Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Am J Respir Crit Care Med ; 207(8): 1055-1069, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36913491

ABSTRACT

Rationale: Genetic studies suggest that SOX17 (SRY-related HMG-box 17) deficiency increases pulmonary arterial hypertension (PAH) risk. Objectives: On the basis of pathological roles of estrogen and HIF2α (hypoxia-inducible factor 2α) signaling in pulmonary artery endothelial cells (PAECs), we hypothesized that SOX17 is a target of estrogen signaling that promotes mitochondrial function and attenuates PAH development via HIF2α inhibition. Methods: We used metabolic (Seahorse) and promoter luciferase assays in PAECs together with the chronic hypoxia murine model to test the hypothesis. Measurements and Main Results: Sox17 expression was reduced in PAH tissues (rodent models and from patients). Chronic hypoxic pulmonary hypertension was exacerbated by mice with conditional Tie2-Sox17 (Sox17EC-/-) deletion and attenuated by transgenic Tie2-Sox17 overexpression (Sox17Tg). On the basis of untargeted proteomics, metabolism was the top pathway altered by SOX17 deficiency in PAECs. Mechanistically, we found that HIF2α concentrations were increased in the lungs of Sox17EC-/- and reduced in those from Sox17Tg mice. Increased SOX17 promoted oxidative phosphorylation and mitochondrial function in PAECs, which were partly attenuated by HIF2α overexpression. Rat lungs in males displayed higher Sox17 expression versus females, suggesting repression by estrogen signaling. Supporting 16α-hydroxyestrone (16αOHE; a pathologic estrogen metabolite)-mediated repression of SOX17 promoter activity, Sox17Tg mice attenuated 16αOHE-mediated exacerbations of chronic hypoxic pulmonary hypertension. Finally, in adjusted analyses in patients with PAH, we report novel associations between a SOX17 risk variant, rs10103692, and reduced plasma citrate concentrations (n = 1,326). Conclusions: Cumulatively, SOX17 promotes mitochondrial bioenergetics and attenuates PAH, in part, via inhibition of HIF2α. 16αOHE mediates PAH development via downregulation of SOX17, linking sexual dimorphism and SOX17 genetics in PAH.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Male , Rats , Female , Mice , Animals , Hypertension, Pulmonary/metabolism , Endothelial Cells/metabolism , Lung , Pulmonary Artery , Hypoxia/complications , Estrogens , Pulmonary Arterial Hypertension/metabolism , Familial Primary Pulmonary Hypertension/complications , HMGB Proteins/metabolism , SOXF Transcription Factors/genetics
2.
J Am Heart Assoc ; 10(15): e019488, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34315227

ABSTRACT

Background We have previously reported important strain differences in response to SU5416 (SU, a vascular endothelial growth factor receptor 2 inhibitor) in rats and have identified a specific colony of Sprague-Dawley rats that are hyperresponsive (SDHR) to SU alone and develop severe pulmonary arterial hypertension (PAH) with a single injection of SU, even in the absence of hypoxia. Interestingly, SDHR rats exhibit incomplete penetrance of the severe PAH phenotype with an "all-or-none" response to SU alone, which provides a unique opportunity to assess the influence of female sex and sex hormones on susceptibility to PAH after endothelial injury in a genetically prone model. Methods and Results SDHR rats were injected with SU (20 mg/kg SC) and, in the absence of hypoxia, 72% of male but only 27% of female rats developed severe PAH at 7 weeks, which was associated with persistent endothelial cell apoptosis. This sex difference in susceptibility for severe PAH was abolished by ovariectomy. Estradiol replacement, beginning 2 days before SU (prevention), inhibited lung endothelial cell apoptosis and completely abrogated severe PAH phenotype in both male and ovariectomized female rats, while progesterone was only protective in ovariectomized female rats. In contrast, delayed treatment of SDHR rats with established PAH with estradiol or progesterone (initiated at 4 weeks post-SU) failed to reduce lung endothelial cell apoptosis or improve PAH phenotype. Conclusions Female sex hormones markedly reduced susceptibility for the severe PAH phenotype in response to SU alone in a hyperresponsive rat strain by abolishing SU-induced endothelial cell apoptosis, but did not reverse severe PAH in established disease.


