Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Cell Mol Med ; 22(9): 4183-4196, 2018 09.
Article in English | MEDLINE | ID: mdl-29921042

ABSTRACT

Necroptosis, a form of cell loss involving the RIP1-RIP3-MLKL axis, has been identified in cardiac pathologies while its inhibition is cardioprotective. We investigated whether the improvement of heart function because of ischaemic preconditioning is associated with mitigation of necroptotic signaling, and these effects were compared with a pharmacological antinecroptotic approach targeting RIP1. Langendorff-perfused rat hearts were subjected to ischaemic preconditioning with or without a RIP1 inhibitor (Nec-1s). Necroptotic signaling and the assessment of oxidative damage and a putative involvement of CaMKII in this process were analysed in whole tissue and subcellular fractions. Ischaemic preconditioning, Nec-1s and their combination improved postischaemic heart function recovery and reduced infarct size to a similar degree what was in line with the prevention of MLKL oligomerization and translocation to the membrane. On the other hand, membrane peroxidation and apoptosis were unchanged by either approach. Ischaemic preconditioning failed to ameliorate ischaemia-reperfusion-induced increase in RIP1 and RIP3 while pSer229-RIP3 levels were reduced only by Nec-1s. In spite of the additive phosphorylation of CaMKII and PLN because of ditherapy, the postischaemic contractile force and relaxation was comparably improved in all the intervention groups while antiarrhythmic effects were observed in the ischaemic preconditioning group only. Necroptosis inhibition seems to be involved in cardioprotection of ischaemic preconditioning and is comparable but not intensified by an anti-RIP1 agent. Changes in oxidative stress nor CaMKII signaling are unlikely to explain the beneficial effects.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/therapy , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Apoptosis/drug effects , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Gene Expression Regulation , Heart/drug effects , Heart/physiopathology , Male , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Necrosis/genetics , Necrosis/metabolism , Necrosis/pathology , Necrosis/prevention & control , Organ Culture Techniques , Oxidative Stress , Phosphorylation/drug effects , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Rats , Rats, Wistar , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction
2.
Can J Physiol Pharmacol ; 95(10): 1204-1212, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28683229

ABSTRACT

Currently, there are no satisfactory interventions to protect the heart against the detrimental effects of ischemia-reperfusion injury. Although ischemic preconditioning (PC) is the most powerful form of intrinsic cardioprotection, its application in humans is limited to planned interventions, due to its short duration and technical requirements. However, many organs/tissues are capable of producing "remote" PC (RPC) when subjected to brief bouts of ischemia-reperfusion. RPC was first described in the heart where brief ischemia in one territory led to protection in other area. Later on, RPC started to be used in patients with acute myocardial infarction, albeit with ambiguous results. It is hypothesized that the connection between the signal triggered in remote organ and protection induced in the heart can be mediated by humoral and neural pathways, as well as via systemic response to short sublethal ischemia. However, although RPC has a potentially important clinical role, our understanding of the mechanistic pathways linking the local stimulus to the remote organ remains incomplete. Nevertheless, RPC appears as a cost-effective and easily performed intervention. Elucidation of protective mechanisms activated in the remote organ may have therapeutic and diagnostic implications in the management of myocardial ischemia and lead to development of pharmacological RPC mimetics.


Subject(s)
Ischemic Preconditioning, Myocardial/methods , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Animals , Disease Models, Animal , Humans , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Regional Blood Flow , Signal Transduction , Time Factors , Treatment Outcome
3.
Cureus ; 9(4): e1201, 2017 Apr 29.
Article in English | MEDLINE | ID: mdl-28560127

