Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
NPJ Precis Oncol ; 3: 24, 2019.
Article in English | MEDLINE | ID: mdl-31602400

ABSTRACT

Metastatic outcomes depend on the interactions of metastatic cells with a specific organ microenvironment. Our previous studies have shown that triple-negative breast cancer (TNBC) MDA-MB-231 cells passaged in astrocyte-conditioned medium (ACM) show proclivity to form brain metastases, but the underlying mechanism is unknown. The combination of microarray analysis, qPCR, and ELISA assay were carried out to demonstrate the ACM-induced expression of angiopoietin-like 4 (ANGPTL4) in TNBC cells. A stable ANGPTL4-knockdown MDA-MB-231 cell line was generated by ANGPTL4 short-hairpin RNA (shRNA) and inoculated into mice via left ventricular injection to evaluate the role of ANGPTL4 in brain metastasis formation. The approaches of siRNA, neutralizing antibodies, inhibitors, and immunoprecipitation were used to demonstrate the involved signaling molecules. We first found that ACM-conditioned TNBC cells upregulated the expression of ANGPTL4, a secreted glycoprotein whose effect on tumor progression is known to be tumor microenvironment- and tumor-type dependent. Knockdown of ANGPTL4 in TNBC MDA-MB-231 cells with shRNA decreased ACM-induced tumor cell metastatic growth in the brain and attributed to survival in a mouse model. Furthermore, we identified that astrocytes produced transforming growth factor-beta 2 (TGF-ß2), which in part is responsible for upregulation of ANGPTL4 expression in TNBC through induction of SMAD signaling. Moreover, we identified that tumor cells communicate with astrocytes, where tumor cell-derived interleukin-1 beta (IL-1ß) and tumor necrosis factor alpha (TNF-α) increased the expression of TGF-ß2 in astrocytes. Collectively, these findings indicate that the invading TNBC cells interact with astrocytes in the brain microenvironment that facilitates brain metastases of TNBC cells through a TGF-ß2/ANGPTL4 axis. This provides groundwork to target ANGPTL4 as a treatment for breast cancer brain metastases.

2.
Front Oncol ; 5: 154, 2015.
Article in English | MEDLINE | ID: mdl-26236688

ABSTRACT

High-grade serous carcinoma (HGSC) is the most lethal ovarian cancer histotype. The fallopian tube secretory epithelial cells (FTSECs) are a proposed progenitor cell type. Genetically altered FTSECs form tumors in mice; however, a spontaneous HGSC model has not been described. Apart from a subpopulation of genetically predisposed women, most women develop ovarian cancer spontaneously, which is associated with aging and lifetime ovulations. A murine oviductal cell line (MOE(LOW)) was developed and continuously passaged in culture to mimic cellular aging (MOE(HIGH)). The MOE(HIGH) cellular model exhibited a loss of acetylated tubulin consistent with an outgrowth of secretory epithelial cells in culture. MOE(HIGH) cells proliferated significantly faster than MOE(LOW), and the MOE(HIGH) cells produced more 2D foci and 3D soft agar colonies as compared to MOE(LOW) MOE(HIGH) were xenografted into athymic female nude mice both in the subcutaneous and the intraperitoneal compartments. Only the subcutaneous grafts formed tumors that were negative for cytokeratin, but positive for oviductal markers, such as oviductal glycoprotein 1 and Pax8. These tumors were considered to be poorly differentiated carcinoma. The differential molecular profiles between MOE(HIGH) and MOE(LOW) were determined using RNA-Seq and confirmed by protein expression to uncover pathways important in transformation, like the p53 pathway, the FOXM1 pathway, WNT signaling, and splicing. MOE(HIGH) had enhanced protein expression of c-myc, Cyclin E, p53, and FOXM1 with reduced expression of p21. MOE(HIGH) were also less sensitive to cisplatin and DMBA, which induce lesions typically repaired by base-excision repair. A model of spontaneous tumorogenesis was generated starting with normal oviductal cells. Their transition to cancer involved alterations in pathways associated with high-grade serous cancer in humans.

