Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
1.
J Cardiothorac Vasc Anesth ; 34(9): 2501-2512, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31685419

ABSTRACT

Ischemic heart disease, the leading cause of death worldwide, may result in devastating perioperative ischemia and infarction. The underlying pathophysiology, precipitating factors, and approach to prevention differ between patients presenting for noncardiac surgery, developing acute coronary syndrome versus stable angina. The first half of this article reviews the pathophysiology of acute coronary syndrome and stable angina. Acute coronary syndrome, otherwise known as Type 1 myocardial infarction, includes unstable angina, non-ST segment elevated myocardial infarction and ST segment elevated myocardial infarction. Acute coronary syndrome occurs as a result of vulnerable plaque rupture with subsequent varying degrees of thrombus formation, arterial spasm, and thus coronary occlusion. Stable angina, on the other hand, results from a myocardial oxygen delivery and demand mismatch in the setting of fixed coronary stenosis. After this discussion, the review article considers how both apply to perioperative myocardial infarctions and myocardial injury after noncardiac surgery. This article furthermore argues why myocardial oxygen delivery demand mismatch (Type 2) myocardial infarction is the most likely underlying pathophysiology responsible for perioperative myocardial infarctions. Being aware of this and knowledgeable about Type 2 infarctions may enable anesthetic providers to better predict the majority of triggers contributing to, and thus decreasing the incidence of, perioperative myocardial infarctions.


Subject(s)
Acute Coronary Syndrome , Myocardial Infarction , Myocardial Ischemia , Plaque, Atherosclerotic , Thrombosis , Humans , Myocardial Infarction/etiology , Myocardial Ischemia/etiology
2.
Heliyon ; 5(10): e02659, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31720456

ABSTRACT

Research focus recently shifted to mitochondrial dynamics and the role of fusion and fission in cardioprotection. The aim of this study was to evaluate (i) the function and dynamics of mitochondria isolated from hearts exposed to ischaemia/reperfusion (I/R) (ii) the effects of melatonin, a powerful cardioprotectant, on mitochondrial dynamics in I/R. Isolated perfused rat hearts were stabilized for 30 min, subjected to 20 min global ischaemia, followed by 30 min reperfusion. Tissue was collected, mitochondria isolated for measurement of mitochondrial oxidative function and lysates from mitochondrial and cytosolic fractions prepared for western blotting. Melatonin (0.3 or 50 µM) was administered for 10 min immediately before the onset of ischaemia and for 10 min at the onset of reperfusion. Infarct size was assessed after 35 min regional ischaemia/60 min reperfusion using triphenyltetrazolium staining. The results show that reperfusion significantly reduced mitochondrial QO2 (states 3 and 4), with minor effects by melatonin. Cytosolic Beclin 1 and the LC3 II/I ratio were reduced by ischaemia and increased by reperfusion. Both ischaemia and reperfusion reduced mitochondrial PINK1 and Parkin levels, while reperfusion increased p62. An alternative mitophagy pathway mediated by Rab9 is activated during myocardial ischaemia/reperfusion. Ischaemia reduced and reperfusion increased cytosolic ULK1 expression, associated with redistribution of Rab9 and Drp1 between the cytosol and mitochondria. Melatonin significantly reduced mitochondrial p62 expression upon reperfusion. Throughout the protocol, melatonin significantly (i) increased cytosolic total (t) and phospho (p) ULK1, and Rab9 levels (ii) increased the cytosolic and reduced the mitochondrial pDrp1 levels and p/t Drp1 ratio, suggesting inhibition of mitochondrial fission. Fusion was affected to a lesser extent. Cardioprotection by melatonin is associated with substantial effects on mitophagy, the significance thereof remains to be established.

3.
BMC Physiol ; 17(1): 3, 2017 Mar 17.
Article in English | MEDLINE | ID: mdl-28302152

ABSTRACT

BACKGROUND: Reports on the effect of age and obesity on myocardial ischaemia/reperfusion (I/R) injury and ischaemic preconditioning are contradictory. The aim of this study was to re-evaluate the effects of age and diet-induced obesity (DIO) on myocardial I/R injury and preconditioning potential. METHODS: Four groups of Wistar male rats were used: age-matched controls (AMC) receiving standard rat chow for (i) 16 weeks and (ii) 16 months respectively; DIO rats receiving a sucrose-supplemented diet for (iii) 16 weeks and (iv) 16 months respectively. The ages of groups (i) and (iii) were 22 weeks ("young") and groups (ii) and (iv) 17 months ("middle-aged") at time of experimentation. Isolated perfused working hearts were subjected to 35 min regional ischaemia/1 h reperfusion. Endpoints were infarct size (tetrazolium staining) and functional recovery. Hearts were preconditioned by 3 × 5 min ischaemia/5 min reperfusion. Results were processed using GraphPad Prism statistical software. RESULTS: Age did not affect baseline heart function before induction of ischaemia and I/R damage as indicated by infarct size and similar values were obtained in hearts from both age groups. Age also had no effect on functional recovery of hearts during reperfusion after regional ischaemia in AMC rats, but cardiac output during reperfusion was better in hearts from middle-aged than young DIO rats. The diet reduced infarct size in hearts from young rats (% of area at risk: AMC: 32.4 ± 3.6; DIO: 20.7 ± 2.9, p < 0.05), with no differences in hearts from middle-aged rats (AMC: 24.6 ± 4.6; DIO: 28.3 ± 13.5, p = NS). Compared to their respective AMC, diet-induced obesity had no significant effect on functional recovery of hearts from both age groups after exposure to regional ischaemia. When exposed to the more severe stress of global ischaemia, the functional recovery potential of middle-aged DIO rats appeared to be impeded compared to hearts of young DIO rats, while age had no effect on the functional recovery of AMC hearts. Preconditioning reduced infarct size in hearts from young control rats and both middle-aged groups, but not from young DIO rats. Age had a significant effect on functional recovery in preconditioning: it was improved in hearts from young control and DIO rats, but depressed in both middle-aged groups. CONCLUSIONS: The data showed that middle-age and obesity had no effect on baseline myocardial function and did not increase susceptibility to I/R damage upon exposure to regional ischaemia. On the contrary, obesity reduced I/R damage in young rats. Preconditioned aging hearts showed a decreased infarct size, but a reduction in functional recovery.