ABSTRACT

Statins reduce infarct size (IS) in ischemia-reperfusion injury of the myocardium. Inhibition of cyclooxygenase-2 (COX-2) attenuates this benefit. We investigated the effect of two widely used non-selective non-steroidal anti-inflammatory drugs (NSAIDs) with different degree of anti-COX-2 activity on atorvastatin-mediated preconditioning. Wistar rats received oral atorvastatin (10 mg∙kg-1∙day-1), naproxen (10 mg∙kg-1∙day-1), diclofenac (8 mg∙kg-1∙day-1), atorvastatin+naproxen, atorvastatin+diclofenac or water for three days. Hearts were then excised and perfused in the Langendorff system. Area at risk (AR) and IS were determined after 30 min of regional ischemia and 120 min of reperfusion. Atorvastatin reduced IS by 51.3% compared with controls (14.7 ± 3.9% vs. 30.2 ± 4.6% of the AR; P < 0.001). Naproxen and diclofenac alone did not alter IS compared to control. Diclofenac completely abrogated atorvastatin-mediated protection of the myocardium. Naproxen significantly attenuated but did not eliminate the IS reducing the effect of atorvastatin when compared with controls (P = 0.038). The difference in IS between the atorvastatin+naproxen group and the atorvastatin+diclofenac group showed a strong trend in reaching statistical significance (P = 0.058), but was not found to be significant. Our results suggest relatively small, but noticeable differences among non-selective NSAIDs in their potential to attenuate statin-mediated preconditioning.

4.
Curr Pharm Des ; 22(42): 6451-6458, 2016.
Article in English | MEDLINE | ID: mdl-27526788

ABSTRACT

BACKGROUND: It is known that statins possess beneficial cardioprotective effects irrespective of lipidlowering action and that cardiac injury due ischemia/reperfusion is associated with Ca2+ dysregulation resulting in contractile dysfunction. OBJECTIVE: With this background, we tested a hypothesis that simvastatin influences signaling of Ca2+/calmodulindependent protein kinase IIδ (CaMKIIδ), a protein kinase regulating both Ca2+ homeostasis and thick filament function, and thereby might underlie the mitigation of ischemia/reperfusion (I/R)-induced cardiac dysfunction. METHOD: Isolated hearts of control and simvastatin-treated (p.o. 10 mg/kg, 5 days) rats were subjected to global I and R and Western blotting was used to study the expression/activation of certain signaling proteins. RESULTS: Simvastatin treatment did not modify the plasma lipid levels; however, it recovered depressed cardiac performance and reduced reperfusion arrhythmias without affecting the activation of CaMKIIδ through phosphorylation of Thr287. Activation of its downstreams, such as phospholamban (PLN) and cardiac myosin-binding protein C (cMyBP-C) at Thr17 and Ser282, respectively, was in accordance with the levels of pThr287-CaMKIIδ. Total expression of these proteins, however, did not follow the same pattern and was either unchanged (CaMKIIδ, cMYBP-C) or increased (PLN). Likewise, PLN/SERCA2a (sarco/endoplasmic reticulum Ca2+-ATPase 2a) ratio in I/R hearts was unaffected by the treatment. On the other hand, simvastatin reversed the increased protein expression of protein phosphatase 1ß (PP1ß), but not protein phosphatase 2A (PP2A), in I/R hearts. CONCLUSION: A lower rate of dephosphorylation and thereby a delay in inactivation of phosphorylated proteins due to a decrease in PP1ß, rather than effects on phosphorylation of CaMKIIδ and its downstreams, such as PLN and cMyBP-C, may underlie beneficial effects of simvastatin in I/R hearts.


Subject(s)
Calcium/metabolism , Cardiotonic Agents/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Muscle Proteins/metabolism , Myocardial Reperfusion Injury/metabolism , Simvastatin/pharmacology , Administration, Oral , Animals , Lipids/blood , Male , Myofibrils/drug effects , Myofibrils/metabolism , Rats , Rats, Wistar , Simvastatin/administration & dosage
5.
Data Brief ; 7: 730-4, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27054186

ABSTRACT

Content of particular proteins indicating cellular injury due to apoptosis and necrosis has been investigated in ischemic/reperfused (IR) hearts and ischemic/reperfused hearts treated with CaMKII inhibitor and/or AT1 receptor inhibitor. This data article provides information in support of the original research article "Oxidative activation of CaMKIIδ in acute myocardial ischemia/reperfusion injury: a role of angiotensin AT1 receptor-NOX2 signaling axis" [1].