3.
J Lipid Res ; 55(10): 2093-102, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24958911

ABSTRACT

Microsomal epoxide hydrolase (EPHX1, EC 3.3.2.9) is a highly abundant α/ß-hydrolase enzyme that is known for its catalytical epoxide hydrolase activity. A wide range of EPHX1 functions have been demonstrated including xenobiotic metabolism; however, characterization of its endogenous substrates is limited. In this study, we present evidence that EPHX1 metabolizes the abundant endocannabinoid 2-arachidonoylglycerol (2-AG) to free arachidonic acid (AA) and glycerol. The EPHX1 metabolism of 2-AG was demonstrated using commercially available EPHX1 microsomes as well as PC-3 cells overexpressing EPHX1. Conversely, EPHX1 siRNA markedly reduced the EPHX1 expression and 2-AG metabolism in HepG2 cells and LNCaP cells. A selective EPHX1 inhibitor, 10-hydroxystearamide, inhibited 2-AG metabolism and hydrolysis of a well-known EPHX1 substrate, cis-stilbene oxide. Among the inhibitors studied, a serine hydrolase inhibitor, methoxy-arachidonyl fluorophosphate, was the most potent inhibitor of 2-AG metabolism by EPHX1 microsomes. These results demonstrate that 2-AG is an endogenous substrate for EPHX1, a potential role of EPHX1 in the endocannabinoid signaling and a new AA biosynthetic pathway.


Subject(s)
Arachidonic Acids/metabolism , Endocannabinoids/metabolism , Epoxide Hydrolases/metabolism , Glycerides/metabolism , Microsomes/enzymology , Signal Transduction/physiology , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Hep G2 Cells , Humans , Signal Transduction/drug effects
4.
J Steroids Horm Sci ; 5(3): 136, 2014.
Article in English | MEDLINE | ID: mdl-25844270

ABSTRACT

OBJECTIVE: Progesterone (P4) plays a central role in women's health. Synthetic progestins are used clinically in hormone replacement therapy (HRT), oral contraceptives, and for the treatment of endometriosis and infertility. Unfortunately, synthetic progestins are associated with side effects, including cardiovascular disease and breast cancer. Botanical dietary supplements are widely consumed for the alleviation of a variety of gynecological issues, but very few studies have characterized natural compounds in terms of their ability to bind to and activate progesterone receptors (PR). Kaempferol is a flavonoid that functions as a non-steroidal selective progesterone receptor modulator (SPRM) in vitro. This study investigated the molecular and physiological effects of kaempferol in the ovariectomized rat uteri. METHODS: Since genistein is a phytoestrogen that was previously demonstrated to increase uterine weight and proliferation, the ability of kaempferol to block genistein action in the uterus was investigated. Analyses of proliferation, steroid receptor expression, and induction of well-established PR-regulated targets Areg and Hand2 were completed using histological analysis and qPCR gene induction experiments. In addition, kaempferol in silico binding analysis was completed for PR. The activation of estrogen and androgen receptor signalling was determined in vitro. RESULTS: Molecular docking analysis confirmed that kaempferol adopts poses that are consistent with occupying the ligand-binding pocket of PRA. Kaempferol induced expression of PR regulated transcriptional targets in the ovariectomized rat uteri, including Hand2 and Areg. Consistent with progesterone-l ke activity, kaempferol attenuated genistein-induced uterine luminal epithelial proliferation without increasing uterine weight. Kaempferol signalled without down regulating PR expression in vitro and in vivo and without activating estrogen and androgen receptors. CONCLUSION: Taken together, these data suggest that kaempferol is a unique natural PR modulator that activates PR signaling in vitro and in vivo without triggering PR degradation.

5.
Methods Enzymol ; 499: 149-65, 2011.
Article in English | MEDLINE | ID: mdl-21683253

ABSTRACT

Clade B serpin family of proteins regulate a variety of cellular functions including cell adhesion and motility. One key member of the clade B serpin family is maspin (SERPINB5). Maspin is classified as a type II tumor suppressor that regulates cell adhesion and invasion. It is expressed in normal mammary epithelial cells but is reduced in benign breast tumors and absent in invasive breast carcinomas. Although maspin regulates cell apoptosis, cell adhesion, migration, and invasion in breast cancer cell culture systems, mouse models are necessary to verify this in vivo. In this chapter, we review the development of transgenic and syngeneic mouse models to study the role of maspin in mammary tumorigenesis and in normal mammary development.