Subject(s)
Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Obesity/metabolism , Aging , Animals , Disease Models, Animal , Disease Susceptibility , Ischemic Preconditioning, Myocardial/methods , Male , Myocardial Reperfusion Injury/complications , Obesity/complications , Rats, Wistar
4.
Cardiovasc Drugs Ther ; 30(3): 237-46, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27180786

ABSTRACT

PURPOSE: There is an ongoing search for new drugs and drug targets to treat diseases like Alzheimer's disease, cancer and type 2 diabetes (T2D). Both obesity and T2D are characterized by the development of a cardiomyopathy associated with increased hypertension and compensatory left ventricular hypertrophy. Small, specific glycogen synthase kinase-3 (GSK-3) inhibitors were developed to replace lithium chloride for use in psychiatric disorders. In addition, they were advocated as treatment for T2D since GSK-3 inhibition improves blood glucose handling. However, GSK-3 is a regulator of hypertrophic signalling in the heart via phosphorylation of NFATc3 and ß-catenin respectively. In view of this, we hypothesized that chronic inhibition of GSK-3 will induce myocardial hypertrophy or exacerbate existing hypertrophy. METHODS: Rats with obesity-induced prediabetes were treated orally with GSK-3 inhibitor (CHIR118637 (CT20026)), 30 mg/kg/day for the last 8 weeks of a 20-week diet high in sugar content vs a control diet. Biometric and biochemical parameters were measured, echocardiography performed and localization and co-localization of NFATc3 and GATA4 determined in cardiomyocytes. RESULTS: Obesity initiated myocardial hypertrophy, evidenced by increased ventricular mass (1.158 ± 0.029 vs 0.983 ± 0.03 g) and enlarged cardiomyocytes (18.86 ± 2.25 vs 14.92 ± 0.50um(2)) in association with increased end-diastolic diameter (EDD = 8.48 ± 0.11 vs 8.15 ± 0.10 mm). GSK-3 inhibition (i) increased ventricular mass only in controls (1.075 ± 0.022 g) and (ii) EDD in both groups (controls: 8.63 ± 0.07; obese: 8.72 ± 0.15 mm) (iii) localized NFATc3 and GATA4 peri-nuclearly. CONCLUSION: Indications of onset of myocardial hypertrophy in both control and obese rats treated with a GSK-3 inhibitor were found. It remains speculation whether these changes were adaptive or maladaptive.


Subject(s)
Cardiomegaly/etiology , Diabetes Mellitus/drug therapy , Diabetic Cardiomyopathies/etiology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Hypoglycemic Agents/adverse effects , Obesity/drug therapy , Animals , Blood Glucose/analysis , Cardiomegaly/diagnostic imaging , Cardiomegaly/metabolism , Cell Size/drug effects , Diabetes Mellitus/diagnostic imaging , Diabetes Mellitus/metabolism , Diabetic Cardiomyopathies/diagnostic imaging , Diabetic Cardiomyopathies/metabolism , Echocardiography , GATA4 Transcription Factor/metabolism , Heart/diagnostic imaging , Hypoglycemic Agents/therapeutic use , Insulin/blood , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NFATC Transcription Factors/metabolism , Obesity/complications , Obesity/diagnostic imaging , Obesity/metabolism , Rats, Wistar
5.
Eur J Nutr ; 55(6): 2063-73, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26286349

ABSTRACT

PURPOSE: The consumption of foods rich in dietary fiber and polyunsaturated fatty acids such as nuts can contribute to a healthy diet. Therefore, the formation of fermentation end-products which might exert chemopreventive effects regarding colon cancer was investigated after an in vitro simulated digestion and fermentation of nuts using human fecal microbiota. METHODS: Fermentation supernatants (FS) and pellets (FP) were obtained after an in vitro fermentation of hazelnuts, almonds, macadamia, pistachios and walnuts. Short-chain fatty acids (SCFA) and bile acids (BA) in FS as well as fatty acids in FP were analyzed via gas chromatography. Malondialdehyde (MDA) levels in FS were determined photometrically. RESULTS: Fermentation of nuts resulted in 1.9- to 2.8-fold higher concentrations of SCFA compared to the control and a shift of molar ratios toward butyrate production. In vitro fermentation resulted in the formation of vaccenic acid (C18:1t11, 32.1 ± 3.2 % FAME; fatty acid methyl ester) and conjugated linoleic acid (c9,t11 CLA, 2.4 ± 0.7 % FAME) exclusively in fermented walnut samples. Concentrations of secondary BA deoxycholic-/iso-deoxycholic acid (6.8-24.1-fold/4.9-10.9-fold, respectively) and levels of MDA (1.3-fold) were significantly reduced in fermented nut samples compared to the control. CONCLUSION: This is the first study that demonstrates the ability of the human fecal microbiota to convert polyunsaturated fatty acids from walnuts to c9,t11 CLA as a potential chemopreventive metabolite. In addition, the production of butyrate and reduction in potential carcinogens such as secondary BA and lipid peroxidation products might contribute to the protective effects of nuts regarding colon cancer development.


