's action.
Collapse
Affiliation(s)
- Tomáš Filipský
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | | | | | | | | |
Collapse
|
25
|
Lele SS, Mukhopadhyay BN, Mardikar MM, Patel TA, Vasavada AK, Banker DN, Kapasi KD, Chauhan VC, Chawla KC, Raju SR, Hiremath SS, Chinchole SS, Rajapurkar MM. Impact of catalytic iron on mortality in patients with acute coronary syndrome exposed to iodinated radiocontrast-The Iscom Study. Am Heart J 2013; 165:744-51. [PMID: 23622911 DOI: 10.1016/j.ahj.2013.02.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 02/18/2013] [Indexed: 11/15/2022]
Abstract
BACKGROUND Catalytic iron (CI) mediates vascular injury by generating reactive oxygen species. We evaluated role of CI in predicting mortality in patients with acute coronary syndrome (ACS) and studied association of contrast nephropathy with CI levels. METHODS We investigated 806 patients with ACS undergoing contrast exposure for a cardiac procedure who were followed up for 30 days. RESULTS Overall mortality was 1.6% at 30 days. Catalytic iron at baseline predicted mortality with CI levels significantly higher in those who died, 0.45 μmol/L (0.37, 0.68) compared with survivors 0.31 μmol/L (0.21, 0.40); P = .004. Catalytic iron was associated with increased risk of death in the highest quartile compared with lower 3 quartiles (hazard ratio 7.88, P = .001) after adjustment for age, diabetes, ST deviation, Killip class, ejection fraction, baseline creatinine, hemoglobin level, and troponin. Fifty-five patients (6.8%) developed contrast nephropathy. Patients with contrast nephropathy had a 27% increase in median CI levels from baseline up to 48 hours compared with a marginal 2.9% increase in those without contrast nephropathy (0.37, 0.14 μmol/L to 0.47, 0.20 μmol/L versus 0.35, 0.12 μmol/L to 0.36, 0.14 μmol/L, P < .0001). Patients with contrast nephropathy had significantly higher mortality compared with those without contrast nephropathy (9.1% vs 1.1%, P = .001). CONCLUSION High baseline CI levels predicted mortality in patients with ACS. Occurrence of contrast nephropathy was associated with rise in CI levels and higher mortality. Therapeutic options to buffer or chelate CI may have beneficial effects on mortality in this setting.
Collapse
Affiliation(s)
- Suhas S Lele
- Department of Cardiology, Bhailal Amin General Hospital, Baroda, Gujarat, India.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Říha M, Karlíčková J, Filipský T, Macáková K, Hrdina R, Mladěnka P. Novel method for rapid copper chelation assessment confirmed low affinity of D-penicillamine for copper in comparison with trientine and 8-hydroxyquinolines. J Inorg Biochem 2013; 123:80-7. [PMID: 23563391 DOI: 10.1016/j.jinorgbio.2013.02.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 02/06/2013] [Accepted: 02/28/2013] [Indexed: 12/11/2022]
Abstract
Copper is an essential trace element involved in many physiological processes. Since disorder of copper homeostasis is observed in various pathologies, copper chelators may represent a promising therapeutic tool. This study was aimed at: 1) formation of an in vitro methodology for screening of copper chelators, and 2) detailed analysis of the interaction of copper with clinically used D-penicillamine (D-PEN), triethylenetetramine (trientine), experimentally tested 8-hydroxyquinolines, and the disodium salt of EDTA as a standard chelator. Methodology based on bathocuproinedisulfonic acid disodium salt (BCS), usable at (patho)physiologically relevant pHs (4.5-7.5), enabled assessment of both cuprous and cupric ions chelation and comparison of the relative affinities of the tested compounds for copper. In the case of potent chelators, the stoichiometry could be estimated too. Clioquinol, chloroxine and EDTA formed very stable complexes with Cu(+)/Cu(2+) at all tested pHs, while copper complexes with trientine were stable only under neutral or slightly acidic conditions. Non-substituted 8-hydroxyquinoline was a less efficient copper chelator, but still unequivocally more potent than D-PEN. Both 8-hydroxyquinoline and D-PEN chelation potencies, similarly to that of trientine, were pH-dependent and decreased with pH. Moreover, only D-PEN was able to reduce cupric ions. Conclusively, BCS assay represents a rapid, simple and precise method for copper chelation measurement. In addition, lower binding affinity of D-PEN compared with 8-hydroxyquinolines and trientine was demonstrated.
Collapse
Affiliation(s)
- Michal Říha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | | | | | | | | | | |
Collapse
|
27
|
Zacharski LR, DePalma RG, Shamayeva G, Chow BK. The statin-iron nexus: anti-inflammatory intervention for arterial disease prevention. Am J Public Health 2013; 103:e105-12. [PMID: 23409890 DOI: 10.2105/ajph.2012.301163] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVES We postulated the existence of a statin-iron nexus by which statins improve cardiovascular disease outcomes at least partially by countering proinflammatory effects of excess iron stores. METHODS Using data from a clinical trial of iron (ferritin) reduction in advanced peripheral arterial disease, the Iron and Atherosclerosis Study, we compared effects of ferritin levels versus high-density lipoprotein to low-density lipoprotein ratios (both were randomization variables) on clinical outcomes in participants receiving and not receiving statins. RESULTS Statins increased high-density lipoprotein to low-density lipoprotein ratios and reduced ferritin levels by noninteracting mechanisms. Improved clinical outcomes were associated with lower ferritin levels but not with improved lipid status. CONCLUSIONS There are commonalities between the clinical benefits of statins and the maintenance of physiologic iron levels. Iron reduction may be a safe and low-cost alternative to statins.
