Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Cardiovasc Drugs Ther ; 31(2): 119-132, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28447181

ABSTRACT

PURPOSE: We assessed whether (1) dapagliflozin (Dapa, an SGLT2-inhibitor) attenuates the deterioration of heart function Nlrp3 and inflammasome activation in diabetic mice. (2) The effects can be augmented with saxagliptin (Saxa), a DDP4-inhibitor. (3) Dapa effect is possibly SGLT2-independent on cardiofibroblasts in vitro. METHODS: Type 2 diabetic (BTBR ob/ob) and wild-type (WT) mice received vehicle, Dapa, or Dapa+Saxa for 8 weeks. Glucose tolerance test and echocardiogram were performed. Cardiofibroblasts from WT and BTBR hearts were incubated with Dapa and exposed to LPS. RESULTS: Left ventricular ejection fraction (LVEF) was 81 ± 1% in the WT and 53 ± 1% in the T2D-cont mice. Dapa and Dapa+Saxa improved LVEF to 68 ± 1 and 74.6 ± 1% in the BTBR mice (p < 0.001). The mRNA levels of NALP3, ASC, IL-1ß, IL-6, caspase-1, and TNFα were significantly higher in the BTBR compared to the WT hearts; and Dapa and Dapa+Saxa significantly attenuated these levels. Likewise, protein levels of NLRP3, TNFα, and caspase-1 were higher in the BTBR compared to the WT hearts and Dapa, and to a greater extent Dapa+Saxa, attenuated the increase in the BTBR mice. Collagen-1 and collagen-3 mRNA levels significantly increased in the BTBR mice and these increases were attenuated by Dapa and Dapa+Saxa. P-AMPK/total-AMPK ratio was significantly lower in the BTBR mice than in the WT mice. Dapa and Dapa+Saxa equally increased the ratio in the BTBR mice. This in vitro study showed that NALP3, ASC, IL-1ß, and caspase-1 mRNA levels were higher in the BTBR cardiofibroblasts and attenuated with Dapa. The effect was AMPK-dependent and SGLT1-independent. CONCLUSIONS: Dapa attenuated the activation of the inflammasome, fibrosis, and deterioration of LVEF in BTBR mice. The anti-inflammatory, anti-fibrotic effects are likely SGLT2- and glucose-lowering-independent, as they were replicated in the in vitro model. The effects on remodeling were augmented when Saxa was added to Dapa. Yet, adding Saxa to Dapa did not result in a greater effect on myocardial fibrosis and collagen levels.


Subject(s)
Adamantane/analogs & derivatives , Benzhydryl Compounds/pharmacology , CARD Signaling Adaptor Proteins/antagonists & inhibitors , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/prevention & control , Dipeptides/pharmacology , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Inflammasomes/antagonists & inhibitors , Kidney Tubules, Proximal/drug effects , Myocytes, Cardiac/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors , AMP-Activated Protein Kinases/metabolism , Adamantane/pharmacology , Animals , Apoptosis/drug effects , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/enzymology , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Drug Therapy, Combination , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibrosis , Inflammasomes/genetics , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Kidney Tubules, Proximal/metabolism , Male , Mice, Inbred C57BL , Mice, Obese , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction/drug effects , Sodium-Glucose Transporter 2/metabolism , Stroke Volume/drug effects , Time Factors , Ventricular Function, Left/drug effects
2.
Arterioscler Thromb Vasc Biol ; 35(8): 1805-14, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26044583

ABSTRACT

OBJECTIVE: In addition to P2Y12 receptor antagonism, ticagrelor inhibits adenosine cell uptake. Prior data show that 7-day pretreatment with ticagrelor limits infarct size. We explored the acute effects of ticagrelor and clopidogrel on infarct size and potential long-term effects on heart function. APPROACH AND RESULTS: Rats underwent 30-minute ischemia per 24-hour reperfusion. (1) Ticagrelor (10 or 30 mg/kg) or clopidogrel (12.5 mg/kg) was given via intraperitoneal injection 5 minutes before reperfusion. (2) Rats received ticagrelor acute (intraperitoneal; 30 mg/kg), chronic (oral; 300 mg/kg per day) for 4 weeks starting 1 day after reperfusion or the combination (acute+chronic). Another group received clopidogrel (intraperitoneal [12.5 mg/kg]+oral [62.5 mg/kg per day]) for 4 weeks. (1) Ticagrelor dose-dependently reduced infarct size, 10 mg/kg (31.5%±1.8%; P<0.001) and 30 mg/kg (21.4%±2.6%; P<0.001) versus control (45.3±1.7%), whereas clopidogrel had no effect (42.4%±2.6%). Ticagrelor, but not clopidogrel, increased myocardial adenosine levels, increased phosphorylation of Akt, endothelial NO synthase, and extracellular-signal-regulated kinase 1/2 4 hours after reperfusion and decreased apoptosis. (2) After 4 weeks, left ventricular ejection fraction was reduced in the vehicle-treated group (44.8%±3.5%) versus sham (77.6%±0.9%). All ticagrelor treatments improved left ventricular ejection fraction, acute (69.5%±1.6%), chronic (69.2%±1.0%), and acute+chronic (76.3%±1.2%), whereas clopidogrel had no effect (37.4%±3.7%). Ticagrelor, but not clopidogrel, attenuated fibrosis and decreased collagen-III mRNA levels 4 weeks after ischemia/reperfusion. Ticagrelor, but not clopidogrel, attenuated the increase in proinflammatory tumor necrosis factor-α, interleukin-1ß, and interleukin-18, and increased anti-inflammatory 15-epi-lipoxin-A4 levels. CONCLUSIONS: Ticagrelor, but not clopidogrel, administered just before reperfusion protects against reperfusion injury. This acute treatment or chronic ticagrelor for 4 weeks or their combination improved heart function, whereas clopidogrel, despite achieving a similar degree of platelet inhibition, had no effect.


