188 results on '"Lecour, S"'
Search Results
152. HDL protects against ischemia reperfusion injury by preserving mitochondrial integrity.
- Author
-
Frias MA, Pedretti S, Hacking D, Somers S, Lacerda L, Opie LH, James RW, and Lecour S
- Subjects
- Animals, Cell Survival physiology, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocardial Infarction pathology, Myocardial Reperfusion Injury pathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Phosphorylation physiology, STAT3 Transcription Factor genetics, Signal Transduction physiology, Tumor Necrosis Factor-alpha genetics, Cholesterol, HDL metabolism, Mitochondria metabolism, Mitochondrial Membranes metabolism, Myocardial Infarction metabolism, Myocardial Reperfusion Injury metabolism
- Abstract
Objective: High density lipoproteins (HDL) protect against ischemia reperfusion injury (IRI). However the precise mechanisms are not clearly understood. The novel intrinsic prosurvival signaling pathway named survivor activating factor enhancement (SAFE) path involves the activation of tumor necrosis factor (TNF) alpha and signal transducer and activator of transcription 3 (STAT3). SAFE plays a crucial role in cardioprotection against IRI. We propose that HDL protect against IRI via activation of the SAFE pathway and modulation of the mitochondrial permeability transition pore (mPTP) opening., Methods and Results: Isolated mouse hearts were subjected to global ischemia (35 min) followed by reperfusion (45 min). HDL were given during the first 7 min of reperfusion. In control hearts, the post-reperfusion infarct size was 41.3 ± 2.3%. Addition of HDL during reperfusion reduced the infarct size in a dose-dependent manner (HDL 200 μg protein/ml: 25.5 ± 1.6%, p < 0.001 vs. control). This protective effect was absent in TNF deficient mice (TNF-KO) or cardiomyocyte-STAT3 deficient mice (STAT3-KO). Similarly, HDL, given as a preconditioning stimulus, improved cell survival and inhibited mPTP opening in isolated cardiomyocytes subjected to simulated ischemia. These protective responses were inhibited in cardiomyocytes from TNF-KO and STAT3-KO mice., Conclusion: Our data demonstrate that HDL protect against IRI by inhibition of mPTP opening, an effect mediated via activation of the SAFE pathway., (Copyright © 2013 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
153. Translating cardioprotection for patient benefit: position paper from the Working Group of Cellular Biology of the Heart of the European Society of Cardiology.
- Author
-
Hausenloy DJ, Erik Bøtker H, Condorelli G, Ferdinandy P, Garcia-Dorado D, Heusch G, Lecour S, van Laake LW, Madonna R, Ruiz-Meana M, Schulz R, Sluijter JP, Yellon DM, and Ovize M
- Subjects
- Animals, Cardiopulmonary Bypass, Cardiopulmonary Resuscitation, Collateral Circulation, Coronary Artery Bypass, Coronary Circulation, Coronary Disease complications, Disease Models, Animal, Heart Transplantation, Humans, Signal Transduction, Coronary Disease therapy, Myocardial Reperfusion Injury prevention & control
- Abstract
Coronary heart disease (CHD) is the leading cause of death and disability worldwide. Despite current therapy, the morbidity and mortality for patients with CHD remains significant. The most important manifestations of CHD arise from acute myocardial ischaemia-reperfusion injury (IRI) in terms of cardiomyocyte death and its long-term consequences. As such, new therapeutic interventions are required to protect the heart against the detrimental effects of acute IRI and improve clinical outcomes. Although a large number of cardioprotective therapies discovered in pre-clinical studies have been investigated in CHD patients, few have been translated into the clinical setting, and a significant number of these have failed to show any benefit in terms of reduced myocardial infarction and improved clinical outcomes. Because of this, there is currently no effective therapy for protecting the heart against the detrimental effects of acute IRI in patients with CHD. One major factor for this lack of success in translating cardioprotective therapies into the clinical setting can be attributed to problems with the clinical study design. Many of these clinical studies have not taken into consideration the important data provided from previously published pre-clinical and clinical studies. The overall aim of this ESC Working Group Cellular Biology of the Heart Position Paper is to provide recommendations for optimizing the design of clinical cardioprotection studies, which should hopefully result in new and effective therapeutic interventions for the future benefit of CHD patients.
- Published
- 2013
- Full Text
- View/download PDF
154. Loss of cardioprotection with ischemic preconditioning in aging hearts: role of sirtuin 1?
- Author
-
Adam T, Sharp S, Opie LH, and Lecour S
- Subjects
- Animals, Male, Myocardial Reperfusion Injury prevention & control, Rats, Rats, Long-Evans, Aging physiology, Ischemic Preconditioning, Myocardial, Sirtuin 1 physiology
- Abstract
The effectiveness of ischemic preconditioning (IPC) to protect the heart against ischemia/reperfusion injury (IRI) declines with age. The deacetylase protein sirtuin 1 (Sirt 1) confers myriad functions including longevity and cardioprotection against IRI. As such, Sirt 1 may be a potential candidate to explain the protective effect of IPC. We aim to explore the role of Sirt 1 in the loss of the cardioprotective effect of IPC with age. Isolated hearts from young (9 weeks) and older (12-18 months) Long-Evans rats were subjected to 30 minutes of global ischemia and 60 minutes of reperfusion. Preconditioning stimuli were applied with either 2 cycles of 5-minute ischemia/reperfusion or with the potent Sirt 1 agonist resveratrol (RSV, 10 µmol/L) for 15 minutes followed by a 10-minute washout before the sustained ischemia. Both IPC and RSV significantly enhanced the functional recovery of young hearts by 168% (P < .001 vs control) and 65% (P < .01 vs control), respectively, and concomitantly reduced the infarct size by 65% and 45%, but the effect was blunted in older hearts. Administration of the selective Sirt 1 inhibitor III to young hearts did not alter the protective effect of IPC. Following ischemia/reperfusion, higher Sirt 1 deacetylase activity was detected in older hearts compared to young hearts (0.48 ± 0.13 arbitrary units [AU] vs 0.17 ± 0.03 AU, P < .01) and IPC did not alter Sirt 1 deacetylase activity. In conclusion, although Sirt 1 deacetylase activity is increased with age during ischemia/reperfusion, our data suggest that the loss of the cardioprotective effect of IPC in older animals is likely to be independent of Sirt 1.
- Published
- 2013
- Full Text
- View/download PDF
155. Trials, tribulations and speculation! Report from the 7th Biennial Hatter Cardiovascular Institute Workshop.
- Author
-
Bell R, Beeuwkes R, Bøtker HE, Davidson S, Downey J, Garcia-Dorado D, Hausenloy DJ, Heusch G, Ibanez B, Kitakaze M, Lecour S, Mentzer R, Miura T, Opie L, Ovize M, Ruiz-Meana M, Schulz R, Shannon R, Walker M, Vinten-Johansen J, and Yellon D
- Subjects
- Animals, Cardiotonic Agents therapeutic use, Humans, Ischemic Postconditioning, Ischemic Preconditioning, Myocardial, Mitochondria, Heart physiology, Acute Coronary Syndrome therapy, Myocardial Infarction therapy, Myocardial Reperfusion Injury prevention & control
- Published
- 2012
- Full Text
- View/download PDF
156. Different lipid profiles according to ethnicity in the Heart of Soweto study cohort of de novo presentations of heart disease.
- Author
-
Sliwa K, Lyons JG, Carrington MJ, Lecour S, Marais AD, Raal FJ, and Stewart S
- Subjects
- Female, Heart Diseases blood, Humans, Male, Middle Aged, Retrospective Studies, Risk Factors, South Africa epidemiology, Black People, Heart Diseases ethnology, Lipid Metabolism, Lipids blood, Registries
- Abstract
Background: Historically, sub-Saharan Africa has reported low levels of atherosclerotic cardiovascular disease (CVD). However as these populations undergo epidemiological transition, this may change., Methods: This was an observational cohort study performed at Chris Hani Baragwanath Hospital in Soweto, South Africa. As part of the Heart of Soweto study, a clinical registry captured detailed clinical data on all de novo cases of structural and functional heart disease presenting to the Cardiology unit during the period 2006 to 2008. We examined fasting lipid profiles in 2 182 patients (of 5 328 total cases) according to self-reported ethnicity. The study cohort comprised 1 823 patients of African descent (61% female, aged 56 ± 16 years), 142 white Europeans (36% female, aged 57 ± 13 years), 133 Indians (51% female, aged 59 ± 12 years) and 87 of mixed ancestry (40% female, aged 56 ± 12 years)., Results: Consistent with different patterns in heart disease aetiology, there were clear differences in total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) and triglycerides across ethnicities (p < 0.001): patients of African descent had the lowest TC and LDL-C levels and Indians the highest. However, there were no significant differences in high-density lipoprotein cholesterol (HDL-C) levels between ethnicities (p = 0.20). Adjusting for age, gender and body mass index, patients of African descent were significantly less likely to record a TC of > 4.5 mmol/l (OR 0.33, 95% CI: 0.25-0.41) compared to all ethnic groups (all p < 0.001)., Conclusions: These data confirm important blood lipid differentials according to ethnicity in patients diagnosed with heart disease in Soweto, South Africa. Such disparities in CVD risk factors may justify the use of specialised prevention and management protocols.
