Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Curr Vasc Pharmacol ; 11(1): 105-11, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22303912

ABSTRACT

BACKGROUND: Epoxyeicosatrienoic acids (EETs) have been shown to play a role in cardiovascular protection by reducing ischemia reperfusion injury, producing anti-inflammatory effects, and promoting angiogenesis. EETs are regulated through conversion to less active corresponding diols by soluble epoxide hydrolase (sEH). Inhibition of sEH enhances the beneficial properties of EETs and has been investigated as a possible treatment for cardiovascular diseases. CONTENT: sEH inhibitors (sEHIs) have anti-inflammatory effects by stabilizing anti-inflammatory EETs. Additionally, sEHIs strongly inhibit and reverse cardiac hypertrophy. sEHIs have been shown to protect myocardial cells from ischemiareperfusion injury, treat atherosclerosis and prevent the development of hypertension. sEHIs promote blood vessels to release bradykinin via an EET-mediated STAT3 signaling pathway to elicit tolerance to ischemia. SUMMARY: Inhibition of sEH has been shown to improve several aspects of cardiovascular diseases, including inflammation, hypertension, cardiac hypertrophy and atherosclerosis. For this reason, sEHIs are promising new pharmaceutical for the treatment of cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/enzymology , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Epoxide Hydrolases/metabolism , Myocytes, Cardiac/drug effects , Animals , Cardiovascular Diseases/metabolism , Humans , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism
2.
Int J Cardiol ; 167(4): 1298-304, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-22525341

ABSTRACT

BACKGROUND: Epoxyeicosatrienoic acids (EETs) are natural angiogenic mediators regulated by soluble epoxide hydrolase (sEH). Inhibitors of sEH can stabilize EETs levels and were reported to reduce atherosclerosis and inhibit myocardial infarction in animal models. In this work, we investigated whether increasing EETs with the sEH inhibitor t-AUCB would increase angiogenesis related function in endothelial progenitor cells (EPCs) from patients with acute myocardial infarction (AMI). METHODS AND RESULTS: EPCs were isolated from 50 AMI patients and 50 healthy subjects (control). EPCs were treated with different concentrations of t-AUCB for 24h with or without peroxisome proliferator activated receptor γ (PPARγ) inhibitor GW9662. Migration of EPCs was assayed in trans-well chambers. Angiogenesis assays were performed using a Matrigel-Matrix in vitro model. The expression of vascular endothelial growth factor (VEGF), hypoxia-inducible factor 1α (HIF-1α) mRNA and protein in EPCs was measured by real-time PCR or Western blot, respectively. Also, the concentration of EETs in the culture supernatant was detected by ELISA. The activity of EPCs in the AMI patient group was reduced compared to healthy controls. Whereas increasing EET levels with t-AUCB promoted a dose dependent angiogenesis and migration in EPCs from AMI patients. Additionally, the t-AUCB dose dependently increased the expression of the angiogenic factors VEGF and HIF-α. Lastly, we provide evidence that these effects were PPARγ dependent. CONCLUSION: The results demonstrate that the sEH inhibitor positively modulated the functions of EPCs in patients with AMI through the EETs-PPARγ pathway. The present study suggests the potential utility of sEHi in the therapy of ischemic heart disease.


Subject(s)
Benzoates/pharmacology , Endothelial Cells/physiology , Epoxide Hydrolases/antagonists & inhibitors , Myocardial Infarction/enzymology , PPAR gamma/physiology , Stem Cells/physiology , Urea/analogs & derivatives , Aged , Anilides/pharmacology , Cells, Cultured , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/metabolism , Female , Humans , Male , Middle Aged , Myocardial Infarction/blood , PPAR gamma/antagonists & inhibitors , Stem Cells/drug effects , Urea/pharmacology
3.
Eur Respir J ; 42(2): 350-61, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23180589

