Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Cardiovasc Diabetol ; 12: 154, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24148218

ABSTRACT

BACKGROUND: The dipeptidyl peptidase-4 (DPP-4) inhibitors Sitagliptin and Vildagliptin lower blood glucose by augmenting endogenous levels of glucagon-like peptide-1 (GLP-1), an incretin which also confers cardioprotection. As such, we hypothesized that treatment with DPP-4 inhibitors are also cardioprotective. METHODS: In ex vivo experiments: Male Sprague-Dawley rats were randomized to receive by oral gavage either Vildagliptin (20 mg/kg/day), Sitagliptin (100 mg/kg/day), or water for 2 weeks. Excised hearts were Langendorff-perfused with buffer containing either 5 mmol/L or 11 mmol/L glucose and subjected to 35 minutes ischaemia/120 minutes reperfusion. In in vivo experiments: Male young Wistar and Sprague-Dawley rats, middle aged Wistar and Goto-Kakizaki diabetic rats were randomized to receive by oral gavage either Sitagliptin (100 mg/kg/day), or water for 2 weeks. Rats were then subjected to 30 minutes ischaemia/120 minutes reperfusion and infarct size ascertained. RESULTS: Two weeks pre-treatment with either Vildagliptin or Sitagliptin reduced ex vivo myocardial infarction (MI) size in hearts perfused with buffer containing 11 mmol/L glucose but not 5 mmol/L glucose. This effect was abolished by Exendin 9-39 (GLP-1 receptor antagonist) and H-89 (PKA antagonist). Treatment of perfused hearts with native GLP-1 was also glucose-sensitive, reducing MI size, at glucose concentrations 7, 9, and 11 mmol/L but not at 5 mmol/L. Finally, Sitagliptin reduced in vivo MI size in middle aged Wistar (7-8 mmol/L glucose) and Goto-Kakizaki (9-10 mmol/L glucose) rats where blood glucose was elevated, but not in young Wistar (5 mmol/L glucose) or Sprague-Dawley (5 mmol/L glucose) rats, where blood glucose was normal. CONCLUSIONS: We find that chronic treatment with DPP-4 inhibitors reduced MI size, via the GLP-1 receptor-PKA pathway, in a glucose-dependent manner. Glucose-sensitive cardioprotection of endogenous GLP-1 in diabetic patients may in part explain why intensive control of serum glucose levels has been associated with increased cardiovascular risk.


Subject(s)
Adamantane/analogs & derivatives , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Glucagon-Like Peptide 1/drug effects , Glucose/metabolism , Heart/drug effects , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Nitriles/pharmacology , Pyrazines/pharmacology , Pyrrolidines/pharmacology , Triazoles/pharmacology , Adamantane/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Glucagon-Like Peptide 1/physiology , Glucagon-Like Peptide-1 Receptor , Male , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Glucagon/antagonists & inhibitors , Severity of Illness Index , Signal Transduction , Sitagliptin Phosphate , Vildagliptin
2.
PLoS Biol ; 11(9): e1001666, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24086110

ABSTRACT

Cardiomyocytes are vulnerable to hypoxia in the adult, but adapted to hypoxia in utero. Current understanding of endogenous cardiac oxygen sensing pathways is limited. Myocardial oxygen consumption is determined by regulation of energy metabolism, which shifts from glycolysis to lipid oxidation soon after birth, and is reversed in failing adult hearts, accompanying re-expression of several "fetal" genes whose role in disease phenotypes remains unknown. Here we show that hypoxia-controlled expression of the transcription factor Hand1 determines oxygen consumption by inhibition of lipid metabolism in the fetal and adult cardiomyocyte, leading to downregulation of mitochondrial energy generation. Hand1 is under direct transcriptional control by HIF1α. Transgenic mice prolonging cardiac Hand1 expression die immediately following birth, failing to activate the neonatal lipid metabolising gene expression programme. Deletion of Hand1 in embryonic cardiomyocytes results in premature expression of these genes. Using metabolic flux analysis, we show that Hand1 expression controls cardiomyocyte oxygen consumption by direct transcriptional repression of lipid metabolising genes. This leads, in turn, to increased production of lactate from glucose, decreased lipid oxidation, reduced inner mitochondrial membrane potential, and mitochondrial ATP generation. We found that this pathway is active in adult cardiomyocytes. Up-regulation of Hand1 is protective in a mouse model of myocardial ischaemia. We propose that Hand1 is part of a novel regulatory pathway linking cardiac oxygen levels with oxygen consumption. Understanding hypoxia adaptation in the fetal heart may allow development of strategies to protect cardiomyocytes vulnerable to ischaemia, for example during cardiac ischaemia or surgery.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Energy Metabolism/genetics , Lipid Metabolism/genetics , Myocardium/metabolism , Oxygen Consumption/genetics , Adenosine Triphosphate/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia/genetics , Cell Line , Gene Expression Regulation, Developmental , Heart/embryology , Heart/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Potential, Mitochondrial/genetics , Mice , Mice, Transgenic , Mitochondria/metabolism , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Transcriptional Activation
3.
Cardiovasc Res ; 99(4): 694-704, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23723063