Subject(s)
Adenosine/analogs & derivatives , Cardiotonic Agents/pharmacology , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Purinergic P2Y Receptor Antagonists/pharmacology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Adenosine/administration & dosage , Adenosine/pharmacology , Administration, Oral , Animals , Apoptosis/drug effects , Cardiotonic Agents/administration & dosage , Clopidogrel , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrosis , Injections, Intraperitoneal , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Purinergic P2Y Receptor Antagonists/administration & dosage , Rats, Sprague-Dawley , Signal Transduction/drug effects , Ticagrelor , Ticlopidine/analogs & derivatives , Ticlopidine/pharmacology , Time Factors
3.
Cardiovasc Drugs Ther ; 28(5): 447-57, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25106875

ABSTRACT

PURPOSE: Statins increase the incidence of new onset diabetes. Prolonged statin therapy upregulates PTEN expression. PTEN levels are also elevated in diabetic animals. Activation of protein kinase A by cAMP decreases PTEN expression. We assessed whether prolonged treatment with rosuvastatin (ROS) induces glucose intolerance by upregulating Phosphatase and Tensin Homologue on Chromosome 10 (PTEN) in mice receiving normal (ND) or Western Diet (WD) and whether concomitant treatment with cilostazol (CIL, a phosphodiesterase-3 inhibitor) attenuates the effects. METHODS: PTEN(loxp/cre) or PTEN(+/-) mice received ND or WD without or with ROS (10 mg/kg/day). Wild-type mice received ND or WD without or with ROS, CIL (10 mg/kg/day), or ROS+CIL for 30 days. Fasting insulin and glucose tolerance test were measured as well as PTEN and P-AKT levels in skeletal muscle. RESULTS: Serum glucose after intraperitoneal injection of glucose was higher in PTEN(loxp/cre) mice receiving WD or ROS and especially WD+ROS. Levels were lower in PTEN(+/-) mice compared to PTEN(loxp/cre) in each treatment group. CIL decreased glucose levels in mice receiving WD, ROS and their combination. Insulin levels were higher in the WD+ROS group. CIL decreased insulin in mice receiving WD+ROS. WD, ROS and especially their combination increased PTEN and decreased P-AKT levels. CIL attenuated the effect of WD, ROS and their combination. CONCLUSIONS: Long-term ROS can induce diabetes by upregulating PTEN. CIL attenuates these changes. Partial knockdown of PTEN also ameliorates ROS-induced insulin resistance. Further studies are needed to assess the effects of increasing cAMP levels to prevent the induction of diabetes by statins.


Subject(s)
Diabetes Mellitus, Type 2/chemically induced , Fluorobenzenes/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Insulin Resistance/genetics , PTEN Phosphohydrolase/biosynthesis , Pyrimidines/adverse effects , Sulfonamides/adverse effects , Up-Regulation/drug effects , Animals , Blood Glucose/drug effects , Blood Glucose/genetics , Cilostazol , Diabetes Mellitus, Type 2/blood , Diet, Western , Fluorobenzenes/administration & dosage , Fluorobenzenes/antagonists & inhibitors , Fluorobenzenes/pharmacology , Gene Knockdown Techniques , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Insulin/blood , Mice , Muscle, Skeletal/metabolism , Phosphodiesterase 3 Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/administration & dosage , Pyrimidines/antagonists & inhibitors , Pyrimidines/pharmacology , Rosuvastatin Calcium , Sulfonamides/administration & dosage , Sulfonamides/antagonists & inhibitors , Sulfonamides/pharmacology , Tetrazoles/pharmacology
4.
Hypertension ; 61(5): 1008-13, 2013 May.
Article in English | MEDLINE | ID: mdl-23460283

ABSTRACT

Nebivolol is a selective ß1-blocker with nitric oxide-enhancing effects. MicroRNAs are small noncoding RNA molecules that downregulate gene expression. We compared the effects of nebivolol and atenolol, a first generation ß1-selective blocker, on left ventricular hypertrophy, fibrosis, and function and microRNA expression in a rodent model of hypertension. Dahl salt-sensitive rats received either low-salt chow (control) or AIN-76A high-salt (8% NaCl) diet and randomized to vehicle (high-salt), nebivolol (20 mg/kg per day), or atenolol (50 mg/kg per day) for 8 weeks. High-salt induced left ventricular hypertrophy and fibrosis and decreased the expression of miR-27a, -29a, and -133a. Nebovolol attenuated deterioration of left ventricular systolic function, remodeling, and fibrosis more than atenolol, despite similar effects on heart rate and blood pressure. Nebivolol, but not atenolol, prevented the decrease in miR-27a and -29a induced by high-salt. Nebivolol and atenolol equally attenuated the decrease in miR-133a. In vitro overexpression of miR-27a,-29a, and -133a inhibited cardiomyocyte hypertrophy and reduced collagen expression. Both miR-27a and -29a target Sp1, and miR-133a targets Cdc42. Pharmacological inhibition of Sp1 and Cdc42 decreased myocardial fibrosis and hypertrophy. Our data support a differential microRNAs expression profile in salt-induced hypertension. Nebivolol substantially attenuated cardiac remodeling, hypertrophy, and fibrosis more than atenolol. These effects are related to attenuation of the hypertension-induced decrease in miR-27a and -29a (with a subsequent decrease in Sp1 expression) and miR-133a (with a subsequent decrease in Cdc42).


Subject(s)
Antihypertensive Agents/pharmacology , Atenolol/pharmacology , Benzopyrans/pharmacology , Ethanolamines/pharmacology , Hypertension/metabolism , MicroRNAs/drug effects , MicroRNAs/metabolism , Myocardium/metabolism , Myocardium/pathology , Adrenergic beta-Antagonists/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cells, Cultured , Disease Models, Animal , Fibrosis , Hypertension/physiopathology , Immunoglobulins/metabolism , In Vitro Techniques , Male , Nebivolol , Pilot Projects , Rats , Rats, Inbred Dahl , Rats, Sprague-Dawley , cdc42 GTP-Binding Protein/metabolism
5.
Am J Physiol Heart Circ Physiol ; 304(1): H131-41, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23103492