6.
Eur J Pharmacol ; 771: 114-22, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26694801

ABSTRACT

During ischemia/reperfusion (IR), increased activation of angiotensin AT1 receptors recruits NADPH oxidase 2 (NOX2) which contributes to oxidative stress. It is unknown whether this stimulus can induce oxidative activation of Ca(2+)/calmodulin-dependent protein kinase IIδ (CaMKIIδ) leading into the aggravation of cardiac function and whether these effects can be prevented by angiotensin AT1 receptors blockade. Losartan, a selective AT1 blocker, was used. Its effects were compared with effects of KN-93, an inhibitor of CaMKIIδ. Global IR was induced in Langendorff-perfused rat hearts. Protein expression was evaluated by immunoblotting and lipoperoxidation was measured by TBARS assay. Losartan improved LVDP recovery by 25%; however, it did not reduce reperfusion arrhythmias. Oxidized CaMKIIδ (oxCaMKIIδ) was downregulated at the end of reperfusion compared to before ischemia and losartan did not change these levels. Phosphorylation of CaMKIIδ mirrored the pattern of changes in oxCaMKIIδ levels. Losartan did not prevent the higher lipoperoxidation due to IR and did not influence NOX2 expression. Inhibition of CaMKII ameliorated cardiac IR injury; however, this was not accompanied with changes in the levels of either active form of CaMKIIδ in comparison to the angiotensin AT1 receptor blockade. In spite of no changes of oxCaMKIIδ, increased cardiac recovery of either therapy was abolished when combined together. This study showed that oxidative activation of CaMKIIδ is not elevated at the end of R phase. NOX2-oxCAMKIIδ signaling is unlikely to be involved in cardioprotective action of angiotensin AT1 receptor blockade which is partially abolished by concomitant CaMKII inhibition.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Myocardial Reperfusion Injury/enzymology , NADPH Oxidases/drug effects , Receptor, Angiotensin, Type 1/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 1/antagonists & inhibitors , Down-Regulation , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Lipid Peroxidation/drug effects , Losartan/pharmacology , Male , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Phosphorylation , Rats , Rats, Wistar , Sulfonamides/pharmacology
7.
Mol Cell Biochem ; 388(1-2): 269-76, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24347176

ABSTRACT

While Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) has been suggested to be an important protein regulating heart function upon ischemia/reperfusion (I/R), the mechanisms responsible are not fully known. Furthermore, it is not known whether CaMKII activation can modulate necroptosis, a recently described form of programmed cell death. In order to investigate these issues, Langendroff-perfused rat hearts were subjected to global ischemia and reperfusion, and CaMKII inhibition was achieved by adding the CaMKII inhibitor KN-93 (0.5 µmol/dm(3)) to the perfusion solution before the induction of ischemia. Immunoblotting was used to detect changes in expression of proteins modulating both necroptotic and apoptotic cell death. CaMKII inhibition normalized I/R induced increases in expression of necroptotic RIP1 and caspase-8 along with proteins of the intrinsic apoptotic pathway, namely cytochrome c and caspase-9. In addition, it increased the Bcl-2/Bax ratio and reduced caspase-3 and cleaved PARP1 content suggesting reduction of cell death. These changes coexisted with improvement of postischemic contractile function. On the other hand, there was no correlation between levels of pT287-CaMKIIδ and LVDP recovery after I/R. These results demonstrate for the first time that CaMKII inhibition may mitigate cardiac contractile dysfunction, at least partially, by limiting the contents of not only apoptotic, but also necroptotic proteins. Phosphorylation of CaMKII seems unlikely to determine the degree of postischemic recovery of contractile function.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Myocardial Contraction/physiology , Myocardial Reperfusion Injury/pathology , Necrosis/pathology , Animals , Benzylamines/pharmacology , Caspase 3/biosynthesis , Caspase 8/biosynthesis , Caspase 9/biosynthesis , Cytochromes c/biosynthesis , Heart Ventricles/metabolism , Male , Nuclear Pore Complex Proteins/biosynthesis , Phosphorylation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , RNA-Binding Proteins/biosynthesis , Rats , Rats, Wistar , Sulfonamides/pharmacology , bcl-2-Associated X Protein/biosynthesis
8.
Can J Physiol Pharmacol ; 91(8): 608-16, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23889688