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is the third leading cause of death. The statin drugs may have therapeutic potential in respiratory diseases such as COPD, but whether they prevent bronchial epithelial injury is unknown. We hypothesised that simvastatin attenuates acute tobacco smoke-induced neutrophilic lung inflammation and airway epithelial injury. Spontaneously hypertensive rats were given simvastatin (20 mg·kg(-1) i.p.) daily for either 7 days prior to tobacco smoke exposure and during 3 days of smoke exposure, or only during tobacco smoke exposure. Pretreatment with simvastatin prior to and continued throughout smoke exposure reduced the total influx of leukocytes, neutrophils and macrophages into the lung and airways. Simvastatin attenuated tobacco smoke-induced cellular infiltration into lung parenchymal and airway subepithelial and interstitial spaces. 1 week of simvastatin pretreatment almost completely prevented smoke-induced denudation of the airway epithelial layer, while simvastatin given only concurrently with the smoke exposure had no effect. Simvastatin may be a novel adjunctive therapy for smoke-induced lung diseases, such as COPD. Given the need for statin pretreatment there may be a critical process of conditioning that is necessary for statins' anti-inflammatory effects. Future work is needed to elucidate the mechanisms of this statin protective effect.


Subject(s)
Epithelium/pathology , Pulmonary Disease, Chronic Obstructive/prevention & control , Pulmonary Disease, Chronic Obstructive/therapy , Simvastatin/pharmacology , Smoke/adverse effects , Animals , Anti-Inflammatory Agents/pharmacology , Bronchoalveolar Lavage Fluid , Cholesterol/chemistry , Inflammation/prevention & control , Inflammation/therapy , Leukocytes/drug effects , Macrophages/drug effects , Male , Monomeric GTP-Binding Proteins/metabolism , Neutrophils/drug effects , Oxidative Stress , Rats , Rats, Inbred SHR , Respiratory Function Tests , Nicotiana/adverse effects , Treatment Outcome , rho GTP-Binding Proteins/metabolism
4.
PLoS One ; 7(3): e33304, 2012.
Article in English | MEDLINE | ID: mdl-22457750

ABSTRACT

Chronic obstructive pulmonary disease (COPD) kills approximately 2.8 million people each year, and more than 80% of COPD cases can be attributed to smoking. Leukocytes recruited to the lung contribute to COPD pathology by releasing reactive oxygen metabolites and proteolytic enzymes. In this work, we investigated where leukocytes enter the lung in the early stages of COPD in order to better understand their effect as a contributor to the development of COPD. We simultaneously evaluated the parenchyma and airways for neutrophil accumulation, as well as increases in the adhesion molecules and chemokines that cause leukocyte recruitment in the early stages of tobacco smoke induced lung disease. We found neutrophil accumulation and increased expression of adhesion molecules and chemokines in the bronchial blood vessels that correlated with the accumulation of leukocytes recovered from the lung. The expression of adhesion molecules and chemokines in other vascular beds did not correlate with leukocytes recovered in bronchoalveolar lavage fluid (BALF). These data strongly suggest leukocytes are recruited in large measure through the bronchial circulation in response to tobacco smoke. Our findings have important implications for understanding the etiology of COPD and suggest that pharmaceuticals designed to reduce leukocyte recruitment through the bronchial circulation may be a potential therapy to treat COPD.


Subject(s)
Bronchi/pathology , Disease Models, Animal , Hypertension/pathology , Leukocytes/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Animals , Bronchi/blood supply , Bronchoalveolar Lavage Fluid , Hypertension/complications , Male , Pulmonary Disease, Chronic Obstructive/complications , Rats , Rats, Inbred SHR , Smoke , Nicotiana
5.
Biochem Biophys Res Commun ; 410(3): 494-500, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21683067