Subject(s)
Apoptosis , Endothelial Cells/metabolism , Gonadal Steroid Hormones/metabolism , Indoles , Penetrance , Pulmonary Arterial Hypertension/chemically induced , Pyrroles , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Apoptosis/drug effects , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Estradiol/pharmacology , Estrogen Replacement Therapy , Female , Genetic Predisposition to Disease , Male , Ovariectomy , Phenotype , Progesterone/pharmacology , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/prevention & control , Rats, Sprague-Dawley , Severity of Illness Index , Sex Factors
4.
Pulm Circ ; 9(4): 2045894019883613, 2019.
Article in English | MEDLINE | ID: mdl-31700608

ABSTRACT

Micro-computed tomography (micro-CT) is used in pre-clinical research to generate high-resolution three-dimensional (3D) images of organs and tissues. When combined with intravascular contrast agents, micro-CT can provide 3D visualization and quantification of vascular networks in many different organs. However, the lungs present a particular challenge for contrast perfusion due to the complexity and fragile nature of the lung microcirculation. The protocol described here has been optimized to achieve consistent lung perfusion of the microvasculature to vessels < 20 microns in both normal and pulmonary arterial hypertension rats. High-resolution 3D micro-CT imaging can be used to better visualize changes in 3D architecture of the lung microcirculation in pulmonary vascular disease and to assess the impact of therapeutic strategies on microvascular structure in animal models of pulmonary arterial hypertension.

6.
Circ Res ; 124(11): 1551-1567, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31120820

ABSTRACT

The past 2 decades have witnessed a >40% improvement in mortality for patients with heart failure and left ventricular systolic dysfunction. 1 This success has coincided with the stepwise availability of drugs that target neurohormonal activation: ß-adrenergic receptor blockers (ß-blockers), ACE (angiotensin-converting enzyme) inhibitors and ANG (angiotensin) II blockers, neprilysin inhibitors, and aldosterone antagonists. Our understanding of right heart failure (RHF) has lagged behind and many proven targeted therapies for left heart failure do not appear to provide similar benefits for RHF. Until recently, the right ventricle (RV) has often been viewed as less important than the left ventricle and in contemporary literature received the moniker "The Forgotten Ventricle". Recent advances in echocardiography and magnetic resonance imaging have enabled detailed assessments of RV anatomy and physiology in both health and disease allowing us to more accurately describe the clinical sequelae and end-organ manifestations of RHF. RV function is now recognized as one of the most important predictors of prognosis in many cardiovascular disease states. 2 Despite the significance of RV function to survival, there are no clinically approved therapies that directly nor selectively improve RV function. As well, relative to our understanding of left heart failure, the basis for RHF remains poorly understood. This article aims to condense the current knowledge on RV adaptation and failure, review current management strategies for RHF, and explore evolving therapeutic approaches.


Subject(s)
Cardiovascular Agents/therapeutic use , Heart Failure/drug therapy , Hemodynamics/drug effects , Hypertension, Pulmonary/drug therapy , Ventricular Dysfunction, Right/drug therapy , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects , Cardiovascular Agents/adverse effects , Disease Progression , Heart Failure/etiology , Heart Failure/mortality , Heart Failure/physiopathology , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/mortality , Hypertension, Pulmonary/physiopathology , Risk Factors , Stem Cell Transplantation , Treatment Outcome , Ventricular Dysfunction, Right/etiology , Ventricular Dysfunction, Right/mortality , Ventricular Dysfunction, Right/physiopathology
8.
Cardiovasc Res ; 115(4): 788-799, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30357319