ABSTRACT

AIMS: Old age and diabetes are risk factors that often coexist increasing the vulnerability of the heart to the lethal effects of ischaemia-reperfusion injury (IRI). However, to our knowledge, no investigations have examined IRI and cardioprotective signalling in animal models bearing these co-morbidities concomitantly. The ability of the heart to recover following IRI is greatly dependent on its innate cardioprotective potential, in which a central role is played by Akt. We aimed to investigate in an aging diabetic rat model, the susceptibility of the heart to IRI, the achievability of ischaemic preconditioning (IPC) against this lethal event, and the changes in Akt signalling, as the main prosurvival intracellular pathway. METHODS AND RESULTS: Our data showed that the isolated hearts of aged, diabetic Goto-Kakizaki rats were more susceptible to sub-lethal injury and not amenable to cardioprotection via IPC, compared with younger diabetic rat hearts. Western blot analysis of the heart tissue suggested a chronic up-regulation of Akt phosphorylation, and reduced expression of the mitochondrial regulator PGC-1α and of the anti-oxidant enzyme catalase, potentially due to the Akt up-regulation. Moreover, no further activation of Akt could be achieved following IPC. CONCLUSION: An increased susceptibility to IRI in the aged, diabetic heart could be a consequence of impaired Akt signalling due to chronic Akt phosphorylation. Additional Akt phosphorylation required for IPC protection may therefore not be possible in the aged, diabetic rat heart and may explain why this cardioprotective manoeuvre cannot be achieved in these hearts.


Subject(s)
Aging/physiology , Diabetes Mellitus/physiopathology , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/etiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Animals , Blood Glucose/analysis , Glycated Hemoglobin/analysis , Humans , Male , Myocardial Infarction/etiology , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenotype , Phosphorylation , Rats , Rats, Wistar , Transcription Factors/physiology
4.
PLoS One ; 8(4): e62400, 2013.
Article in English | MEDLINE | ID: mdl-23638067

ABSTRACT

OBJECTIVES: Mutations in PTEN inducible kinase-1 (PINK1) induce mitochondrial dysfunction in dopaminergic neurons resulting in an inherited form of Parkinson's disease. Although PINK1 is present in the heart its exact role there is unclear. We hypothesized that PINK1 protects the heart against acute ischemia reperfusion injury (IRI) by preventing mitochondrial dysfunction. METHODS AND RESULTS: Over-expressing PINK1 in HL-1 cardiac cells reduced cell death following simulated IRI (29.2±5.2% PINK1 versus 49.0±2.4% control; N = 320 cells/group P<0.05), and delayed the onset of mitochondrial permeability transition pore (MPTP) opening (by 1.3 fold; P<0.05). Hearts excised from PINK1+/+, PINK1+/- and PINK1-/- mice were subjected to 35 minutes regional ischemia followed by 30 minutes reperfusion. Interestingly, myocardial infarct size was increased in PINK1-/- hearts compared to PINK1+/+ hearts with an intermediate infarct size in PINK1+/- hearts (25.1±2.0% PINK1+/+, 38.9±3.4% PINK1+/- versus 51.5±4.3% PINK1-/- hearts; N>5 animals/group; P<0.05). Cardiomyocytes isolated from PINK1-/- hearts had a lower resting mitochondrial membrane potential, had inhibited mitochondrial respiration, generated more oxidative stress during simulated IRI, and underwent rigor contracture more rapidly in response to an uncoupler when compared to PINK1+/+ cells suggesting mitochondrial dysfunction in hearts deficient in PINK1. CONCLUSIONS: We show that the loss of PINK1 increases the heart's vulnerability to ischemia-reperfusion injury. This may be due, in part, to increased mitochondrial dysfunction. These findings implicate PINK1 as a novel target for cardioprotection.