- Published
- 2012
- Full Text
- View/download PDF
157. Interplay between SAFE and RISK pathways in sphingosine-1-phosphate-induced cardioprotection.
- Author
-
Somers SJ, Frias M, Lacerda L, Opie LH, and Lecour S
- Subjects
- Animals, Cardiotonic Agents pharmacology, Extracellular Signal-Regulated MAP Kinases metabolism, Forkhead Box Protein O1, Forkhead Transcription Factors metabolism, Lysophospholipids pharmacology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury pathology, Proto-Oncogene Proteins c-akt metabolism, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, Signal Transduction drug effects, Sphingosine pharmacology, Sphingosine therapeutic use, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha metabolism, Cardiotonic Agents therapeutic use, Lysophospholipids therapeutic use, Myocardial Infarction drug therapy, Myocardial Reperfusion Injury drug therapy, Sphingosine analogs & derivatives
- Abstract
Purpose: We studied the role of two powerful molecular signalling mechanisms involved in the cardioprotective effect of sphingosine-1-phosphate (S1P), a major component of high density lipoprotein (HDL) against myocardial ischaemic-reperfusion injury, namely the RISK pathway (Akt/Erk), including its downstream target FOXO-1 and, the SAFE pathway (TNF/STAT-3)., Methods: Control hearts from wildtype, TNF deficient (TNF(-/-)) or cardiomyocyte STAT-3 deficient (STAT-3(-/-)) male mice were perfused on a Langendorff apparatus (35 min global ischaemia and 45 min reperfusion). S1P (10 nM) was given at the onset of reperfusion for the first 7 min, with/without STAT-3 or Akt inhibitors, AG490 and wortmannin (W), respectively., Results: S1P reduced myocardial infarct size in wildtype hearts (39.3±4.4% in control vs 17.3±3.1% in S1P-treated hearts; n≥6; p<0.05) but not in STAT-3(-/-) or TNF(-/-) mice (34.2±4.3% in STAT-3(-/-) and 34.1±2.0% in TNF(-/-) mice; n≥6; p=ns vs. their respective control). Both STAT-3 and Akt inhibitors abolished the protective effects of S1P (33.7±3.3% in S1P + AG490 and 36.6±4.9% in S1P + W; n=6; p=ns vs. their respective control). Increased nuclear levels of phosphorylated STAT-3 (pSTAT-3), Akt and FOXO-1 were observed at 15 min reperfusion in wildtype mice with Western Blot analysis (53% STAT-3, 47% Akt, 41% FOXO-1; p<0.05 vs control) but not in STAT-3-/- mice or in wiltype hearts treated with the Akt inhibitor. Interestingly, an activation of pSTAT-3 was noticed in the mitochondria at 7 min but not 15 min of reperfusion., Conclusions: In conclusion, S1P activates both the SAFE and RISK pathways, therefore suggesting a dual protective signalling in S1P-induced cardioprotection.
- Published
- 2012
- Full Text
- View/download PDF
158. Lowering the alcohol content of red wine does not alter its cardioprotective properties.
- Author
-
Lamont K, Blackhurst D, Albertyn Z, Marais D, and Lecour S
- Subjects
- Analysis of Variance, Animals, Antioxidants analysis, Blood Pressure drug effects, Cardiotonic Agents, Heart Rate drug effects, In Vitro Techniques, Male, Myocardial Ischemia physiopathology, Myocardial Reperfusion Injury physiopathology, Rats, Rats, Long-Evans, Antioxidants pharmacology, Ethanol pharmacology, Myocardial Reperfusion Injury prevention & control, Wine
- Abstract
Background: Many epidemiological, clinical and laboratory studies suggest that chronic and moderate consumption of red wine benefits cardiovascular health, because of the alcoholic content or the polyphenols/flavonoids., Aims: The antioxidant and cardioprotective properties of a French red wine (cabernet sauvignon, 12% alcohol by volume) were compared with those of the same wine subjected to reverse osmosis for partial removal of alcohol (6% alcohol by volume)., Methods: Antioxidant capacity was assessed in vitro using the oxygen radical absorbance capacity (ORAC) assay. To test the cardioprotective effect of 12% v. 6% wine, the drinking water of rats used for controls was supplemented with red wine (12% or 6%). After 10 days, hearts were isolated on a Langendorff system and subjected to 30 minutes of global ischaemia plus 30 minutes of reperfusion (I/R)., Results: No differences in antioxidant capacity were observed between wine of 12% and 6% alcohol content (n=8 per group). Control hearts subjected to I/R presented a rate pressure product (heart rate x left ventricular developed pressure, expressed as a percentage of baseline value) of 16±4% (mean±standard error). Pretreatment with wine 12% or 6% improved the rate pressure product to 40±6% and 43±6%, respectively (p<0.05 v. control)., Conclusion: Our findings suggest that the reduction of alcohol content from 12% to 6% in wine did not alter its antioxidant and cardioprotective properties. Moderate and regular consumption of lower alcohol content wines may confer beneficial effects without the risks associated with traditional wines of higher alcohol content.
- Published
- 2012
- Full Text
- View/download PDF
159. High density lipoprotein/sphingosine-1-phosphate-induced cardioprotection: Role of STAT3 as part of the SAFE pathway.
- Author
-
Frias MA, Lecour S, James RW, and Pedretti S
- Abstract
High density lipoprotein (HDL) cholesterol has beneficial effects beyond its atheroprotective function in reverse cholesterol transport, including cardioprotection against ischemia reperfusion (IR) injuries. Two major constituents of HDL, namely the structural protein apolipoprotein AI (apoAI) and the sphingolipid sphingosine-1-phosphate (S1P) appear to contribute to this cardioprotective effect via the activation of intrinsic prosurvival signaling pathways that still remain to be clarified. Recently, a powerful prosurvival signaling pathway, termed the survivor activating factor enhancement (SAFE) pathway, which involves the activation of signal transducer and activator of transcription 3 (STAT3) and tumor necrosis factor α (TNF), has been shown to protect against ischemia-reperfusion injuries. The present review summarizes the evidence for the roles of HDL and S1P in cardioprotection and discusses the signaling pathways that have been implicated. It thus provides support for our contention that S1P should be considered in potential formulations of reconstituted HDL (reHDL) that may be tested for cardioprotection against coronary artery disease via the activation of the SAFE pathway.
- Published
- 2012
- Full Text
- View/download PDF
160. Influence of tumour necrosis factor alpha on the outcome of ischaemic postconditioning in the presence of obesity and diabetes.
- Author
-
Lacerda L, Opie LH, and Lecour S
- Subjects
- Animals, Cardiomegaly etiology, Diabetes Mellitus, Type 1 blood, Diabetes Mellitus, Type 1 physiopathology, Dietary Carbohydrates adverse effects, Disease Susceptibility, Heart physiopathology, Hyperglycemia etiology, Hyperinsulinism etiology, In Vitro Techniques, Leptin blood, Mice, Mice, Knockout, Myocardial Reperfusion Injury complications, Myocardial Reperfusion Injury physiopathology, Myocardium pathology, Obesity blood, Obesity etiology, Obesity physiopathology, Organ Size, Severity of Illness Index, Streptozocin, Tumor Necrosis Factor-alpha genetics, Diabetes Mellitus, Type 1 complications, Ischemic Postconditioning, Myocardial Ischemia complications, Myocardial Reperfusion Injury prevention & control, Obesity complications, Tumor Necrosis Factor-alpha metabolism
- Abstract
Obesity and diabetes contribute to cardiovascular disease and alter cytokine profile. The cytokine, tumour necrosis factor alpha (TNFα), activates a protective signalling cascade during ischaemic postconditioning (IPostC). However, most successful clinical studies with IPostC have not included obese and/or diabetic patients. We aimed to investigate the influence of TNFα on the outcome of IPostC in obese or diabetic mice. TNF knockout or wildtype mice were fed for 11 weeks with a high carbohydrate diet (HCD) to induce modest obesity. Diabetes was induced in a separate group by administration of a single intraperitoneal injection of streptozotocin. Hearts were then isolated and subjected to ischaemia (35 min of global ischaemia) followed by 45 min of reperfusion. HCD increased body weight, plasma insulin and leptin levels while the glucose level was unchanged. In streptozotocin-treated mice, blood glucose, plasma leptin and insulin were altered. Control, obese or diabetic mice were protected with IPostC in wiltype animals. In TNF knockout mice, IPostC failed to protect control and diabetic hearts while a slight protection was observed in obese hearts. Our data confirm a bidirectional role for TNFα associated with the severity of concomitant comorbidities and suggest that diabetic and/or modestly obese patients may still benefit from IPostC.
- Published
- 2012
- Full Text
- View/download PDF
161. Delayed postconditioning: cardioprotection at the limit?
- Author
-
Opie LH and Lecour S
- Subjects
- Animals, Male, Ischemic Postconditioning methods, Ischemic Preconditioning, Myocardial methods, Myocardial Infarction prevention & control, Myocardial Reperfusion Injury prevention & control
- Published
- 2011
- Full Text
- View/download PDF
162. Is red wine a SAFE sip away from cardioprotection? Mechanisms involved in resveratrol- and melatonin-induced cardioprotection.
- Author
-
Lamont KT, Somers S, Lacerda L, Opie LH, and Lecour S
- Subjects
- Animals, Blotting, Western, In Vitro Techniques, Male, Mice, Myocardium, Phosphorylation drug effects, Rats, Rats, Wistar, Receptors, Tumor Necrosis Factor, Type II genetics, Receptors, Tumor Necrosis Factor, Type II metabolism, Resveratrol, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, Melatonin pharmacology, Myocardial Infarction prevention & control, Stilbenes pharmacology, Wine
- Abstract
Epidemiological studies suggest that regular moderate consumption of red wine confers cardioprotection but the mechanisms involved in this effect remain unclear. Recent studies demonstrate the presence of melatonin in wine. We propose that melatonin, at a concentration found in red wine, confers cardioprotection against ischemia-reperfusion injury. Furthermore, we investigated whether both melatonin and resveratrol protect via the activation of the newly discovered survivor activating factor enhancement (SAFE) prosurvival signaling pathway that involves the activation of tumor necrosis factor alpha (TNFα) and the signal transducer and activator of transcription 3 (STAT3). Isolated perfused male mouse (wild type, TNFα receptor 2 knockout mice, and cardiomyocyte-specific STAT3-deficient mice) or rat hearts (Wistars) were subjected to ischemia-reperfusion. Resveratrol (2.3 mg/L) or melatonin (75 ng/L) was perfused for 15 min with a 10-min washout period prior to an ischemia-reperfusion insult. Infarct size was measured at the end of the protocol, and Western blot analysis was performed to evaluate STAT3 activation prior to the ischemic insult. Both resveratrol and melatonin, at concentrations found in red wine, significantly reduced infarct size compared with control hearts in wild-type mouse hearts (25 ± 3% and 25 ± 3% respectively versus control 69 ± 3%, P < 0.001) but failed to protect in TNF receptor 2 knockout or STAT3-deficient mice. Furthermore, perfusion with either melatonin or resveratrol increased STAT3 phosphorylation prior to ischemia by 79% and 50%, respectively (P < 0.001 versus control). Our data demonstrate that both melatonin and resveratrol, as found in red wine, protect the heart in an experimental model of myocardial infarction via the SAFE pathway., (© 2011 John Wiley & Sons A/S.)