ABSTRACT

AIMS: The ability of the right ventricle (RV) to adapt to increased afterload is the major determinant of survival in patients with pulmonary hypertension (PH). In this study, we explored the effect of genetic background on RV adaptation and survival in a rat model of severe pulmonary arterial hypertension (PAH). METHODS AND RESULTS: PH was induced by a single injection of SU5416 (SU) in age-matched Sprague Dawley (SD) or Fischer rats, followed by a 3-week exposure to chronic hypoxia (SUHx). SD and Fischer rats exhibited similar elevations in RV systolic pressure, number of occlusive pulmonary vascular lesions, and RV hypertrophy (RV/LV+S) in response to SUHx. However, no Fischer rats survived beyond 7 weeks compared with complete survival for SD rats. This high early mortality of Fischer rats was associated with significantly greater RV dilatation and reduced ejection fraction, cardiac output, and exercise capacity at 4 weeks post-SU. Moreover, microarray analysis revealed that over 300 genes were uniquely regulated in the RV in the severe PAH model in the Fischer compared with SD rats, mainly related to angiogenesis and vascular homoeostasis, fatty acid metabolism, and innate immunity. A focused polymerase chain reaction array confirmed down-regulation of angiogenic genes in the Fischer compared with SD RV. Furthermore, Fischer rats demonstrated significantly lower RV capillary density compared with SD rats in response to SUHx. CONCLUSION: Fischer rats are prone to develop RV failure in response to increased afterload. Moreover, the high mortality in the SUHx model of severe PAH was caused by a failure of RV adaptation associated with lack of adequate microvascular angiogenesis, together with metabolic and immunological responses in the hypertrophied RV.


Subject(s)
Heart Failure/etiology , Hypertension, Pulmonary/complications , Hypertrophy, Right Ventricular/etiology , Ventricular Dysfunction, Right/etiology , Ventricular Function, Right , Ventricular Remodeling , Adaptation, Physiological , Animals , Disease Models, Animal , Exercise Tolerance , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Male , Myocytes, Cardiac/metabolism , Neovascularization, Physiologic , Rats, Inbred F344 , Rats, Sprague-Dawley , Severity of Illness Index , Signal Transduction , Species Specificity , Transcriptome , Ventricular Dysfunction, Right/genetics , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/physiopathology
9.
Br J Pharmacol ; 175(20): 3976-3989, 2018 10.
Article in English | MEDLINE | ID: mdl-30098019

ABSTRACT

BACKGROUND AND PURPOSE: Pulmonary arterial hypertension (PAH) is a life-threatening disease that leads to progressive pulmonary hypertension, right heart failure and death. Parenteral prostaglandins (PGs), including treprostinil, a prostacyclin analogue, represent the most effective medical treatment for severe PAH. We investigated the effect of treprostinil on established severe PAH and underlying mechanisms using the rat SU5416 (SU, a VEGF receptor-2 inhibitor)-chronic hypoxia (Hx) model of PAH. EXPERIMENTAL APPROACH: Male Sprague Dawley rats were injected with SU (20 mg·kg-1 , s.c.) followed by 3 weeks of Hx (10% O2 ) to induce severe PAH. Four weeks post-SU injection, baseline right ventricular (RV) systolic pressure (RVSP) was measured, and the rats were randomized to receive vehicle or treprostinil treatment (Trep-100: 100 ng·kg-1 ·min-1 or Trep-810: 810 ng·kg-1 ·min-1 ). Following 3 weeks of treatment, haemodynamic and echocardiographic assessments were performed, and tissue samples were collected for protein expression and histological analysis. KEY RESULTS: At week 7, no difference in RVSP or RV hypertrophy was observed between vehicle and Trep-100; however, Trep-810 significantly reduced RVSP and RV hypertrophy. Trep-810 treatment significantly improved cardiac structure and function. Further, a short-term infusion of treprostinil in rats with established PAH at 4 weeks post-SU produced an acute, dose-dependent reduction in RVSP consistent with a vasodilator effect. However, chronic Trep-810 treatment did not alter media wall thickness, degree of vascular occlusion or total vessel count in the lungs. CONCLUSIONS AND IMPLICATIONS: Treprostinil exerts therapeutic benefits in PAH through decreased vascular resistance and improved cardiac structure and function; however, treprostinil treatment does not have direct impact vascular remodelling.