ABSTRACT

Glucagon-like peptide (GLP)-1 receptor activation increases intracellular cAMP with downstream activation of PKA. Cilostazol (CIL), a phosphodiesterase-3 inhibitor, prevents cAMP degradation. We assessed whether CIL amplifies the exenatide (EX)-induced increase in myocardial cAMP levels and PKA activity and augments the infarct size (IS)-limiting effects of EX in db/db mice. Mice fed a Western diet received oral CIL (10 mg/kg) or vehicle by oral gavage 24 h before surgery. One hour before surgery, mice received EX (1 µg/kg sc) or vehicle. Additional mice received H-89, a PKA inhibitor, alone or with CIL + EX. Mice underwent 30 min of coronary artery occlusion and 24 h of reperfusion. Both EX and CIL increased myocardial cAMP levels and PKA activity. Levels were significantly higher in the EX + CIL group. Both EX and CIL reduced IS. IS was the smallest in the CIL + EX group. H-89 completely blocked the IS-limiting effects of EX + CIL. EX + CIL decreased phosphatase and tensin homolog on chromosome 10 upregulation and increased Akt and ERK1/2 phosphorylation after ischemia-reperfusion. These effects were blocked by H-89. In conclusion, EX and CIL have additive effects on IS limitation in diabetic mice. The additive effects are related to cAMP-induced PKA activation, as H-89 blocked the protective effect of CIL + EX.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Myocardial Infarction/prevention & control , Myocardium/pathology , Peptides/pharmacology , Phosphodiesterase 3 Inhibitors/pharmacology , Tetrazoles/pharmacology , Venoms/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Blotting, Western , Cholesterol/blood , Cilostazol , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Disease Models, Animal , Enzyme Activation , Exenatide , Glucagon-Like Peptide-1 Receptor , Glycated Hemoglobin/metabolism , Isoquinolines/pharmacology , Lipoxins/metabolism , Male , Mice , Myocardial Infarction/blood , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Myocardium/metabolism , PTEN Phosphohydrolase/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Triglycerides/blood , Up-Regulation
6.
Cardiovasc Drugs Ther ; 26(6): 445-56, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22936458

ABSTRACT

PURPOSE: We assessed whether phosphodiesterase-III inhibition with cilostazol (Cil) augments the infarct size (IS)-limiting effects of MK0626 (MK), a dipeptidyl-peptidase-4 (DPP4) inhibitor, by increasing intracellular cAMP in mice with type-2 diabetes. METHODS: Db/Db mice received 3-day MK (0, 1, 2 or 3 mg/kg/d) with or without Cil (15 mg/kg/d) by oral gavage and were subjected to 30 min coronary artery occlusion and 24 h reperfusion. RESULTS: Cil and MK at 2 and 3 mg/kg/d significantly reduced IS. Cil and MK had additive effects at all three MK doses. IS was the smallest in the MK-3+Cil. MK in a dose dependent manner and Cil increased cAMP levels (p < 0.001). cAMP levels were higher in the combination groups at all MK doses. MK-2 and Cil increased PKA activity when given alone; however, PKA activity was significantly higher in the MK-2+Cil group than in the other groups. Both MK-2 and Cil increased myocardial levels of Ser(133) P-CREB, Ser(523) P-5-lipoxygenase, Ser(473)P-Akt and Ser(633) P-eNOS. These levels were significantly higher in the MK-2+Cil group. Myocardial PTEN (Phosphatase and tensin homolog on chromosome ten) levels were significantly higher in the Db/Db mice compared to nondiabetic mice. MK-2 and Cil normalized PTEN levels. PTEN levels tended to be lower in the combination group than in the MK and Cil alone groups. CONCLUSION: MK and Cil have additive IS-limiting effects in diabetic mice. The additive effects are associated with an increase in myocardial cAMP levels and PKA activity with downstream phosphorylation of Akt, eNOS, 5-lipoxygenase and CREB and downregulation of PTEN expression.


Subject(s)
Cyclic AMP/metabolism , Diabetes Mellitus, Type 2/metabolism , Myocardial Infarction/drug therapy , Phosphodiesterase 3 Inhibitors/pharmacology , Tetrazoles/pharmacology , Triazoles/pharmacology , Animals , Blood Glucose , Cilostazol , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Glucagon-Like Peptide 1 , Glycated Hemoglobin , Immunoblotting , Lipids/blood , Lipoxins/metabolism , Male , Membrane Proteins/metabolism , Mice , Myocardium/metabolism , PTEN Phosphohydrolase/metabolism , Phosphodiesterase 3 Inhibitors/administration & dosage , Triazoles/administration & dosage
7.
Basic Res Cardiol ; 106(6): 925-52, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21892746

ABSTRACT

Heart disease and stroke account for 65% of the deaths in people with diabetes mellitus (DM). DM and hyperglycemia cause systemic inflammation, endothelial dysfunction, a hypercoagulable state with impaired fibrinolysis and increased platelet degranulation, and reduced coronary collateral blood flow. DM also interferes with myocardial protection afforded by preconditioning and postconditioning. Newer anti-diabetic agents should not only reduce serum glucose and HbA1c levels, but also improve cardiovascular outcomes. The older sulfonylurea agent, glyburide, abolishes the benefits of ischemic and pharmacologic preconditioning, but newer sulfonylurea agents, such as glimepiride, may not interfere with preconditioning. GLP-1 analogs and sitagliptin, an oral dipeptidyl peptidase IV inhibitor, limit myocardial infarct size in animal models by increasing intracellular cAMP levels and activating protein kinase A, whereas metformin protects the heart by activating AMP-activated protein kinase. Both thiazolidinediones (rosiglitazone and pioglitazone) limit infarct size in animal models. The protective effect of pioglitazone is dependent on downstream activation of cytosolic phospholipase A(2) and cyclooxygenase-2 with subsequent increased production of 15-epi-lipoxin A(4), prostacyclin and 15-d-PGJ(2). We conclude that agents used to treat DM have additional actions that have been shown to affect the ability of the heart to protect itself against ischemia-reperfusion injury in preclinical models. However, the effects of these agents in doses used in the clinical setting to minimize ischemia-reperfusion injury and to affect clinical outcomes in patients with DM have yet to be shown. The clinical implications as well as the mechanisms of protection should be further studied.