Subject(s)
Myocardium/metabolism , Protein Kinases/deficiency , Reperfusion Injury/enzymology , Animals , Cell Line , Disease Susceptibility , Gene Knockout Techniques , Membrane Potential, Mitochondrial , Mice , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress , Oxygen/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
5.
J Cardiovasc Pharmacol Ther ; 18(3): 263-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23263382

ABSTRACT

AIMS: The diabetic heart is resistant to the myocardial infarct-limiting effects of ischemic preconditioning (IPC). This may be in part due to the downregulation of the phosphatidylinositol 3'-kinase-Akt pathway, an essential component of IPC protection. We hypothesized that treating the diabetic heart with the sulfonylurea, glimepiride, which has been reported to activate Akt, may lower the threshold required to protect the diabetic heart by IPC. METHODS: Goto-Kakizaki rats (a type II lean model of diabetes) received glimepiride (20 mg/kg per d, by oral gavage) or vehicle for (a) 3 months (chronic treatment) or (b) 24 hours (subacute treatment). In the third group, glimepiride (10 µmol/L) was administered only to the isolated hearts on the Langendorff apparatus (acute treatment). All hearts were subjected to 35 minutes ischemia and 120 minutes reperfusion ex vivo, at the end of which infarct size was determined by tetrazolium staining. Preconditioning treatment comprised 1 (IPC-1) or 3 (IPC-3) cycles of 5 minutes global ischemia and 10 minutes reperfusion. RESULTS: The diabetic heart was found to be resistant to IPC such that 3-IPC cycles, instead of the usual 1-IPC cycle, were required for cardioprotection. However, pretreatment with glimepiride lowered the threshold for IPC such that both 1 and 3 cycles of IPC elicited cardioprotection: chronic glimepiride treatment (IPC-1 31.9% ± 3.8% and IPC-3 33.5% ± 2.4% vs 43.9% ± 1.4% control, P < .05; N > 6 per group); subacute glimepiride treatment (IPC-1 31.1% ± 3.0% and IPC-3 29.3% ± 3.3% vs 42.2% ± 2.3% control, P < .05 N > 6 per group); and acute glimepiride treatment (IPC-1 28.2% ± 3.7% and IPC-3 24.6% ± 5.4% vs 41.9% ± 5.4% control, P < .05; N > 6 per group). This effect of glimepiride was independent of changes in blood glucose. CONCLUSIONS: We report for the first time that glimepiride treatment facilitates the cardioprotective effect elicited by IPC in the diabetic heart.


Subject(s)
Cardiotonic Agents/therapeutic use , Diabetic Cardiomyopathies/drug therapy , Hypoglycemic Agents/therapeutic use , Ischemic Preconditioning, Myocardial , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/prevention & control , Sulfonylurea Compounds/therapeutic use , Animals , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/pharmacology , Combined Modality Therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/therapy , Enzyme Activators/administration & dosage , Enzyme Activators/pharmacology , Enzyme Activators/therapeutic use , Glycated Hemoglobin/analysis , Heart/drug effects , Hyperglycemia/prevention & control , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardium/enzymology , Myocardium/metabolism , Myocardium/pathology , Perfusion , Proto-Oncogene Proteins c-akt/agonists , Random Allocation , Rats , Rats, Inbred Strains , Sulfonylurea Compounds/administration & dosage , Sulfonylurea Compounds/pharmacology
6.
Cardiovasc Drugs Ther ; 27(1): 5-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23192487