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors regulating cardiac lipid metabolism and energy homeostasis. Although the activation of PPARs has been implicated in cardioprotection, the molecular mechanisms are largely unexplored. In this study, we aimed to investigate the effect of the PPAR-α agonist WY-14643 (WY), mimicking a delayed effect of preconditioning in rat hearts exposed to acute ischaemia-reperfusion (I/R) 24 h later, and to define whether antioxidative and antiapoptotic mechanisms are involved. Treatment with WY markedly attenuated post-ischaemic contractile dysfunction (as evidenced by the reduced infarct size), the higher left ventricular developed pressure (LVDP) recovery, and the decreased occurrence of arrhythmias. These effects were abolished in the presence of the PPAR-α antagonist MK886. Heme oxygenase-1, a key antioxidative enzyme implicated in cytoprotection, was upregulated in response to WY at baseline, but was markedly reduced after I/R, indicating reduced oxidative stress. WY treatment was also associated with decreased mRNA levels and enzymatic activity of matrix metalloproteinase-2, and increased ratios of Bcl-2:Bax proteins. These results indicate that PPAR-α activation by its selective ligand WY may confer delayed preconditioning-like protection in rat hearts subjected to I/R by modulating oxidative stress, activation of matrix metalloproteinase-2, and expression of Bcl-2 and Bax.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Myocardial Reperfusion Injury , PPAR alpha/agonists , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Pyrimidines/pharmacology , Animals , Heart Function Tests , In Vitro Techniques , Male , Myocardial Contraction/drug effects , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Wistar , Time Factors , Ventricular Function, Left/drug effects
9.
Can J Physiol Pharmacol ; 90(8): 1135-44, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22809038

ABSTRACT

Peroxisome proliferator-activated receptors (PPAR) regulate the expression of genes involved in lipid metabolism, energy production, and inflammation. Their role in ischaemia-reperfusion (I/R) is less clear, although research indicates involvement of PPARs in some forms of preconditioning. This study aimed to explore the effects of PPAR-α activation on the I/R injury and potential cardioprotective downstream mechanisms involved. Langendorff-perfused hearts of rats pretreated with the selective PPAR-α agonist WY-14643 (WY, pirinixic acid; 3 mg·(kg body mass)·day(-1); 5 days) were subjected to 30 min ischaemia - 2 h reperfusion with or without the phosphatidylinositol 3-kinase (PI3K)-Akt inhibitor wortmannin for the evaluation of functional (left ventricular developed pressure, LVDP) recovery, infarct size (IS), and reperfusion-induced arrhythmias. A 2-fold increase in baseline PPAR-α mRNA levels (qPCR) in the WY-treated group and higher post-I/R PPAR-α levels compared with those in untreated controls were accompanied by similar changes in the expression of PPAR-α target genes PDK4 and mCPT-1, regulating glucose and fatty acid metabolism, and by enhanced Akt phosphorylation. Post-ischaemic LVDP restoration in WY-treated hearts reached 60% ± 9% of the pre-ischaemic values compared with 24% ± 3% in the control hearts (P < 0.05), coupled with reduced IS and incidence of ventricular fibrillation that was blunted by wortmannin. Results indicate that PPAR-α up-regulation may confer preconditioning-like protection via metabolic effects. Downstream mechanisms of PPAR-α-mediated cardioprotection may involve PI3K-Akt activation.