Subject(s)
Heart/drug effects , Hypoglycemic Agents/pharmacology , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/physiopathology , Animals , Diabetes Mellitus/drug therapy , Humans
8.
Basic Res Cardiol ; 106(3): 431-46, 2011 May.
Article in English | MEDLINE | ID: mdl-21360043

ABSTRACT

Pioglitazone (PIO), a PPAR-γ agonist, limits myocardial infarct size by activating Akt and upregulating cytosolic phospholipase A(2) (cPLA(2)) and cyclooxygenase (COX)-2. However, PIO has several PPAR-γ-independent effects. We assessed whether PIO limits myocardial infarct size in PPAR-γ-knockout mice, attenuates hypoxia-reoxygenation injury and upregulates P-Akt, cPLA(2), and COX-2 expression in PPAR-γ-knockout cardiomyocytes. Cardiac-specific inducible PPAR-γ knockout mice were generated by crossing αMHC-Cre mice to PPAR-γ(loxp/loxp) mice. PPAR-γ deletion was achieved after 7 days of intraperitoneal tamoxifen (20 mg/kg/day) administration. Mice received PIO (10 mg/kg/day), or vehicle, for 3 days and underwent coronary occlusion (30 min) followed by reperfusion (4 h). We assessed the area at risk by blue dye and infarct size by TTC. Cultured adult cardiomyocytes of PPAR-γ(loxp/loxp/cre) mice without or with pretreatment with tamoxifen were incubated with or without PIO and subjected to 2 h hypoxia/2 h reoxygenation. Cardiac-specific PPAR-γ knockout significantly increased infarct size. PIO reduced infarct size by 51% in PPAR-γ knockout mice and by 55% in mice with intact PPAR-γ. Deleting the PPAR-γ gene increased cell death in vitro. PIO reduced cell death in cells with and without intact PPAR-γ. PIO similarly increased myocardial Ser-473 P-Akt, cPLA(2), and COX-2 levels after hypoxia/reoxygenation in cells with and without intact PPAR-γ. PIO limited infarct size in mice in a PPAR-γ-independent manner. PIO activated Akt, increased the expression of cPLA(2) and COX-2, and protected adult cardiomyocytes against the effects of hypoxia/reoxygenation independent of PPAR-γ activation.


Subject(s)
Cyclooxygenase 2/biosynthesis , Hypoglycemic Agents/pharmacology , Myocardial Infarction/prevention & control , Phospholipases A2, Cytosolic/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Thiazolidinediones/pharmacology , Animals , Enzyme Activation/drug effects , Gene Expression , Immunoblotting , Immunohistochemistry , Mice , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , PPAR gamma/metabolism , Pioglitazone , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Up-Regulation
9.
Physiol Genomics ; 43(10): 534-42, 2011 May 01.
Article in English | MEDLINE | ID: mdl-20959496

ABSTRACT

MicroRNAs (miRNAs) are small (∼22 nt) noncoding single-stranded RNA molecules that downregulate gene expression. Studies have shown that miRNAs control diverse aspects of heart disease, including hypertrophy, remodeling, heart failure, and arrhythmia. Recently, several studies have suggested that miRNAs contribute to ischemia-reperfusion injury by altering key signaling elements, thus making them potential therapeutic targets. By altering the expression of various key elements in cell survival and apoptosis [such as phosphoinositide 3-kinase (PI3K), phosphatase and tensin homolog deleted on chromosome 10 (PTEN), Bcl-2, Mcl-1, heat shock protein (HSP)60, HSP70, HSP20, programmed cell death 4 (Pdcd4), LRRFIP1, Fas ligand (FasL), Sirt-1, etc.], miRNAs alter the response to ischemia-reperfusion injury. Studies using various in vivo, ex vivo, and in vitro models have suggested the possible involvement of miR-1, miR-21, miR-29, miR-92a, miR-133, miR-199a, and miR-320 in ischemia-reperfusion injury and/or remodeling after myocardial infarction. Thus miRNAs could be potential therapeutic targets for the treatment of heart disease. Inhibiting miRNAs by antisense strategies or pharmacological approaches is likely to emerge as an alternative and safe method for conferring short- and intermediate-term protection against ischemia-reperfusion injury.


Subject(s)
MicroRNAs/physiology , Myocardial Reperfusion Injury/genetics , Animals , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism
10.
Ann N Y Acad Sci ; 1207: 76-82, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20955429

ABSTRACT

Statins and antiplatelet agents are currently used as therapeutic agents for patients with acute myocardial infarction. Statins limit myocardial infarct size by activating phosphatidylinositol-3-kinase (PI3K), ecto-5'-nucleotidase, Akt/endothelial nitric oxide synthase (eNOS), and the downstream effectors inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Inhibition of PI3K, adenosine receptors, eNOS, iNOS, or COX-2 abrogates the protective effects of statins. At >5 mg/kg, aspirin attenuates the myocardial infarct-size-limiting effect of statins. In contrast, the combination of low-dose atoravastatin with either the phosphodiesterase-III inhibitor cilostazol or the adenosine reuptake inhibitor dipyridamole synergistically limits infarct size. Low-dose aspirin with dipyridamole started during ischemia augmented the infarct-size-limiting effects of simvastatin. In contrast, high-dose aspirin blocked the protective effect of simvastatin. The combination of dipyridamole with low-dose aspirin and simvastatin resulted in the smallest infarct size. According to the most current data available, we believe that antiplatelet regimens may require modification for patients who are receiving statins.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Myocardial Reperfusion Injury/prevention & control , Platelet Aggregation Inhibitors/administration & dosage , Aspirin/administration & dosage , Cilostazol , Dipyridamole/administration & dosage , Drug Interactions , Humans , Models, Cardiovascular , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Simvastatin/administration & dosage , Tetrazoles/administration & dosage
11.
Cardiovasc Drugs Ther ; 24(5-6): 391-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20640495

ABSTRACT

PURPOSE: Statins protect against ischemia-reperfusion injury and limit myocardial infarct size (IS). This effect is dependent on increased generation of adenosine by ecto-5' nucleotidase and downstream activation of cyclooxygenase-2 (COX2). Dipyridamole (DIP) augments the IS-limiting effects of statins by blocking the cellular reuptake of adenosine; whereas aspirin (ASA) attenuates the effect by inhibiting COX2. We studied the effect of acute administration of DIP, ASA and their combination on the IS-limiting effect of simvastatin (SIM). METHODS: Rats received oral SIM (10 mg/kg/d) or vehicle for 3 days. Rats underwent 30 min of coronary artery occlusion and 4 h reperfusion. After 5 min of ischemia rats received i.v. DIP (5 mg/kg), ASA (20 mg/kg or 2 mg/kg) or DIP+ASA (2 mg/kg) or vehicle alone. Ischemia area at risk (AR) was assessed by blue dye and IS by TTC. Myocardial samples were analyzed for the activation of Akt, ERK 1/2, endothelial nitric oxide synthase (eNOS), and cyclic-AMP-response-element-binding-protein (CREB). RESULTS: SIM limited IS. High- or low-dose ASA alone had no effect on IS. DIP alone or with low-dose ASA significantly reduced IS. Low-dose ASA did not attenuate the SIM effect, whereas high-dose ASA completely blocked the effect. The combination of DIP+low-dose ASA+SIM resulted in the smallest IS. Both SIM and DIP+low-dose ASA augmented Akt phosphorylation and their effect was additive. Both SIM and DIP+low-dose ASA augmented eNOS, ERK 1/2 and CREB phosphorylation. CONCLUSIONS: During acute myocardial ischemia, DIP alone or with low-dose ASA limits IS and does not attenuate the IS-limiting effect of SIM as high-dose ASA.