ABSTRACT

PURPOSE: Clinical and experimental investigations demonstrated that metformin, a widely used anti-diabetic drug, exhibits cardioprotective properties against myocardial infarction. Interestingly, metformin was previously shown to increase the expression of PGC-1α a key controller of energy metabolism in skeletal muscle, which is down-regulated in diabetic conditions. We hypothesized that chronic treatment with metformin could protect the aged, diabetic heart against ischemia-reperfusion injury (IRI) by up-regulating PGC-1α and improving the impaired functionality of diabetic mitochondria. METHODS: Following 4 weeks of metformin (300 mg/kg) administered in the drinking water, 12 month-old diabetic Goto Kakizaki and non-diabetic Wistar rat hearts were assigned for infarct measurement following 35 min ischemia and 60 min reperfusion or for electron microscopy (EM) and Western blotting (WB) investigations. RESULTS: Metformin elicited a cardioprotective effect in both non-diabetic and diabetic hearts. In contrast with the diabetic non-treated hearts, the diabetic hearts treated with metformin showed more organized and elongated mitochondria and demonstrated a significant increase in phosphorylated AMPK and in PGC-1α expression. CONCLUSIONS: In summary these results show for the first time that chronic metformin treatment augments myocardial resistance to ischemia-reperfusion injury, by an alternative mechanism in addition to the lowering of blood glucose. This consisted of a positive effect on mitochondrial structure possibly via a pathway involving AMPK activation and PGC-1α. Thus, metformin prescribed chronically to patients may lead to a basal state of cardioprotection thereby potentially limiting the occurrence of myocardial damage by cardiovascular events.


Subject(s)
Blood Glucose/metabolism , Cardiotonic Agents/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Myocardial Infarction/drug therapy , AMP-Activated Protein Kinase Kinases , Aging/blood , Aging/metabolism , Aging/pathology , Animals , Blotting, Western , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/pharmacology , Dose-Response Relationship, Drug , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Male , Metformin/administration & dosage , Metformin/pharmacology , Microscopy, Electron , Mitochondria, Heart/drug effects , Mitochondria, Heart/ultrastructure , Myocardial Infarction/enzymology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/enzymology , Myocardium/metabolism , Myocardium/ultrastructure , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Kinases/metabolism , RNA-Binding Proteins/biosynthesis , Rats , Rats, Wistar , Transcription Factors/biosynthesis
7.
Cardiol Res Pract ; 2012: 845698, 2012.
Article in English | MEDLINE | ID: mdl-22462028

ABSTRACT

Diabetes mellitus is a major risk factor for ischemic heart disease (IHD). Patients with diabetes and IHD experience worse clinical outcomes, suggesting that the diabetic heart may be more susceptible to ischemia-reperfusion injury (IRI). In contrast, the animal data suggests that the diabetic heart may be either more, equally, or even less susceptible to IRI. The conflicting animal data may be due to the choice of diabetic and/or IRI animal model. Ischemic conditioning, a phenomenon in which the heart is protected against IRI by one or more brief nonlethal periods of ischemia and reperfusion, may provide a novel cardioprotective strategy for the diabetic heart. Whether the diabetic heart is amenable to ischemic conditioning remains to be determined using relevant animal models of IRI and/or diabetes. In this paper, we review the limitations of the current experimental models used to investigate IRI and cardioprotection in the diabetic heart.

8.
J Cell Mol Med ; 16(8): 1739-49, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22117619

ABSTRACT

Phosphatidyl-inositol-3-kinase (PI3K)-Akt pathway is essential for conferring cardioprotection in response to ischaemic preconditioning (IPC) stimulus. However, the role of the individual Akt isoforms expressed in the heart in mediating the protective response to IPC is unknown. In this study, we investigated the specific contribution of Akt1 and Akt2 in cardioprotection against ischaemia-reperfusion (I-R) injury. Mice deficient in Akt1 or Akt2 were subjected to in vivo regional myocardial ischaemia for 30 min. followed by reperfusion for 2 hrs with or without a prior IPC stimulus. Our results show that mice deficient in Akt1 were resistant to protection with either one or three cycles of IPC stimulus (42.7 ± 6.5% control versus 38.5 ± 1.9% 1 χ IPC, N = 6, NS; 41.4 ± 6.3% control versus 32.4 ± 3.2% 3 χ IPC, N = 10, NS). Western blot analysis, performed on heart samples taken from Akt1(-/-) mice subjected to IPC, revealed an impaired phosphorylation of GSK-3ß, a downstream effector of Akt, as well as Erk1/2, the parallel component of the reperfusion injury salvage kinase pathway. Akt2(-/-) mice, which exhibit a diabetic phenotype, however, were amenable to protection with three but not one cycle of IPC (46.4 ± 5.6% control versus 35.9 ± 5.0% in 1 χ IPC, N = 6, NS; 47.0 ± 6.0% control versus 30.8 ± 3.3% in 3 χ IPC, N = 6; *P = 0.039). Akt1 but not Akt2 is essential for mediating a protective response to an IPC stimulus. Impaired activation of GSK-3ß and Erk1/2 might be responsible for the lack of protective response to IPC in Akt1(-/-) mice. The rise in threshold for protection in Akt2(-/-) mice might be due to their diabetic phenotype.