Subject(s)
Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/physiopathology , PPAR alpha/physiology , Phosphatidylinositol 3-Kinase/physiology , Proto-Oncogene Proteins c-akt/physiology , Androstadienes/pharmacology , Animals , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Chymases/biosynthesis , Disease Models, Animal , Male , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/metabolism , PPAR alpha/biosynthesis , Peroxisome Proliferators/antagonists & inhibitors , Peroxisome Proliferators/pharmacology , Peroxisome Proliferators/therapeutic use , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinases/biosynthesis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects , Up-Regulation/physiology , Wortmannin
10.
Can J Physiol Pharmacol ; 90(8): 1127-34, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22571766

ABSTRACT

Although Ca(2+)/calmodulin-dependent protein kinase II delta (CaMKIIδ) has been implicated in development of different phenotypes of myocardial ischaemia-reperfusion injury, its involvement in arrhythmogenesis and cardiac stunning is not sufficiently elucidated. Moreover, the mechanisms by which CaMKIIδ mediates disturbances in excitation-contraction coupling, are not exactly known. To investigate this, KN-93 (0.5 µmol/L), a CaMKII inhibitor, was administered before induction of global ischaemia and reperfusion in isolated Langendorff-perfused rat hearts. Expression of CaMKIIδ and the sarcollemal Ca(2+)-cycling proteins, known to be activated during reperfusion, was analyzed using immunoblotting. KN-93 reduced reperfusion-induced ectopic activity and the incidence of ventricular fibrillation. Likewise, the severity of arrhythmias was lower in KN-treated hearts. During the pre-ischaemia phase, neither inotropic nor chronotropic effects were elicited by KN-93, whereas post-ischaemic contractile recovery was significantly improved. Ischaemia-reperfusion increased the expression of CaMKIIδ and sodium-calcium exchanger (NCX1) proteins without any influence on the protein content of alpha 1c, a pore-forming subunit of L-type calcium channels (LTCCs). On the other hand, inhibition of CaMKII normalized changes in the expression of CaMKIIδ and NCX1. Taken together, CaMKIIδ seems to regulate its own turnover and to be an important component of cascade integrating NCX1, rather than LTCCs that promote ischaemia-reperfusion-induced contractile dysfunction and arrhythmias.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/biosynthesis , Heart/physiopathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Sarcolemma/metabolism , Up-Regulation/physiology , Animals , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/drug therapy , Benzylamines/pharmacology , Benzylamines/therapeutic use , Calcium Channels, L-Type/biosynthesis , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Disease Models, Animal , Heart/drug effects , In Vitro Techniques , Male , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Rats , Rats, Wistar , Sodium-Calcium Exchanger/biosynthesis , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Up-Regulation/drug effects
11.
Gen Physiol Biophys ; 30(4): 329-41, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22131314

ABSTRACT

Peroxisome proliferator-activated receptors (PPAR), ligand-activated transcription factors, belong to the nuclear hormone receptor superfamily regulating expression of genes involved in different aspects of lipid metabolism, inflammation and cardiac energy production. Activation of PPAR-α isoform by its natural ligands, fatty acids (FA) and eicosanoids, promotes mitochondrial FA oxidation as the primary ATP-generating pathway. On the other hand, PPAR-γ regulates lipid anabolism or storage, while, until recently, the function of PPAR-ß/δ has been less explored. Under conditions associated with acute or chronic oxygen deprivation, PPAR-α modulates expression of genes that determine substrate switch (FA vs. glucose) aimed at maintenance of basic cardiac function. Although PPAR-α and PPAR-γ synthetic agonists, hypolipidemic and antidiabetic drugs, have been reported to protect the heart against ischemia/reperfusion injury, it is still a matter of debate whether PPAR activation plays a beneficial or detrimental role in myocardial response to ischemia, in particular, in pathological conditions. This article reviews some findings demonstrating the impact of PPAR activation on cardiac resistance to ischemia in normal and pathologically altered heart. Specifically, it addresses the issue of susceptibility to ischemia in the diabetic myocardium, with particular regards to the role of PPAR. Finally, involvement of PPAR in the mechanisms of lipid-independent cardioprotective effects of some hypolipidemic drugs is also discussed.