Subject(s)
Breast Neoplasms/metabolism , Serpins/metabolism , Animals , Breast Neoplasms/genetics , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Serpins/genetics
6.
J Biol Chem ; 286(28): 24599-607, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21606500

ABSTRACT

Maspin is a non-inhibitory serine protease inhibitor (serpin) that influences many cellular functions including adhesion, migration, and invasion. The underlying molecular mechanisms that facilitate these actions are still being elucidated. In this study we determined the mechanism by which maspin mediates increased MCF10A cell adhesion. Utilizing competition peptides and mutation analyses, we discovered two unique regions (amino acid residues 190-202 and 260-275) involved in facilitating the increased adhesion function of maspin. In addition, we demonstrate that the urokinase-type plasminogen activator (uPA)/uPA receptor (uPAR) complex is required for the localization and adhesion function of maspin. Finally, we showed that maspin, uPAR, and ß1 integrin co-immunoprecipitate, suggesting a novel maspin-uPA-uPAR-ß1 integrin mega-complex that regulates mammary epithelial cell adhesion.


Subject(s)
Epithelial Cells/metabolism , Integrin beta1/metabolism , Multiprotein Complexes/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Serpins/metabolism , Cell Adhesion/physiology , Cell Line, Transformed , Epithelial Cells/cytology , Humans , Integrin beta1/genetics , Multiprotein Complexes/genetics , Receptors, Urokinase Plasminogen Activator/genetics , Serpins/genetics
7.
Prostaglandins Other Lipid Mediat ; 94(1-2): 34-43, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21167293

ABSTRACT

Endocannabinoids (ECs), anandamide (AEA) and 2-arachidonoylglycerol (2-AG), inhibit proliferation of carcinoma cells. Several enzymes hydrolyze ECs to reduce endogenous EC concentrations and produce eicosanoids that promote cell growth. In this study, we determined the effects of EC hydrolysis inhibitors and a putative EC, 2-arachidonylglyceryl ether (noladin ether, NE) on proliferation of prostate carcinoma (PC-3, DU-145, and LNCaP) cells. PC-3 cells had the least specific hydrolysis activity for AEA and administration of AEA effectively inhibited cell proliferation. The proliferation inhibition was blocked by SR141716A (a selective CB1R antagonist) but not SR144528 (a selective CB2R antagonist), suggesting a CB1R-mediated inhibition mechanism. On the other hand, specific hydrolysis activity for 2-AG was high and 2-AG inhibited proliferation only in the presence of EC hydrolysis inhibitors. NE inhibited proliferation in a concentration-dependent manner; however, SR141716A, SR144528 and pertussis toxin did not block the NE-inhibited proliferation, suggesting a CBR-independent mechanism of NE. A peroxisome proliferator-activated receptor gamma (PPARγ) antagonist GW9662 did not block the NE-inhibited proliferation, suggesting that PPARγ was not involved. NE also induced cell cycle arrest in G(0)/G(1) phase in PC-3 cells. NE inhibited the nuclear translocation of nuclear factor-kappa B (NF-κB p65) and down-regulated the expression of cyclin D1 and cyclin E in PC-3 cells, suggesting the NF-κB/cyclin D and cyclin E pathways are involved in the arrest of G1 cell cycle and inhibition of cell growth. These results indicate therapeutic potentials of EC hydrolysis inhibitors and the enzymatically stable NE in prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Endocannabinoids , Glycerides/pharmacology , Prostatic Neoplasms/metabolism , Anilides/pharmacology , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Cyclin E/metabolism , Humans , Male , NF-kappa B/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Prostate/pathology , Prostatic Neoplasms/pathology
8.
Int J Cancer ; 123(6): 1318-26, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18566995