Collapse
Affiliation(s)
- Leo R Zacharski
- Research Service, Veterans Affairs Hospital, White River Junction, VT 05009, USA.
| | | | | | | |
Collapse
|
28
|
Intralipid, a clinically safe compound, protects the heart against ischemia-reperfusion injury more efficiently than cyclosporine-A. Anesthesiology 2012; 117:836-46. [PMID: 22814384 DOI: 10.1097/aln.0b013e3182655e73] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND We have recently shown that postischemic administration of intralipid protects the heart against ischemia-reperfusion injury. Here we compared the cardioprotective effects of intralipid with cyclosporine-A, a potent inhibitor of the mitochondrial permeability transition pore opening. METHODS In vivo rat hearts or isolated Langendorff-perfused mouse hearts were subjected to ischemia followed by reperfusion with intralipid (0.5%, 1% and 2% ex-vivo, and 20% in vivo), cyclosporine-A (0.2 μM, 0.8 μM, and 1.5 μM ex- vivo and 10 mg/kg in vivo), or vehicle. The hemodynamic function, infarct size, calcium retention capacity, mitochondrial superoxide production, and phosphorylation levels of protein kinase B (Akt)/glycogen synthase kinase-3β (GSK-3β) were measured. The values are mean ± SEM. RESULTS Administration of intralipid at reperfusion significantly reduced myocardial infarct size compared with cyclosporine-A in vivo (infarct size/area at risk)%: 22.9 ± 2.5% vs. 35.2 ± 3.5%; P = 0.030, n = 7/group). Postischemic administration of intralipid at its optimal dose (1%) was more effective than cyclosporine-A (0.8 μM) in protecting the ex vivo heart against ischemia-reperfusion injury, as the rate pressure product at the end of reperfusion was significantly higher (mmHg · beats/min: 12,740 ± 675 [n = 7] vs. 9,203 ± 10,781 [n = 5], P = 0.024), and the infarct size was markedly smaller (17.3 ± 2.9 [n = 7] vs. 29.2 ± 2.7 [n = 5], P = 0.014). Intralipid was as efficient as cyclosporine-A in inhibiting the mitochondrial permeability transition pore opening (calcium retention capacity = 280 ± 8.2 vs. 260.3 ± 2.9 nmol/mg mitochondria protein in cyclosporine-A, P = 0.454, n = 6) and in reducing cardiac mitochondrial superoxide production. Unlike intralipid, which increased phosphorylation of Akt (6-fold) and GSK-3β (5-fold), cyclosporine-A had no effect on the activation of these prosurvival kinases. CONCLUSIONS Although intralipid inhibits the opening of the mitochondrial permeability transition pore as efficiently as cyclosporine-A, intralipid is more effective in reducing the infarct size and improving the cardiac functional recovery.
Collapse
|
29
|
Macáková K, Mladěnka P, Filipský T, Říha M, Jahodář L, Trejtnar F, Bovicelli P, Proietti Silvestri I, Hrdina R, Saso L. Iron reduction potentiates hydroxyl radical formation only in flavonols. Food Chem 2012; 135:2584-92. [PMID: 22980846 DOI: 10.1016/j.foodchem.2012.06.107] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 06/13/2012] [Accepted: 06/27/2012] [Indexed: 10/28/2022]
Abstract
Flavonoids, substantial components of the human diet, are generally considered to be beneficial. However, they may possess possible pro-oxidative effects, which could be based on their reducing potential. The aims of this study were to evaluate the ability of 26 flavonoids to reduce ferric ions at relevant pH conditions and to find a possible relationship with potentiation of hydroxyl radical production. A substantial ferric ions reduction was achieved under acidic conditions, particularly by flavonols and flavanols with the catecholic ring B. Apparently corresponding bell-shaped curves displaying the pro-oxidant effect of flavonols quercetin and kaempferol on iron-based Fenton reaction were documented. Several flavonoids were efficient antioxidants at very low concentrations but rather inefficient or pro-oxidative at higher concentrations. Flavonols, morin and rutin were progressively pro-oxidant, while 7-hydroxyflavone and hesperetin were the only flavonoids with dose-dependent inhibition of hydroxyl radical production. Conclusively, administration of flavonoids may lead to unpredictable consequences with few exceptions.
Collapse
Affiliation(s)
- Kateřina Macáková
- Department of Pharmaceutical Botany and Ecology, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Chan W, Taylor AJ, Ellims AH, Lefkovits L, Wong C, Kingwell BA, Natoli A, Croft KD, Mori T, Kaye DM, Dart AM, Duffy SJ. Effect of iron chelation on myocardial infarct size and oxidative stress in ST-elevation-myocardial infarction. Circ Cardiovasc Interv 2012; 5:270-8. [PMID: 22496085 DOI: 10.1161/circinterventions.111.966226] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Experimental studies suggest that deferoxamine (DFO) limits the generation of reactive oxygen species by chelating redox-active iron and thereby may reduce ischemia-reperfusion injury and myocardial infarct (MI) size. We investigated whether DFO administered before reperfusion by primary percutaneous coronary intervention (PPCI) would ameliorate oxidative stress and MI size. METHODS AND RESULTS We randomly assigned 60 patients with ST-elevation-MI to receive an intravenous bolus of DFO (500 mg) immediately before PPCI followed by a 12-hour infusion (50 mg/kg of body weight) (n=28) or normal saline bolus and infusion (placebo group, n=32). MI size was measured by contrast-enhanced cardiac MRI (CMRI; day 3±1), creatine kinase and troponin I area-under-the-curve, and severity of wall motion abnormality on echocardiography. Clinical follow-up including repeat CMRI and echocardiography were performed at 3 months (100±17 days). Oxidative stress was assessed by plasma F(2)-isoprostane levels. DFO and placebo groups were well balanced with respect to baseline characteristics, symptom- and door-to-balloon times, pre-PPCI coronary patency, and infarct-related artery location. Serum iron levels were decreased with DFO treatment after PPCI compared with placebo (3.0±2.5 versus 12.6±5.5 μmol/L, P<0.0001), which persisted until the end of the infusion. In DFO-treated patients, there was a significant reduction in plasma F(2)-isoprostane levels immediately after PPCI (2878±1461 versus 2213±579 pmol/L, P=0.04). However, there was no difference in CMRI-determined infarct size (DFO, 17.4±10.8%, versus placebo, 18.6±10.2%; P=0.73), myocardial salvage index at 3 days or at 3 months, or the area-under-the-curve for creatine kinase or troponin I. CONCLUSIONS Adjunctive DFO treatment after the onset of ischemia and continued periprocedurally ameliorates oxidative stress without limiting infarct size. CLINICAL TRIAL REGISTRATION URL: http://www.anzctr.org.au/. Unique identifier: ACTRN12608000308392.