Subject(s)
Ischemic Preconditioning, Myocardial , Proto-Oncogene Proteins c-akt/metabolism , Aging/pathology , Animals , Hemodynamics , Hyperglycemia/enzymology , Hyperglycemia/pathology , Immunoblotting , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/enzymology , Myocardium/pathology , Proto-Oncogene Proteins c-akt/deficiency , Signal Transduction , Survival Analysis
9.
J Mol Cell Cardiol ; 50(6): 940-50, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21385587

ABSTRACT

In the late 19th century, a number of investigators were working on perfecting isolated heart model, but it was Oscar Langendorff who, in 1895, pioneered the isolated perfused mammalian heart. Since that time, the Langendorff preparation has evolved and provided a wealth of data underpinning our understanding of the fundamental physiology of the heart: its contractile function, coronary blood flow regulation and cardiac metabolism. In more recent times, the procedure has been used to probe pathophysiology of ischaemia/reperfusion and disease states, and with the dawn of molecular biology and genetic manipulation, the Langendorff perfused heart has remained a stalwart tool in the study of the impact upon the physiology of the heart by pharmacological inhibitors and targeted deletion or up-regulation of genes and their impact upon intracellular signalling and adaption to clinically relevant stressful stimuli. We present here the basic structure of the Langendorff system and the fundamental experimental rules which warrant a viable heart preparation. In addition, we discuss the use of the isolated retrograde perfused heart in the model of ischaemia-reperfusion injury ex-vivo, and its applicability to other areas of study. The Langendorff perfusion apparatus is highly adaptable and this is reflected not only in the procedure's longevity but also in the number of different applications to which it has been turned.


Subject(s)
Coronary Circulation/physiology , Perfusion , Animals , Humans , Perfusion/methods
10.
Am J Physiol Heart Circ Physiol ; 300(6): H2123-34, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21421816

ABSTRACT

AMPK activation during ischemia helps the myocardium to cope with the deficit of energy production. As AMPK activity is considered to be impaired in diabetes, we hypothesized that enhancing AMPK activation during ischemia above physiological levels would protect the ischemic diabetic heart through AMPK activation and subsequent inhibition of mitochondrial permeability transition pore (mPTP) opening. Isolated perfused hearts from normoglycemic Wistar or diabetic Goto-Kakizaki (GK) rats (n ≥ 6/group) were subjected to 35 min of ischemia in the presence of 10, 20, and 40 µM of A-769662, a known activator of AMPK, followed by 120 min of reperfusion with normal buffer. Myocardial infarction and AMPK phosphorylation were assessed. The effect of A-769662 on mPTP opening in adult cardiomyocytes isolated from both strains was also determined. A-769662 at 20 µM reduced infarct size in both Wistar (30.5 ± 2.7 vs. 51.8 ± 3.9% vehicle; P < 0.001) and GK hearts (22.7 ± 3.0 vs. 48.5 ± 4.7% vehicle; P < 0.001). This protection was accompanied by a significant increase in AMPK and GSK-3ß phosphorylation. In addition, A-769662 significantly inhibited mPTP opening in both Wistar and GK cardiomyocytes subjected to oxidative stress. We demonstrate that AMPK activation during ischemia via A-769662 reduces myocardial infarct size in both the nondiabetic and diabetic rat heart. Furthermore, this cardioprotective effect appears to be mediated through inhibition of mPTP opening. Our findings suggest that improving AMPK activation during ischemia can be another mechanism for protecting the ischemic heart.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus/metabolism , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , AMP-Activated Protein Kinases/drug effects , Animals , Biphenyl Compounds , Cells, Cultured , Disease Models, Animal , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Male , Microtubule-Associated Proteins/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation , Pyrones/pharmacology , Rats , Rats, Inbred Strains , Rats, Wistar , Thiophenes/pharmacology
11.
J Cardiovasc Pharmacol Ther ; 16(1): 63-71, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21183727