ABSTRACT

The hydrolysis of endocannabinoids has profound effects on the function of the endocannabinoid signaling system in the regulation of prostate carcinoma cells. Prostate carcinoma cells exhibit a wide range of hydrolysis activity for 2-arachidonoylglycerol (2-AG), the major endocannabinoid. However, enzyme(s) responsible for 2-AG hydrolysis and their functions in prostate cancer have not been characterized. In this study, we demonstrated that fatty acid amide hydrolase (FAAH) was differentially expressed in normal and prostate carcinoma cells. In PC-3 cells, overexpression of FAAH resulted in increased FAAH protein, 2-AG hydrolysis, cell invasion and cell migration. Conversely, small-interfering RNA (siRNA) knockdown of FAAH in LNCaP cells decreased FAAH protein, 2-AG hydrolysis and cell invasion. Furthermore, CAY10401, a FAAH inhibitor, decreased cell invasion and it enhanced the reduction of invasion in FAAH siRNA-transfected LNCaP cells. Immunohistochemistry staining of commercial tissue microarrays (TMAs) demonstrated FAAH staining in 109 of 157 cores of prostate adenocarcinomas but weak staining in 1 of 8 cores of normal prostate tissues. These results suggest that FAAH regulates 2-AG hydrolysis and invasion of prostate carcinoma cells and is potentially involved in prostate tumorigenesis.


Subject(s)
Adenocarcinoma/enzymology , Amidohydrolases/biosynthesis , Prostatic Neoplasms/enzymology , Amidohydrolases/drug effects , Arachidonic Acids/metabolism , Blotting, Western , Cell Movement/physiology , Chromatography, Liquid , Endocannabinoids , Enzyme Inhibitors/pharmacology , Gene Expression , Glycerides/metabolism , Humans , Immunohistochemistry , Male , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Electrospray Ionization , Tissue Array Analysis , Transfection
9.
Int J Cancer ; 121(5): 984-91, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17443494

ABSTRACT

Endogenous 2-arachidonoylglycerol (2-AG) is antiinvasive in androgen-independent prostate carcinoma (PC-3) cells. Invasion of PC-3 cells is also inhibited by exogenously added noladin ether, a non-hydrolyzable analog of 2-AG. In contrast, exogenous 2-AG has the opposite effect. Cell invasion significantly increased with high concentrations of exogenous 2-AG. In PC-3 cells, arachidonic acid (AA) and 12-hydroxyeicosatetraenoic acid (12-HETE) concentrations increased along with exogenously added 2-AG, and 12-HETE concentrations increased with exogenously added AA. Invasion of PC-3 cells also increased with exogenously added AA and 12(S)-HETE but not 12(R)-HETE. The exogenous 2-AG-induced invasion of PC-3 cells was inhibited by 3-octylthio-1,1,1-trifluoropropan-2-one (OTFP, an inhibitor of 2-AG hydrolysis) and baicalein (a 12-LO inhibitor). Western blot and RT-PCR analyses indicated expression of 12-HETE producing lipoxygenases (LOs), platelet-type 12-LO (P-12-LO) and leukocyte-type 12-LO (L-12-LO), in PC-3 cells. These results suggest that exogenous 2-AG induced, rather inhibited, cell invasion because of its rapid hydrolysis to free AA, and further metabolism by 12-LO of AA to 12(S)-HETE, a promoter of PC cell invasion. The results also suggest that PC-3 cells and human prostate stromal (WPMY-1) cells released free AA, 2-AG, and 12-HETE. In the microenvironment of the PC cells, this may contribute to the cell invasion. The 2-AG hydrolysis and concentration of 2-AG in microenvironment are critical for PC cell's fate. Therefore, inhibitors of 2-AG hydrolysis could potentially serve as therapeutic agents for the treatment of prostate cancer. (c) 2007 Wiley-Liss, Inc.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonic Acids/metabolism , Glycerides/metabolism , Prostatic Neoplasms/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Arachidonic Acid/pharmacology , Arachidonic Acids/pharmacology , Blotting, Western , Chromatography, Liquid , Endocannabinoids , Glycerides/pharmacology , Humans , Hydrolysis , Male , Neoplasm Invasiveness , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Electrospray Ionization , Stromal Cells/metabolism
10.
Am J Physiol Heart Circ Physiol ; 290(2): H500-5, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16214838