- Published
- 2011
- Full Text
- View/download PDF
163. Reperfusion injury salvage kinase and survivor activating factor enhancement prosurvival signaling pathways in ischemic postconditioning: two sides of the same coin.
- Author
-
Hausenloy DJ, Lecour S, and Yellon DM
- Subjects
- Animals, Humans, Mitochondrial Membrane Transport Proteins metabolism, Myocardial Reperfusion Injury prevention & control, STAT3 Transcription Factor metabolism, Tumor Necrosis Factor-alpha, Ischemic Postconditioning, Myocardial Reperfusion Injury metabolism, Phosphotransferases metabolism, Signal Transduction
- Abstract
The discovery of ischemic postconditioning (IPost) has rejuvenated the field of cardioprotection. As an interventional strategy to be applied at the onset of myocardial reperfusion, the transition of IPost from a bench-side curiosity to potential clinical therapy has been impressively rapid. Its existence also confirms the existence of lethal myocardial reperfusion injury in man, suggesting that 40%-50% of the final reperfused myocardial infarct may actually be due to myocardial reperfusion injury. Intensive analysis of the signal transduction pathways underlying IPost has identified similarities with the signaling pathways underlying its preischemic counterpart, ischemic preconditioning. In this article, the reperfusion injury salvage kinase pathway and the more recently described survivor activating factor enhancement pathway, two apparently distinct signaling pathways that actually interact to convey the IPost stimulus from the cell surface to the mitochondria, where many of the prosurvival and death signals appear to converge. The elucidation of the reperfusion signaling pathways underlying IPost may result in the identification of novel pharmacological targets for cardioprotection.
- Published
- 2011
- Full Text
- View/download PDF
164. Effects of sphingosine-1-phosphate on acute contractile heart failure (ACHF).
- Author
-
Deshpande GP, McCarthy J, Mardikar H, Lecour S, and Opie L
- Subjects
- Animals, Heart Failure physiopathology, Heart Rate drug effects, Heart Ventricles drug effects, Myocardial Contraction drug effects, Myocardial Reperfusion Injury drug therapy, Rats, Signal Transduction drug effects, Sphingosine pharmacology, Heart Failure drug therapy, Lysophospholipids pharmacology, Sphingosine analogs & derivatives
- Published
- 2010
- Full Text
- View/download PDF
165. Ethanolamine is a novel STAT-3 dependent cardioprotective agent.
- Author
-
Kelly RF, Lamont KT, Somers S, Hacking D, Lacerda L, Thomas P, Opie LH, and Lecour S
- Subjects
- Amidohydrolases antagonists & inhibitors, Amidohydrolases metabolism, Animals, Benzamides pharmacology, Carbamates pharmacology, Disease Models, Animal, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Janus Kinases antagonists & inhibitors, Janus Kinases metabolism, Lysophospholipids metabolism, Male, Mice, Mice, Knockout, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury pathology, Myocardium pathology, Phosphorylation, Rats, Rats, Wistar, STAT3 Transcription Factor deficiency, STAT3 Transcription Factor genetics, Sphingosine analogs & derivatives, Sphingosine metabolism, Tyrphostins pharmacology, Cardiovascular Agents pharmacology, Ethanolamine pharmacology, Myocardial Infarction prevention & control, Myocardial Reperfusion Injury prevention & control, Myocardium metabolism, STAT3 Transcription Factor metabolism
- Abstract
Ethanolamine is a biogenic amine found naturally in the body as part of membrane lipids and as a metabolite of the cardioprotective substances, sphingosine-1-phosphate (S1P) and anandamide. In the brain, ethanolamine, formed from the breakdown of anandamide protects against ischaemic apoptosis. However, the effects of ethanolamine in the heart are unknown. Signal transducer and activator of transcription 3 (STAT-3) is a critical prosurvival factor in ischaemia/reperfusion (I/R) injury. Therefore, we investigated whether ethanolamine protects the heart via activation of STAT-3. Isolated hearts from wildtype or cardiomyocyte specific STAT-3 knockout (K/O) mice were pre-treated with ethanolamine (Etn) (0.3 mmol/L) before I/R insult. In vivo rat hearts were subjected to 30 min ischaemia/2 h reperfusion in the presence or absence of 5 mg/kg S1P and/or the FAAH inhibitor, URB597. Infarct size was measured at the end of each protocol by triphenyltetrazolium chloride staining. Pre-treatment with ethanolamine decreased infarct size in isolated mouse or rat hearts subjected to I/R but this infarct sparing effect was lost in cardiomyocyte specific STAT-3 deficient mice. Pre-treatment with ethanolamine increased nuclear phosphorylated STAT-3 [control 0.75 ± 0.08 vs. Etn 1.50 ± 0.09 arbitrary units; P < 0.05]. Our findings suggest a novel cardioprotective role for ethanolamine against I/R injury via activation of STAT-3.
- Published
- 2010
- Full Text
- View/download PDF
166. Translating novel strategies for cardioprotection: the Hatter Workshop Recommendations.
- Author
-
Hausenloy DJ, Baxter G, Bell R, Bøtker HE, Davidson SM, Downey J, Heusch G, Kitakaze M, Lecour S, Mentzer R, Mocanu MM, Ovize M, Schulz R, Shannon R, Walker M, Walkinshaw G, and Yellon DM
- Subjects
- Age Factors, Animals, Disease Models, Animal, Humans, Sex Factors, Species Specificity, Treatment Outcome, Cardiovascular Agents therapeutic use, Ischemic Postconditioning, Ischemic Preconditioning, Myocardial, Myocardial Infarction prevention & control, Myocardial Ischemia therapy, Myocardial Reperfusion Injury prevention & control, Translational Research, Biomedical
- Abstract
Ischemic heart disease (IHD) is the leading cause of death worldwide. Novel cardioprotective strategies are therefore required to improve clinical outcomes in patients with IHD. Although a large number of novel cardioprotective strategies have been discovered in the research laboratory, their translation to the clinical setting has been largely disappointing. The reason for this failure can be attributed to a number of factors including the inadequacy of the animal ischemia-reperfusion injury models used in the preclinical cardioprotection studies and the inappropriate design and execution of the clinical cardioprotection studies. This important issue was the main topic of discussion of the UCL-Hatter Cardiovascular Institute 6th International Cardioprotection Workshop, the outcome of which has been published in this article as the "Hatter Workshop Recommendations". These have been proposed to provide guidance on the design and execution of both preclinical and clinical cardioprotection studies in order to facilitate the translation of future novel cardioprotective strategies for patient benefit.
- Published
- 2010
- Full Text
- View/download PDF
167. TNFα protects cardiac mitochondria independently of its cell surface receptors.
- Author
-
Lacerda L, McCarthy J, Mungly SF, Lynn EG, Sack MN, Opie LH, and Lecour S
- Subjects
- Adenosine Triphosphate metabolism, Animals, Antioxidants pharmacology, Cell Hypoxia, Cell Respiration, Enzyme Inhibitors pharmacology, Male, Membrane Potential, Mitochondrial, Mice, Mice, 129 Strain, Mice, Knockout, Mitochondria, Heart drug effects, Mitochondrial Membrane Transport Proteins metabolism, Mitochondrial Permeability Transition Pore, Myocytes, Cardiac metabolism, Reactive Oxygen Species metabolism, Receptors, Tumor Necrosis Factor, Type I deficiency, Receptors, Tumor Necrosis Factor, Type I genetics, Receptors, Tumor Necrosis Factor, Type II deficiency, Receptors, Tumor Necrosis Factor, Type II genetics, Sphingolipids metabolism, Mitochondria, Heart metabolism, Receptors, Tumor Necrosis Factor, Type I metabolism, Receptors, Tumor Necrosis Factor, Type II metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
Our novel proposal is that TNFα exerts a direct effect on mitochondrial respiratory function in the heart, independently of its cell surface receptors. TNFα-induced cardioprotection is known to involve reactive oxygen species (ROS) and sphingolipids. We therefore further propose that this direct mitochondrial effect is mediated via ROS and sphingolipids. The protective concentration of TNFα (0.5 ng/ml) was added to isolated heart mitochondria from black 6 × 129 mice (WT) and double TNF receptor knockout mice (TNFR1&2(-/-)). Respiratory parameters and inner mitochondrial membrane potential were analyzed in the presence/absence of two antioxidants, N-acetyl-L: -cysteine or N-tert-butyl-α-(2-sulfophenyl)nitrone or two antagonists of the sphingolipid pathway, N-oleoylethanolamine (NOE) or imipramine. In WT, TNFα reduced State 3 respiration from 279.3 ± 3 to 119.3 ± 2 (nmol O₂/mg protein/min), increased proton leak from 15.7 ± 0.6% (control) to 36.6 ± 4.4%, and decreased membrane potential by 20.5 ± 3.1% compared to control groups. In TNFR1&2(-/-) mice, TNFα reduced State 3 respiration from 205.2 ± 4 to 75.7 ± 1 (p < 0.05 vs. respective control). In WT mice, both antioxidants added with TNFα restored State 3 respiration to 269.2 ± 2 and 257.6 ± 2, respectively. Imipramine and NOE also restored State 3 respiration to 248.4 ± 2 and 249.0 ± 2, respectively (p < 0.01 vs. TNFα alone). Similarly, both antioxidant and inhibitors of the sphingolipid pathway restored the proton leak to pre-TNF values. TNFα-treated mitochondria or isolated cardiac muscle fibers showed an increase in respiration after anoxia-reoxygenation, but this effect was lost in the presence of an antioxidant or NOE. Similar data were obtained in TNFR1&2(-/-) mice. TNFα exerts a protective effect on respiratory function in isolated mitochondria subjected to an anoxia-reoxygenation insult. This effect appears to be independent of its cell surface receptors, but is likely to be mediated by ROS and sphingolipids.