Subject(s)
Butyrates/chemistry , Fermentation , Linoleic Acids, Conjugated/chemistry , Nuts/chemistry , Bile Acids and Salts/metabolism , Colonic Neoplasms/prevention & control , Corylus/chemistry , Fatty Acids, Unsaturated/chemistry , Feces/microbiology , Gastrointestinal Microbiome , Humans , Hydrogen-Ion Concentration , Juglans/chemistry , Macadamia/chemistry , Malondialdehyde/chemistry , Oleic Acids/chemistry , Pistacia/chemistry , Prunus dulcis/chemistry , Thiobarbituric Acid Reactive Substances/analysis
6.
Cardiovasc Drugs Ther ; 28(5): 489-91, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25022545

ABSTRACT

AIMS: In isolated rat heart perfusion experiments, drug administration occurs via retrograde perfusion. This can be done in the non-recirculating mode (coronary effluent is discarded), or recirculating mode (coronary effluent is collected and reused). It was recently observed in our lab while using sanguinarine, an MKP-1 inhibitor, that there were differences in outcomes depending on the mode of recirculation used. METHODS AND RESULTS: Hearts from control (C); diet-induced obese (DIO) Wistar rats and their age matched controls (AMC) were perfused on the rig. Hearts received buffer (control) , insulin, sanguinarine, insulin + sanguinarine combination or methanol (vehicle) for 15 mins pre- and 10 mins post-ischemia in either a non- or re-circulating manner. Hearts were subjected to 15 mins global ischemia and 30 mins reperfusion. Mechanical function was documented pre- and post-ischemia . When not-recirculated , sanguinarine alone and in combination with insulin in C, DIO and AMC groups, caused a significant decrease in functional recovery during reperfusion. However, when the coronary effluent was recirculated, hearts perfused with sanguinarine or sanguinarine + insulin exhibited a significant recovery in function when compared with their non-recirculation counterparts (p < 0.01). No differences were seen with either control, insulin nor vehicle hearts. CONCLUSION: Sanguinarine elicited a vast improvement in perfusion outcomes when recirculated compared to non-recirculation . Since this was seen during perfusion only when sanguinarine was present, it is possible that recirculating reperfusion of the drug caused profound changes in its composition. More investigation is needed into the mechanisms involved. Thus caution should be exercised by researchers when designing a perfusion protocol for drug research.


Subject(s)
Benzophenanthridines/therapeutic use , Heart/drug effects , In Vitro Techniques , Isoquinolines/therapeutic use , Perfusion , Recovery of Function/drug effects , Reperfusion Injury/drug therapy , Animals , Cardiotonic Agents/therapeutic use , Coronary Circulation/drug effects , Drug Therapy, Combination , Insulin/administration & dosage , Insulin/therapeutic use , Male , Rats
7.
Br J Anaesth ; 111(5): 768-75, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23801744

ABSTRACT

BACKGROUND: Clinical temperature management remains challenging. Choosing the right sensor location to determine the core body temperature is a particular matter of academic and clinical debate. This study aimed to investigate the relationship of measured temperatures at different sites during surgery in deep hypothermic patients. METHODS: In this prospective single-centre study, we studied 24 patients undergoing cardiothoracic surgery: 12 in normothermia, 3 in mild, and 9 in deep hypothermia. Temperature recordings of a non-invasive heat flux sensor at the forehead were compared with the arterial outlet temperature of a heart-lung machine, with the temperature on a conventional vesical bladder thermistor and, for patients undergoing deep hypothermia, with oesophageal temperature. RESULTS: Using a linear model for sensor comparison, the arterial outlet sensor showed a difference among the other sensor positions between -0.54 and -1.12°C. The 95% confidence interval ranged between 7.06 and 8.82°C for the upper limit and -8.14 and -10.62°C for the lower limit. Because of the hysteretic shape, the curves were divided into phases and fitted into a non-linear model according to time and placement of the sensors. During cooling and warming phases, a quadratic relationship could be observed among arterial, oesophageal, vesical, and cranial temperature recordings, with coefficients of determination ranging between 0.95 and 0.98 (standard errors of the estimate 0.69-1.12°C). CONCLUSION: We suggest that measured surrogate temperatures as indices of the cerebral temperature (e.g. vesical bladder temperature) should be interpreted with respect to the temporal and spatial dispersion during cooling and rewarming phases.