ABSTRACT

Cytochrome P-450 (CYP) omega-hydroxylases and their arachidonic acid (AA) metabolite, 20-hydroxyeicosatetraenoic acid (20-HETE), produce a detrimental effect on ischemia-reperfusion injury in canine hearts, and the inhibition of CYP omega-hydroxylases markedly reduces myocardial infarct size expressed as a percentage of the area at risk (IS/AAR, %). In this study, we demonstrated that a specific CYP omega-hydroxylase inhibitor, N-methylsulfonyl-12,12-dibromododec-11-enamide (DDMS), markedly reduced 20-HETE production during ischemia-reperfusion and reduced myocardial infarct size compared with control [19.5 +/- 1.0% (control), 9.6 +/- 1.5% (0.40 mg/kg DDMS), 4.0 +/- 2.0% (0.81 mg/kg DDMS), P < 0.01]. In addition, 20-hydroxyeicosa-6(Z),15(Z)-dienoic acid (20-HEDE, a putative 20-HETE antagonist) significantly reduced myocardial infarct size from control [10.3 +/- 1.3% (0.032 mg/kg 20-HEDE) and 5.9 +/- 1.9% (0.064 mg/kg 20-HEDE), P < 0.05]. We further demonstrated that one 5-min period of ischemic preconditioning (IPC) reduced infarct size to a similar extent as that observed with the high doses of DDMS and 20-HEDE, and the higher dose of DDMS given simultaneously with IPC augmented the infarct size reduction [9.9 +/- 2.8% (IPC) to 2.5 +/- 1.4% (0.81 mg/kg DDMS), P < 0.05] to a greater degree than that observed with either treatment alone. These results suggest an important negative role for endogenous CYP omega-hydroxylases and their product, 20-HETE, to exacerbate myocardial injury in canine myocardium. Furthermore, for the first time, this study demonstrates that the effect of IPC and the inhibition of CYP omega-hydroxylase synthesis (DDMS) or its actions (20-HEDE) may have additive effects in protecting the canine heart from ischemia-reperfusion injury.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Ischemic Preconditioning, Myocardial , Mixed Function Oxygenases/antagonists & inhibitors , Myocardial Infarction/pathology , Amides/pharmacology , Animals , Arachidonic Acid/metabolism , Cardiotonic Agents/pharmacology , Coronary Circulation/drug effects , Cytochrome P-450 Enzyme System/metabolism , Dogs , Female , Hemodynamics/drug effects , Hydroxyeicosatetraenoic Acids/antagonists & inhibitors , Hydroxyeicosatetraenoic Acids/pharmacology , Male , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Sulfones/pharmacology
11.
Biochem Biophys Res Commun ; 332(4): 1028-33, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15919052

ABSTRACT

Endogenous 2-arachidonoylglycerol (2-AG) inhibits invasion of androgen-independent prostate cancer cells. Blocking cellular hydrolysis of 2-AG to increase its endogenous concentration results in a decrease in cell invasion. A series of compounds containing a trifluoromethyl ketone (TFK) moiety or the methyl analog (known to inhibit carboxylesterases) were investigated for their ability to inhibit 2-AG hydrolysis and prostate cancer cell invasion. Compounds containing a thioether beta to a TFK moiety inhibited 2-AG hydrolysis as well as cell invasion in a concentration-dependent manner. Inhibition of 2-AG hydrolysis increased concomitantly with inhibitor alkyl chain length from 4- to 12-carbons while inhibition of cell invasion exhibited a maximum at 8- to 10-carbons of the compounds. These results demonstrate a new series of 2-AG hydrolysis inhibitors as a potential therapeutic approach for prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Arachidonic Acids/metabolism , Enzyme Inhibitors/pharmacology , Glycerides/metabolism , Prostatic Neoplasms/pathology , Cannabinoid Receptor Modulators/metabolism , Cell Line, Tumor , Chromatography, Liquid , Collagen/chemistry , Dose-Response Relationship, Drug , Drug Combinations , Endocannabinoids , Humans , Hydrolysis , Ketones/chemistry , Laminin/chemistry , Male , Models, Chemical , Neoplasm Invasiveness , Proteoglycans/chemistry , Spectrometry, Mass, Electrospray Ionization , Sulfides/chemistry
12.
Cancer Res ; 64(24): 8826-30, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15604240