Subject(s)
Aspirin/pharmacology , Dipyridamole/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myocardial Infarction/prevention & control , Simvastatin/pharmacology , Animals , CREB-Binding Protein/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
12.
Cardiovasc Drugs Ther ; 24(2): 107-20, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20379769

ABSTRACT

PURPOSE: We assessed the effects of TAK-491 (a newly designed potent and selective ARB) alone and in combination with pioglitazone (PIO) on myocardial infarct size (IS). METHODS: Rats received TAK-491 (0.3, 1, 3, or 10 mgkg(-1)d(-1)), PIO (1.0 or 2.5 mgkg(-1)d(-1)), or PIO 2.5 mgkg(-1)d(-1) with TAK-491 (1 or 3 mgkg(-1)d(-1)) for 4 days. On day 5 rats underwent 30-minute coronary artery occlusion and 4-hour reperfusion. Area at risk (AR) was assessed by blue dye and IS by TTC. Left ventricular (LV) dimensions and function was assessed by echocardiography 35 days after infarction. RESULTS: TAK (1.0-10 mgkg(-1)d(-1)), PIO (1.0 to 2.5 mgkg(-1)d(-1)), PIO2.5+TAK1.0, and PIO2.5+TAK3.0 significantly reduced IS. IS was the smallest in the TAK 10.0, followed by PIO+TAK 3.0. The protective effects of TAK and PIO were additive, as IS was smaller in the PIO2.5+TAK1.0 than in PIO 2.5 alone (p = 0.008) or TAK1.0 alone (p = 0.002); and in PIO2.5+TAK3.0 than in PIO alone (p < 0.001) or TAK3.0 alone (p < 0.001). TAK, PIO and their combination tended to attenuate LV remodeling and improved LV function 35 days after infarction; however, the differences among individual groups were not statistically significant. Both TAK-491 and PIO increased calcium-dependent nitric oxide synthase activity, whereas only PIO increased COX2 expression and activity. Both PIO and TAK-491 increased Akt, ERK 1/2 and eNOS phosphorylation and inhibited BAX activation. CONCLUSIONS: TAK-491 and PIO independently limited myocardial IS in a dose-dependent fashion; and the effects were additive. The mechanism of protection and the role of TAK-491 in this clinical setting should be further investigated.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Myocardial Infarction/prevention & control , Thiazolidinediones/therapeutic use , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Apoptosis/drug effects , Blood Pressure/physiology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Drug Therapy, Combination , Heart Rate/physiology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/pathology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Male , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/drug effects , Pioglitazone , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Thiazolidinediones/pharmacology , Ventricular Function, Left/physiology , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Am J Physiol Heart Circ Physiol ; 298(5): H1454-65, 2010 May.
Article in English | MEDLINE | ID: mdl-20207816

ABSTRACT

Pioglitazone (PIO) and glucagon-like peptide-1 (GLP-1) analogs limit infarct size (IS) in experimental models. The effects of the dipeptidyl-peptidase-IV inhibitors, which increase the endogenous levels of GLP-1, on myocardial protection, are unknown. We studied whether sitagliptin (SIT) and PIO have additive effects on IS limitation in the mouse. Mice received 3-day or 14-day oral SIT (300 mg.kg(-1).day(-1)), PIO (5 mg.kg(-1).day(-1)), SIT + PIO, or vehicle. In addition, mice received intravenous H-89 [20 mg/kg, a protein kinase A (PKA) inhibitor] or vehicle 1 h before ischemia. Rats underwent 30 min myocardial ischemia and 4 h reperfusion. SIT, PIO, and SIT + PIO for 3 days significantly reduced IS (24.3 +/- 2.7, 23.0 +/- 0.8, and 14.7 +/- 0.9%) compared with controls (46.2 +/- 2.8%). H-89 completely blocked the effect of SIT and partially blocked the PIO effect. SIT, but not PIO, increased cAMP levels. PKA activity was increased by PIO and to a greater extent by SIT. PIO, but not SIT, increased cytosolic phospholipase A(2) and cyclooxygenase-2 activity. Accordingly, 6-keto-PGF(1alpha) and 15-deoxy-PGJ(2) increased by PIO but not SIT. In contrast, SIT, and to a lesser extent PIO, increased 15-epi-lipoxin A(4) levels. H-89 completely blocked the effect of SIT and PIO on 15-epi-lipoxin A(4) levels. PIO, and to a greater extent SIT, increased endothelial nitric oxide synthase and cAMP response element-binding protein phosphorylation, an effect that was blocked by H-89. With a 14-day pretreatment experiment, IS was 46.4 +/- 1.0% in the control group, 16.9 +/- 0.6% in SIT (P < 0.001), 19.1 +/- 1.1% in PIO (P = 0.014), and 12.9 +/- 0.7% in SIT + PIO (P < 0.001). We found that SIT and PIO limit IS using different pathways. The protective effect of SIT is via cAMP-dependent PKA activation, whereas PIO mediates its effects via both PKA-dependent and -independent pathways.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Hypoglycemic Agents/therapeutic use , Myocardial Infarction/drug therapy , Pyrazines/therapeutic use , Thiazolidinediones/therapeutic use , Triazoles/therapeutic use , Animals , Blood Glucose/metabolism , Blotting, Western , Body Weight/drug effects , Culture Media , Cyclic AMP/metabolism , Eicosanoids/metabolism , Glucagon-Like Peptide 1/metabolism , Hypertrophy, Left Ventricular/pathology , In Vitro Techniques , Mice , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Organ Size/drug effects , Phospholipases A2/metabolism , Pioglitazone , Prostaglandin-Endoperoxide Synthases/metabolism , Protective Agents/therapeutic use , Sitagliptin Phosphate
14.
Cardiovasc Res ; 87(3): 535-44, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20164119