- Published
- 2010
- Full Text
- View/download PDF
168. Ischaemic postconditioning protects against reperfusion injury via the SAFE pathway.
- Author
-
Lacerda L, Somers S, Opie LH, and Lecour S
- Subjects
- Androstadienes pharmacology, Animals, Disease Models, Animal, Extracellular Signal-Regulated MAP Kinases antagonists & inhibitors, Extracellular Signal-Regulated MAP Kinases metabolism, Flavonoids pharmacology, Glycogen Synthase Kinase 3 metabolism, Glycogen Synthase Kinase 3 beta, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury pathology, Myocardium pathology, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Phosphorylation, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Receptors, Tumor Necrosis Factor, Type I genetics, Receptors, Tumor Necrosis Factor, Type I metabolism, Receptors, Tumor Necrosis Factor, Type II deficiency, Receptors, Tumor Necrosis Factor, Type II genetics, STAT3 Transcription Factor antagonists & inhibitors, STAT3 Transcription Factor deficiency, STAT3 Transcription Factor genetics, Tumor Necrosis Factor-alpha deficiency, Tumor Necrosis Factor-alpha genetics, Tyrphostins pharmacology, Wortmannin, Myocardial Infarction prevention & control, Myocardial Reperfusion Injury prevention & control, Myocardium metabolism, Receptors, Tumor Necrosis Factor, Type II metabolism, STAT3 Transcription Factor metabolism, Signal Transduction drug effects, Tumor Necrosis Factor-alpha metabolism
- Abstract
Aims: Ischaemic postconditioning (IPostC) is a powerful protective phenomenon that activates prosurvival intrinsic signalling cascades to limit reperfusion injury. We propose that IPostC confers its infarct-sparing effect via activation of the newly described prosurvival Survivor Activating Factor Enhancement (SAFE) pathway, which involves the activation of the cytokine tumour necrosis factor alpha (TNFalpha) and signal transducer and activator of transcription-3 (STAT-3)., Methods and Results: Isolated ischaemic/reperfused hearts from TNF knockout, TNF receptor-1 knockout, TNF receptor-2 knockout, cardiomyocyte-specific STAT-3-deficient mice or their respective wild-type, (TNF-WT) or (STAT-3-WT), were postconditioned by ischaemic episodes (IPostC) or with exogenous TNFalpha (0.5 microg/L) (TNF-PostC) at the onset of reperfusion. IPostC reduced infarct size (IS) in TNF-WT and TNFR1(-/-) hearts (by 33 and 27%, respectively, P < 0.05), whereas hearts from TNF(-/-) or TNFR2(-/-) failed to be postconditioned. TNF-PostC reduced IS by 37% (P < 0.05) in STAT-3-WT hearts but failed to protect cardiac-specific STAT-3(-/-) hearts. Administration of wortmannin, an inhibitor of PI-3 kinase/Akt, or PD98059, an inhibitor of extracellular regulated kinase 1/2 (Erk1/2), during the postconditioning stimulus did not abolish the infarct-sparing effect of TNF-PostC. AG490, an inhibitor of STAT-3, abrogated the protective effect of TNFalpha. Western blot analysis did not demonstrate the involvement of Akt or Erk1/2 in TNF-PostC, whereas STAT-3 phosphorylation was increased in both IPostC and TNF-PostC., Conclusion: The protective effect of the SAFE pathway is shown in IPostC, with the activation of TNFalpha, its receptor type 2, and STAT-3. This signalling cascade is activated independently of the well-known Reperfusion Injury Salvage Kinases (RISK) pathway, which involves the kinases Akt and Erk1/2.
- Published
- 2009
- Full Text
- View/download PDF
169. Activation of the protective Survivor Activating Factor Enhancement (SAFE) pathway against reperfusion injury: Does it go beyond the RISK pathway?
- Author
-
Lecour S
- Subjects
- Animals, Humans, Models, Biological, STAT3 Transcription Factor metabolism, Tumor Necrosis Factor-alpha metabolism, Tumor Necrosis Factor-alpha physiology, Myocardial Reperfusion Injury metabolism, Signal Transduction physiology
- Abstract
Lethal reperfusion injury is now recognized as a major limitation of current reperfusion therapy by primary percutaneous coronary intervention for acute myocardial infarction. Interestingly, the heart itself is capable of activating an intrinsic protective signaling programme to limit cell death during reperfusion. Tumor necrosis factor alpha (TNFalpha) is a cytokine generally thought to contribute to myocardial dysfunction in ischemia/reperfusion or heart failure. We review evidence that TNFalpha can paradoxically initiate the activation of a novel protective pathway against reperfusion injuries that we have named the Survivor Activating Factor Enhancement (SAFE) pathway. This path requires the activation of the signal transducer and activator of transcription 3 (STAT-3) and it can successfully lessen cardiomyocyte death at the time of reperfusion, independently of the activation of the already well-described Reperfusion Injury Salvage Kinase (RISK) pathway (which includes activation of Akt and Erk 1/2). Emerging knowledge on this novel protective path is presented here with the aim of unravelling its interaction with the RISK pathway and its potential human application to protect against lethal reperfusion injury.
- Published
- 2009
- Full Text
- View/download PDF
170. Multiple protective pathways against reperfusion injury: a SAFE path without Aktion?
- Author
-
Lecour S
- Subjects
- Animals, Cannabinoids metabolism, Clinical Trials as Topic, Enzyme Activation, Humans, Reperfusion Injury therapy, Proto-Oncogene Proteins c-akt metabolism, Reperfusion Injury enzymology, Reperfusion Injury prevention & control, Signal Transduction
- Published
- 2009
- Full Text
- View/download PDF
171. Signal transducer and activator of transcription 3 is involved in the cardioprotective signalling pathway activated by insulin therapy at reperfusion.
- Author
-
Fuglesteg BN, Suleman N, Tiron C, Kanhema T, Lacerda L, Andreasen TV, Sack MN, Jonassen AK, Mjøs OD, Opie LH, and Lecour S
- Subjects
- Animals, Cell Death drug effects, Cell Survival drug effects, Disease Models, Animal, Enzyme Inhibitors pharmacology, Male, Mice, Mice, Mutant Strains, Myocardial Reperfusion Injury pathology, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, Oxygen pharmacology, Phosphorylation, Proto-Oncogene Proteins c-akt metabolism, Rats, Rats, Wistar, STAT3 Transcription Factor antagonists & inhibitors, STAT3 Transcription Factor genetics, Tyrphostins pharmacology, Cardiotonic Agents pharmacology, Insulin pharmacology, Myocardial Reperfusion Injury drug therapy, Myocardial Reperfusion Injury metabolism, STAT3 Transcription Factor metabolism, Signal Transduction physiology
- Abstract
Objective: To evaluate the significance of the JAK-STAT pathway in insulin-induced cardioprotection from reperfusion injury., Methods: In isolated perfused rat hearts subjected to insulin therapy (0.3 mU/ml) +/- AG490 (5 microM, JAK-STAT inhibitor), the phosphorylation state of STAT3 and Akt was determined after 15 min of reperfusion. Infarct size was measured after 120 min of reperfusion. Isolated cardiac myocytes from wild type (WT) and cardiac specific STAT3 deficient mice were treated with insulin at reoxygenation following simulated ischemia (SI, 26 h). Cell viability was measured after 120 min of reoxygenation following SI, whereas phosphorylation state of Akt was measured after 15 min of reoxygenation following SI., Results: Insulin given at reperfusion led to phosphorylation of STAT3 and Akt both of which were inhibited by AG490. AG490 also blocked the insulin-dependent decrease in infarct size, supporting a role for JAK-STAT in cardioprotection. In addition, insulin protection from SI was blocked in myocytes from the STAT3 deficient mice, or in WT mice treated with AG490. Furthermore, insulin failed to phosphorylate Akt in the STAT3 deficient cardiomyocytes., Conclusion: Insulin-induced cardioprotection at reperfusion occurs through activation of STAT3. Inhibiting STAT3 by AG490, or STAT3 depletion in cardiac myocytes affects activation of Akt, suggesting close interaction between STAT3 and Akt in the cardioprotective signalling pathway activated by insulin treatment at reperfusion.
- Published
- 2008
- Full Text
- View/download PDF
172. TNFalpha is required to confer protection in an in vivo model of classical ischaemic preconditioning.
- Author
-
Deuchar GA, Opie LH, and Lecour S
- Subjects
- Animals, Dose-Response Relationship, Drug, Male, Myocardial Infarction drug therapy, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury pathology, Rats, Rats, Wistar, Time Factors, Tumor Necrosis Factor-alpha antagonists & inhibitors, Cardiotonic Agents metabolism, Cardiotonic Agents pharmacology, Ischemic Preconditioning, Myocardial, Myocardial Reperfusion Injury drug therapy, Receptors, Tumor Necrosis Factor, Tumor Necrosis Factor-alpha metabolism, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Although Tumor Necrosis Factor alpha (TNFalpha) is used as a preconditioning mimetic in vitro, its role in ischaemic preconditioning (IPC) has not been clearly defined. Here, we propose to use an in vivo model (that takes into account the activation of leukocytes which may affect levels of TNFalpha) to demonstrate that i) TNFalpha acts as a trigger in IPC and ii) the dose-dependent nature of this cardioprotective effect of TNFalpha. Male Wistar rats were subjected to 30 min of left coronary artery occlusion (index ischaemia), followed by 24 h reperfusion. In the presence or absence of a soluble TNFalpha receptor (sTNFalpha-R), preconditioning was induced by 3 cycles of ischaemia (3 min)/reperfusion (5 min) (IPC) or various doses (0.05-4 microg/kg) of exogenous TNFalpha. Following 24 h reperfusion, infarct size (IS, expressed as % of the area at risk (AAR)) was assessed. Tissue levels of TNFalpha from the AAR, following IPC and TNFalpha stimulus were determined using Western Blot. IPC caused decrease in IS (4.5+/-1.3% vs 30.8+/-4.3% in ischaemic rats; P<0.001) and increase of TNFalpha levels following the IPC stimulus. The protective effect of IPC was abrogated in the presence of the sTNFalpha-R. In addition, exogenous TNFalpha dose-dependently reduced IS with maximal protection at a dose of 0.1 microg/kg (IS=12.6%, P<0.01 vs ischaemic). In conclusion our data provide strong evidence for a role of TNFalpha during the trigger phase of IPC. In addition, exogenous TNFalpha mimics IPC by providing a dose-dependent cardioprotective effect against ischaemia-reperfusion injury in vivo.