Subject(s)
Myocardial Ischemia/pathology , Peroxisome Proliferator-Activated Receptors/physiology , Animals , Cardiotonic Agents/pharmacology , Diabetes Complications/metabolism , Heart/physiology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation , Ligands , Models, Biological , Myocardial Ischemia/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Reperfusion Injury/metabolism , Tissue Distribution , Transcriptional Activation
12.
Can J Physiol Pharmacol ; 88(4): 465-71, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20555415

ABSTRACT

Quercetin is a plant-derived bioflavonoid with potentially beneficial effects on the cardiovascular system. Studies focused on the efficiency of flavonoids against ischemia-reperfusion (I/R) injury have demonstrated that quercetin exerts robust protective effects in renal, cerebral, and hepatic I/R models. However, there is only limited evidence about the effect of quercetin on myocardial I/R injury. Therefore, the aim of the current study was to examine the effect of quercetin on isolated rat heart during ischemia and reperfusion. Rat hearts perfused according to Langendorff at 37 degrees C were examined during 25 min global ischemia followed by 120 min reperfusion. Quercetin (15 micromol/L) was administered either 15 min before ischemia (group Q1), or during the entire reperfusion period (group Q2). Changes in functional parameters of the hearts were measured during the initial 40 min of reperfusion. At the end of the experiment, the hearts were stained with tetrazolium to estimate the size of infarction (IS). Our study showed that quercetin improved postischemic recovery of functional parameters of isolated hearts in both treated groups. This improvement was manifested by significantly higher values of left ventricular developed pressure (LVDP) and the maximal rates of pressure development and fall (+(dP/dt)max and -(dP/dt)max) and by significantly lower increase of end-diastolic pressure. Coronary flow was not significantly changed during reperfusion in the group treated before ischemia, but was significantly increased in the group treated during reperfusion. Quercetin also significantly reduced IS in both groups, more markedly in postischemically treated group. We conclude that acute quercetin treatment exerts significant positive effects on isolated hearts during I/R injury. These results are consistent with the beneficial effects of quercetin and other flavonoids on the cardiovascular system.


Subject(s)
Cardiotonic Agents/therapeutic use , Heart/drug effects , Myocardial Reperfusion Injury/drug therapy , Quercetin/therapeutic use , Animals , Blood Pressure/drug effects , Cardiotonic Agents/pharmacology , Heart/physiopathology , Male , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/physiopathology , Quercetin/pharmacology , Rats , Rats, Wistar , Ventricular Pressure/drug effects
13.
Can J Physiol Pharmacol ; 87(12): 1028-36, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20029539