ABSTRACT

Endocannabinoids have been implicated in cancer. Increasing endogenous 2-arachidonoylglycerol (2-AG) by blocking its metabolism inhibits invasion of androgen-independent prostate cancer (PC-3 and DU-145) cells. Noladin ether (a stable 2-AG analog) and exogenous CB1 receptor agonists possess similar effects. Conversely, reducing endogenous 2-AG by inhibiting its synthesis or blocking its binding to CB1 receptors with antagonists increases the cell invasion. 2-AG and noladin ether decrease protein kinase A activity in these cells, indicating coupling of the CB1 receptor to downstream effectors. The results suggest that cellular 2-AG, acting through the CB1 receptor, is an endogenous inhibitor of invasive prostate cancer cells.


Subject(s)
Arachidonic Acids/physiology , Glycerides/physiology , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/pathology , Androgens/physiology , Arachidonic Acids/antagonists & inhibitors , Arachidonic Acids/biosynthesis , Arachidonic Acids/metabolism , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclohexanones/pharmacology , Endocannabinoids , Glycerides/antagonists & inhibitors , Glycerides/biosynthesis , Glycerides/metabolism , Humans , Hydrolysis , Lipoprotein Lipase/antagonists & inhibitors , Male , Neoplasm Invasiveness , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/biosynthesis , Receptor, Cannabinoid, CB1/physiology
13.
Circ Res ; 95(8): e65-71, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15388642

ABSTRACT

Cytochrome P450s (CYP) and their arachidonic acid (AA) metabolites have important roles in regulating vascular tone, but their function and specific pathways involved in modulating myocardial ischemia-reperfusion injury have not been clearly established. Thus, we characterized the effects of several selective CYPomega-hydroxylase inhibitors and a CYPomega-hydroxylase metabolite of AA, 20-hydroxyeicosatetraenoic acid (20-HETE), on the extent of ischemia-reperfusion injury in canine hearts. During 60 minutes of ischemia and particularly after 3 hours of reperfusion, 20-HETE was produced at high concentrations. A nonspecific CYP inhibitor, miconazole, and 2 specific CYPomega-hydroxylase inhibitors, 17-octadecanoic acid (17-ODYA) and N-methylsulfonyl-12,12-dibromododec-11-enamide (DDMS), markedly inhibited 20-HETE production during ischemia-reperfusion and produced a profound reduction in myocardial infarct size (expressed as a percent of the area at risk) (19.6+/-1.7% [control], 8.4+/-2.5% [0.96 mg/kg miconazole], 5.9+/-2.2% [0.28 mg/kg 17-ODYA], and 10.8+/-1.8% [0.40 mg/kg DDMS], P<0.05, respectively). Conversely, exogenous 20-HETE administration significantly increased infarct size (26.9+/-1.9%, P<0.05). Several CYPomega-hydroxylase isoforms, which are known to produce 20-HETE such as CYP4A1, CYP4A2, and CYP4F, were demonstrated to be present in canine heart tissue and their activity was markedly inhibited by incubation with 17-ODYA. These results indicate an important endogenous role for CYPomega-hydroxylases and in particular their product, 20-HETE, in exacerbating myocardial injury in canine myocardium. The full text of this article is available online at http://circres.ahajournals.org.


Subject(s)
Amides/therapeutic use , Cytochrome P-450 Enzyme Inhibitors , Fatty Acids, Unsaturated/therapeutic use , Miconazole/therapeutic use , Mixed Function Oxygenases/antagonists & inhibitors , Myocardial Ischemia/enzymology , Myocardial Reperfusion Injury/enzymology , Sulfones/therapeutic use , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , Amides/pharmacology , Animals , Arachidonic Acids/metabolism , Cytochrome P-450 Enzyme System/physiology , Dogs , Fatty Acids, Unsaturated/pharmacology , Female , Hydroxyeicosatetraenoic Acids/biosynthesis , Hydroxyeicosatetraenoic Acids/blood , Hydroxyeicosatetraenoic Acids/toxicity , Isoenzymes/antagonists & inhibitors , Isoenzymes/physiology , Male , Miconazole/pharmacology , Microsomes/enzymology , Mixed Function Oxygenases/physiology , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Ischemia/pathology , Myocardial Ischemia/prevention & control , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Sulfones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...