- Published
- 2007
- Full Text
- View/download PDF
173. TNFalpha-induced cytoprotection requires the production of free radicals within mitochondria in C2C12 myotubes.
- Author
-
Lacerda L, Smith RM, Opie L, and Lecour S
- Subjects
- Cell Hypoxia drug effects, Cell Survival drug effects, Cells, Cultured, Flow Cytometry methods, Fluoresceins pharmacology, Glycine analogs & derivatives, Glycine pharmacology, Humans, Ischemic Preconditioning, Membrane Potentials drug effects, Mitochondrial Membranes metabolism, Muscle Fibers, Skeletal cytology, Myoblasts cytology, Myoblasts metabolism, Sulfhydryl Compounds pharmacology, Time Factors, Cytoprotection drug effects, Mitochondria, Muscle metabolism, Muscle Fibers, Skeletal metabolism, Reactive Oxygen Species metabolism, Tumor Necrosis Factor-alpha pharmacology
- Abstract
We previously reported that tumour necrosis factor alpha (TNFalpha) can mimic classic ischemic preconditioning (IPC) in both cells and heart. However, the signalling pathways involved remain incompletely understood. One potential protective pathway could be TNFalpha-induced reactive oxygen species (ROS). We hypothesized that TNFalpha cytoprotection occurs through the generation of ROS which originate within the mitochondria. C(2)C(12) myotubes were preconditioned with either a short period of hypoxia (IPC) or a low concentration of TNFalpha (0.5 ng/ml) prior to a simulated ischemic insult. ROS generation was evaluated on cells stained with dichlorofluorescin diacetate (DCFH-DA) by flow cytometry. The source of TNFalpha-induced ROS was examined with Mitotracker Red CM-H(2)XRos. The bioenergetics of the mitochondria were evaluated by investigation of the respiratory parameters and the inner mitochondrial membrane potential. Pretreatment with TNFalpha improved cell viability compared with the simulated ischemic control (TNFalpha: 75 +/- 1% versus 34 +/- 1% for the control: p<0.001). The ROS scavenger, N-2-mercaptopropionyl-glycine (MPG), reduced the viability of TNFalpha-stimulated cells to 15 +/- 1% (p<0.001 versus TNFalpha). Similar results were obtained with IPC. TNFalpha stimulation increased ROS production mainly in the mitochondria, and this increase was abolished in the presence of MPG. Addition of TNFalpha to the cells increased State 2 respiration and modestly depolarised the membrane potential prior to the ischemic insult. In conclusion, TNFalpha-induced ROS generation can occur within the mitochondria, resulting in temporal mitochondrial perturbations which may initiate the cytoprotective effect of TNFalpha.
- Published
- 2006
- Full Text
- View/download PDF
174. Ceramide-induced preconditioning involves reactive oxygen species.
- Author
-
Lecour S, Owira P, and Opie LH
- Subjects
- Animals, Antioxidants pharmacology, Catalase metabolism, In Vitro Techniques, Male, Myocardial Infarction enzymology, Myocardial Infarction metabolism, Rats, Rats, Long-Evans, Sphingosine pharmacology, Superoxide Dismutase metabolism, Tiopronin pharmacology, Ischemic Preconditioning, Myocardial, Myocardial Infarction prevention & control, Reactive Oxygen Species metabolism, Sphingosine analogs & derivatives
- Abstract
Introduction: Ceramide induces programmed cell death and it is thought to contribute to cardiac ischemia/reperfusion (I/R) injury. In contrast, we have demonstrated that administration of low doses of ceramide engenders cardiac preconditioning (PC). Ceramide is known to generate reactive oxygen species (ROS) in cells. Since mechanisms triggering the ceramide-induced cardioprotection remain unknown, we investigated the role of ROS in the genesis of this protective mechanism., Methods: Using an isolated Langendorff-perfused rat heart model, four groups (n > or = 6 in each group) were considered: Control hearts underwent 30 min index regional ischemia and 120 min of reperfusion. In the ceramide group, hearts were preconditioned with c2-ceramide 1 microM for 7 min followed by 10 min washout prior to the I/R insult. In additional groups, MPG (1 mM), a synthetic antioxidant was given for 15 min alone or bracketing the ceramide perfusion. In each group, infarct size was determined at the end of the reperfusion period and superoxide dismutases (CuZnSOD and MnSOD) and catalase activities were evaluated., Results: Ceramide preconditioning reduced the infarct/area at risk (I/AAR) ratio (8.3 +/- 1.1% for ceramide vs. 36.4 +/- 1.2% for control, p < 0.001). Perfusion with MPG abolished the preconditioning effect of ceramide (I/AAR ratio = 36.7 +/- 4.9%). Ceramide was also associated with a 29% and 38% increase in catalase and CuZnSOD activities, respectively, compared with control group., Conclusion: Production of reactive oxygen species following ceramide preconditioning of the ischemic-reperfused heart appears to play a role in the cardioprotective effect of ceramide.
- Published
- 2006
- Full Text
- View/download PDF
175. Ceramide attenuates hypoxic cell death via reactive oxygen species signaling.
- Author
-
Lecour S, Van der Merwe E, Opie LH, and Sack MN
- Subjects
- Animals, Antioxidants pharmacology, Cell Line, Cell Survival drug effects, Cyclooxygenase 2 physiology, Mitochondria physiology, NADPH Oxidases physiology, Rats, Ceramides pharmacology, Cytoprotection drug effects, Hypoxia pathology, Reactive Oxygen Species metabolism, Signal Transduction physiology
- Abstract
We have previously demonstrated that tumor necrosis factor alpha (TNFalpha), a cytokine known to be induced by ischemia, independently promotes preconditioning in part via ceramide generation. As reactive oxygen species (ROS) signaling is evoked by ischemic preconditioning, by TNFalpha and by ceramide we reasoned that ceramide-induced preconditioning is ROS-mediated. Fibroblastic L-cells were subjected to 8 hours simulated ischemia and were preconditioned by pretreatment with cell permeable c2 ceramide (1 microM) with or without the antioxidant N-mercaptopropionyl glycine (MPG; 1 mM). Pretreatment with ceramide reduced lactate dehydrogenase release at the end of the simulated ischemia but this cytoprotective effect was lost in the presence of MPG. Concurrent temporal ROS generation was measured using confocal microscopy on cells stained with dichlorofluorescein diacetate (DCF-DA). Ceramide increased ROS production after 30 minutes and this induction was decreased by MPG. Incubation of ceramide with cyclooxygenase-2 inhibitor, NS 398 (10 microM), or with a mitochondrial respiratory chain inhibitor, rotenone (10 microM) reduced the cytoprotective effect of ceramide in parallel with a partial diminution in ROS generation. In contrast, inhibition of other ROS-producing systems including nitric oxide synthase, xanthine oxidase, or NADPH oxidase failed to modulate ceramide-induced cytoprotection. Collectively, these data demonstrate that ceramide induces a cell survival program through ROS signaling activated, in part, via cyclooxygenase and the mitochondrial respiratory chain.
- Published
- 2006
- Full Text
- View/download PDF
176. Free radicals trigger TNF alpha-induced cardioprotection.
- Author
-
Lecour S, Rochette L, and Opie L
- Subjects
- Animals, Antioxidants pharmacology, Electron Spin Resonance Spectroscopy, Free Radicals analysis, Glycine pharmacology, Male, Myocardial Ischemia metabolism, Perfusion, Rats, Rats, Inbred Lew, Sulfhydryl Compounds pharmacology, Tumor Necrosis Factor-alpha metabolism, Free Radicals metabolism, Glycine analogs & derivatives, Ischemic Preconditioning, Myocardial, Myocardial Reperfusion Injury prevention & control, Myocardium metabolism, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Objective: Tumor Necrosis Factor alpha (TNF alpha) induces programmed cell death and contributes to cardiac ischemia/reperfusion injury. Paradoxically, we have recently demonstrated that low doses of TNF alpha can induce cardiac preconditioning (PC). We hypothesized that the production of free radicals participates in this cardioprotective program., Methods: Control isolated rat hearts underwent 30 min regional ischemia and 120 min of reperfusion. A second group of hearts received a low dose of TNF alpha (0.5 ng/ml) for 7 min followed by 10 min washout prior to I/R. In other groups, the antioxidant N-2-mercaptopropionyl glycine (MPG) (1 mM) was given for 15 min prior to I/R alone or during TNF alpha perfusion. Infarct size was determined at the end of the reperfusion period. Ventricular catalase and superoxide dismutase activities were assessed as an index of oxidative stress and free radical production was directly measured by the oxidation of 1-hydroxy-3-carboxy-pyrrolidine (CP-H) to paramagnetic 3-carboxy-proxyl (CP.) using electron spin resonance spectroscopy., Results: TNF alpha reduced the infarct/area at risk (I/AAR) ratio (7.2+/-1.7% vs. 36.5+/-1.7% for controls, p<0.05). MPG reduced the cardioprotective effect of TNF alpha (I/AAR ratio: 20.5+/-3.3%, p<0.05). TNF alpha-perfusion increased catalase activity in the ventricles (15.8+/-1.2 I.U./mg for controls vs. 19.9+/-1.1 I.U/mg for TNF alpha, p<0.05). Proof of formation of free radicals was increased CP formation in the coronary effluent during TNF alpha infusion (24.2+/-4.5 for TNF alpha vs. 11.9+/-1.5 arbitrary units for controls, p<0.05), with decreased CP after addition of MPG., Conclusions: Our data provide firm evidence for a production and role of free radicals in TNF alpha-induced cardioprotection.