ABSTRACT

Peroxisome proliferator-activated receptors (PPAR), which are key transcriptional regulators of lipid metabolism and energy production, have been suggested to play an important role in myocardial ischaemia-reperfusion (I/R) injury. Their role in cardioprotection, however, is not yet fully elucidated. Statins have shown beneficial effects on I/R damage beyond lipid lowering, and some of their cardioprotective cholesterol-independent effects may be related to the regulation of PPAR. To clarify this issue, we explored a potential link between a response to I/R and changes in cardiac PPARalpha protein and gene expression in simvastatin-treated normocholesterolaemic rats. After 5 days of treatment with simvastatin (10 mg/kg per day, p.o.), Langendorff-perfused hearts were subjected to 30 min regional ischaemia (occlusion of the left anterior descending coronary artery) or global ischaemia and 2 h reperfusion for the evaluation of the infarct size (triphenyltetrazolium chloride and planimetry; as percentage of risk area), ischaemic arrhythmias, and postischaemic contractile recovery. Baseline PPARalpha mRNA and protein levels were increased by 3-fold and 2-fold, respectively, in simvastatin-treated hearts compared with the untreated controls. Simvastatin-treated hearts exhibited smaller size of infarction (11.5% +/- 0.4% vs. 33.7% +/- 4% in controls; p < 0.01), improved postischaemic contractile recovery, and lower severity of arrhythmias during ischaemia and early reperfusion. Enhanced resistance to I/R injury was associated with preservation of mRNA and protein levels of PPARalpha in contrast to their marked downregulation in controls. In conclusion, statin-induced changes in the expression of PPARalpha may contribute to attenuation of myocardial I/R injury and thus suggest the involvement of cardioprotective mechanisms independent of inhibition of HMG-CoA reductase.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myocardial Ischemia/physiopathology , Peroxisome Proliferator-Activated Receptors/biosynthesis , Simvastatin/pharmacology , Animals , Arrhythmias, Cardiac/physiopathology , Cholesterol/blood , Gene Expression/genetics , Gene Expression/physiology , Immunoblotting , Male , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , PPAR alpha/biosynthesis , PPAR alpha/drug effects , PPAR alpha/physiology , Peroxisome Proliferator-Activated Receptors/drug effects , Peroxisome Proliferator-Activated Receptors/physiology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
14.
Can J Physiol Pharmacol ; 87(12): 1055-62, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20029542

ABSTRACT

Opening of mitochondrial KATP channels (mitoKATP) has been reported to underlie protection against ischaemia-reperfusion injury induced by ischaemic preconditioning (I-PC); however, the molecular mechanisms of its antiarrhythmic effect have not been fully elucidated. We explored the involvement of phosphatidylinositol 3-kinase (PI3K)/Akt in the PC-like effect of mitoKATP opener diazoxide with particular regard to its role in protection against ischaemia-induced arrhythmias. Langendorff-perfused rat hearts were subjected to 30 min LAD occlusion with or without a prior 15 min of perfusion with diazoxide (50 micromol/L) given either alone (D-PC) or in combination with the PI3K/Akt inhibitor wortmannin (100 nmol/L). In an additional protocol, ischaemia was followed by 2 h reperfusion for infarct size (IS) determination (tetrazolium staining). The total number of premature ventricular complexes over the whole period of ischaemia, episodes of ventricular tachycardia and its duration were significantly lower in the D-PC group than in the non-preconditioned controls (158 +/- 20, 2 +/- 0.6 and 4.6 +/- 1.8 s vs. 551 +/- 61, 11 +/- 2 and 42 +/- 8 s, respectively; p < 0.05), concomitant with a 62% reduction in the size of infarction. Wortmannin modified neither arrhythmogenesis nor IS in the non-preconditioned hearts. Bracketing of diazoxide with wortmannin did not reverse the antiarrhythmic protection, whereas the IS-limiting effect was blunted. The results indicate that in contrast with the positive role of PI3K/Akt in protection against lethal myocardial injury, its activity is not involved in suppression of ischaemia-induced arrhythmias conferred by mitoKATP opening in the rat heart.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Mitochondria, Heart/physiology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/physiopathology , Phosphatidylinositol 3-Kinases/physiology , Potassium Channels/physiology , Proto-Oncogene Proteins c-akt/physiology , Androstadienes/pharmacology , Animals , Arrhythmias, Cardiac/drug therapy , Diazoxide/pharmacology , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/drug therapy , Phosphatidylinositol 3-Kinases/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Rats , Rats, Wistar , Vasodilator Agents/pharmacology , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...