Collapse
Affiliation(s)
- William Chan
- Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Filipský T, Mladěnka P, Macáková K, Hrdina R, Saso L, Marchetti F, Pettinari C. In vitro characteristics of 1-phenyl-3-methyl-4-acylpyrazol-5-ones iron chelators. Biochimie 2012; 94:125-31. [DOI: 10.1016/j.biochi.2011.09.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 09/26/2011] [Indexed: 01/19/2023]
|
32
|
Raedschelders K, Ansley DM, Chen DDY. The cellular and molecular origin of reactive oxygen species generation during myocardial ischemia and reperfusion. Pharmacol Ther 2011; 133:230-55. [PMID: 22138603 DOI: 10.1016/j.pharmthera.2011.11.004] [Citation(s) in RCA: 288] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 11/04/2011] [Indexed: 02/07/2023]
Abstract
Myocardial ischemia-reperfusion injury is an important cause of impaired heart function in the early postoperative period subsequent to cardiac surgery. Reactive oxygen species (ROS) generation increases during both ischemia and reperfusion and it plays a central role in the pathophysiology of intraoperative myocardial injury. Unfortunately, the cellular source of these ROS during ischemia and reperfusion is often poorly defined. Similarly, individual ROS members tend to be grouped together as free radicals with a uniform reactivity towards biomolecules and with deleterious effects collectively ascribed under the vague umbrella of oxidative stress. This review aims to clarify the identity, origin, and progression of ROS during myocardial ischemia and reperfusion. Additionally, this review aims to describe the biochemical reactions and cellular processes that are initiated by specific ROS that work in concert to ultimately yield the clinical manifestations of myocardial ischemia-reperfusion. Lastly, this review provides an overview of several key cardioprotective strategies that target myocardial ischemia-reperfusion injury from the perspective of ROS generation. This overview is illustrated with example clinical studies that have attempted to translate these strategies to reduce the severity of ischemia-reperfusion injury during coronary artery bypass grafting surgery.
Collapse
Affiliation(s)
- Koen Raedschelders
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine. The University of British Columbia, Vancouver, BC, Canada.
| | | | | |
Collapse
|
33
|
Postresuscitation syndrome: potential role of hydroxyl radical-induced endothelial cell damage. Crit Care Med 2011; 39:1712-20. [PMID: 21494109 DOI: 10.1097/ccm.0b013e3182186d42] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE After out of hospital cardiac arrest, it has been reported that endothelium dysfunction may occur during the postresuscitation syndrome. However, the consequences of the reperfusion phase on endothelial reactive oxygen species production and redox homeostasis have not been explored in out of hospital cardiac arrest patients. DESIGN Prospective, observational study. SETTING Medical intensive care unit in a university hospital. PATIENTS Twenty successfully resuscitated out of hospital cardiac arrest patients, seven septic shock patients, and ten healthy volunteers. INTERVENTION Plasma was collected from patients at admission and 12, 24, 36, 48, and 72 hrs after cardiac arrest. We studied the production of reactive oxygen species and cell survival during plasma perfusion using perfused endothelial cells (human umbilical vein endothelial cells) as a model. Cell antioxidant response was studied by measuring superoxide dismutase, glutathione peroxidase, and glutathione reductase activities and reduced and oxidized glutathione levels. Mitochondrial respiratory chain activity was assessed by measuring complex I, II, III, and IV activities and anaerobic glycolysis by measuring glucose-6-phosphate dehydrogenase activity. MEASUREMENTS AND MAIN RESULTS Using perfused endothelial cells as a model, we demonstrate that plasma from out of hospital cardiac arrest patients induced on naive human umbilical vein endothelial cells a significant and massive cell death compared to plasma from septic shock patients and healthy volunteers. An increase of reactive oxygen species production with a decrease in antioxidant defenses (superoxide dismutase, glutathione peroxidase, and glutathione reductase activities, reduced and oxidized glutathione levels) was observed. The metabolic consequence of plasma exposure showed that mitochondrial respiratory chain activity was significantly impaired and anaerobic glycolysis was significantly increased. Inhibiting hydroxyl radical production significantly decreased cell death, suggesting that plasma from out of hospital cardiac arrest induced significant cell death by triggering the Fenton reaction. CONCLUSION Plasma from out of hospital cardiac arrest induces major endothelial toxicity with an acute pro-oxidant state in the cells and impairment of mitochondrial respiratory chain activity. This toxicity could be due to hydroxyl radical production by activation of the Fenton reaction.
Collapse
|
34
|
Mladěnka P, Macáková K, Filipský T, Zatloukalová L, Jahodář L, Bovicelli P, Silvestri IP, Hrdina R, Saso L. In vitro analysis of iron chelating activity of flavonoids. J Inorg Biochem 2011; 105:693-701. [PMID: 21450273 DOI: 10.1016/j.jinorgbio.2011.02.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 02/01/2011] [Accepted: 02/07/2011] [Indexed: 10/18/2022]
Abstract
Flavonoids have been demonstrated to possess miscellaneous health benefits which are, at least partly, associated with iron chelation. In this in vitro study, 26 flavonoids from different subclasses were analyzed for their iron chelating activity and stability of the formed complexes in four patho/physiologically relevant pH conditions (4.5, 5.5, 6.8, and 7.5) and compared with clinically used iron chelator deferoxamine. The study demonstrated that the most effective iron binding site of flavonoids represents 6,7-dihydroxy structure. This site is incorporated in baicalein structure which formed, similarly to deferoxamine, the complexes with iron in the stoichiometry 1:1 and was not inferior in all tested pH to deferoxamine. The 3-hydroxy-4-keto conformation together with 2,3-double bond and the catecholic B ring were associated with a substantial iron chelation although the latter did not play an essential role at more acidic conditions. In agreement, quercetin and myricetin possessing all three structural requirements were similarly active to baicalein or deferoxamine at the neutral conditions, but were clearly less active in lower pH. The 5-hydroxy-4-keto site was less efficient and the complexes of iron in this site were not stable at the acidic conditions. Isolated keto, hydroxyl, methoxyl groups or an ortho methoxy-hydroxy groups were not associated with iron chelation at all.