- Published
- 2005
- Full Text
- View/download PDF
177. Direct demonstration of nitric oxide formation in organs of rabbits treated by transdermal glyceryl trinitrate using an in vivo spin trapping technique.
- Author
-
Clermont G, Lecour S, Vergely C, Zeller M, Perrin C, Maupoil V, Bouchot O, and Rochette L
- Subjects
- Animals, Aorta, Thoracic drug effects, Aorta, Thoracic metabolism, Blood Pressure drug effects, Blood Pressure physiology, Dosage Forms, Ferrous Compounds chemistry, Heart Rate drug effects, Heart Rate physiology, Heart Ventricles chemistry, Heart Ventricles drug effects, Heart Ventricles metabolism, Hemoglobins chemistry, Injections, Intravenous, Kidney chemistry, Kidney drug effects, Kidney metabolism, Liver chemistry, Liver drug effects, Liver metabolism, Nitrates blood, Nitric Oxide pharmacokinetics, Nitrites blood, Nitroglycerin administration & dosage, Nitroglycerin metabolism, Organ Specificity drug effects, Organ Specificity physiology, Rabbits, Sorbitol agonists, Sorbitol metabolism, Sorbitol pharmacology, Spin Labels, Spleen chemistry, Spleen drug effects, Spleen metabolism, Thiocarbamates agonists, Thiocarbamates metabolism, Thiocarbamates pharmacology, Time Factors, Vasodilator Agents administration & dosage, Vasodilator Agents metabolism, Vasodilator Agents pharmacokinetics, Vena Cava, Inferior drug effects, Vena Cava, Inferior metabolism, Administration, Cutaneous, Nitric Oxide biosynthesis, Nitric Oxide blood, Nitroglycerin pharmacokinetics, Sorbitol analogs & derivatives, Spin Trapping methods
- Abstract
Glyceryl trinitrate (GTN) is commonly delivered by a patch for the treatment of angina pectoris. The idea is now generally accepted that GTN requires a biotransformation process that activates the drug, in particular through nitric oxide (NO) generation. However, the pharmacokinetics of NO delivery from GTN still remains obscure. The objective of this study was to assess GTN-derived NO formation in vascular tissues and organs in rabbit given GTN patches. NO levels were evaluated in rabbits after 3 h of treatment with a 10 mg GTN patch (GTN group; n = 7) or a placebo patch (CTL; n = 7). Nitrosylhaemoglobin (HbNO) was evaluated by electron spin resonance (ESR) spectroscopy in red cell suspension. In vivo spin trapping technique using FeMGD as a spin trap, associated with ESR was used to quantify NO in tissues. The NO-spin trap complex, which is a relatively stable product, has been measured in several tissues. The ESR spectrum corresponding to HbNO was not found in red cell of GTN or CTL rabbits. The spectrum corresponding to the NO-spin trap complex was observed in all analysed tissues of CTL rabbits. The signal was significantly increased in liver, renal medulla, heart left ventricle and spleen of GTN-treated rabbits, and to a lesser extent in right ventricle and lung. No difference was shown between NO-spin trap levels measured in aorta or inferior vena cava from GTN or CTL rabbits. These data suggest that GTN patch treatment induced NO release, and that tissue-specific differences in transdermal GTN-derived NO exist. The GTN-NO pathway appears to be largely involved in organs such as the liver, kidney and heart.
- Published
- 2003
- Full Text
- View/download PDF
178. [Sphingolipid signaling: a potential pathway for TNF-alpha induced preconditioning].
- Author
-
Lecour S, Sack MN, and Opie LH
- Subjects
- Ceramides metabolism, Humans, Myocardial Ischemia metabolism, Myocardial Ischemia prevention & control, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury prevention & control, Ischemic Preconditioning, Myocardial, Signal Transduction physiology, Sphingolipids metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
Although cytokine activation has long been recognized to associate with cardiac ischemia and reperfusion, the concept that these cytokines may enhance some cardioprotective mechanisms has only recently been considered. Ischemic preconditioning is a biologic phenomenon that activates innate cytoprotective programs in the heart. Ischemic preconditioning has been described where a transient non-lethal ischemic "trigger" or endogenous molecules produced/released by ischemia enables the tissue to become more resistant/tolerant to subsequent potentially lethal ischemia. The mechanisms and signalling events involved in this cytoprotective program still remain obscure. Recently, it has been suggested that cytokine activation including tumour necrosis factor (TNF alpha) may play a key role in the preconditioned phenotype. Moreover, new studies have given the evidence that the exploration of cytokine-activated sphingolipid signalling pathways may enhance our understanding of the preconditioning program.
- Published
- 2003
179. Evidence for the extrapulmonary localization of inhaled nitric oxide.
- Author
-
Lecour S, Clermont G, du Toit E, Gilson L, Maupoil V, Lowe S, Dupuis P, Girard C, and Rochette L
- Subjects
- Administration, Inhalation, Animals, Aorta metabolism, Biomarkers blood, Blood Pressure drug effects, Cyclic GMP blood, Dose-Response Relationship, Drug, Injections, Intravenous, Kidney metabolism, Liver metabolism, Lung metabolism, Models, Animal, Models, Cardiovascular, Nitrates blood, Nitric Oxide metabolism, Nitrites blood, Rats, Rats, Wistar, Sorbitol administration & dosage, Sorbitol metabolism, Spin Labels, Thiocarbamates administration & dosage, Thiocarbamates metabolism, Time Factors, Nitric Oxide administration & dosage, Sorbitol analogs & derivatives
- Abstract
Inhaled nitric oxide (NO) has emerged as a promising pulmonary vasodilator to treat pulmonary hypertension associated with heart disease and ventilation/perfusion mismatching. However, the pharmacokinetics of inhaled NO still remains obscure and its cardiopulmonary selectivity appears to be increasingly under debate. In the present study measured NO content and levels of cyclic guanosine 3',5'monophosphate (cGMP), a mediator of NO-induced vasodilation, in a variety of organs from rats subjected to NO inhalation. Electron spin resonance spectroscopy associated to a spin trapping technique using N-methyl D-glucamine dithiocarbamate (FeMGD) was used to directly quantify NO levels in the lung, kidney, liver, aorta, and heart from anesthetized Wistar rats subjected to various doses (0, 20, 50, 100, or 200 ppm) and various times (0, 30, 45, or 75 minutes) of inhaled NO. Inhaled NO at a dose of 100 and 200 ppm significantly increased the NO-FeMGD complex in all organs studied. An increase of cGMP was detected in the lung and the aorta after inhaled NO for 45 minutes at the dose of 50 ppm. No changes in NO levels and its metabolites were shown between 30 and 75 minutes of inhaled NO. The results show that inhaled NO at a dose of 100 ppm or more increases NO levels in other organs beside the lung, strongly suggesting that inhaled NO would be more than a pulmonary vasodilator and its selectivity remains to be reconsidered when used for therapeutic purposes.
- Published
- 2003
- Full Text
- View/download PDF
180. Innate immunity and cardiac preconditioning: a putative intrinsic cardioprotective program.
- Author
-
Smith RM, Lecour S, and Sack MN
- Subjects
- Animals, Humans, Immune Tolerance, Lipopolysaccharides pharmacology, Membrane Glycoproteins metabolism, Models, Animal, Receptors, Cell Surface metabolism, Time Factors, Toll-Like Receptors, Cytokines physiology, Drosophila Proteins, Ischemic Preconditioning, Myocardial, Myocardial Ischemia immunology, Myocardium immunology, Signal Transduction physiology
- Abstract
Ischemic preconditioning is thought to evoke cell survival programs in the heart in large part via the activation of G(I)-protein coupled receptor signal transduction pathways. However, the identification and characterization of G(I)-protein coupled receptor independent pathways would enable researchers to pursue novel cellular events that could direct or promote preconditioning. In this regard recent work has begun to explore the role of the innate immune system in intrinsic cardioprotection against both viral myocarditis and ischemia. Interestingly, cytokines such as TNFalpha, IL-1beta and leukemia inhibitory factor, which are components of innate immunity, have been shown to mimic ischemic preconditioning. Thus as the innate immune system functions via a diverse array of G(I)-protein independent receptors, the study of this immunological system in the heart may provide new insight into mechanisms driving and promoting ischemic preconditioning. We propose that innate immunity is indeed an integral part of ischemic preconditioning. In this review, we provide an overview of the innate immune system, describe the studies whereby cytokines mimic ischemic preconditioning and finally postulate some mechanisms whereby innate immunity may promote cardioprotection as a component of preconditioning.
- Published
- 2002
- Full Text
- View/download PDF
181. Intrarenal detection of nitric oxide using electron spin resonance spectroscopy in hypertensive lipopolysaccharide-treated rats.