ABSTRACT

Experimental studies have linked the adipocytokines with acute cardioprotection. Whether the adipocytokine, resistin, confers protection is, however, debatable. In the current study, the actions of resistin, administered at reperfusion, were investigated in in vivo and in vitro rodent and in vitro human models of myocardial ischemia-reperfusion (I/R) injury. Resistin did not reduce infarct size in Langendorff-perfused rat hearts or murine hearts perfused in vivo. Resistin also did not protect human atrial muscle subjected to hypoxia-reoxygenation. Although cyclosporin A delayed mitochondrial permeability transition pore (MPTP) opening in murine cardiomyocytes, resistin was ineffective. Western blot analysis revealed that resistin treatment was associated with enhanced phosphorylation of Akt, at both the serine-473 (+ 51.9%, P = .01) and threonine-308 (+107%, P < .01) phosphorylation sites, although not to the extent seen with ischemic preconditioning (+132.5%, P = .002 and +389.1%, P < .01, respectively). We conclude that resistin administered at reperfusion at concentrations/doses equivalent to normal (upper end) and pathological serum levels does not protect against I/R injury or inhibit MPTP opening.


Subject(s)
Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/prevention & control , Resistin/pharmacology , Resistin/therapeutic use , Aged , Aged, 80 and over , Animals , Cells, Cultured , Female , Heart/drug effects , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
12.
Basic Res Cardiol ; 105(6): 677-86, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20865418

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.


Subject(s)
Cardiovascular Agents/therapeutic use , Ischemic Postconditioning , Ischemic Preconditioning, Myocardial , Myocardial Infarction/prevention & control , Myocardial Ischemia/therapy , Myocardial Reperfusion Injury/prevention & control , Translational Research, Biomedical , Age Factors , Animals , Disease Models, Animal , Humans , Sex Factors , Species Specificity , Treatment Outcome
13.
Cardiovasc Drugs Ther ; 24(1): 25-32, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20229055

ABSTRACT

PURPOSE: AMPK plays a crucial role in the regulation of the energy metabolism of the heart. During ischaemia, AMPK activation is a known adaptative prosurvival mechanism that helps to maintain the energy levels of the myocardium. However, it still remains unclear if activation of AMPK during reperfusion is beneficial for the heart. Two known AMPK activators (metformin and AICAR) were used to verify the hypothesis that a transitory activation of AMPK at reperfusion may exert cardioprotection, as reflected in a reduction in myocardial infarct size. METHODS: Perfused rat hearts were subjected to 35 min ischaemia and 120 min reperfusion. Metformin (50 microM) or AICAR (0.5 mM) were added for 15 min at the onset of reperfusion alone or with Compound C (CC, 10 microM), an AMPK inhibitor. Infarct size and alpha-AMPK phosphorylation were measured. RESULTS: Metformin significantly reduced infarct size from 47.8 +/- 1.7% in control to 31.4 +/- 2.9%, an effect abolished by CC when the drugs were given concomitantly. Similarly, AICAR also induced a significant reduction in infarct size to 32.3 +/- 4.8%, an effect also abrogated by CC. However, metformin's protection was not abolished if CC was administered later in reperfusion. In addition, alpha-AMPK phosphorylation was significantly increased in the metformin treated group during the initial 30 min of reperfusion. CONCLUSIONS: Our data demonstrated that, in our ex vivo model of myocardial ischaemia-reperfusion injury, AMPK activation in early reperfusion is associated with a reduction in infarct size.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Enzyme Activation/drug effects , Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , AMP-Activated Protein Kinases/antagonists & inhibitors , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Aminoimidazole Carboxamide/therapeutic use , Animals , Heart/drug effects , Heart/physiology , Hemodynamics/physiology , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Infarction/etiology , Infarction/pathology , Male , Metformin/administration & dosage , Metformin/pharmacology , Metformin/therapeutic use , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Rats , Rats, Wistar , Ribonucleotides/administration & dosage , Ribonucleotides/pharmacology , Ribonucleotides/therapeutic use
14.
J Cardiovasc Pharmacol ; 53(5): 373-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19295441