Subject(s)
Body Temperature/physiology , Circulatory Arrest, Deep Hypothermia Induced , Adult , Aged , Aged, 80 and over , Algorithms , Anesthesia, General , Blood Physiological Phenomena , Cardiac Surgical Procedures , Echocardiography, Transesophageal , Esophagus/physiology , Female , Forehead/physiology , Heart Diseases/surgery , Humans , Linear Models , Male , Middle Aged , Monitoring, Intraoperative , Nonlinear Dynamics , Prospective Studies , Skin Temperature , Thoracic Surgical Procedures , Urinary Bladder/physiology
8.
Acta Physiol (Oxf) ; 206(1): 6-19, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22741552

ABSTRACT

AIM: To investigate the effects of dietary creatine supplementation alone and in combination with exercise on basal cardiac function, susceptibility to ischaemia/reperfusion injury and mitochondrial oxidative function. There has been an increase in the use of creatine supplementation among sports enthusiasts, and by clinicians as a therapeutic agent in muscular and neurological diseases. The effects of creatine have been studied extensively in skeletal muscle, but not in the myocardium. METHODS: Male Wistar rats were swim-trained for 8 weeks, 5 days per week. Hearts were excised and either freeze-clamped for biochemical analysis or perfused on the isolated heart perfusion system to assess function and ischaemia/reperfusion tolerance. Mechanical function was documented in working heart and retrograde mode. The left coronary artery was ligated and infarct size determined. Mitochondrial oxidative capacity was quantified. RESULTS: Aortic output recovery of hearts from the sedentary controls (CSed) was significantly higher than those from creatine-supplemented sedentary (CrSed), creatine-supplemented exercised (CrEx) as well as control exercised (CEx) groups. Ischaemic contracture of hearts from CrEx was significantly higher than that of CSed. There were no differences in infarct size and mitochondrial oxygen consumption. CONCLUSION: This study suggests that creatine supplementation has no effects on basal cardiac function but reduces myocardial tolerance to ischaemia in hearts from exercise-trained animals, by increasing the ischaemic contracture and decreasing reperfusion aortic output. Exercise training alone also significantly decreased aortic output recovery. However, the exact mechanisms for these adverse myocardial effects are unknown and need further investigation.


Subject(s)
Creatine/therapeutic use , Mitochondria, Heart/metabolism , Oxygen Consumption/drug effects , Physical Conditioning, Animal/physiology , Reperfusion Injury/drug therapy , Animals , Creatine/administration & dosage , Dietary Supplements , Male , Mitochondria, Heart/drug effects , Myocardial Infarction , Organ Culture Techniques , Oxidative Stress , Phosphorylation , Random Allocation , Rats , Rats, Wistar
9.
Acta Physiol (Oxf) ; 205(2): 209-23, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22226301

ABSTRACT

The metabolic syndrome (MetS) is a cluster of metabolic abnormalities associated with increased risk for cardiovascular diseases. Apart from its powerful antioxidant properties, the pineal gland hormone melatonin has recently attracted the interest of various investigators as a multifunctional molecule. Melatonin has been shown to have beneficial effects in cardiovascular disorders including ischaemic heart disease and hypertension. However, its role in cardiovascular risk factors including obesity and other related metabolic abnormalities is not yet established, particularly in humans. New emerging data show that melatonin may play an important role in body weight regulation and energy metabolism. This review will address the role of melatonin in the MetS focusing on its effects in obesity, insulin resistance and leptin resistance. The overall findings suggest that melatonin should be exploited as a therapeutic tool to prevent or reverse the harmful effects of obesity and its related metabolic disorders.


Subject(s)
Body Weight/physiology , Energy Metabolism/physiology , Melatonin/metabolism , Metabolic Syndrome/metabolism , Obesity/metabolism , Humans , Insulin Resistance/physiology , Metabolic Syndrome/physiopathology , Obesity/physiopathology
10.
J Ethnopharmacol ; 137(1): 298-304, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21645608

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Diabetes mellitus is rampantly increasing and the need for therapeutics is crucial. In recognition of this, untested antidiabetic agents are flooding the market. Diavite™ which is a product consisting solely of the dried and ground pods of Prosopis glandulosa (Torr.) [Fabaceae] is currently marketed as a food supplement with glucose stabilizing properties. However, these are anecdotal claims lacking scientific evidence. The aim of this study was to determine the efficacy of Prosopis glandulosa as an antidiabetic agent. MATERIALS AND METHODS: Male Wistar rats were rendered (a) type 1 diabetic after an intraperitoneal injection of STZ (40 mg/kg) and (b) insulin resistant after a 16-week high caloric diet (DIO). Zucker fa/fa ZDF rats were used in a pilot study. Half of each group of animals was placed on Prosopis glandulosa treatment (100mg/kg/day) for 8 weeks and the remaining animals served as age-matched controls. At the time of sacrifice, blood was collected for glucose and insulin level determination, the pancreata of the STZ rats were harvested for histological analysis and cardiomyocytes prepared from the DIO and Zucker fa/fa hearts for determination of insulin sensitivity. RESULTS: Type 1 diabetic model: Prosopis glandulosa treatment resulted in significant increased insulin levels (p<0.001), which was accompanied by a significant decrease in blood glucose levels (p<0.05). Additionally, Prosopis glandulosa treatment resulted in increased small ß-cells (p<0.001) in the pancreata. The body weight of the STZ animals decreased significantly after STZ injection, with Prosopis glandulosa treatment partially preventing this. Zucker fa/fa rats: Prosopis glandulosa treatment significantly reduced fasting glucose levels (p<0.01) and improved IPGTT, when comparing treated to untreated animals. DIO insulin resistant model: Prosopis glandulosa treatment resulted in an increased basal (p<0.01) and insulin-stimulated (p<0.05) glucose uptake by cardiomyocytes prepared from this group. CONCLUSIONS: The present study showed that Prosopis glandulosa treatment moderately lowers glucose levels in different animal models of diabetes, stimulates insulin secretion, leads to the formation of small ß-cells and improves insulin sensitivity of isolated cardiomyocytes.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/pharmacology , Insulin Resistance , Plant Preparations/pharmacology , Prosopis , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/physiopathology , Dose-Response Relationship, Drug , Glucose Tolerance Test , Insulin/blood , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Rats, Wistar , Rats, Zucker , Time Factors
11.
Med Hypotheses ; 77(1): 52-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21482032