ABSTRACT

Excess leukocyte recruitment to the lung plays a central role in the development or exacerbation of several lung inflammatory diseases including chronic obstructive pulmonary disease. Epoxyeicosatrienoic acids (EETs) are cytochrome P-450 metabolites of arachidonic acid reported to have multiple biological functions, including blocking of leukocyte recruitment to inflamed endothelium in cell culture through reduction of adhesion molecule expression. Inhibition of the EET regulatory enzyme, soluble epoxide hydrolase (sEH) also has been reported to have anti-inflammatory effects in vivo including reduced leukocyte recruitment to the lung. We tested the hypothesis that the in vivo anti-inflammatory effects of sEH inhibitors act through the same mechanisms as the in vitro anti-inflammatory effects of EETs in a rat model of acute inflammation following exposure to tobacco smoke. Contrary to previously published data, we found that sEH inhibition did not reduce tobacco smoke-induced leukocyte recruitment to the lung. Furthermore, sEH inhibition did not reduce tobacco smoke-induced adhesion molecule expression in the lung vasculature. Similarly, concentrations of EETs greater than or equal to their reported effective dose did not reduce TNFα induced expression of the adhesion molecules. These results suggest that the anti-inflammatory effects of sEH inhibitors are independent of leukocyte recruitment and EETs do not reduce the adhesion molecules responsible for leukocyte recruitment in vitro. This demonstrates that the widely held belief that sEH inhibition prevents leukocyte recruitment via EET prevention of adhesion molecule expression is not consistently reproducible.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Leukocytes/drug effects , Lung/physiopathology , Animals , Bronchi/blood supply , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , Leukocytes/enzymology , Leukocytes/physiology , Lung/drug effects , Lung/pathology , Male , Pneumonia/chemically induced , Pneumonia/pathology , Pneumonia/physiopathology , Rats , Rats, Inbred SHR , Smoking/adverse effects , Nicotiana/adverse effects
6.
J Cardiovasc Pharmacol ; 52(4): 314-23, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18791465

ABSTRACT

To determine whether sEH inhibitors influence atherosclerotic lesion formation, we used an established murine model of accelerated atherogenesis, ApoE knockout (-/-) mice. The sEH inhibitor, 1-adamantan-3-(5-(2-(2-ethylethoxy)ethoxy)pentyl)urea (AEPU) was delivered in drinking water. All animals were fed an atherogenic diet while simultaneously infused with angiotensin II by osmotic minipump to induce atherosclerosis. In AEPU-treated animals, there was a 53% reduction in atherosclerotic lesions in the descending aortae as compared to control aortae. AEPU and its major metabolites were detected in the plasma of animals which received it. As expected from the inhibition of sEH, a significant increase in linoleic and arachidonic acid epoxides, as well as an increase in individual 11,12-EET/DHET and 14,15-EET/DHET ratios, were observed. The reduction in atherosclerotic lesion area was inversely correlated with 11,12- and 14,15- EET/DHET ratios, suggesting that the reduction corresponds to the inhibition of sEH. Our data suggest that orally-available sEH inhibitors may be useful in the treatment of patients with atherosclerotic cardiovascular disease.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/metabolism , Epoxide Hydrolases/antagonists & inhibitors , Angiotensin II/metabolism , Animals , Atherosclerosis/chemically induced , Atherosclerosis/pathology , Diet, Atherogenic , Disease Models, Animal , Epoxide Hydrolases/chemistry , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Molecular Weight , Pilot Projects , Solubility
7.
Proc Natl Acad Sci U S A ; 103(49): 18733-8, 2006 Dec 05.
Article in English | MEDLINE | ID: mdl-17130447

ABSTRACT

Sustained cardiac hypertrophy represents one of the most common causes leading to cardiac failure. There is emerging evidence to implicate the involvement of NF-kappaB in the development of cardiac hypertrophy. However, several critical questions remain unanswered. We tested the use of soluble epoxide hydrolase (sEH) inhibitors as a means to enhance the biological activities of epoxyeicosatrienoic acids (EETs) to treat cardiac hypertrophy. sEH catalyzes the conversion of EETs to form the corresponding dihydroxyeicosatrienoic acids. Previous data have suggested that EETs may inhibit the activation of NF-kappaB-mediated gene transcription. We directly demonstrate the beneficial effects of several potent sEH inhibitors (sEHIs) in cardiac hypertrophy. Specifically, we show that sEHIs can prevent the development of cardiac hypertrophy using a murine model of pressure-induced cardiac hypertrophy. In addition, sEHIs reverse the preestablished cardiac hypertrophy caused by chronic pressure overload. We further demonstrate that these compounds potently block the NF-kappaB activation in cardiac myocytes. Moreover, by using in vivo electrophysiologic recordings, our study shows a beneficial effect of the compounds in the prevention of cardiac arrhythmias that occur in association with cardiac hypertrophy. We conclude that the use of sEHIs to increase the level of the endogenous lipid epoxides such as EETs may represent a viable and completely unexplored avenue to reduce cardiac hypertrophy by blocking NF-kappaB activation.