ABSTRACT

Metformin improves cardiovascular outcomes in patients with type 2 diabetes compared with other glucose-lowering drugs. Experimental studies have shown that metformin can increase the intracellular concentration of adenosine monophosphate, which is a major determinant of the intracellular formation of adenosine. We hypothesize that metformin, given at reperfusion, can limit myocardial infarct size due to increased adenosine receptor stimulation. Isolated perfused hearts from Sprague-Dawley rats were subjected to 35 minutes of regional ischemia and 120 minutes of reperfusion. Perfusion with metformin (50 microM) for the first 15 minutes of reperfusion reduced infarct size (percent area at risk) from 42% +/- 2% to 19% +/- 4% (n >or= 6; P < 0.01), which was blocked by a concomitant perfusion with the adenosine receptor antagonist 8-p-sulfophenyltheophylline (100 microM; 43% +/- 3%) or nitrobenzylthioinosine (a blocker of transmembranous adenosine transport; 1 microM; 45% +/- 5%). In addition, intravenous administration of metformin (5 mg/kg) reduced infarct size in a rat in situ model of myocardial infarction (34% +/- 6% vs. 62% +/- 5%; P < 0.01), which was completely abolished by 8-p-sulfophenyltheophylline (61% +/- 3%). We conclude that metformin, given at reperfusion, reduces infarct size in a rat model of myocardial infarction, which is critically dependent on adenosine receptor stimulation, probably via increased intracellular formation of adenosine.


Subject(s)
Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Myocardial Reperfusion Injury/prevention & control , Purinergic P1 Receptor Agonists , Adenosine/metabolism , Adenosine/pharmacology , Animals , Biological Transport/drug effects , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/therapeutic use , In Vitro Techniques , Male , Metformin/administration & dosage , Metformin/therapeutic use , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/pathology , Purinergic P1 Receptor Antagonists , Rats , Rats, Sprague-Dawley , Theophylline/analogs & derivatives , Theophylline/pharmacology , Thioinosine/analogs & derivatives , Thioinosine/pharmacology
17.
Basic Res Cardiol ; 103(6): 560-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18604624

ABSTRACT

Activation of the PI3K/Akt pathway protects the heart from ischemia-reperfusion injury (IRI). The phosphatase PTEN is the main negative regulator of this pathway. We hypothesized that reduced PTEN levels could protect against IRI. Isolated perfused mouse hearts from PTEN(+/-) and their littermates PTEN(+/+) (WT), were subjected to 35 min global ischemia and 30 min reperfusion, with and without 2, 4 or 6 cycles ischemic preconditioning (IPC). The end point was infarct size, expressed as a percentage of the myocardium at risk (I/R%). PTEN and Akt levels were determined using Western blot analysis. Unexpectedly, there were no significant differences in infarction between PTEN(+/-) and WT (42.1 +/- 5.0% Vs. 45.6 +/- 3.3%). However, the preconditioning threshold was significantly reduced in the PTEN(+/-) Vs. WT, with 4 cycles of IPC being sufficient to reduce I/R%, compared to 6 cycles in the WT (4 cycles IPC: 29.8. +/- 3.69% in PTEN(+/-) Vs. 45.5. +/- 5.08% in WT, P < 0.01). In addition, the ratio between the phospho/total Akt (Ser473 and Thr308) was slightly but significantly increased in the PTEN(+/-) indicating an upregulation of PI3K/Akt pathway. Interestingly, the levels of the other phosphatases that may negatively regulate the PI3K/Akt pathway (PP2A, SHIP2 and PHLPP) were not significantly different between littermates and PTEN(+/-). In conclusion, PTEN haploinsufficiency alone does not induce cardioprotection in this model; however, it reduces the threshold of protection induced by IPC.


Subject(s)
Haplotypes/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Animals , Disease Models, Animal , Genotype , Ischemic Preconditioning, Myocardial , Male , Mice , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
18.
Basic Res Cardiol ; 103(3): 274-84, 2008 May.
Article in English | MEDLINE | ID: mdl-18080084