Collapse
Affiliation(s)
- Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Hradec Králové, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Wang Y, Wu M, Al-Rousan R, Liu H, Fannin J, Paturi S, Arvapalli RK, Katta A, Kakarla SK, Rice KM, Triest WE, Blough ER. Iron-induced cardiac damage: role of apoptosis and deferasirox intervention. J Pharmacol Exp Ther 2011; 336:56-63. [PMID: 20947636 DOI: 10.1124/jpet.110.172668] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
Excess cardiac iron levels are associated with cardiac damage and can result in increased morbidity and mortality. Here, we hypothesize that elevations in tissue iron can activate caspase-dependent signaling, which leads to increased cardiac apoptosis and fibrosis, and that these alterations can be attenuated by iron chelation. Using an iron-overloaded gerbil model, we show that increased cardiac iron is associated with reduced activation of Akt (Ser473 and Thr308), diminished phosphorylation of the proapoptotic regulator Bad (Ser136), and an increased Bax/Bcl-2 ratio. These iron-overload-induced alterations in Akt/Bad phosphorylation and Bax/Bcl-2 ratio were coupled with increased activation of the downstream caspase-9 (40/38- and 17-kDa fragments) and apoptosis executioner caspase-3 (19- and 17-kDa fragments), which were accompanied by evidence of elevated cytoskeletal α-fodrin cleavage (150- and 120-kDa fragments), discontinuity of myocardial membrane dystrophin immunoreactivity, increases in the number of terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL)-positive cells (nucleic DNA fragmentation), and cardiac fibrosis. We demonstrate that the administration of deferasirox, a tridentate iron chelator, is associated with diminished tissue iron deposition, attenuated activation of caspases, reduced α-fodrin cleavage, improved membrane integrity, decreased TUNEL reactivity, and attenuated cardiac fibrosis. These results suggest that the activation of caspase-dependent signaling may play a role in the development of iron-induced cardiac apoptosis and fibrosis, and deferasirox, via a reduction in cardiac tissue iron levels, may be useful for decreasing the extent of iron-induced cardiac damage.
Collapse
Affiliation(s)
- Yeling Wang
- Center for Diagnostic Nanosystems, Marshall University, 1 John Marshall Drive, Huntington, WV 25755-1090, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Khan M, Meduru S, Mostafa M, Khan S, Hideg K, Kuppusamy P. Trimetazidine, administered at the onset of reperfusion, ameliorates myocardial dysfunction and injury by activation of p38 mitogen-activated protein kinase and Akt signaling. J Pharmacol Exp Ther 2010; 333:421-9. [PMID: 20167841 PMCID: PMC2872960 DOI: 10.1124/jpet.109.165175] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Accepted: 02/17/2010] [Indexed: 01/28/2023] Open
Abstract
Trimetazidine [1-(2,3,4-trimethoxybenzyl)piperazine; TMZ] is an anti-ischemic cardiac drug; however, its efficacy and mechanism of cardioprotection upon reperfusion are largely unknown. The objective of this study was to determine whether TMZ, given before reperfusion, could attenuate myocardial reperfusion injury. Ischemia/reperfusion (I/R) was induced in rat hearts by ligating the left anterior descending (LAD) coronary artery for 30 min followed by 48 h of reperfusion. TMZ (5 mg/kg b.wt.) was administered 5 min before reperfusion. The study used three experimental groups: control (-I/R; -TMZ), I/R (+I/R; -TMZ), and TMZ (+I/R; +TMZ). Echocardiography and EPR oximetry were used to assess cardiac function and oxygenation, respectively. The ejection fraction, which was significantly depressed in the I/R group (62 +/- 5 versus 84 +/- 3% in control), was restored to 72 +/- 3% in the TMZ group. Myocardial pO2 in the TMZ group returned to baseline levels (approximately 20 mm Hg) within 1 h of reperfusion, whereas the I/R group showed a significant hyperoxygenation even after 48 h of reperfusion. The infarct size was significantly reduced in the TMZ group (26 +/- 3 versus 47 +/- 5% in I/R). TMZ treatment significantly attenuated superoxide levels in the tissue. Tissue homogenates showed a significant increase in p38 and p-Akt and decrease in caspase-3 levels in the TMZ group. In summary, the results demonstrated that TMZ is cardioprotective when administered before reperfusion and that this protection appears to be mediated by activation of p38 mitogen-activated protein kinase and Akt signaling. The study emphasizes the importance of administering TMZ before reflow to prevent reperfusion-mediated cardiac injury and dysfunction.