Subject(s)
Antihypertensive Agents/therapeutic use , Epoprostenol/analogs & derivatives , Hypertension, Pulmonary/drug therapy , Vasodilator Agents/therapeutic use , Angiogenesis Inhibitors , Animals , Epoprostenol/therapeutic use , Hemodynamics/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/physiopathology , Hypoxia/chemically induced , Hypoxia/drug therapy , Hypoxia/physiopathology , Indoles , Male , Protein Kinase Inhibitors , Pyrroles , Rats, Sprague-Dawley , Vascular Remodeling/drug effects , Vascular Remodeling/physiology , Ventricular Function, Right/drug effects
13.
Am J Respir Cell Mol Biol ; 54(4): 461-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26291195

ABSTRACT

We assessed the pulmonary hemodynamic response to vascular endothelial growth factor receptor, type 2, inhibition using SU5416 (SU) with and without chronic hypoxia (CH) in different background strains and colonies of rats. A single subcutaneous injection of SU (20 mg/kg) or vehicle was administered to different substrains of Sprague-Dawley (SD) rats, and they were compared with Lewis and Fischer rats, with and without exposure to CH (10% O2 for 3 wk). Remarkably, a unique colony of SD rats from Charles River Laboratories, termed the SD-hyperresponsive type, exhibited severe pulmonary arterial hypertension (PAH) with SU alone, characterized by increased right ventricular systolic pressure, right ventricular/left ventricular plus septal weight ratio, and arteriolar occlusive lesions at 7-8 weeks (all P < 0.0001 versus vehicle). In contrast, the other SD substrain from Harlan Laboratories, termed SD-typical type, as well as Fischer rats, developed severe PAH only when exposed to SU and CH, whereas Lewis rats showed only a minimal response. All SD-typical type rats survived for up to 13 weeks after SU/CH, whereas SD-hyperresponsive type rats exhibited mortality after SU and SU/CH (35% and 50%, respectively) at 8 weeks. Fischer rats exposed to SU/CH exhibited the greatest mortality at 8 weeks (78%), beginning as early as 4 weeks after SU and preceded by right ventricle enlargement. Of note, a partial recovery of PAH after 8 weeks was observed in the SD-typical type substrain only. In conclusion, variation in strain, even between colonies of the same strain, has a remarkable influence on the nature and severity of the response to SU, consistent with an important role for genetic modifiers of the PAH phenotype.


Subject(s)
Disease Models, Animal , Hypertension, Pulmonary/pathology , Indoles/therapeutic use , Pyrroles/therapeutic use , Animals , Hypertension, Pulmonary/drug therapy , Hypoxia , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Rats, Sprague-Dawley , Species Specificity
14.
J Cardiovasc Pharmacol ; 61(3): 258-63, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23403888

ABSTRACT

BACKGROUND: Caveolins (Cav) are structural proteins that insert into the plasma membrane to form caveolae that can bind molecules important in cardiac signal transduction and function. Cytochrome P450 epoxygenases can metabolize arachidonic acid to epoxyeicosatrienoic acids (EETs) which have known cardioprotective effects. Subsequent metabolism of EETs by soluble epoxide hydrolase reduces the protective effect. AIMS: (1) To assess the effect of ischemia-reperfusion injury on expression and subcellular localization of caveolins. (2) To study the effect of EETs on caveolins. METHODS: Hearts from soluble epoxide hydrolase null (KO) and littermate control (WT) mice were perfused in Langendorff mode and subjected to 20 minutes ischemia followed by 40 minutes reperfusion. Immunohistochemistry, immunoblot, and electron microscopy were performed to study localization of caveolins and changes in ultrastructure. RESULTS: In WT heart, Cav-1 and Cav-3 were present in cardiomyocyte and capillary endothelial cell at baseline. After ischemia, Cav-1 but not Cav-3, disappeared from cardiomyocyte; moreover, caveolae were absent and mitochondrial cristae were damaged. Improved postischemic functional recovery observed in KO or WT hearts treated with 11,12-EET corresponded to higher Cav-1 expression and maintained caveolae structure. In addition, KO mice preserved the Cav-1 signaling after ischemia that lost in WT mice. CONCLUSIONS: Taken together, our data suggest that ischemia-reperfusion injury causes loss of Cav-1 and caveolins, and EETs-mediated cardioprotection involves preservation of Cav-1.