ABSTRACT

Autophagy is a conserved catabolic process for long-lived proteins and organelles and is primarily responsible for nonspecific degradation of redundant or faulty cell components. Although autophagy has been described as the cell's major adaptive strategy in response to metabolic challenges, its influence on the cell's energy profile is poorly understood. In the myocardium, autophagy is active at basal levels and is crucial for maintaining its contractile function. Defects in the autophagic machinery cause cardiac dysfunction and heart failure. In this paper we propose that (1) autophagy contributes significantly to the metabolic balance sheet of the heart. (2) Increased autophagy contributes to an improved myocardial energy profile through changing the cardiac substrate preference. (3) Substrates generated through autophagy give rise to an alternative for ATP production with an oxygen-sparing effect. These elements identify autophagy in a new context of myocardial metabolic interregulation, which we discuss in the settings of myocardial infarction, heart failure and the diabetic heart. It is hoped that the hypothesis presented can lead to new insights aimed at exploiting autophagy to improve existing metabolic-based therapy in heart disease.


Subject(s)
Autophagy , Heart Diseases/immunology , Energy Metabolism , Heart Diseases/metabolism , Humans
12.
Exp Cell Res ; 317(10): 1437-53, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21420401

ABSTRACT

Ischemic cell injury leads to cell death. Three main morphologies have been described: apoptosis, cell death with autophagy and necrosis. Their inherent dynamic nature, a point of no return (PONR) and molecular overlap have been stressed. The relationship between a defined cell death type and the severity of injury remains unclear. The functional role of autophagy and its effects on cell death onset is largely unknown. In this study we report a differential induction of cell death, which is dependent on the severity and duration of an ischemic insult. We show that mild ischemia leads to the induction of autophagy and apoptosis, while moderate or severe ischemia induces both apoptotic and necrotic cell death without increased autophagy. The autophagic response during mild injury was associated with an ATP surge. Real-time imaging and Fluorescence Resonance Energy Transfer (FRET) revealed that increased autophagy delays the PONR of both apoptosis and necrosis significantly. Blocking autophagy shifted PONR to an earlier point in time. Our results suggest that autophagic activity directly alters intracellular metabolic parameters, responsible for maintaining mitochondrial membrane potential and cellular membrane integrity. A similar treatment also improved functional recovery in the perfused rat heart. Taken together, we demonstrate a novel finding: autophagy is implicated only in mild injury and positions the PONR in cell death.


Subject(s)
Apoptosis , Autophagy , Disease Models, Animal , Ischemia/pathology , Necrosis , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Cells, Cultured , Flow Cytometry , Fluorescence Resonance Energy Transfer , Fluorescent Antibody Technique , Heart/physiology , Male , Membrane Potential, Mitochondrial , Myoblasts/cytology , Myoblasts/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Rats , Rats, Wistar
13.
Mol Cell Biochem ; 349(1-2): 21-31, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21153603

ABSTRACT

Exogenous insulin therapy improves endothelial function in insulin resistant patients, indirectly indicating that nitric oxide synthase activity and NO production may be impaired. Insulin stimulates production of NO by activating a signaling pathway including insulin receptor substrate-1, phosphatidylinositol-3-kinase and protein kinase B (PKB/Akt). Angiotensin II type I (AT1) receptor-evoked oxidative stress is implicated in the inactivation of NO, impairing endothelium-dependent vasodilatation. Blocking the actions of Angiotensin II with an AT1 receptor antagonist (Losartan), has beneficial effects in patients with insulin resistance or type 2 diabetes mellitus. This study investigated whether elevated Angiotensin II influences myocardial insulin resistance, insulin signaling and NO production in a rat model of diet-induced obesity (DIO) by antagonizing the actions of the AT1 receptor with Losartan. Isolated, perfused hearts, Western blotting and flow-cytometric methods were utilized to determine myocardial function, expression and phosphorylation of key proteins and NO production, respectively. Results showed that hearts from DIO rats are insulin resistant (higher serine phosphorylation of IRS-1, lower insulin-stimulated phosphorylation of PKB/Akt and eNOS, lower NO production) and had poorer functional recovery and larger infarct development after ischaemia/reperfusion. Losartan improved the impaired functional recovery, and NO production and enhanced eNOS expression and phosphorylation and reduced infarct size in hearts from the DIO animals. Data obtained from Losartan treatment also revealed that Angiotensin II signaling modulates myocardial PKB/Akt expression. We conclude that Angiotensin II signaling exacerbates inhibition of NO production in insulin resistance and that this can be improved by AT1 antagonism.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Insulin Resistance , Losartan/pharmacology , Myocardium/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cardiac Output/drug effects , Diet , In Vitro Techniques , Male , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/cytology , Obesity/etiology , Obesity/metabolism , Obesity/physiopathology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Signal Transduction
14.
Physiol Res ; 60(2): 291-301, 2011.
Article in English | MEDLINE | ID: mdl-21114368