Collapse
Affiliation(s)
- Mahmood Khan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | |
Collapse
|
37
|
Zoroddu MA, Medici S, Peana M. Metal-chelating properties of carvedilol: an antihypertensive drug with antioxidant activity. J COORD CHEM 2009. [DOI: 10.1080/00958970903267280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
| | - Serenella Medici
- a Department of Chemistry , University of Sassari , Via Vienna 2, Sassari 07100, Italy
| | - Massimiliano Peana
- a Department of Chemistry , University of Sassari , Via Vienna 2, Sassari 07100, Italy
| |
Collapse
|
38
|
Andreadou I, Iliodromitis EK, Farmakis D, Kremastinos DT. To prevent, protect and save the ischemic heart: antioxidants revisited. Expert Opin Ther Targets 2009; 13:945-56. [DOI: 10.1517/14728220903039698] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Mladĕnka P, Kalinowski DS, Hašková P, Bobrovová Z, Hrdina R, Šimůnek T, Nachtigal P, Semecký V, Vávrová J, Holečková M, Palicka V, Mazurová Y, Jansson PJ, Richardson DR. The Novel Iron Chelator, 2-Pyridylcarboxaldehyde 2-Thiophenecarboxyl Hydrazone, Reduces Catecholamine-Mediated Myocardial Toxicity. Chem Res Toxicol 2008; 22:208-17. [DOI: 10.1021/tx800331j] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Pr̆emysl Mladĕnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Danuta S. Kalinowski
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Pavlína Hašková
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Zuzana Bobrovová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Radomír Hrdina
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Tomáš Šimůnek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Petr Nachtigal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Vladimír Semecký
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Jaroslava Vávrová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Magdaléna Holečková
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Vladimir Palicka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Yvona Mazurová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Patric J. Jansson
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| | - Des R. Richardson
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Department of Biological and Medical Science, Faculty of Pharmacy in Hradec Králové, Institute of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Králové, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Czech Republic, and Iron Metabolism and Chelation Program, Bosch
| |
Collapse
|
40
|
Mohan IK, Khan M, Wisel S, Selvendiran K, Sridhar A, Carnes CA, Bognar B, Kálai T, Hideg K, Kuppusamy P. Cardioprotection by HO-4038, a novel verapamil derivative, targeted against ischemia and reperfusion-mediated acute myocardial infarction. Am J Physiol Heart Circ Physiol 2008; 296:H140-51. [PMID: 18978191 DOI: 10.1152/ajpheart.00687.2008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Many cardiac interventional procedures, such as coronary angioplasty, stenting, and thrombolysis, attempt to reintroduce blood flow (reperfusion) to an ischemic region of myocardium. However, the reperfusion is accompanied by a complex cascade of cellular and molecular events resulting in oxidative damage, termed myocardial ischemia-reperfusion (I/R) injury. In this study, we evaluated the ability of HO-4038, an N-hydroxypiperidine derivative of verapamil, on the modulation of myocardial tissue oxygenation (Po(2)), I/R injury, and key signaling molecules involved in cardioprotection in an in vivo rat model of acute myocardial infarction (MI). MI was created in rats by ligating the left anterior descending coronary artery (LAD) for 30 min followed by 24 h of reperfusion. Verapamil or HO-4038 was infused through the jugular vein 10 min before the induction of ischemia. Myocardial Po(2) and the free-radical scavenging ability of HO-4038 were measured using electron paramagnetic resonance spectroscopy. HO-4038 showed a significantly better scavenging ability of reactive oxygen radicals compared with verapamil. The cardiac contractile functions in the I/R hearts were significantly higher recovery in HO-4038 compared with the verapamil group. A significant decrease in the plasma levels of creatine kinase and lactate dehydrogenase was observed in the HO-4038 group compared with the verapamil or untreated I/R groups. The left ventricular infarct size was significantly less in the HO-4038 (23 +/- 2%) compared with the untreated I/R (36 +/- 4%) group. HO-4038 significantly attenuated the hyperoxygenation (36 +/- 1 mmHg) during reperfusion compared with the untreated I/R group (44 +/- 2 mmHg). The HO-4038-treated group also markedly attenuated superoxide production, increased nitric oxide generation, and enhanced Akt and Bcl-2 levels in the reperfused myocardium. Overall, the results demonstrated that HO-4038 significantly protected hearts against I/R-induced cardiac dysfunction and damage through the combined beneficial actions of calcium-channel blocking, antioxidant, and prosurvival signaling activities.
Collapse
Affiliation(s)
- Iyyapu K Mohan
- Davis Heart and Lung Research Institute, The Ohio State Univ., Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Direct administration of rutin does not protect against catecholamine cardiotoxicity. Toxicology 2008; 255:25-32. [PMID: 18992299 DOI: 10.1016/j.tox.2008.09.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Revised: 09/23/2008] [Accepted: 09/24/2008] [Indexed: 11/22/2022]
Abstract
High levels of catecholamines are cardiotoxic and may trigger acute myocardial infarction (AMI). Similarly, the synthetic catecholamine isoprenaline (ISO) evokes a pathological state similar to AMI. During AMI there is a marked increase of free iron and copper which are crucial catalysts of reactive oxygen species formation. Rutin, a natural flavonoid glycoside possessing free radical scavenging and iron/copper chelating activity, may therefore be potentially useful in reduction of catecholamine cardiotoxicity as was previously demonstrated after its long-term peroral administration. Male Wistar:Han rats received rutin (46 or 11.5 mg kg(-1) i.v.) alone or with necrogenic dose of ISO (100 mg kg(-1) s.c.). Haemodynamic parameters were measured 24h after drug application together with analysis of blood, myocardial content of elements and histological examination. Results were confirmed by cytotoxicity studies using cardiomyoblast cell line H9c2. Rutin in a dose of 46 mg kg(-1) aggravated ISO-cardiotoxicity while the dose of 11 mg kg(-1) had no effect. These unexpected results were in agreement with in vitro experiments, where co-incubation with larger concentrations of rutin significantly augmented ISO cytotoxicity. Our results, in contrast to previous studies in the literature, suggest that the reported positive effects of peroral administration of rutin were unlikely to have been mediated by rutin per se but probably by its metabolite(s) or by some other, at this moment, unknown adaptive mechanism(s), which merit further investigation.