Subject(s)
Caveolae/metabolism , Caveolin 1/metabolism , Caveolin 3/metabolism , Eicosanoids/metabolism , Epoxide Hydrolases/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Animals , Biological Transport , Blotting, Western , Caveolae/ultrastructure , Epoxide Hydrolases/genetics , Heart/physiopathology , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocardium/ultrastructure , Perfusion , Signal Transduction
15.
Article in English | MEDLINE | ID: mdl-22922020

ABSTRACT

Cardioprotective effects of epoxyeicosatrienoic acids (EETs) have been demonstrated in models of young mice with either the cardiomyocyte specific over-expression of cytochrome P450 2J2 (CYP2J2 Tr) or deletion of soluble epoxide hydrolase (sEH null). In this study we examined differences in EET-induced cardioprotection in young (2 months) and aged (12 months) CYP2J2 Tr and sEHnull mice using Langendorff isolated perfused heart model. Improved postischemic functional recovery was observed in both young and aged sEH null mice compared to age matched WT. Conversely, the cardioprotective effect observed in young CYP2J2 Tr was lost in aged CYP2J2 Tr mice. The loss of cardioprotection in aged CYP2J2 Tr was regained following perfusion with the sEH inhibitor t-AUCB. Data demonstrated increased levels of leukotoxin diol (DiHOME) and oxidative stress as well decreased protein phosphatase 2A (PP2A) activation in aged CYP2J2 Tr. In conclusion, inhibition of sEH and EET-induced cardioprotection is maintained in aged mice. However, the loss of protective effects observed in aged CYP2J2 Tr might be attributed to increased levels of DiHOME, oxidative stress and/or decreased PP2A activity.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Benzoates/pharmacology , Cardiotonic Agents/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Epoxide Hydrolases/antagonists & inhibitors , Heart/drug effects , Myocardial Reperfusion Injury/prevention & control , Urea/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/metabolism , 8,11,14-Eicosatrienoic Acid/pharmacology , Age Factors , Animals , Cells, Cultured , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme System/genetics , Epoxide Hydrolases/deficiency , Female , Gene Expression Regulation , Heart/physiopathology , Male , Mice , Mice, Knockout , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Organ Culture Techniques , Oxidative Stress , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Stearic Acids/metabolism , Urea/pharmacology
16.
Can J Physiol Pharmacol ; 90(6): 811-23, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22624559

ABSTRACT

Epoxyeicosatrienoic acids (EETs) are active metabolites of arachidonic acid that are inactivated by soluble epoxide hydrolase enzyme (sEH) to dihydroxyeicosatrienoic acid. EETs are known to render cardioprotection against ischemia reperfusion (IR) injury by maintaining mitochondrial function. We investigated the effect of a novel sEH inhibitor (sEHi) in limiting IR injury. Mouse hearts were perfused in Langendorff mode for 40 min and subjected to 20 min of global no-flow ischemia followed by 40 min of reperfusion. Hearts were perfused with 0.0, 0.1, 1.0 and 10.0 µmol·L(-1) of the sEHi N-(2-chloro-4-methanesulfonyl-benzyl)-6-(2,2,2-trifluoro-ethoxy)-nicotinamide (BI00611953). Inhibition of sEH by BI00611953 significantly improved postischemic left-ventricular-developed pressure and reduced infarct size following IR compared with control hearts, and similar to hearts perfused with 11,12-EETs (1 µmol·L(-1)) and sEH(-/-) mice. Perfusion with the putative EET receptor antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE, 10 µmol·L(-1)), or the plasma membrane K(ATP) channels (pmK(ATP)) inhibitor (glibenclamide, 10 µmol·L(-1)) abolished the improved recovery by BI00611953 (1 µmol·L(-1)). Mechanistic studies in H9c2 cells demonstrated that BI0611953 decreased ROS generation, caspase-3 activity, proteasome activity, increased HIF-1∝ DNA binding, and delayed the loss of mitochondrial membrane potential (ΔΨ(m)) caused by anoxia-reoxygenation. Together, our data demonstrate that the novel sEHi BI00611953, a nicotinamide-based compound, provides significant cardioprotection against ischemia reperfusion injury.