ABSTRACT

AIMS: MicroRNAs (miRNAs) regulate various cardiac processes including cell proliferation and apoptosis. Pioglitazone (PIO), a peroxisome proliferator-activated receptor (PPAR)-gamma agonist, protects against myocardial ischaemia-reperfusion (IR) injury. We assessed the effects of PPAR-gamma activation on myocardial miRNA levels and the role of miRNAs in IR injury. METHODS AND RESULTS: We evaluated the expression changes of miRNAs in the rat heart after PIO administration using miRNA arrays and then confirmed the result by northern blot. miR-29a and c levels decreased remarkably after 7-day treatment with PIO. In H9c2 cells, the effects of PIO and rosiglitazone on miR-29 expression levels were blocked by a selective PPAR-gamma inhibitor GW9662. Downregulation of miR-29 by antisense inhibitor or by PIO protected H9c2 cells from simulated IR injury, indicated as increased cell survival and decreased caspase-3 activity. In contrast, overexpressing miR-29 promoted apoptosis and completely blocked the protective effect of PIO. Antagomirs against miR-29a or -29c significantly reduced myocardial infarct size and apoptosis in hearts subjected to IR injury. Western blot analyses demonstrated that Mcl-2, an anti-apoptotic Bcl-2 family member, was increased by miR-29 inhibition. CONCLUSION: Downregulation of miR-29 protected hearts against IR injury. The modulation of miRNAs can be achieved by pharmacological intervention. These findings provide a rationale for the development of miRNA-based strategies for the attenuation of IR injury.


Subject(s)
MicroRNAs/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Oligonucleotides, Antisense/metabolism , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Anilides/pharmacology , Animals , Apoptosis/drug effects , Blotting, Northern , Blotting, Western , Caspase 3/metabolism , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Down-Regulation , Gene Expression Profiling/methods , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Oligonucleotide Array Sequence Analysis , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Phosphorylation , Pioglitazone , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rosiglitazone , Time Factors , Transfection , bcl-2-Associated X Protein/metabolism
15.
Cardiovasc Drugs Ther ; 22(6): 429-36, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18825491

ABSTRACT

BACKGROUND: Many patients with type 2 diabetes mellitus receive several oral hypoglycemic agents, including sulfonylurea drugs. Intravenous glyburide (Glyb), a sulfonylurea agent, blocks the protective effects of "ischemic" and pharmacologic preconditioning in various animal models without affecting myocardial infarct size when administered alone. However, there are conflicting results when other sulfonylurea drugs are used. Pioglitazone (PIO) reduces infarct size in the rat. We asked whether oral Glyb and glimepiride (Glim) affect the infarct size-limiting effects of PIO. METHODS: Sprague-Dawley rats received 3-day oral treatment with: PIO (5 mg/kg/day); PIO + Glyb (10 mg/kg/day); PIO + Glim (4 mg/kg/day) or water alone (experiment 1) or PIO (5 mg/kg/day) with or without 5-hydroxydecanoate (5HD, 10 mg/kg), a specific mitochondrial ATP-sensitive K+ channels inhibitor, administered intravenously 30 min before coronary artery ligation. PIO, Glyb and Glim were administered by oral gavage. Sugar 5% was added to water to prevent hypoglycemia. Rats underwent 30 min coronary artery occlusion and 4 h reperfusion (n = 6 in each group). Ischemic area at risk was assessed by blue dye and infarct size by triphenyl-tetrazolium-chloride. RESULTS: Body weight and the size of the area at risk were comparable among groups. Infarct size (% of the area at risk) was significantly smaller in the PIO (14.3 +/- 1.1%; p < 0.001) and PIO + Glim (13.2 +/- 0.8%; p < 0.001) groups than in the control group (37.7 +/- 1.2%). Glyb completely blocked the effect of PIO (43.0 +/- 1.7%; p < 0.001). Glim did not affect the protective effect of PIO (p = 0.993). 5HD blocked the protective effect of PIO (infarct size 48.5 +/- 0.8% versus 14.8 +/- 0.6%, respectively; p < 0.0001). In conclusion, the infarct size limiting effects of PIO are dependent on activation of mitochondrial ATP-sensitive K+ channels. Oral Glyb, but not Glim, blocks the infarct size limiting effects of PIO. It is plausible that Glyb affects other pleiotropic effects of PIO and thus may attenuate favorable effects on cardiovascular outcomes. In contrast, Glim does not attenuate the protective effect of PIO.


Subject(s)
Glyburide/pharmacology , Myocardial Infarction/drug therapy , Sulfonylurea Compounds/pharmacology , Thiazolidinediones/antagonists & inhibitors , Thiazolidinediones/therapeutic use , Administration, Oral , Animals , Body Weight/drug effects , Body Weight/physiology , Coronary Vessels/injuries , Data Interpretation, Statistical , Decanoic Acids/pharmacology , Diabetes Mellitus/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Glyburide/therapeutic use , Hydroxy Acids/pharmacology , Injections, Intravenous , Intubation, Gastrointestinal , KATP Channels/antagonists & inhibitors , Ligation/methods , Mitochondria, Heart , Myocardial Infarction/physiopathology , Myocardial Ischemia/etiology , Pioglitazone , Rats , Rats, Sprague-Dawley , Sulfonylurea Compounds/therapeutic use , Thiazolidinediones/pharmacology , Ventricular Remodeling/drug effects
16.
J Immunol ; 181(5): 3515-23, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18714024