- Author
-
Lecour S, Chevet D, Maupoil V, Moisant M, Bernard C, Zahnd JP, Touchard G, Briot F, and Rochette L
- Subjects
- Animals, Electron Spin Resonance Spectroscopy methods, Hypertension chemically induced, Hypertension pathology, Kidney chemistry, Kidney pathology, Kidney ultrastructure, Male, NG-Nitroarginine Methyl Ester administration & dosage, Nitric Oxide analysis, Rats, Rats, Wistar, Sepsis chemically induced, Sepsis metabolism, Sepsis pathology, Hypertension metabolism, Kidney metabolism, Lipopolysaccharides administration & dosage, Nitric Oxide metabolism
- Abstract
The inhibition of nitric oxide (NO) synthesis by chronic administration of NG-nitro-l-arginine methyl ester (l-NAME) in rats is responsible for systemic hypertension. However, the mechanisms involved in this hypertension remain unclear. The effects of chronic l-NAME on kidney and blood NO production were studied in rats in a state of endotoxic shock due to lipopolysaccharide (LPS). A nitric oxide spin trapping technique using electron spin resonance (ESR) spectroscopy has been used to identify and measure the production of NO in the kidney. This method is based on the trapping of nitric oxide by a metal-chelator complex consisting of N-methyl-d-glucamine dithiocarbamate (MGD) and reduced iron (Fe2+) forming a water-soluble NO-FeMGD complex detected by ESR. After LPS injection (14 mg/kg, IV, 6 h before the sacrifice) to rats pretreated with l-NAME (10 mg/kg/d over 14 days), the NO-FeMGD complex was evaluated in the kidney (arbitrary units [AU]/g of kidney) and the density of polynuclear neutrophils was counted by light microscopy. Chronic inhibition of NO synthase by l-NAME, a nonspecific inhibitor, was responsible for a decrease of the NO-FeMGD complex levels in the kidney (24.9 +/- 1.6 AU versus 13.8 +/- 1.3 AU). LPS administration was responsible for a large increase in both NO-FeMGD complex and neutrophil levels in the kidney of normotensive rats (332.6 +/- 12.8 AU versus 24.9 +/- 1.6 AU for NO-FeMGD complex and 1.36 +/- 0.41 versus 0.11 +/- 0.03 for neutrophils). Conversely, LPS administration in hypertensive, l-NAME-pretreated rats was linked to a smaller increase in the NO-FeMGD complex (85.1 +/- 7.9 AU versus 332.6 +/- 12.8 AU) and a larger increase in glomerular neutrophils (2.48 +/- 0.36 versus 1.36 +/- 0.41) compared with normotensive rats. These results are in agreement with a direct implication of NO during LPS-and l-NAME-induced kidney injuries.
- Published
- 2002
- Full Text
- View/download PDF
182. Identification of a novel role for sphingolipid signaling in TNF alpha and ischemic preconditioning mediated cardioprotection.
- Author
-
Lecour S, Smith RM, Woodward B, Opie LH, Rochette L, and Sack MN
- Subjects
- Animals, Male, Rats, Rats, Long-Evans, Tumor Necrosis Factor-alpha metabolism, Ventricular Function, Ischemic Preconditioning, Myocardial, Myocardial Ischemia prevention & control, Signal Transduction physiology, Sphingolipids metabolism, Tumor Necrosis Factor-alpha therapeutic use
- Abstract
TNF alpha administration mimics ischemic preconditioning and neutralizing antibodies to TNF alpha and IL-1 beta abolish exercise-induced preconditioning. However, the pharmacology of TNF alpha's cardioprotective effects and associated downstream signaling events has not been delineated. We evaluated the temporal and dose specific requirements of TNF alpha to function as a preconditioning mimetic. Furthermore we postulated that the preconditioning effect of TNF alpha might be orchestrated via sphingolipid signaling. The cardioprotective effect of TNF alpha and the role of sphingolipid signaling were assessed using a classical preconditioning protocol in the isolated perfused rat heart with the measurement of infarct size and contractile function modulation in response to index ischemia and reperfusion. Recombinant TNF alpha at an optimal dose of 0.5 ng/ml mimicked ischemic preconditioning by reducing infarct size by 60%v non-preconditioned ischemia-reperfusion controls (P<0.01). The infarct sparing effect of TNF alpha required a wash-out period prior to the index ischemic-reperfusion. Moreover, the classic ischemic preconditioning antagonist such as 5-hydroxydecanoate abolished TNF alpha preconditioning. An inhibitor of the sphingolipid signaling pathway, N-oleoylethanolamine (NOE, 1 microm) attenuated ischemic and TNF alpha preconditioning. Likewise, cell-permeable C(2)-ceramide and sphingosine 1-phosphate (sphingolipid signaling intermediates) both reproduced the preconditioning cardioprotective phenotype. Finally, TNF alpha and ceramide conferred preconditioning-like cardioprotection against post-ischemic contractile dysfunction and this cardioprotective effect was attenuated by NOE. In contrast, NOE did not reverse ischemic preconditioning enhanced post-ischemic contractile function. In conclusion, TNF alpha activates preconditioning-like tolerance against infarction and contractile dysfunction. This cardioprotection is mediated, in part, via activation of novel sphingolipid signaling intermediates., (Copyright 2002 Academic Press.)
- Published
- 2002
- Full Text
- View/download PDF
183. Systemic free radical activation is a major event involved in myocardial oxidative stress related to cardiopulmonary bypass.
- Author
-
Clermont G, Vergely C, Jazayeri S, Lahet JJ, Goudeau JJ, Lecour S, David M, Rochette L, and Girard C
- Subjects
- Aged, Electron Spin Resonance Spectroscopy, Female, Free Radicals, Humans, Male, Middle Aged, Myocardial Reperfusion Injury etiology, Uric Acid blood, Cardiopulmonary Bypass adverse effects, Myocardium metabolism, Oxidative Stress
- Abstract
Background: Cardiopulmonary bypass (CPB) can induce deleterious effects that could be triggered in part by radical oxygen species; however, their involvement in the course of surgery has been elusive. The aim of this study was to evaluate the time course and origin of radical oxygen species release, myocardial or not, in patients undergoing coronary artery surgery involving CPB., Methods: Blood samples were taken from periphery and coronary sinus of patients during CPB, and oxidative stress was evaluated by direct and indirect approaches. Direct detection of alkyl and alkoxyl radicals was assessed by electron spin resonance spectroscopy associated with the spin-trapping technique using alpha-phenyl-N-tert-butylnitrone., Results: The authors showed that the spin adduct concentration was not influenced by anesthesia and pre-CPB surgery. A rapid systemic increase of plasma spin adduct concentration occurred after starting CPB, and it stayed at a high concentration until the end of CPB. At the beginning of reperfusion period, radical oxygen species release was accelerated in the coronary sinus; however, it was not significant. A positive correlation was found between alpha-phenyl-N-tert-butylnitrone adduct concentrations and (1) the duration of CPB and (2) concentration of postoperative creatine phosphokinase of muscle band (CPK MB). Plasma vitamin E and C, ascorbyl radical, uric acid, thiol, plasma antioxidant status, and thiobarbituric acid reacting substances were also measured but did not give relevant indications, except for uric acid, which seemed to be consumed by the heart during reperfusion., Conclusion: The results indicate that a systemic production of free radicals occurs during CPB that may overwhelm the production related to reperfusion of the ischemic heart. This systemic oxidative stress is likely to participate in secondary myocardial damage.
- Published
- 2002
- Full Text
- View/download PDF
184. Levels of nitric oxide in the heart after experimental myocardial ischemia.
- Author
-
Lecour S, Maupoil V, Zeller M, Laubriet A, Briot T, and Rochette L
- Subjects
- Animals, Coronary Vessels injuries, Electron Spin Resonance Spectroscopy, Guanidines pharmacology, Hemoglobins metabolism, Ligation, Male, Nitrates blood, Nitric Oxide Synthase antagonists & inhibitors, Nitrites blood, Rats, Rats, Wistar, Spin Labels, Chelating Agents metabolism, Myocardial Ischemia metabolism, Myocardium metabolism, Nitric Oxide metabolism, Sorbitol analogs & derivatives, Sorbitol metabolism, Thiocarbamates metabolism
- Abstract
The effect of myocardial ischemia on nitric oxide (NO) production is controversial in part because of indirect NO quantification. In the present study, direct quantification of NO was investigated in an in vivo rat model of myocardial ischemia (MI). A NO spin-trapping technique using electron spin resonance (ESR) spectroscopy was used to study NO production in the ischemic and in the nonischemic area of the rat heart 2, 8, or 24 h after left main coronary artery ligation. The method was based on the trapping of NO by a metal-chelator complex consisting of N-methyl-D-glucamine-dithiocarbamate (MGD) and Fe(II) to form a stable NO-FeMGD complex that gives rise to a characteristic triplet ESR spectrum. This metal-chelator complex was administered half an hour before sacrifice of the rats. A large and time-dependent increase of the ESR signal corresponding to the NO-FeMGD complex was observed 8 h (11.6 +/- 0.9 arbitrary units [AU]) and 24 h (29.7 +/- 2.9 AU) in the ischemic area after MI. On the contrary, no ESR triplet was observed in the nonischemic region of the heart and in sham-operated rats. NO blood derivative levels (nitrosylhemoglobin and plasma nitrites and nitrates) were unchanged compared with sham-operated rats. Previous administration of aminoguanidine, a NO synthase inhibitor, in animals subjected to a 24-h ischemia resulted in a complete abolition in the NO-FeMGD spectrum in the ischemic area. These findings directly demonstrated an increase of the NO-FeMGD levels during in vivo myocardial ischemia that appeared to be specifically localized in the ischemic area.
- Published
- 2001
- Full Text
- View/download PDF
185. Alteration in plasma antioxidant capacities in chronic renal failure and hemodialysis patients: a possible explanation for the increased cardiovascular risk in these patients.