Subject(s)
Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Mitochondria, Heart/drug effects , Myocardial Reperfusion Injury/drug therapy , Reperfusion Injury/drug therapy , Animals , Arachidonic Acid/metabolism , Caspase 3/metabolism , Cells, Cultured , Epoxide Hydrolases/metabolism , Heart/drug effects , Hypoxia/drug therapy , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/enzymology , Myocardium/metabolism , Proteasome Endopeptidase Complex/metabolism , Rats , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism
17.
J Mol Cell Cardiol ; 53(1): 43-52, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22561102

ABSTRACT

AIMS: Epoxyeicosatrienoic acids (EETs) are cytochrome P450 epoxygenase metabolites of arachidonic acid that have known cardioprotective properties. While the mechanism(s) remains unknown, evidence suggests that phosphoinositide 3-kinase (PI3K) and sarcolemmal ATP-sensitive potassium channels (pmK(ATP)) are important. However the role of specific PI3K isoforms and corresponding intracellular mechanisms remains unknown. METHODS AND RESULTS: To study this, mice hearts were perfused in Langendorff mode for 40 min of baseline and subjected to 20 or 30 min of global no-flow ischemia followed by 40 min of reperfusion. C57BL6 mice perfused with 11,12-EET (1 µM) had improved postischemic recovery, whereas co-perfusion with PI3Kα inhibitor, PI-103 (0.1 µM), abolished the EET-mediated effect. In contrast, blocking of PI3Kß or PI3Kγ isoforms failed to inhibit EET-mediated cardioprotection. In addition to the improved post-ischemic recovery, increased levels of p-Akt, decreased calcineurin activity and decreased translocation of proapoptotic protein BAD to mitochondria were noted in EET-treated hearts. Perfusion of 11,12-EET to Kir6.2 deficient mice (pmK(ATP)) failed to improve postischemic recovery, decrease calcineurin activity and translocation of proapoptotic protein BAD, however increased levels of p-Akt were still observed. Patch-clamp experiments demonstrated that 11,12-EET could not activate pmK(ATP) currents in myocytes pre-treated with PI-103. Mechanistic studies in H9c2 cells demonstrate that 11,12-EET limits anoxia-reoxygenation triggered Ca(2+) accumulation and maintains mitochondrial ΔΨm compared to controls. Both PI-103 and glibenclamide (10 µM, pmK(ATP) inhibitor) abolished EET cytoprotection. CONCLUSION: Together our data suggest that EET-mediated cardioprotection involves activation of PI3Kα, upstream of pmK(ATP), which prevents Ca(2+) overload and maintains mitochondrial function.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Cardiotonic Agents/pharmacology , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Heart/drug effects , KATP Channels/metabolism , Myocardium/metabolism , Sarcolemma/metabolism , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Calcium/metabolism , Cell Line , Hypoxia , Isoenzymes/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Sarcolemma/enzymology
18.
J Geriatr Cardiol ; 8(3): 159-67, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22783302

ABSTRACT

The average human life span has markedly increased in modern society largely attributed to advances in medical and therapeutic sciences that have successfully reduced important health risks. However, advanced age results in numerous alterations to cellular and subcellular components that can impact the overall health and function of an individual. Not surprisingly, advanced age is a major risk factor for the development of heart disease in which elderly populations observe increased morbidity and mortality. Even healthy individuals that appear to have normal heart function under resting conditions, actually have an increased susceptibility and vulnerability to stress. This is confounded by the impact that stress and disease can have over time to both the heart and vessels. Although, there is a rapidly growing body of literature investigating the effects of aging on the heart and how age-related alterations affect cardiac function, the biology of aging and underlying mechanisms remain unclear. In this review, we summarize effects of aging on the heart and discuss potential theories of cellular aging with special emphasis on mitochondrial dysfunction.