Subject(s)
Cardiomegaly/drug therapy , Cardiomegaly/prevention & control , Enzyme Inhibitors/therapeutic use , Epoxide Hydrolases/antagonists & inhibitors , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Solubility
8.
J Pharmacol Exp Ther ; 316(2): 815-21, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16221742

ABSTRACT

Epoxyeicosatrienoic acid(s) (EET) have variable hemodynamic, anti-inflammatory, and growth regulatory effects, and inhibitors of their regulatory enzyme, soluble epoxide hydrolase (sEH), can mimic these effects. For this reason, sEH inhibitors are being studied as potential pharmaceuticals for the treatment of hypertension, atherosclerosis, and inflammatory diseases. We now show that a highly selective urea-based sEH inhibitor 1-cyclohexyl-3-dodecyl urea (CDU) attenuates human aortic vascular smooth muscle (HVSM) cell proliferation independently of any effect on sEH. CDU also inhibits endothelial cells when stimulated with basic fibroblast growth factor or serum. In addition, we demonstrate that EET, as well as several newer generation sEH inhibitors and a urea-based weak sEH inhibitor, do not affect proliferation in HVSM cells. Structure-activity relationships demonstrate that the addition of an acid group to the dodecyl carbon chain, changing the cyclohexyl group to an adamantyl group, and shortening the carbon chain to two carbons all abolish the antiproliferative effect. Our finding that a highly selective urea-based inhibitor of sEH can alter biology independently of its putative target enzyme suggests that there may be other useful properties of this class of compounds unrelated to their influence on epoxyeicosanoids. In addition, our results show that caution should be used when attempting to infer conclusions of EET biology based solely on the effects these inhibitors in tissue culture models, especially when used at micromolar concentrations.


Subject(s)
Cell Proliferation/drug effects , Epoxide Hydrolases/antagonists & inhibitors , Muscle, Smooth, Vascular/cytology , Urea/analogs & derivatives , Aorta/cytology , Cells, Cultured , Dose-Response Relationship, Drug , Eicosanoic Acids/antagonists & inhibitors , Humans , Molecular Structure , Muscle, Smooth, Vascular/enzymology , Solubility , Structure-Activity Relationship , Urea/pharmacology
9.
Am J Physiol Renal Physiol ; 286(4): F720-6, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14665429

ABSTRACT

Epoxyeicosatrienoic acids are cytochrome P-450 metabolites of arachidonic acid with multiple biological functions, including the regulation of vascular tone, renal tubular transport, cellular proliferation, and inflammation. Epoxyeicosatrienoic acids are converted by soluble epoxide hydrolase into the corresponding dihydroxyeicosatrienoic acids, and epoxyeicosatrienoic acid hydration is regarded as one mechanism whereby their biological effects are eliminated. Previous animal studies indicate that soluble epoxide hydrolase plays an important role in the regulation of renal eicosanoid levels and systemic blood pressure. To begin to elucidate the mechanism of these effects, we determined the cellular localization of soluble epoxide hydrolase in human kidney by examining biopsies taken from patients with a variety of non-end-stage renal diseases, as well as those without known renal disease. Immunohistochemical staining of acetone-fixed kidney biopsy samples revealed that soluble epoxide hydrolase was preferentially expressed in the renal vasculature with relatively low levels in the surrounding tubules. Expression of soluble epoxide hydrolase was evident in renal arteries of varying diameter and was localized mostly in the smooth muscle layers of the arterial wall. Western blot analysis and functional assays confirmed the expression of soluble epoxide hydrolase in the human kidney. There were no obvious differences in soluble epoxide hydrolase expression between normal and diseased human kidney tissue in the samples examined. Our results indicate that soluble epoxide hydrolase is present in the human kidney, being preferentially expressed in the renal vasculature, and support an essential role for this enzyme in renal hemodynamic regulation and its potential utility as a target for therapeutic intervention.