ABSTRACT

The 5-lipoxygenase (5LO) produces leukotriene B(4) and 15-epilipoxin-A(4) (15-epi-LXA(4)). Phosphorylation at Ser(523) by protein kinase A (PKA) prevents 5LO shift to the perinuclear membrane. Atorvastatin and pioglitazone up-regulate 15-epi-LXA(4) production in the heart. We assessed whether phosphorylation of 5LO by PKA determines whether 5LO interacts with the membranous cytosolic phospholipase A(2) (cPLA(2)) to produce leukotriene B(4) or with cyclooxygenase-2 (COX2) to produce 15-epi-LXA(4). Rats received either pioglitazone, atorvastatin, pioglitazone plus atorvastatin, vehicle, or LPS. Rat myocardial cells were incubated with pioglitazone plus atorvastatin, pioglitazone plus atorvastatin plus H-89 (PKA inhibitor), H-89, or vehicle for 8 h. Pioglitazone and atorvastatin did not affect total 5LO expression. However, both increased 5LO levels in the cytosolic fraction. H-89 caused a shift of 5LO to the membranous fraction in atorvastatin- and pioglitazone-treated rats. Pioglitazone and atorvastatin increased phospho-5LO levels. H-89 attenuated this increase. Both pioglitazone and atorvastatin increased COX2 levels in the cytosolic fraction and the membranous fraction. H-89 prevented this increase. Pioglitazone and atorvastatin increased cPLA(2) expression in the membranous fraction. This effect was not attenuated by H-89. Pioglitazone plus atorvastatin increased 15-epi-LXA(4) levels. H-89 attenuated the effect of pioglitazone plus atorvastatin. Pioglitazone plus atorvastatin plus H-89 increased leukotriene B(4) levels. Coimmunoprecipitation showed that without H-89, atorvastatin and pioglitazone induced an interaction between 5LO and COX2 in the cytosolic fraction, whereas when H-89 was added, 5LO interacted with cPLA(2) on the membranous fraction. The 5LO phosphorylation determines whether 15-epi-LXA(4) (anti-inflammatory) or leukotriene B(4) (inflammatory mediator) is produced.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Heptanoic Acids/pharmacology , Leukotriene B4/biosynthesis , Lipoxins/biosynthesis , Pyrroles/pharmacology , Thiazolidinediones/pharmacology , Animals , Atorvastatin , Gene Expression Regulation/drug effects , Heptanoic Acids/administration & dosage , Immunologic Factors/genetics , Isoquinolines/pharmacology , Male , Myocardium/cytology , Myocardium/immunology , Phosphorylation , Pioglitazone , Pyrroles/administration & dosage , Rats , Rats, Sprague-Dawley , Serine/metabolism , Sulfonamides/pharmacology , Thiazolidinediones/administration & dosage
17.
J Cardiovasc Pharmacol Ther ; 13(1): 72-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18287593

ABSTRACT

Statins reduce infarct size by upregulating nitric oxide synthases and PGI2 production. In this article, the infarct size-limiting effect of low-dose simvastatin + ezetimibe, ezetimibe, and high-dose statins were compared. Rats received 3-day water, atorvastatin (10 mg/kg/d), simvastatin (10 mg/kg/d), simvastatin (2 mg/kg/d), simvastatin (2 mg/kg/d) + ezetimibe (1 mg/kg/d), or ezetimibe. Rats underwent 30-minute coronary artery occlusion and 4-hour reperfusion. Atorvastatin and simvastatin 10 reduced infarct size, whereas simvastatin 2, ezetimibe, and simvastatin 2 + ezetimibe had no effect. Atorvastatin and simvastatin 10 increased nitric oxide synthases activity, whereas simvastatin-2, ezetimibe, and simvastatin-2 + ezetimibe had only a small effect. Atorvastatin and simvastatin 10 significantly increased myocardial 6-ketoprostaglandin F(1 alpha) levels, whereas simvastatin 2, ezetimibe, and simvastatin 2 + ezetimibe had no effect. High-dose statin is required to decrease infarct size, upregulate myocardial nitric oxide synthases activities, and increase 6-keto prostaglandin F(1 alpha) levels. Combination of ezetimibe and low-dose statin is ineffective in modulating myocardial biochemical changes associated with cardioprotection.


Subject(s)
Anticholesteremic Agents/pharmacology , Azetidines/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myocardial Infarction/drug therapy , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Anticholesteremic Agents/administration & dosage , Atorvastatin , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Ezetimibe , Heptanoic Acids/administration & dosage , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Male , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Pyrroles/administration & dosage , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Simvastatin/administration & dosage , Simvastatin/pharmacology , Up-Regulation/drug effects
18.
Am J Physiol Heart Circ Physiol ; 292(6): H2891-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17277020

ABSTRACT

We assessed whether aspirin (acetylsalicylic acid, ASA), administered before reperfusion, abrogates the infarct size (IS)-limiting effect of atorvastatin (ATV). Statins reduce IS. This dose-dependent effect is mediated by upregulation of cycloxygenase-2 (COX2) and PGI(2) production. Administration of selective COX2-inhibitors either with ATV for 3 days or immediately before coronary occlusion blocks the IS-limiting effect of ATV. Sprague-Dawley rats received 3-day ATV (10 mg x kg(-1) x day(-1)) or water alone. Rats underwent 30 min coronary artery occlusion and 4 h reperfusion (IS protocol, n=8 in each group), or rats underwent 30 min coronary artery occlusion and 10 min reperfusion (enzyme expression and activity protocol, n=4 in each group). Immediately before reperfusion rats received intravenous ASA (5, 10, or 20 mg/kg) or saline. Area-at-risk (AR) was assessed by blue dye and IS by triphenyltetrazolium chloride. ATV reduced IS (10.1 +/- 1.4% of the AR) compared with controls (31.0 +/- 2.2%). Intravenous ASA alone did not affect IS (29.0 +/- 2.6%); however, ASA dose dependently (5, 10, and 20 mg/kg) attenuated the protective effect of ATV on IS (15.8 +/- 0.9%, 22.0 +/- 1.6%, and 23.7 +/- 3.8%, respectively). ASA dose dependently blocked the upregulation of COX2 by ATV. COX2 activity was as follows: control, 8.93 +/- 0.90 pg/mg; ATV, 75.85 +/- 1.08 pg/mg; ATV + ASA5, 34.39 +/- 1.48 pg/mg; ATV + ASA10, 19.87 +/- 1.10 pg/mg; and ATV + ASA20, 9.36 +/- 0.94 pg/mg. ASA, administered before reperfusion in doses comparable to those used in the clinical setting, abrogates the IS-limiting effect of ATV in a model with mechanical occlusion of the coronary artery. This potential adverse interaction should be further investigated in the clinical setting of acute coronary syndromes.