19.
J Cardiovasc Pharmacol ; 55(1): 67-73, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19834332

ABSTRACT

Arachidonic acid, a polyunsaturated fatty acid, can be metabolized to cardioprotective epoxyeicosatrienoic acids (EETs) by cytochrome P450 epoxygenases, which are subsequently hydrolyzed to less bioactive dihydroxyeicosatrienoic acids by soluble epoxide hydrolase (sEH). To study the effects of pharmacological inhibitor of sEH (sEHi), C57BL6 mice hearts were perfused in Langendorff mode for 40 minutes of baseline and subjected to 30 minutes of global no-flow ischemia followed by 40 minutes of reperfusion. Hearts were perfused with the sEHi, trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (t-AUCB; 0.05, 0.1, 0.5, and 1 microM). To study the mechanism(s), hearts were perfused with 0.1 microM t-AUCB in the presence or absence of putative EET receptor antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (10 microM) or phosphatidylinositol 3-kinase (PI3K) inhibitors wortmannin (200 nM) or LY294002 (5 microM).Infarct size was determined at the end of 2-hour reperfusion by 2,3,5-triphenyltetrazolium chloride staining. Inhibition of sEH by t-AUCB significantly improved postischemic left ventricular developed pressure (LVDP) recovery and reduced the infarct size after ischemia and reperfusion, as compared with control hearts. Perfusion with 14,15-epoxyeicosa-5(Z)-enoic acid, wortmannin or LY294002 before ischemia abolished the cardioprotective phenotype; however, co-perfusion of both t-AUCB and 11,12-EET did not result in an additive effect on improved LVDP recovery. Together, our data suggest that pharmacological inhibition of sEH by t-AUCB is cardioprotective.


Subject(s)
Benzoates/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Urea/analogs & derivatives , Animals , Arachidonic Acid/metabolism , Benzoates/administration & dosage , Dose-Response Relationship, Drug , Eicosanoids/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/physiopathology , Receptors, Eicosanoid/antagonists & inhibitors , Urea/administration & dosage , Urea/pharmacology
20.
IUBMB Life ; 61(10): 954-60, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19787709

ABSTRACT

The cytochrome P450 monooxygenase system (CYP) is a multigene superfamily of heme-thiolate enzymes, which are important in the metabolism of foreign and endogenous compounds. Genetic variations, drug interactions, or pathophysiological factors can lead to reduced, absent, or increased enzymatic activity. This altered CYP activity greatly influences an individual's response to therapeutic treatment. What is not known is the impact of these changes on the many functional roles of CYP in physiological and pathophysiological processes of the heart. Many extrahepatic tissues, like heart, contain active P450 enzymes but lack information regarding their role in cellular injury or homeostasis. Much of our current knowledge about cardiac CYP has been limited to studies investigating the role of fatty acid metabolites in heart. Traditional risk factors including diabetes, smoking, and hypertension have well established links to cardiovascular disease. And new evidence strongly suggests exposure to chemicals and other environmental agents has a profound impact on the cardiovascular system. These risk factors can independently affect the expression and activity of CYP enzymes. Therefore, altered CYP activity is important from a detoxification as well as a bioactivation perspective. Considering CYP, interactions are greatly dependent on inherited differences or acquired changes in enzyme activity further research into their potential impact on pathogenesis, risk assessment, and therapy of heart disease is warranted. This review explores the expression of CYP isoforms, their functional roles, and the effects of genetic variation in the heart.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Myocardium/metabolism , Animals , Cardiovascular System/metabolism , Forecasting , Gene Expression Regulation, Enzymologic , Humans , Pharmacogenetics , Polymorphism, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Substrate Specificity/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...