- Author
-
Clermont G, Lecour S, Lahet J, Siohan P, Vergely C, Chevet D, Rifle G, and Rochette L
- Subjects
- Aged, Analysis of Variance, Ascorbic Acid blood, Cardiovascular Diseases blood, Case-Control Studies, Cholesterol blood, Electron Spin Resonance Spectroscopy, Female, Free Radicals analysis, Humans, Kidney Failure, Chronic blood, Kidney Failure, Chronic therapy, Male, Middle Aged, Nitrates blood, Nitric Oxide blood, Nitrites blood, Oxidative Stress, Renal Dialysis, Risk Factors, Sulfhydryl Compounds analysis, Uric Acid blood, Vitamin E blood, Antioxidants analysis, Cardiovascular Diseases etiology, Kidney Failure, Chronic complications
- Abstract
Objective: The high incidence of cardiovascular diseases in chronic renal failure (CRF) and hemodialyzed (HD) patients is now well established and the involvement of oxidative stress has been hypothesized in these phenomena. The aim of our study was to evaluate the level of oxidative stress in healthy controls (CTL) compared with CRF and HD patients before (pre-HD) and after (post-HD) the dialysis session, carried out on a high biocompatible polyacrylonitrile membrane AN69., Methods: Several indicators of the extracellular redox status were evaluated in plasma. The ascorbyl free radical (AFR) was directly measured using electron spin resonance spectroscopy (ESR) and expressed with respect to the vitamin C level to obtain a direct index of oxidative stress. Indirect plasma parameters such as vitamin E, thiol and uric acid levels were also quantified. The plasma antioxidant status (PAS) was evaluated by the allophycocyanin test. Nitric oxide (NO) stable-end metabolites: nitrites and nitrates (NO(x)), were measured in plasma., Results: In CRF patients, vitamin C and thiol levels were low, and the AFR/vitamin C ratio high compared with the CTL. On the other hand, PAS and uric acid levels were shown to be higher in CRF patients. After the dialysis session, vitamin C level decreased and AFR/vitamin C ratio increased. The thiol levels were shown to be increased, in return PAS and uric acid levels were significantly lower after the dialysis session. NO(x) levels rose during CRF, but were significantly decreased after the dialysis procedure. No differences in vitamin E status were observed between CTL, CRF and HD patients., Conclusion: Our study demonstrates that profound disturbances in the extracellular redox system occur during the course of chronic renal failure and hemodialysis, and may provide an explanation for the cardiovascular complications in these patients.
- Published
- 2000
- Full Text
- View/download PDF
186. Electron spin resonance detection of nitric oxide generation in major organs from LPS-treated rats.
- Author
-
Lecour S, Maupoil V, Siri O, Tabard A, and Rochette L
- Subjects
- Analysis of Variance, Animals, Aorta metabolism, Chelating Agents metabolism, Electron Spin Resonance Spectroscopy methods, Glycated Hemoglobin metabolism, Heart drug effects, Iron metabolism, Lipopolysaccharides pharmacology, Male, Nitrates blood, Nitric Oxide analysis, Nitric Oxide metabolism, Nitrites blood, Rats, Rats, Wistar, Sorbitol analogs & derivatives, Sorbitol metabolism, Spin Labels, Thiocarbamates metabolism, Time Factors, Tissue Distribution, Myocardium metabolism, Nitric Oxide biosynthesis
- Abstract
The increased production of nitric oxide (NO) has been implicated as the basis for myocardial dysfunction and the lack of response to vasoconstrictors during endotoxin shock induced by lipopolysaccharide (LPS). Our objective was to evaluate and compare NO production in major organs of rats treated with LPS, 1 or 14 mg/kg. A NO spin-trapping technique using electron spin resonance (ESR) spectroscopy has been used to study NO production in the liver, the kidney, the aorta, and the heart. The method was based on the trapping of NO by a metal-chelator complex consisting of N-methyl-D-glucamine dithiocarbamate (MGD) and reduced iron (Fe2+) to form a stable [(MGD)2-Fe2+-NO] complex, giving rise to a characteristic triplet ESR spectrum with g = 2.04 and aN = 12.65 G: Iron was quantified in the different organs to study the [(MGD)2-Fe2+] complex distribution. Six hours after intravenous injection of 1 or 14 mg/kg of LPS, we observed large increases in the [(MGD)2-Fe2+-NO] adduct signal in the liver, the kidney, and in the aorta, strongly suggesting an increased production of NO in these organs. The [(MGD)2-Fe2+-NO] adduct was also detected in the heart, 6 h after injection of LPS. Moreover, we observed dose-dependent increases in [(MGD)2-Fe2+-NO] adduct in the heart, whereas no changes were observed in the other organs. Concurrently, the [(MGD)2-Fe2+-NO] adduct was not detected in the blood from rats treated with LPS, although circulating nitrosylhemoglobin, nitrite, and nitrate levels increased. The spin-trapping technique allowed us to monitor organ-specific formation of NO after LPS administration and for the first time demonstrated direct NO production in aorta and heart of LPS-treated animals.
- Published
- 1999
- Full Text
- View/download PDF
187. Demonstration of the production of oxygen-centered free radicals during electrolysis using E.S.R. spin-trapping techniques: effects on cardiac function in the isolated rat heart.
- Author
-
Lecour S, Baouali AB, Maupoil V, Chahine R, Abadie C, Javouhey-Donzel A, Rochette L, and Nadeau R
- Subjects
- Animals, Coronary Circulation drug effects, Cyclic N-Oxides, Free Radical Scavengers pharmacology, Free Radicals, In Vitro Techniques, Male, Mannitol pharmacology, Rats, Rats, Wistar, Solutions, Spin Labels, Superoxide Dismutase pharmacology, Electrolysis, Electron Spin Resonance Spectroscopy, Heart drug effects, Heart physiology, Reactive Oxygen Species metabolism, Spin Trapping
- Abstract
The present study was designed to identify the free radicals generated during the electrolysis of the solution used to perfuse isolated rat heart Langendorff preparations. The high reactivity and very short half-life of oxygen free radicals make their detection and identification difficult. A diamagnetic organic molecule (spin trap) can be used to react with a specific radical to produce a more stable secondary radical or "spin adduct" detected by electron spin resonance (ESR). Isovolumic left ventricular systolic pressure (LVSP) and left ventricular end diastolic pressure (LVEDP) were measured by a fluid-filled latex balloon inserted into the left ventricle. The coronary flow was measured by effluent collection. Electrolysis was performed with constant currents of 0.5, 1, 1.5, 3, 5, 7.5, and 10 mA generated by a Grass stimulator and applied to the perfusion solution for 1 min. A group of experiments was done using a 1.5 mA current and a Krebs-Henseleit (K-H) solution containing free radical scavengers (superoxide dismutase (SOD): 100 IU/ml or mannitol: 50 mM). Heart function rapidly declined in hearts perfused with K-H buffer that had been electrolyzed for 1 min. The addition of mannitol (50 mM) to the perfusion solution had no effect on baseline cardiac function before electrolysis while SOD (100 IU/ml) increased the coronary flow. However, SOD was more effective than the mannitol in protecting the heart against decreased of cardiac function, 5 min after the end of electrolysis. Samples of the K-H medium subjected to electrolysis were collected in cuvettes containing a final concentration of 125 mM 5,5-dimethyl-1-pyrroline-N-oxide (DMPO) and analyzed by spectroscopy. The ESR spectrum consisted of a quartet signal (hyperfine couplings aN = aH = 14.9 G) originating from the hydroxyl adduct signal, DMPO-OH. The intensity of the DMPO-OH signal remained stable during the 60 s of electrolysis and the quantity of free radicals induced by electrolysis was directly proportional to the intensity of the current. The addition of mannitol and SOD to the perfusate scavenged the hydroxyl radicals present in the solution, suggesting that both hydroxyl and superoxide radicals were formed during electrolysis.
- Published
- 1998
- Full Text
- View/download PDF
188. In vitro studies of interactions of NO. donor drugs with superoxide and hydroxyl radicals.
- Author
-
Dalloz F, Maupoil V, Lecour S, Briot F, and Rochette L
- Subjects
- Free Radicals metabolism, Hydrogen-Ion Concentration, Molsidomine analogs & derivatives, Molsidomine metabolism, Nitric Oxide biosynthesis, Nitroprusside metabolism, Oxidation-Reduction, Sodium Nitrite metabolism, Hydroxyl Radical metabolism, Nitric Oxide metabolism, Superoxides metabolism
- Abstract
Nitric oxide (NO.) is a free radical characterized by a high spontaneous chemical reactivity with many other molecules including the superoxide radical (O2.-). This complex interaction may generate a peroxynitrite anion (ONOO-), which behaves as an important mediator of oxidative stress in many pathological states. In the present study, in vitro experiments were performed to assess directly the O2.- and hydroxyl (.OH) radical scavenging effects of various NO. donor drugs, i.e. sodium nitroprusside (SNP), sodium nitrite (NaNO2), molsidomine and SIN 1, at pH 7.4, 7 or 6. Concentrations of NO. in the incubation medium containing the different NO. donor drugs were measured by the assay based on the reaction of Fe-N-methyl-D-glucamine dithiocarbamate (MGD) with NO. that yields a stable spin-adduct measured by electron paramagnetic resonance (EPR). O2.- and .OH generation was characterized by EPR spin trapping techniques, using the spin trap 5,5-dimethyl-1-pyrroline-1-oxide (DMPO). These free radicals were generated from the enzymatic system xanthine-xanthine oxidase, in phosphate buffer adjusted at pH 7.4, 7 and 6. Under these experimental conditions, SNP exhibited the strongest superoxide scavenging properties, characterized by IC50 values expressed in the micromolar range, which decreased at low pH. Addition of SNP (800 microM) to solution containing MGD and Fe2+ (5:1) at pH 7 4 produced a three line EPR spectrum which is identified to [(MGD)2-Fe2+-NO]. In control experiments no EPR signal was observed. We obtained the same results with NaNO2 and an augmentation of the spin-adduct level was noted with the prolongation of the incubation period. In return, molsidomine (2 mM) did not produce, in our conditions, a detectable production of NO.. NaNO2 displayed a significant superoxide scavenging effect only at pH 6, whilst neither molsidomine nor SIN 1 had any effect. Therefore, the superoxide scavenging properties of SNP, NaNO2, and molsidomine appeared to be closely related to their potential for NO release, which partially depends on the pH conditions. The behaviour of SIN 1 is more complicated, the speed of oxygen diffusion probably acting as a limiting factor in NO. formation in our conditions. The production of NO. was detected in presence of SIN 1. The intensity of the complex is comparable with the signal founded with NaNO2. By contrast, all molecules exhibited hydroxyl radical scavenging properties, highlighting the capacity of .OH to react with a wide range of molecules. In conclusion, considering the poor chemical reactivity of O2.-, the NO. donor drugs/O2.- interactions suggest a special relationship between these two radical species, which, in certain pathological states, could lead to the generation of cytotoxic end-products with strong oxidizing properties.
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.