Subject(s)
Aspirin/administration & dosage , Cyclooxygenase Inhibitors/administration & dosage , Heptanoic Acids/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Myocardial Infarction/prevention & control , Myocardial Ischemia/complications , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Pyrroles/administration & dosage , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Aspirin/therapeutic use , Atorvastatin , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Heptanoic Acids/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Male , Membrane Proteins/metabolism , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Ischemia/drug therapy , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/enzymology , Pyrroles/therapeutic use , Rats , Rats, Sprague-Dawley
19.
Prostaglandins Other Lipid Mediat ; 83(1-2): 89-98, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17259075

ABSTRACT

Aspirin (ASA) inhibits cycloxygenase-1 and modifies cycloxygenase-2 (COX2) by acetylation at Ser(530), leading to a shift from production of PGH(2), the precursor of prostaglandin, to 15-R-HETE which is converted by 5-lipoxygenase to 15-epi-lipoxin A(4) (15-epi-LXA4), a potent anti-inflammatory mediator. Both atorvastatin (ATV) and pioglitazone (PIO) increase COX2 expression. ATV activates COX2 by S-nitrosylation at Cys(526) to produce 15-epi-LXA4 and 6-keto-PGF(1alpha) (the stable metabolite of PGI(2)). We assessed the effect of ASA on the myocardial production of 15-epi-LXA4 and PGI(2) after induction by lipopolysaccharide (LPS) or PIO+ATV. Sprague-Dawley rats were pretreated with: control; ASA 10 mg/kg; ASA 50 mg/kg; LPS alone; LPS+ASA 10 mg/kg; LPS+ASA 50 mg/kg; LPS+ASA 200 mg/kg; PIO (10 mg/kg/d)+ATV (10 mg/kg/d); PIO+ATV+ASA 10 mg/kg; PIO+ATV+ASA 50 mg/kg; PIO+ATV+ASA 50 mg/kg+1400 W, a specific iNOS inhibitor; or PIO+ATV+1400 W. ASA alone had no effect on myocardial 15-epi-LXA4. LPS increased 15-epi-LXA4 and 6-keto-PGF(1alpha) levels. ASA (50 mg/kg and 200 mg/kg, but not 10 mg/kg) augmented the LPS effect on 15-epi-LXA4 but attenuated the effect on 6-keto-PGF(1alpha). PIO+ATV increased 15-epi-LXA4 and 6-keto-PGF(1alpha) levels. ASA and 1400 W attenuated the effects of PIO+ATV on 15-epi-LXA4 and 6-keto-PGF(1alpha). However, when both ASA and 1400 W were administered with PIO+ATV, there was a marked increase in 15-epi-LXA4, whereas the production of 6-keto-PGF(1alpha) was attenuated. In conclusion, COX2 acetylation by ASA shifts enzyme from producing 6-keto-PGF(1alpha) to 15-epi-LXA4. In contrast, S-nitrosylation by PIO+ASA augments the production of both 15-epi-LXA4 and 6-keto-PGF(1alpha). However, when COX2 is both acetylated and S-nitrosylated, it is inactivated. We suggest potential adverse interactions among statins, thiazolidinediones, and high-dose ASA.


Subject(s)
Aspirin/pharmacology , Heart/drug effects , Heptanoic Acids/pharmacology , Lipopolysaccharides/pharmacology , Lipoxins/biosynthesis , Pyrroles/pharmacology , Thiazolidinediones/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/chemistry , Atorvastatin , Biotin/metabolism , Cyclooxygenase 2/biosynthesis , Enzyme Induction/drug effects , Heptanoic Acids/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Lipoxins/chemistry , Male , Myocardium/enzymology , Nitroso Compounds/metabolism , Pioglitazone , Pyrroles/chemistry , Rats , Rats, Sprague-Dawley , Thiazolidinediones/chemistry
20.
Circulation ; 114(9): 929-35, 2006 Aug 29.
Article in English | MEDLINE | ID: mdl-16908763

ABSTRACT

BACKGROUND: Both statins and thiazolidinediones have antiinflammatory properties. However, the exact mechanisms underlying these effects are unknown. We investigated whether atorvastatin (ATV) and pioglitazone (PIO) increase the myocardial content of lipoxin-A4 and 15(R)-epi-lipoxin-A4 (15-epi-LXA4), both arachidonic acid products with strong antiinflammatory properties. METHODS AND RESULTS: In experiment 1, rats received 3-day pretreatment with water; PIO 2, 5, or 10 mg x kg(-1) x d(-1); ATV 2, 5, or 10 mg x kg(-1) x d(-1); or PIO 10 mg x kg(-1) x d(-1)+ATV 10 mg x kg(-1) x d(-1). In experiment 2, rats received water; PIO 10 mg x kg(-1) x d(-1)+ATV 10 mg x kg(-1) x d(-1); PIO+ATV and valdecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor; PIO+ATV and zileuton, a selective 5-lipoxygenase inhibitor; or zileuton alone. There were 4 rats in each group. Hearts were harvested and analyzed for myocardial lipoxin-A4 and 15-epi-LXA4 levels and for COX-2 and 5-lipoxygenase protein expression. ATV and PIO at 5 and 10 mg x kg(-1) . d(-1) significantly increased myocardial 15-epi-LXA4 levels compared with the sham-treated group (0.51 +/- 0.02 ng/mg). Myocardial 15-epi-LXA4 were significantly higher in the PIO+ATV group (1.29 +/- 0.02 ng/mg; P < 0.001 versus each other group). Both valdecoxib and zileuton abrogated the PIO+ATV increase in 15-epi-LXA4, whereas zileuton alone had no effect. PIO, ATV, and their combination resulted in a small increase in myocardial lipoxin-A4 levels, which was not statistically significant. ATV alone or in combination with PIO markedly augmented COX-2 expression. PIO had a much smaller effect on COX-2 expression. Myocardial expression of 5-lipoxygenase was not altered by PIO, ATV, or their combination. CONCLUSIONS: Both PIO and ATV increase myocardial levels of 15-epi-LXA4, a mediator with antiinflammatory properties. This finding may explain the antiinflammatory properties of both PIO and ATV.


Subject(s)
Heptanoic Acids/pharmacology , Lipoxins/biosynthesis , Myocardium/metabolism , Pyrroles/pharmacology , Thiazolidinediones/pharmacology , Animals , Anti-Inflammatory Agents/metabolism , Atorvastatin , Heart/drug effects , Models, Animal , Pioglitazone , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...