Collapse
|
42
|
Nikas DN, Chatziathanasiou G, Kotsia A, Papamichael N, Thomas C, Papafaklis M, Naka KK, Kazakos N, Milionis HJ, Vakalis K, Katsouras CS, Mpoumpa V, Vougiouklakis T, Michalis L. Effect of intravenous administration of antioxidants alone and in combination on myocardial reperfusion injury in an experimental pig model. CURRENT THERAPEUTIC RESEARCH 2008; 69:423-439. [PMID: 24692817 PMCID: PMC3969921 DOI: 10.1016/j.curtheres.2008.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Several antioxidants have been found to have conflicting results in attenuating myocardial reperfusion injury. These studies were done primarily in experimental protocols that did not approximate clinical situations. OBJECTIVE The aim of this study was to test the efficacy of 3 different antioxidants (ascorbic acid [AA], desferrioxamine, and N-acetylcysteine [NAC]) administered IV alone and in combination in a closed-chest pig model. METHODS Farm-raised domestic male pigs (aged 3-5 months, weight of 30-35 kg) were assigned to 1 of 5 groups to receive treatment as follows: group A, AA 100 mg/kg; group B, desferrioxamine 60 mg/kg; group C, a loading dose of NAC 100 mg/kg for 20 minutes and a 20-mg/kg maintenance dose; group D, all 3 drugs in combination; and group E, normal saline (control group). The infusion of all drugs was started 15 minutes before and completed 5 minutes after reperfusion, except for the administration of NAC, which was terminated 60 minutes postreperfusion. Myocardial ischemia (45 minutes) and reperfusion (210 minutes) were achieved percutaneously by circumflex artery balloon occlusion. Ejection fraction, left ventricular end-diastolic pressure (LVEDP), flow in the infarcted artery, and all ventricular arrhythmias were recorded. Oxidative stress was estimated by serial measurements of thiobarbituric acid reactive substance (TBARS) concentration in coronary sinus blood. Infarct size was assessed as a percentage of the area at risk (I/R ratio) using the tetrazolium red staining method. RESULTS The 25 pigs were divided into 5 groups of 5 pigs each. No significant between-group differences were found in I/R ratio or in oxidative stress (as measured by TBARS concentration). Group C developed significantly more ventricular atrhythmias than the control group (80% vs 0%, P = 0.02). No other differences among groups were found. LVEDP was significantly elevated in all treatment groups (mean LVEDP difference [SD] for group A, 6.0 [1.6] mm Hg; group B, 17.6 [1.9] mm Hg; group C, 3.6 [1.7] mm Hg; group D, 6.8 [3.2] and group E, 5.4 [3.4] mm Hg). LVEDP elevation was found to be significantly higher in group B compared with all the other groups (all, P < 0.001). No significant between-group differences were found in the other parameters measured. CONCLUSION In this experimental pig model, the antioxidants AA, desferrioxamine, and NAC administered alone or in combination did not reduce the deleterious effects of reperfusion injury and specifically the extent of myocardial necrosis.
Collapse
Affiliation(s)
| | | | - Anna Kotsia
- Michaelideion Cardiac Centre, Ioannina Medical School, Ioannina, Greece
| | - Nikos Papamichael
- Department of Cardiology, University Hospital of Ioannina, Ioannina, Greece
| | | | | | - Katerina K. Naka
- Michaelideion Cardiac Centre, Ioannina Medical School, Ioannina, Greece
- Department of Cardiology, University Hospital of Ioannina, Ioannina, Greece
| | - Nikos Kazakos
- Michaelideion Cardiac Centre, Ioannina Medical School, Ioannina, Greece
| | - Haralampos J. Milionis
- Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
| | - Kostas Vakalis
- Michaelideion Cardiac Centre, Ioannina Medical School, Ioannina, Greece
| | - Christos S. Katsouras
- Michaelideion Cardiac Centre, Ioannina Medical School, Ioannina, Greece
- Department of Cardiology, University Hospital of Ioannina, Ioannina, Greece
| | - Vasiliki Mpoumpa
- Department of Forensic Medicine, Ioannina Medical School, Ioannina, Greece
| | | | - Lampros Michalis
- Michaelideion Cardiac Centre, Ioannina Medical School, Ioannina, Greece
- Department of Cardiology, University Hospital of Ioannina, Ioannina, Greece
| |
Collapse
|
43
|
Bloomer SA, Brown KE, Buettner GR, Kregel KC. Dysregulation of hepatic iron with aging: implications for heat stress-induced oxidative liver injury. Am J Physiol Regul Integr Comp Physiol 2008; 294:R1165-74. [DOI: 10.1152/ajpregu.00719.2007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Environmental heat stress is associated with an age-related increase in hepatic oxidative damage and an exaggerated state of oxidative stress. The purpose of this investigation was to evaluate the regulation of hepatic iron after heat stress. A secondary aim was to determine a potential role for iron in heat stress-induced liver injury. Hyperthermia-induced alterations in hepatic iron were evaluated in young (6 mo) and old (24 mo) Fischer 344 rats by exposing them to a two-heat stress protocol. Livers were harvested at several time points after the second heating and assayed for labile and nonheme iron. In the control condition, there was no difference in labile iron between age groups. Both labile iron and storage iron were not altered by hyperthermia in young rats, but both were increased immediately after heating in old rats. To evaluate a role for iron in liver injury, hepatic iron content was manipulated in young and old rats, and then both groups were exposed to heat stress. Iron administration to young rats significantly increased hepatic iron content and ferritin but did not affect markers of lipid peroxidation under control conditions or after heat stress. In old rats, iron chelation with deferoxamine prevented the increase in nonheme iron, labile iron, ferritin, and lipid peroxidation after heat stress. These results suggest that iron may play a role in hepatic injury after hyperthermia. Thus, dysregulation of iron may contribute to the gradual decline in cellular and physiological function that occurs with aging.
Collapse
|
44
|
Raman SV, Winner MW, Tran T, Velayutham M, Simonetti OP, Baker PB, Olesik J, McCarthy B, Ferketich AK, Zweier JL. In vivo atherosclerotic plaque characterization using magnetic susceptibility distinguishes symptom-producing plaques. JACC Cardiovasc Imaging 2008; 1:49-57. [PMID: 19356405 PMCID: PMC2729432 DOI: 10.1016/j.jcmg.2007.09.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 09/18/2007] [Accepted: 09/24/2007] [Indexed: 10/22/2022]
Abstract
OBJECTIVES We investigated the role of iron deposition in atherosclerotic plaque instability using a novel approach of in vivo plaque characterization by a noninvasive, noncontrast magnetic resonance-based T2* measurement. This approach was validated using ex vivo plaque analyses to establish that T2* accurately reflects intraplaque iron composition. BACKGROUND Iron catalyzes free radical production, a key step for lipid peroxidation and atherosclerosis development. The parameter T2* measures tissue magnetic susceptibility, which historically has been used to quantify hepatic and myocardial iron. The T2* measurement has not been used for in vivo plaque characterization in patients with atherosclerosis. METHODS Thirty-nine patients referred for carotid endarterectomy were prospectively enrolled to undergo preoperative carotid magnetic resonance imaging (MRI) and postoperative analysis of the explanted plaque. Clinical history of any symptoms attributable to each carotid lesion was recorded. We could not complete MRI in 4 subjects because of their claustrophobia, and 3 patients scanned before the institution of a neck stabilizer had motion artifact, precluding quantification. RESULTS Symptomatic patients had significantly lower plaque T2* values (20.0 +/- 1.8 ms) compared with asymptomatic patients (34.4 +/- 2.7 ms, p < 0.001). Analytical methods demonstrated similar total iron (138.6 +/- 36.5 microg/g vs. 165.8 +/- 48.3 microg/g, p = NS) but less low molecular weight Fe(III) (7.3 +/- 3.8 microg/g vs. 17.7 +/- 4.0 microg/g, p < 0.05) in the explanted plaques of symptomatic versus asymptomatic patients, respectively, which is consistent with a shift in iron from Fe(III) to greater amounts of T2*-shortening forms of iron. Mass spectroscopy also showed significantly lower calcium (37.5 +/- 10.8 mg/g vs. 123.6 +/- 19.3 mg/g, p < 0.01) and greater copper (3.2 +/- 0.5 microg/g vs. 1.7 +/- 0.1 microg/g, p < 0.01) in plaques from symptomatic patients. CONCLUSIONS In vivo measurement of intraplaque T2* using MRI is feasible and distinguishes symptom-producing from non-symptom-producing plaques in patients with carotid artery atherosclerosis. Symptom-producing plaques demonstrated characteristic changes in iron forms by ex vivo analysis, supporting the dynamic presence of iron in the microenvironment of atherosclerotic plaque.