ABSTRACT

Obesity is increasing at an alarming rate globally. Several studies have shown that premenopausal women have a reduced risk of CV disease and a reduced myocardial susceptibility to ischemia/reperfusion injury. The effect of obesity on myocardial tolerance to ischemia in women has not been established. To determine how obesity affects myocardial susceptibility to ischemia/reperfusion injury in both males and females, we fed male and female Wistar rats a high caloric diet (HCD) or a control rat chow diet (CD) for 18 weeks. Rats were subsequently fasted overnight, anesthetized and blood was collected. In separate experiments, 18-week-fed (HCD and CD) rats underwent 45 min in vivo coronary artery ligation (CAL) followed by 2 hours reperfusion. Hearts were stained with TTC and infarct size determined. Both male and female HCD fed rats had increased body and visceral fat weights. Homeostasis model assessment (HOMA) index values were 13.95+/-3.04 for CD and 33.58+/-9.39 for HCD male rats (p<0.01) and 2.98+/-0.64 for CD and 2.99+/-0.72 for HCD fed female rats. Male HCD fed rats had larger infarct sizes than CD fed littermates (43.2+/-9.3 % vs. 24.4+/-7.6 %, p<0.05). Female HCD and CD diet fed rats had comparable infarct sizes (31.8+/-4.3 % vs. 23.9+/-3.3 %). We conclude that male rats on the HCD became viscerally obese, dyslipidemic and insulin-resistant, while female HCD fed rats became viscerally obese without developing dyslipidemia or insulin resistance. Obesity increased myocardial infarct size in males but not the females.


Subject(s)
Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/physiopathology , Obesity/physiopathology , Animals , Body Weight , Energy Intake/physiology , Female , Insulin Resistance/physiology , Intra-Abdominal Fat/physiopathology , Male , Rats , Rats, Wistar , Sex Factors
15.
Cardiovasc J Afr ; 21(2): 72-8, 2010.
Article in English | MEDLINE | ID: mdl-20532430

ABSTRACT

Activation of AMP-activated protein kinase (AMPK) results in glucose transporter 4 (GLUT4) translocation from the cytosol to the cell membrane, and glucose uptake in the skeletal muscles. This increased activation of AMPK can be stimulated by a pharmacological agent, AICAR (5' -aminoimidazole-4-carboxamide ribonucleoside), which is converted intracellularly into ZMP (5' -aminoimidazole-4-carboxamideribonucleosidephosphate), an AMP analogue. We utilised AICAR and ZMP to study GLUT4 translocation and glucose uptake in isolated cardiomyocytes. Adult ventricular cardiomyocytes were treated with AICAR or ZMP, and glucose uptake was measured via [3H] -2-deoxyglucose accumulation. PKB/Akt, AMPK and acetyl-CoA-carboxylase phosphorylation and GLUT4 translocation were detected by Western blotting or flow cytometry. AICAR and ZMP promoted AMPK phosphorylation. Neither drug increased glucose uptake but on the contrary, inhibited basal glucose uptake, although GLUT4 translocation from the cytosol to the membrane occurred. Using flow cytometry to detect the exofacial loop of the GLUT4 protein, we showed ineffective insertion in the membrane under these conditions. Supplementing with nitric oxide improved insertion in the membrane but not glucose uptake. We concluded that activation of AMPK via AICAR or ZMP was not sufficient to induce GLUT4-mediated glucose uptake in isolated cardiomyocytes. Nitric oxide plays a role in proper insertion of the protein in the membrane but not in glucose uptake.


Subject(s)
Adenylate Kinase/metabolism , Enzyme Activation/genetics , Glucose Transporter Type 4/genetics , Myocytes, Cardiac/enzymology , Translocation, Genetic/genetics , Adenylate Kinase/genetics , Animals , Blotting, Western , Cells, Cultured , Flow Cytometry , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Male , Myocytes, Cardiac/cytology , Rats , Rats, Wistar
16.
Life Sci ; 84(23-24): 838-46, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19361533

ABSTRACT

AIMS: Previous studies suggested that p38 MAPK activation during sustained myocardial ischaemia and reperfusion was harmful. We hypothesize that attenuation of p38MAPK activity via dephosphorylation by the dual-specificity phosphatase MKP-1 should be protective against ischaemia/reperfusion injury. Since the glucocorticoid, dexamethasone, induces the expression of MKP-1, the aim of this study was to determine whether upregulation of this phosphatase by dexamethasone protects the heart against ischaemia/reperfusion injury. MAIN METHODS: Male Wistar rats were treated with dexamethasone (3 mg/kg/day ip) for 10 days, before removal of the hearts for Western blot (ip Dex-P) or perfusion in the working mode (ip Dex+P). Hearts were subjected to 20 min global or 35 min regional ischaemia (36.5 degrees C) and 30 or 120 min reperfusion. In a separate series, dexamethasone (1 microM) was added to the perfusate for 10 min (Pre+Dex) before or after (Rep+Dex) ischaemia. KEY FINDINGS: Dexamethasone, administered intraperitoneally or added directly to the perfusate, significantly improved post-ischaemic functional recovery and reduced infarct size compared to untreated controls (p<0.05). These were associated with enhanced up-regulation of MKP-1 protein expression (arbitrary units (mean+/-SD): Untreated: 1; ip Dex-P: 2.59+/-0.22; ip Dex+P: 1.51+/-0.22; Pre+Dex: 4.11+/-0.73, Rep+15'Dex: 1.51+/-0.14; untreated vs. all groups, p<0.05) and attenuation of p38 MAPK activation (p<0.05) in all dexamethasone-treated groups, except for Rep+10'Dex. ERK and PKB/Akt activation were unchanged. SIGNIFICANCE: Dexamethasone-induced cardioprotection was associated with upregulation of the phosphatase MKP-1 and inactivation of pro-apoptotic p38 MAPK.