ABSTRACT

BACKGROUND: In the majority of studies, metformin has been demonstrated to cardioprotect diabetic patients, the mechanism of which is unclear. We hypothesized that metformin cardioprotects the ischemic heart through the Akt-mediated inhibition of mitochondrial permeability transition pore (mPTP) opening. MATERIALS AND METHODS: Isolated perfused hearts from normoglycemic Wistar or from diabetic Goto-Kakizaki (GK) rats (N > or = 6/group) were subjected to 35 min ischemia and 120 min of reperfusion. Metformin (50 micromol/l) was added for 15 min at reperfusion, alone or with LY294002 (15 micromol/l), a PI3K inhibitor. Infarct size and Akt phosphorylation were measured. Furthermore, the effect of metformin on mPTP opening in adult cardiomyocytes isolated from both strains was determined. RESULTS: Metformin reduced infarct size in both Wistar (35 +/- 2.7% metformin vs. 62 +/- 3.0% control: P < 0.05) and GK hearts (43 +/- 4.7% metformin vs. 60 +/- 3.8% control: P < 0.05). This protection was accompanied by a significant increase in Akt phosphorylation. LY294002 abolished the metformin-induced Akt phosphorylation and the infarct-limiting effect of metformin in Wistar (61 +/- 6.7% metformin + LY294002 vs. 35 +/- 2.7% metformin: P < 0.05) and GK rats (56 +/- 5.7% metformin + LY294002 vs. 43 +/- 4.7% metformin: P < 0.05). In addition, metformin significantly inhibited mPTP opening and subsequent rigor contracture in both Wistar and GK cardiomyocytes subjected to oxidative stress, in a LY-sensitive manner. CONCLUSIONS: We report that metformin given at the time of reperfusion reduces myocardial infarct size in both the non-diabetic and diabetic heart and this protective effect is mediated through PI3K and is associated with Akt phosphorylation. Furthermore, cardioprotection appears to be executed through a PI3K-mediated inhibition of mPTP opening. These findings may explain in part the cardioprotective properties of metformin observed in clinical studies of diabetic patients.


Subject(s)
Diabetes Mellitus/drug therapy , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Mitochondria, Heart/drug effects , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Myocardial Ischemia/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Chromones/pharmacology , Diabetes Mellitus/enzymology , Diabetes Mellitus/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Hypoglycemic Agents/administration & dosage , Ischemic Contracture/enzymology , Ischemic Contracture/prevention & control , Male , Metformin/administration & dosage , Mitochondria, Heart/enzymology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Morpholines/pharmacology , Myocardial Ischemia/enzymology , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar
20.
Cardiovasc Drugs Ther ; 21(4): 253-6, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17541736

ABSTRACT

BACKGROUND AND METHODS: Glucagon Like Peptide-1 (GLP-1), one of the most potent incretin hormones, has potential beneficial actions on the ischaemic and failing heart. This study sought to further identify the mechanisms of action of GLP-1 on the ischaemic heart using an in vitro isolated perfused rat heart model of ischaemic-reperfusion injury (measuring infarct size to area of risk (%)) subjected to 35 min regional ischaemia and 2 h reperfusion. To examine the effect of intact GLP-1 we used an inhibitor of GLP-1 breakdown, Valine pyrrolidide (VP). The downstream target of phosphatidylinositol 3-kinase includes the mTOR/p70s6 kinase pathway which was pharmacologically inhibited by rapamycin. RESULTS AND CONCLUSION: GLP-1 alone did not decrease myocardial infarction (54.4 +/- 3.1%). VP alone did not decrease myocardial infarction (52.5 +/- 4%). GLP-1 in the presence of VP produced significant reduction in myocardial infarction compared to control hearts (28.4 +/- 2.7% vs. 56.4 +/- 3.9% vs. P < 0.05). Inhibiting p70s6 Kinase with rapamycin completely abolished GLP-1 induced protection (57.1 +/- 4.9% vs. 28.4 +/- 2.7% P < 0.05). There was no detectable increase in the phosphorylated p70s6k after either 5 or 10 min of treatment with GLP-1/VP or with VP alone in comparison to control blots. In conclusion we show for the first time that the protective effects of GLP-1 are mediated by intact GLP-1 and can be inhibited by blocking the p70s6 kinase.


Subject(s)
Glucagon-Like Peptide 1/pharmacology , Incretins/pharmacology , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Ribosomal Protein S6 Kinases, 70-kDa/drug effects , Animals , Blotting, Western , Disease Models, Animal , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Male , Myocardial Reperfusion Injury/physiopathology , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/drug effects , Protein Kinases/metabolism , Pyrroles/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Time Factors , Valine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...