Collapse
Affiliation(s)
- Subha V Raman
- Davis Heart and Lung Research Institute and Heart Center, The Ohio State University, Columbus, Ohio 43210, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
After Four Hours of Cold Ischemia and Cardioplegic Protocol, the Heart Can Still Be Rescued With Postconditioning. Transplantation 2007; 84:1474-82. [DOI: 10.1097/01.tp.0000288637.18796.0e] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
Elmes MJ, Gardner DS, Langley-Evans SC. Fetal exposure to a maternal low-protein diet is associated with altered left ventricular pressure response to ischaemia-reperfusion injury. Br J Nutr 2007; 98:93-100. [PMID: 17445339 DOI: 10.1017/s000711450769182x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Rats exposed to protein restriction as fetuses develop hypertension as adults. Hypertension increases the risk of myocardial ischaemia and infarction. We investigated whether rats exposed to low-protein diets in utero are more susceptible to myocardial ischaemia-reperfusion (IR) injury. Pregnant Wistar rats were fed control or low-protein (MLP) diets throughout pregnancy. At 4 and 8 weeks postnatal age systolic blood pressure was determined in the offspring using tail-cuff plethysmography. At 6 months of age, rats were treated with saline or N-acetylcysteine (NAC) for 48 h. Rapidly excised hearts were retro-perfused (Langendorff) to assess isolated cardiac function before (baseline), during 30 min ischaemia (no coronary perfusion) and for 60 min after reinstating coronary perfusion (reperfusion). Hearts were then harvested and treated appropriately for analysis of infarct size. Exposure to the MLP diet in utero significantly increased systolic blood pressure at 4 and 8 weeks of age (6-13 mmHg increase; P < 0.001) and significantly impaired recovery of left ventricular developed pressure after ischaemia at 6 months of age in male offspring only (P < 0.003). Pre-treatment with NAC prevented this impairment of recovery in MLP male offspring and improved recovery in all females. Myocardial infarct size was not different between dietary groups after IR, but NAC pre-treatment significantly reduced the degree of infarction (P < 0.001). In conclusion, an MLP diet throughout gestation significantly impairs recovery of the 6-month-old adult rat heart to IR-induced injury in a sex-specific manner. Undernutrition during development may increase susceptibility to CHD by impairing recovery from coronary events.
Collapse
Affiliation(s)
- Matthew J Elmes
- Division of Nutritional Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, UK.
| | | | | |
Collapse
|
47
|
D'souza B, Vishwanath P, D'souza V. Oxidative injury and antioxidants in coronary artery bypass graft surgery: Off-pump CABG significantly reduces oxidative stress. Clin Chim Acta 2007; 375:147-52. [PMID: 16904092 DOI: 10.1016/j.cca.2006.07.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2005] [Revised: 07/02/2006] [Accepted: 07/03/2006] [Indexed: 10/24/2022]
Abstract
BACKGROUND Coronary artery bypass grafting (CABG) can now be performed with or without cardiopulmonary bypass. The former entails global ischemia followed by reperfusion after declamping, whereas the latter does not. In view of growing evidence that reperfusion is associated with oxidative stress, we studied the extent of oxidative stress and antioxidant status in patients undergoing on-pump and off-pump CABG to determine whether the latter significantly reduces oxidative stress. METHODS Thirty patients were initially enrolled for the study. The inclusion criteria included patients with atherosclerotic triple vessel disease, undergoing elective CABG, with good LV function, no major risk factors for surgery, with all biochemical investigations within normal limits, having stable angina and no history of previous infarct. Patients with valvular heart disease, ventricular aneurysm, heart failure and poor left ventricular function were excluded. These were alternately posted for on-pump and off-pump CABG. Eight patients were excluded as they developed unforeseen complications during the surgery. Out of the remaining 22 patients, 13 underwent off-pump CABG and 9 underwent on-pump CABG. Five blood samples were collected; baseline, 5, 15, 60 min and 24 h after reperfusion. Samples were analyzed for thiobarbituric acid reactive substances (TBARS), glutathione (G-SH) and catalase (CAT). The results were compared with their preanaesthetic levels in both the groups and also with 20 age- and sex-matched normal healthy individuals. RESULTS Lipid peroxidation was significantly increased after reperfusion in patients undergoing on-pump CABG, maximum increase (p<0.0001) was seen 1 h after reperfusion, whereas off-pump CABG reduces oxidative stress. The G-SH levels were significantly decreased after reperfusion in on-pump and off-pump CABG patients, maximum decrease (p<0.0001) was seen 5 min after reperfusion in on-pump CABG. The catalase activity was significantly increased after reperfusion in on-pump and off-pump CABG patients, maximum increase (p<0.0001) was seen 1 h after reperfusion in on-pump CABG. CONCLUSION Significant increase in oxidative stress was seen in patients undergoing on-pump CABG, whereas oxidative stress was less in off-pump CABG patients. The G-SH levels were decreased and Catalase activity was increased significantly in both on-pump and off-pump CABG patients.