Subject(s)
Cardiotonic Agents/therapeutic use , Dexamethasone/therapeutic use , Dual Specificity Phosphatase 1/physiology , Animals , Male , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/prevention & control , Rats , Rats, Wistar
17.
Cardiovasc J Afr ; 20(1): 43-51, 2009.
Article in English | MEDLINE | ID: mdl-19287816

ABSTRACT

Exposure of the heart to one or more short episodes of ischaemia/reperfusion protects the heart against a subsequent prolonged period of ischaemia, as evidenced by a reduction in infarct size and an improvement in functional recovery during reperfusion. Elucidation of the mechanism of this endogenous protection could lead to the development of pharmacological mimetics to be used in the clinical setting. The aim of our studies was therefore to gain more information regarding the mechanism of ischaemic preconditioning, using the isolated perfused working rat heart as model. A preconditioning protocol of 1 x 5 or 3 x 5 min of ischaemia, interspersed with 5 min of reperfusion was found to protect hearts exposed to 25 min of global ischaemia or 35-45 min of regional ischaemia. These models were used throughout our studies. In view of the release of catecholamines by ischaemic tissue, our first aim was to evaluate the role of the alphaadrenergic receptor in ischaemic preconditioning. However, using a multi-cycle ischaemic preconditioning protocol, we could not find any evidence for alpha-1 adrenergic or PKC activation in the mechanism of preconditioning. Cyclic increases in the tissue cyclic nucleotides, cAMP and cGMP were found, however, to occur during a multi-cycle preconditioning protocol, suggesting roles for the beta-adrenergic signalling pathway and nitric oxide (NO) as triggers of cardioprotection. This was substantiated by the findings that (1) administration of the beta-adrenergic agonist, isoproterenol, or the NO donors SNAP or SNP before sustained ischaemia also elicited cardioprotection similar to ischaemic preconditioning; (2) beta-adrenergic blockade or nitric oxide synthase inhibition during an ischaemic preconditioning protocol abolished protection. Effectors downstream of cAMP, such as p38MAPK and CREB, were also demonstrated to be involved in the triggering process. Our next step was to evaluate intracellular signalling during sustained ischaemia and reperfusion. Our results showed that ischaemic preconditioned-induced cardioprotection was associated with a significant reduction in tissue cAMP, attenuation of p38MAPK activation and increased tissue cGMP levels and HSP27 activation, compared to non-preconditioned hearts. The role of the stress kinase p38MAPK was further investigated by using the inhibitor SB203580. Our results suggested that injury by necrosis and apoptosis share activation of p38MAPK as a common signal transduction pathway and that pharmacological targeting of this kinase offers a tenable option to manipulate both these processes during ischaemia/reperfusion injury.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocardial Infarction/prevention & control , Myocardial Ischemia/therapy , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Animals , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Humans , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Ischemia/complications , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardium/enzymology , Nitric Oxide/metabolism , Rats , Receptors, Adrenergic/metabolism , Signal Transduction , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Cardiovasc J Afr ; 19(2): 77-83, 2008.
Article in English | MEDLINE | ID: mdl-18516352

ABSTRACT

Glucagon-like peptide-1 is an incretin hormone proposed to have insulinomimetic effects on peripheral insulin-sensitive tissue. We examined these effects on the heart by using isolated, perfused rat hearts and adult ventricular myocytes. During normoxic perfusion, no effects of escalating concentrations of GLP-1 on either heart rate or left ventricular developed pressure were found. With functional performance as readout, we found that GLP-1 directly protected the heart against damage incurred by global low-flow ischaemia. This protection was sensitive to the presence of iodo-acetate, implicating activation of glycolysis, and was abolished by wortmannin, indicative of PI-3-kinase as mediator of protection. In addition, GLP-1 had an infarct-sparing effect when supported by the presence of the dipeptidyl peptidase-IV inhibitor valine pyrrolidide. GLP-1 could not directly activate protein kinase B (also called Akt) or the extracellular regulated kinases Erk1/2 in hearts or cardiocytes under normoxic conditions, but phosphorylation of the AMP-activated kinase (AMPK) on Thr(172) was enhanced. I n addition, the glycolytic enzyme phosphofructokinase- 2 was activated dose dependently. During reperfusion after ischaemia, modulation of the phosphorylation of PKB/Akt as well as AMPK was evident. GLP-1 therefore directly protected the heart against low-flow ischaemia by enhancing glycolysis, probably via activation of AMP kinase and by modulating the profile of activation of the survival kinase PKB/Akt.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Peptide Fragments/metabolism , Signal Transduction , AMP-Activated Protein Kinases , Animals , Disease Models, Animal , Enzyme Activation , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinases/metabolism , Glycolysis , Heart Rate , In Vitro Techniques , Male , Multienzyme Complexes/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocytes, Cardiac/metabolism , Perfusion , Phosphatidylinositol 3-Kinases/metabolism , Phosphofructokinase-2/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Time Factors , Ventricular Function, Left , Ventricular Pressure
19.
Br J Pharmacol ; 154(1): 41-50, 2008 May.
Article in English | MEDLINE | ID: mdl-18297097