Subject(s)
Arterioles/enzymology , Epoxide Hydrolases/metabolism , Kidney/enzymology , Blotting, Western , Humans , Immunohistochemistry , Kidney/blood supply , Kidney Diseases/enzymology , Solubility
10.
Am J Physiol Cell Physiol ; 284(1): C16-23, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12388104

ABSTRACT

Abnormal vascular smooth muscle (VSM) cell proliferation contributes to the development of atherosclerosis and its associated disorders, including angioplasty restenosis. The tumor-suppressor protein p53 has been linked to the development of atherosclerotic lesions, and its homolog, p73, is proving to have contrasting functions in a variety of tissues. As an outgrowth of our previous finding that p73 is increased in serum-stimulated VSM cells and human atherosclerotic tissue, we examined p73 overexpression in VSM cells to elucidate causality of p73 expression with growth response. Overexpression of p73 results in decreased cell cycle transit and is accompanied by apoptosis. The apoptotic changes in p73 overexpressing VSM cells are independent of p53 and are associated with a decrease in levels of p21(waf1/cip1). In conjunction with our previous data finding that p73 is increased in serum-stimulated VSM cells, this work suggests a role for p73 in vascular proliferative diseases.


Subject(s)
Apoptosis/physiology , DNA-Binding Proteins/biosynthesis , Muscle, Smooth, Vascular/metabolism , Nuclear Proteins/biosynthesis , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , Doxycycline/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Genes, Tumor Suppressor , Humans , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nuclear Proteins/genetics , Rabbits , Rats , Rats, Inbred Lew , Tumor Protein p73 , Tumor Suppressor Proteins
11.
Proc Natl Acad Sci U S A ; 99(4): 2222-7, 2002 Feb 19.
Article in English | MEDLINE | ID: mdl-11842228

ABSTRACT

Atherosclerosis, in its myriad incarnations the foremost killer disease in the industrialized world, is characterized by aberrant proliferation of vascular smooth muscle (VSM) cells in part as a result of the recruitment of inflammatory cells to the blood vessel wall. The epoxyeicosatrienoic acids are synthesized from arachidonic acid in a reaction catalyzed by the cytochrome P450 system and are vasoactive substances. Metabolism of these compounds by epoxide hydrolases results in the formation of compounds that affect the vasculature in a pleiotropic manner. As an outgrowth of our observations that urea inhibitors of the soluble epoxide hydrolase (sEH) reduce blood pressure in spontaneously hypertensive rats as well as the findings of other investigators that these compounds possess antiinflammatory actions, we have examined the effect of sEH inhibitors on VSM cell proliferation. We now show that the sEH inhibitor 1-cyclohexyl-3-dodecyl urea (CDU) inhibits human VSM cell proliferation in a dose-dependent manner and is associated with a decrease in the level of cyclin D1. In addition, cis-epoxyeicosatrienoic acid mimics the growth-suppressive activity of CDU; there is no evidence of cellular toxicity or apoptosis in CDU-treated cells when incubated with 20 microM CDU for up to 48 h. These results, in light of the antiinflammatory and antihypertensive properties of these compounds that have been demonstrated already, suggest that the urea class of sEH inhibitors may be useful for therapy for diseases such as hypertension and atherosclerosis characterized by exuberant VSM cell proliferation and vascular inflammation.


Subject(s)
Epoxide Hydrolases/antagonists & inhibitors , Muscle, Smooth, Vascular/cytology , Blotting, Western , Cell Division/drug effects , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , G1 Phase , Humans , MAP Kinase Signaling System , Muscle, Smooth/cytology , Recombinant Proteins/metabolism , S Phase , Thymidine/pharmacology , Time Factors , Urea/analogs & derivatives , Urea/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...