Collapse
|
48
|
van der A DL, Peeters PHM, Grobbee DE, Roest M, Marx JJM, Voorbij HM, van der Schouw YT. HFE mutations and risk of coronary heart disease in middle-aged women. Eur J Clin Invest 2006; 36:682-90. [PMID: 16968463 DOI: 10.1111/j.1365-2362.2006.01711.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Although heterozygosity for the C282Y mutation in the HFE gene has been associated with an increased risk of cardiovascular events, epidemiological studies remain inconclusive. The aim of the present study was to obtain further evidence as to whether HFE mutations are associated with risk of coronary heart disease (CHD) in middle-aged women. We used data of a cohort of 15 236 Dutch middle-aged women to investigate whether C282Y carriers and H63D carriers are at increased risk of coronary heart disease compared with non-carriers. MATERIALS AND METHODS Women were included in the study between 1993 and 1997 and were followed until 1 January 2000 for cardiovascular events. HFE genotyping was performed on all 211 coronary heart disease cases and a randomly selected sample from the baseline cohort (n = 1526). A weighted Cox proportional hazards model was used to estimate crude, age-adjusted and multivariate adjusted hazard ratios for C282Y and H63D carriership in relation to coronary heart disease. RESULTS Compared with non-carriers, those that carried the C282Y allele were not at increased risk for CHD (HR = 1.25, 95% CI = 0.74-2.09). Neither did we find an association between the H63D mutation and CHD risk (HR = 0.73, 95% CI = 0.43-1.24). CONCLUSIONS Our results are in accordance with similar studies to date, for which we present a meta-analysis. HFE mutations appear not to affect the risk of coronary heart disease.
Collapse
|
49
|
Kevin LG, Novalija E, Stowe DF. Reactive oxygen species as mediators of cardiac injury and protection: the relevance to anesthesia practice. Anesth Analg 2005; 101:1275-1287. [PMID: 16243980 DOI: 10.1213/01.ane.0000180999.81013.d0] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) are central to cardiac ischemic and reperfusion injury. They contribute to myocardial stunning, infarction and apoptosis, and possibly to the genesis of arrhythmias. Multiple laboratory studies and clinical trials have evaluated the use of scavengers of ROS to protect the heart from the effects of ischemia and reperfusion. Generally, studies in animal models have shown such effects. Clinical trials have also shown protective effects of scavengers, but whether this protection confers meaningful clinical benefits is uncertain. Several IV anesthetic drugs act as ROS scavengers. In contrast, volatile anesthetics have recently been demonstrated to generate ROS in the heart, most likely because of inhibitory effects on cardiac mitochondria. ROS are involved in the signaling cascade for cardioprotection induced by brief exposure to a volatile anesthetic (termed "anesthetic preconditioning"). ROS, therefore, although injurious in large quantities, can have a paradoxical protective effect within the heart. In this review we provide background information on ROS formation and elimination relevant to anesthetic and adjuvant drugs with particular reference to the heart. The sources of ROS, the means by which they induce cardiac injury or activate protective signaling pathways, the results of clinical studies evaluating ROS scavengers, and the effects of anesthetic drugs on ROS are each discussed.
Collapse
Affiliation(s)
- Leo G Kevin
- Anesthesiology Research Laboratories, Departments of Anesthesiology and Physiology, Cardiovascular Research Center, The Medical College of Wisconsin, VA Medical Center Research Service, and Department of Biomedical Engineering, Marquette University, Milwaukee, Wisconsin
| | | | | |
Collapse
|
50
|
Philipp S, Cui L, Ludolph B, Kelm M, Schulz R, Cohen MV, Downey JM. Desferoxamine and ethyl-3,4-dihydroxybenzoate protect myocardium by activating NOS and generating mitochondrial ROS. Am J Physiol Heart Circ Physiol 2005; 290:H450-7. [PMID: 16155105 DOI: 10.1152/ajpheart.00472.2005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Protection from a prolyl hydroxylase domain-containing enzyme (PHD) inhibitor, desferoxamine (DFO), was recently reported to be dependent on production of reactive oxygen species (ROS). Ischemic preconditioning triggers the protected state by stimulating nitric oxide (NO) production to open mitochondrial ATP-sensitive K+ (mitoK(ATP)) channels, generating ROS required for protection. We tested whether DFO and a second PHD inhibitor, ethyl-3,4-dihydroxybenzoate (EDHB), might have similar mechanisms. EDHB and DFO increased ROS generation by 50-75% (P < 0.001) in isolated rabbit cardiomyocytes. This increase after EDHB exposure was blocked by N(omega)-nitro-L-arginine methyl ester (L-NAME), an NO synthase (NOS) inhibitor; ODQ, a guanylyl cyclase antagonist; and Rp-8-bromoguanosine-3',5'-cyclic monophosphorothioate Rp isomer, a PKG blocker, thus implicating the NO pathway in EDHB's signaling. Glibenclamide, a nonselective K(ATP) channel blocker, or 5-hydroxydecanoate, a selective mitoK(ATP) channel antagonist, also prevented EDHB's ROS production, as did blockade of mitochondrial electron transport with myxothiazol. NOS is activated by Akt. However, neither wortmannin, an inhibitor of phosphatidylinositol-3-kinase, nor Akt inhibitor blocked EDHB-induced ROS generation, indicating that EDHB initiates signaling downstream of Akt. DFO also increased ROS production, and this effect was blocked by ODQ, 5-hydroxydecanoate, and N-(2-mercaptopropionyl)glycine, an ROS scavenger. DFO increased cardiomyocyte production of nitrite, a metabolite of NO, and this effect was blocked by an inhibitor of NOS. DFO also spared ischemic myocardium in intact hearts. This infarct-sparing effect was blocked by ODQ, L-NAME, and N-(2-mercaptopropionyl)glycine. Hence, DFO and EDHB stimulate NO-dependent activation of PKG to open mitoK(ATP) channels and produce ROS, which act as second messengers to trigger entrance into the preconditioned state.
Collapse
Affiliation(s)
- Sebastian Philipp
- Dept. of Physiology, Univ. of South Alabama College of Medicine, Mobile, AL 36688, USA
| | | | | | | | | | | | | |
Collapse
|