ABSTRACT

BACKGROUND AND PURPOSE: Myocardial reperfusion injury prevents optimal salvage of the ischaemic myocardium, and adjunct therapy that would significantly reduce reperfusion injury is still lacking. We investigated whether (1) the heart could be pre- and/or post-conditioned using levosimendan (levosimendan pre-conditioning (LPC) and levosimendan post-conditioning (LPostC)) and (2) the prosurvival kinases and/or the sarcolemmal or mitochondrial K(ATP) channels are involved. EXPERIMENTAL APPROACH: Isolated guinea pig hearts were treated with two 5 min cycles of levosimendan (0.1 microM) interspersed with vehicle perfusion, or two 5 min cycles of ischaemia/reperfusion, before coronary artery ligation (CAL) for 40 min at 36.5 degrees C. Hearts were treated with mitochondrial or sarcolemmal K(ATP) channel blockers before LPC or LPostC. For post-conditioning, hearts received three 30 s cycles of ischaemia/reperfusion or levosimendan/vehicle. Hearts were pretreated with levosimendan immediately before CAL (without washout). Cardiac function, infarct size and reperfusion injury salvage kinase activity was assessed. KEY RESULTS: LPC and LPostC halved the infarct size compared with controls (P<0.05). Treatment with K(ATP) channel blockers before LPC or LPostC reversed this decrease. Pretreating hearts with levosimendan increased activity of extracellular signal-regulated kinase (ERK) 42/44 on reperfusion and had the most marked infarct-lowering effect (P<0.05). CONCLUSIONS AND IMPLICATIONS: (1) Hearts could be pharmacologically pre- and post-conditioned with levosimendan; (2) levosimendan pretreatment is the most effective way to reduce infarct size, possibly by increasing ERK 42/44 activity; (3) benefits of LPC and LPostC were abolished by both K(ATP) channel blockers and (4) LPC may be useful before elective cardiac surgery, whereas LPostC may be used after acute coronary artery events.


Subject(s)
Cardiotonic Agents/pharmacology , Hydrazones/pharmacology , Ischemic Preconditioning, Myocardial , KATP Channels/physiology , Pyridazines/pharmacology , Signal Transduction/physiology , Animals , Blotting, Western , Cardiac Output/physiology , Coronary Circulation/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Guinea Pigs , In Vitro Techniques , Myocardial Infarction/pathology , Myocardium/pathology , Simendan
20.
Cardiovasc Drugs Ther ; 22(1): 3-17, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18205034

ABSTRACT

PURPOSE: Previous studies from our laboratory showed that activation of p38 MAPK is one of the triggers of ischaemic preconditioning. The signalling events downstream of p38 MAPK and their links to the putative final effectors of preconditioning are not clear. The cAMP responsive element-binding protein (CREB) is also phosphorylated by exposure to short episodes of ischaemia/reperfusion, suggesting a triggering action. The aim of this study was to systematically investigate (1) the signalling pathways leading to CREB phosphorylation during an ischaemic or beta-adrenergic preconditioning protocol (2) changes in CREB phosphorylation during sustained ischaemia and their significance in ischaemia/reperfusion injury. METHODS: The isolated perfused working rat heart was preconditioned by 1 x 5 min global ischaemia or 3 x 5 min global ischaemia and freeze-clamped. Drugs to manipulate CREB activation were added 5 min before onset of ischaemia. Non-preconditioned and preconditioned hearts were subjected to 25 min global or 35 min regional ischaemia, followed by 30 min reperfusion. Infarct sizes were determined using tetrazolium staining. Phosphorylation of CREB was determined by Western blots. RESULTS: Exposure of hearts to 5 min global ischaemia followed by reperfusion, significantly increased CREB phosphorylation This is mediated by, amongst others, release of endogenous catecholamines and adenosine, as indicated by the use of receptor blockers. Events downstream of receptor stimulation were evaluated using inhibitors for PKA (H89), MSK-1 (H89, Ro318220), PKC (bisindolylmaleimide), p38 MAPK (SB203580) and ERK (PD98059). Activation of PKA, PKC, ERK and p38 MAPK is involved in preconditioning-induced CREB phosphorylation. Ischaemia-induced activation of iPLA(2) and cPLA(2) also contribute to CREB phosphorylation as indicated by the use of the inhibitors 4-bromo-enol-lactone (BEL) and AACOF(3,) respectively. Inhibition of CREB phosphorylation by either BEL or AACOF(3) during a preconditioning protocol partially attenuated cardioprotection. CREB phosphorylation was attenuated during sustained global ischaemia of both non-preconditioned and preconditioned hearts. CONCLUSIONS: These data suggest that CREB phosphorylation during an ischaemic preconditioning protocol may contribute to triggering preconditioning, while reduced phosphorylation during sustained ischaemia does not appear to be associated with cardioprotection.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/pathology , Signal Transduction , Adenosine/metabolism , Animals , Blotting, Western , Catecholamines/metabolism , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Phosphorylation , Protein Kinase C/metabolism , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL