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










Publication year range
1.
Channels (Austin) ; 7(6): 493-502, 2013.
Article in English | MEDLINE | ID: mdl-24037327

ABSTRACT

Cardiac ATP-sensitive potassium channels (KATP) are found in both the sarcoplasmic reticulum (sarcKATP) and the inner membrane of mitochondria (mitoKATP). SarcKATP are composed of a pore containing subunit Kir6.2 and a regulatory sulfonylurea receptor subunit (SUR2), but the composition of mitoKATP remains unclear. An unusual intra-exonic splice variant of SUR2 (SUR2A-55) was previously identified in mitochondria of mammalian heart and brain, and by analogy with sarcKATP we proposed SUR2A-55 as a candidate regulatory subunit of mitoKATP. Although SUR2A-55 lacks the first nucleotide binding domain (NBD) and 2 transmembrane domains (TMD), it has a hybrid TMD and retains the second NBD. It resembles a hemi-ABC transporter suggesting it could multimerize to function as a regulatory subunit. A putative mitochondrial targeting signal in the N-terminal domain of SUR2A-55 was removed by truncation and when co-expressed with Kir6.1 and Kir6.2 it targeted to the plasma membrane and yielded KATP currents. Single channel conductance, mean open time, and burst open time of SUR2A-55 based KATP was similar to the full-length SUR2A based KATP. However, the SUR2A-55 KATP were 70-fold less sensitive to block by ATP, and twice as resistant to intracellular Ca (2+) inhibition compared with the SUR2A KATP, and were markedly insensitive to KATP drugs, pinacidil, diazoxide, and glybenclamide. These results suggest that the SUR2A-55 based channels would tend to be open under physiological conditions and in ischemia, and could account for cardiac and mitochondrial phenotypes protective for ischemia.


Subject(s)
Adenosine Triphosphate/metabolism , Electrophysiological Phenomena , KATP Channels/metabolism , Mitochondria/metabolism , Myocardium/cytology , Potassium Channels, Inwardly Rectifying/metabolism , Sulfonylurea Receptors/metabolism , Animals , Calcium/metabolism , Male , Mice , Protein Isoforms/metabolism , Rats
2.
Antioxid Redox Signal ; 18(4): 432-68, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-22897788

ABSTRACT

Reactive oxygen species (ROS) have been associated with various human diseases, and considerable attention has been paid to investigate their physiological effects. Various ROS are synthesized in the mitochondria and accumulate in the cytoplasm if the cellular antioxidant defense mechanism fails. The critical balance of this ROS synthesis and antioxidant defense systems is termed the redox system of the cell. Various cardiovascular diseases have also been affected by redox to different degrees. ROS have been indicated as both detrimental and protective, via different cellular pathways, for cardiac myocyte functions, electrophysiology, and pharmacology. Mostly, the ROS functions depend on the type and amount of ROS synthesized. While the literature clearly indicates ROS effects on cardiac contractility, their effects on cardiac excitability are relatively under appreciated. Cardiac excitability depends on the functions of various cardiac sarcolemal or mitochondrial ion channels carrying various depolarizing or repolarizing currents that also maintain cellular ionic homeostasis. ROS alter the functions of these ion channels to various degrees to determine excitability by affecting the cellular resting potential and the morphology of the cardiac action potential. Thus, redox balance regulates cardiac excitability, and under pathological regulation, may alter action potential propagation to cause arrhythmia. Understanding how redox affects cellular excitability may lead to potential prophylaxis or treatment for various arrhythmias. This review will focus on the studies of redox and cardiac excitation.


Subject(s)
Heart/physiopathology , Action Potentials , Animals , Arrhythmias, Cardiac/physiopathology , Calcium Channels, L-Type/physiology , Calcium Signaling , Humans , Myocardial Contraction , Myocardium/metabolism , Oxidation-Reduction , Potassium Channels/physiology , Reactive Oxygen Species/metabolism , Sodium Channels/physiology
4.
Vascul Pharmacol ; 56(1-2): 106-12, 2012.
Article in English | MEDLINE | ID: mdl-22197897

ABSTRACT

Endothelial 15-lipoxygenase-1 (15-LO-1) metabolites of arachidonic acid (AA), 11,12,15-trihydroxyeicosatrienoic acid (THETA) and 15-hydroxy-11,12-epoxyeicosatrienoic acid (HEETA) and nitric oxide (NO) mediate relaxations to acetylcholine (ACH). However, interactions between NO and the 15-LO-1 pathway have not been explored. Therefore, the effect of physiological and pharmacological concentrations of NO on 15-LO activity and relaxation was studied in rabbit aorta. In indomethacin-treated aortic rings, maximal ACH relaxations of 91.3±4.0%, decreased to 54.5±3.0% by the NO synthase inhibitor, nitro-l-arginine (LNA), to 49.8±3% by the guanylate cyclase (GC) inhibitor, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, to 63.7±4.9% by the lipoxygenase (LO) inhibitor, nordihydroguaiaretic acid (NDGA) and were completely inhibited by the combination of LNA and NDGA. AA relaxations were not affected by GC inhibition but were reduced by LO inhibition. The NO donor, dipropylenetriamine-NONOate (DPTA) caused concentration-related relaxations (EC(50)=4.7×10(-6)M). Aortic metabolism of (14)C-AA to THETA and HEETA was not altered by EC(50) concentrations of DPTA but were reduced 10-fold by 10(-3)M DPTA. In LNA-treated aorta, DPTA (3×10(-6)M) caused relaxations of 38.2.5±4%. Maximum relaxations to ACH did not differ in the presence and absence 3×10(-6)M DPTA (49.5±5% and 44.2±4%, respectively). These results indicate that NO and 15-LO-1 act in parallel to mediate ACH relaxations and NO does not alter 15-LO-1 activity.


Subject(s)
Aorta/drug effects , Arachidonate 15-Lipoxygenase/metabolism , Endothelium, Vascular/drug effects , Muscle, Smooth, Vascular/drug effects , Nitric Oxide/pharmacology , Acetylcholine/metabolism , Alkenes/pharmacology , Animals , Aorta/metabolism , Arachidonic Acid/metabolism , Endothelium, Vascular/metabolism , Guanylate Cyclase/antagonists & inhibitors , Indomethacin/pharmacology , Lipoxygenase Inhibitors/pharmacology , Muscle, Smooth, Vascular/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Rabbits , Vasodilation/drug effects
5.
Am J Physiol Heart Circ Physiol ; 300(3): H725-35, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21193584

ABSTRACT

Arachidonic acid (AA) metabolites function as EDHFs in arteries of many species. They mediate cyclooxygenase (COX)- and nitric oxide (NO)-independent relaxations to acetylcholine (ACh). However, the role of AA metabolites as relaxing factors in mouse arteries remains incompletely defined. ACh caused concentration-dependent relaxations of the mouse thoracic and abdominal aorta and carotid, femoral, and mesentery arteries (maximal relaxation: 57 ± 4%, 72 ± 4%, 82 ± 3%, 80 ± 3%, and 85 ± 3%, respectively). The NO synthase inhibitor nitro-L-arginine (L-NA; 30 µM) blocked relaxations in the thoracic aorta, and L-NA plus the COX inhibitor indomethacin (10 µM) inhibited relaxations in the abdominal aorta and carotid, femoral, and mesenteric arteries (maximal relaxation: 31 ± 10%, 33 ± 5%, 41 ± 8%, and 73 ± 3%, respectively). In mesenteric arteries, NO- and COX-independent relaxations to ACh were inhibited by the lipoxygenase (LO) inhibitors nordihydroguaiaretic acid (NDGA; 10 µM) and BW-755C (200 µM), the K(+) channel inhibitor apamin (1 µM), and 60 mM KCl and eliminated by endothelium removal. They were not altered by the cytochrome P-450 inhibitor N-methylsulfonyl-6-(2-propargyloxyphenyl)hexanamide (20 µM) or the epoxyeicosatrienoic acid antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (10 µM). AA relaxations were attenuated by NDGA or apamin and eliminated by 60 mM KCl. Reverse-phase HPLC analysis revealed arterial [(14)C]AA metabolites that comigrated with prostaglandins, trihydroxyeicosatrienoic acids (THETAs), hydroxyepoxyeicosatrienoic acids (HEETAs), and hydroxyeicosatetraenoic acids (HETEs). Epoxyeicosatrienoic acids were not observed. Mass spectrometry confirmed the identity of 6-keto-PGF(1α), PGE(2), 12-HETE, 15-HETE, HEETAs, 11,12,15-THETA, and 11,14,15-THETA. AA metabolism was blocked by NDGA and endothelium removal. 11(R),12(S),15(S)-THETA relaxations (maximal relaxation: 73 ± 3%) were endothelium independent and blocked by 60 mM KCl. Western immunoblot analysis and RT-PCR of the aorta and mesenteric arteries demonstrated protein and mRNA expression of leukocyte-type 12/15-LO. Thus, in mouse resistance arteries, 12/15-LO AA metabolites mediate endothelium-dependent relaxations to ACh and AA.


Subject(s)
Acetylcholine/metabolism , Arachidonate Lipoxygenases/metabolism , Vasodilation/drug effects , Vasodilator Agents/metabolism , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/pharmacology , Amides/pharmacology , Animals , Apamin/pharmacology , Arteries/metabolism , Arteries/physiopathology , Female , Indomethacin/pharmacology , Male , Masoprocol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Nitroarginine/pharmacology
6.
Am J Physiol Heart Circ Physiol ; 299(6): H1884-90, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20935152

ABSTRACT

The sulfonylurea receptor-2 (SUR2) is a subunit of ATP-sensitive potassium channels (K(ATP)) in heart. Mice with the SUR2 gene disrupted (SUR2m) are constitutively protected from ischemia-reperfusion (I/R) cardiac injury. This was surprising because K(ATP), either sarcolemmal or mitochondrial or both, are thought to be important for cardioprotection. We hypothesized that SUR2m mice have an altered mitochondrial phenotype that protects against I/R. Mitochondrial membrane potential (ΔΨ(m)), tolerance to Ca(2+) load, and reactive oxygen species (ROS) generation were studied by fluorescence-based assays, and volumetric changes in response to K(+) were measured by light scattering in isolated mitochondria. For resting SUR2m mitochondria compared with wild type, the ΔΨ(m) was less polarized (46.1 ± 0.4 vs. 51.9 ± 0.6%), tolerance to Ca(2+) loading was increased (163 ± 2 vs. 116 ± 2 µM), and ROS generation was enhanced with complex I [8.5 ± 1.2 vs. 4.9 ± 0.2 arbitrary fluorescence units (afu)/s] or complex II (351 ± 51.3 vs. 166 ± 36.2 afu/s) substrates. SUR2m mitochondria had greater swelling in K(+) medium (30.2 ± 3.1%) compared with wild type (14.5 ± 0.6%), indicating greater K(+) influx. Additionally, ΔΨ(m) decreased and swelling increased in the absence of ATP in SUR2m, but the sensitivity to ATP was less compared with wild type. When the mitochondria were subjected to hypoxia-reoxygenation, the decrease in respiration rates and respiratory control index was less in SUR2m. ΔΨ(m) maintenance in the SUR2m intact myocytes was also more tolerant to metabolic inhibition. In conclusion, the cardioprotection observed in the SUR2m mice is associated with a protected mitochondrial phenotype resulting from enhanced K(+) conductance that partially dissipated ΔΨ(m). These results have implications for possible SUR2 participation in mitochondrial K(ATP).


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Energy Metabolism , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels/metabolism , Receptors, Drug/metabolism , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cell Hypoxia , Cell Respiration , Genotype , Light , Male , Membrane Potential, Mitochondrial , Mice , Mice, Mutant Strains , Mitochondrial Swelling , Myocardial Reperfusion Injury/metabolism , Phenotype , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Reactive Oxygen Species/metabolism , Receptors, Drug/genetics , Scattering, Radiation , Spectrometry, Fluorescence , Sulfonylurea Receptors , Time Factors
7.
Circ Res ; 105(11): 1083-93, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19797704

ABSTRACT

RATIONALE: Cardioprotective pathways may involve a mitochondrial ATP-sensitive potassium (mitoK(ATP)) channel but its composition is not fully understood. OBJECTIVE: We hypothesized that the mitoK(ATP) channel contains a sulfonylurea receptor (SUR)2 regulatory subunit and aimed to identify the molecular structure. METHODS AND RESULTS: Western blot analysis in cardiac mitochondria detected a 55-kDa mitochondrial SUR2 (mitoSUR2) short form, 2 additional short forms (28 and 68 kDa), and a 130-kDa long form. RACE (Rapid Amplification of cDNA Ends) identified a 1.5-Kb transcript, which was generated by a nonconventional intraexonic splicing (IES) event within the 4th and 29th exons of the SUR2 mRNA. The translated product matched the predicted size of the 55-kDa short form. In a knockout mouse (SUR2KO), in which the SUR2 gene was disrupted, the 130-kDa mitoSUR2 was absent, but the short forms remained expressed. Diazoxide failed to induce increased fluorescence of flavoprotein oxidation in SUR2KO cells, indicating that the diazoxide-sensitive mitoK(ATP) channel activity was associated with 130-kDa-based channels. However, SUR2KO mice displayed similar infarct sizes to preconditioned wild type, suggesting a protective role for the remaining short form-based channels. Heterologous coexpression of the SUR2 IES variant and Kir6.2 in a K(+) transport mutant Escherichia coli strain permitted improved cell growth under acidic pH conditions. The SUR2 IES variant was localized to mitochondria, and removal of a predicted mitochondrial targeting sequence allowed surface expression and detection of an ATP-sensitive current when coexpressed with Kir6.2. CONCLUSIONS: We identify a novel SUR2 IES variant in cardiac mitochondria and provide evidence that the variant-based channel can form an ATP-sensitive conductance and may contribute to cardioprotection.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Alternative Splicing/physiology , Myocardial Ischemia/genetics , Myocytes, Cardiac/physiology , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/genetics , Receptors, Drug/metabolism , Animals , Cells, Cultured , Exons/genetics , Flavoproteins/metabolism , Gene Library , Humans , Mice , Mice, Knockout , Mitochondria/physiology , Myocardial Ischemia/metabolism , Myocytes, Cardiac/cytology , Oxidation-Reduction , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sulfonylurea Receptors
8.
Am J Physiol Heart Circ Physiol ; 296(3): H678-88, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19112096

ABSTRACT

15-Lipoxygenase (15-LO-1) metabolizes arachidonic acid (AA) to 11,12,15-trihydroxyeicosatrienoic acids (THETAs) and 15-hydroxy-11,12-epoxyeicosatrienoic acids (HEETA) that dilate rabbit arteries. Increased endothelial 15-LO-1 expression enhances arterial relaxations to agonists. We tested the effect of hypoxia on 15-LO-1 expression, THETA and HEETA synthesis, and relaxations in rabbit arteries. The incubation of rabbit aortic endothelial cells and isolated aortas in 0.7% O(2) increased 15-LO-1 expression. Rabbits were housed in a hypoxic atmosphere of 12% O(2) for 5 days. 15-LO-1 expression increased in the endothelium of the arteries of rabbits in 12% O(2) compared with room air. THETA and HEETA synthesis was also enhanced in aortas and mesenteric arteries. AA hyperpolarized the smooth muscle cells in indomethacin- and phenylephrine-treated mesenteric arteries of hypoxic rabbits from -29.4 +/- 1 to -50.1 +/- 3 mV. The hyperpolarization to AA was less in arteries of normoxic rabbits (from -26.0 +/- 2 to -37 +/- 2 mV). This AA-induced hyperpolarization was inhibited by the 15-LO inhibitor BW-755C. Nitric oxide and prostaglandin-independent maximum relaxations to acetylcholine (79.7 +/- 2%) and AA (38.3 +/- 4%) were enhanced in mesenteric arteries from hypoxic rabbits compared with the normoxic rabbits (49.7 +/- 6% and 19.9 +/- 2%, respectively). These relaxations were inhibited by BW-755C and nordihydroguaiaretic acid. Therefore, hypoxia increased the relaxations to agonists in the rabbit mesenteric arteries by enhancing endothelial 15-LO-1 expression and synthesis of the hyperpolarizing factors THETA and HEETA.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Arteries/enzymology , Hypoxia/enzymology , Vasodilation , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/metabolism , Animals , Arachidonate 15-Lipoxygenase/genetics , Arachidonic Acid/metabolism , Arteries/drug effects , Arteries/physiopathology , Biological Factors/metabolism , Cyclooxygenase Inhibitors/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Hypoxia/pathology , Hypoxia/physiopathology , Lipoxygenase Inhibitors/pharmacology , Male , Membrane Potentials , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide/metabolism , RNA, Messenger/metabolism , Rabbits , Time Factors , Tunica Intima/pathology , Vasoconstriction , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
9.
Arterioscler Thromb Vasc Biol ; 28(12): 2209-15, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18832753

ABSTRACT

OBJECTIVE: Arachidonic acid (AA) metabolites from 15-lipoxygenase-1 (15-LO-1), trihydroxyeicosatrienoic acid (THETA), and hydroxyepoxyeicosatrienoic acid (HEETA) relax arteries. We studied 15-LO-1 expression, THETA and HEETA synthesis, and their effect on arterial relaxations and blood pressure in hypercholesterolemic nonatherosclerotic rabbits. METHODS AND RESULTS: Immunoblots, RTPCR analysis, and (14)C-AA metabolism revealed that hypercholesterolemia increased 15-LO-1 expression in the endothelium and THETA and HEETA synthesis in the arteries. Isometric tension recording, in presence of nitric oxide synthase (NOS) and cyclooxygenase (COX) inhibitors, showed greater relaxations to acetylcholine (ACH) and AA (max 76.0+/-4.6% and 79.5+/-2.4%, respectively) in aortas from hypercholesterolemic rabbits compared with normal rabbits (max 39.1+/-2.8% and 39.9+/-2.2%, respectively). AA induced greater hyperpolarization in the smooth muscle cells of hypercholesterolemic aortas (-45.85+/-3.0 mV) compared with normal aortas (-31.45+/-1.9 mV). The ACH- and AA-relaxations were inhibited by 15-LO-1 inhibitors. ACH induced hypotensive responses were greater in hypercholesterolemic rabbits in absence (-54.9+/-3.3%) or presence (-48.5+/-3.2%) of NOS and COX-inhibitors compared with control rabbits (-31.6+/-3.3% and -24.3+/-1.6%, respectively). BW755C reduced these responses in hypercholesterolemic rabbits to -29.3+/-2.3%. CONCLUSIONS: Hypercholesterolemia increases endothelial 15-LO-1 expression, THETA and HEETA synthesis and enhances vasorelaxation.


Subject(s)
Arachidonate 15-Lipoxygenase/physiology , Hypercholesterolemia/physiopathology , Hypotension/physiopathology , Vasodilation/physiology , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/metabolism , Acetylcholine/pharmacology , Animals , Aorta/metabolism , Arachidonate 15-Lipoxygenase/genetics , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Arteries/metabolism , Blood Pressure/drug effects , Blood Pressure/physiology , Hypercholesterolemia/genetics , Hypotension/etiology , Hypotension/genetics , In Vitro Techniques , Lipoxygenase Inhibitors/pharmacology , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Vasodilation/drug effects
10.
Am J Physiol Heart Circ Physiol ; 295(1): H89-96, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18456739

ABSTRACT

Arachidonic acid (AA) metabolites from the 15-lipoxygenase-1 (15-LO-1) pathway, trihydroxyeicosatrienoic acids (THETAs) and hydroxy-epoxyeicosatrienoic acids (HEETAs), are endothelium-derived hyperpolarizing factors (EDHFs) and relax rabbit arteries. Rabbit vascular 15-LO-1 expression, THETA and HEETA synthesis, and nitric oxide and prostaglandin-independent relaxations to acetylcholine (ACh) and AA decreased with age (neonates to 16-wk-old). We characterized age-dependent ACh-hypotensive responses in vivo in 1-, 4-, 8-, and 16-wk-old rabbits and the contribution of THETAs and HEETAs to these responses. In anesthetized rabbits, blood pressure responses to ACh (4-4,000 ng/kg) were determined in the presence of vehicle or various inhibitors. ACh responses decreased with age (P > 0.001). In the absence or presence of N(omega)-nitro-l-arginine methyl ester (l-NAME) and indomethacin (Indo), maximum responses in 1 (-54.7 +/- 7.4 and -37.9 +/- 3.9%)- and 4 (-48.8 +/- 2.4 and -35.5 +/- 7.8%)-wk-old rabbits were higher than 8 (-30.0 +/- 2.8 and -26.6 +/- 4.4%)- and 16 (-36.7 +/- 3.5 and -27.3 +/- 10%)-wk-old rabbits. A lipoxygenase inhibitor, BW755C, reduced THETA and HEETA synthesis in mesenteric arteries. In the presence of Indo and N(omega)-nitro-l-arginine, ACh relaxations were reduced by BW755C to a greater extent in the mesenteric arteries from the younger rabbits. In 4-wk-old rabbits treated with l-NAME and Indo, the maximum ACh hypotension was reduced by the potassium channel inhibitors apamin and charybdotoxin to -6.9 +/- 0.9%, by apamin alone to -19.5 +/- 1.4%, and by BW755C to -18.8 +/- 3.5%. The present study indicates that the age-related decrease in ACh-induced hypotension is mediated by the decreased synthesis of the 15-LO-1 metabolites THETAs and HEETAs.


Subject(s)
Acetylcholine/pharmacology , Arachidonate 15-Lipoxygenase/metabolism , Hypotension/metabolism , Mesenteric Arteries/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology , 4,5-Dihydro-1-(3-(trifluoromethyl)phenyl)-1H-pyrazol-3-amine/pharmacology , Age Factors , Aging , Animals , Apamin/pharmacology , Blood Pressure/drug effects , Charybdotoxin/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Eicosanoids/metabolism , Hypotension/physiopathology , In Vitro Techniques , Indomethacin/pharmacology , Lipoxygenase Inhibitors/pharmacology , Mesenteric Arteries/drug effects , Mesenteric Arteries/enzymology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Potassium Channel Blockers/pharmacology , Rabbits
11.
Am J Physiol Heart Circ Physiol ; 294(3): H1348-56, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18192225

ABSTRACT

Arachidonic acid (AA) causes endothelium-dependent smooth muscle hyperpolarizations and relaxations that are mediated by a 15-lipoxygenase-I (15-LO-I) metabolite, 11,12,15-trihydroxyeicosatrienoic acid (11,12,15-THETA). We propose that AA is metabolized sequentially by 15-LO-I and hydroperoxide isomerase to an unidentified hydroxyepoxyeicosatrienoic acid (HEETA), which is hydrolyzed by a soluble epoxide hydrolase (sEH) to 11,12,15-THETA. After incubation of aorta with 14C-labeled AA, metabolites were extracted and the HEETAs were resolved by performing HPLC. Mass spectrometric analyses identified 15-Hydroxy-11,12-epoxyeicosatrienoic acid (15-H-11,12-EETA). Incubation of aortic incubates with methanol and acetic acid trapped the acid-sensitive 15-H-11,12-EETA as methoxydihydroxyeicosatrienoic acids (MDHEs) (367 m/z, M-H). Pretreatment of the aortic tissue with the sEH inhibitor 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA; 10(-6) M) increased the formation of 15-H-11,12-EETA, measured as MDHEs. Thus 15-H-11,12-EETA is an acid- and sEH-sensitive precursor of 11,12,15-THETA. Aortic homogenates and endothelial cells contain a 57-kDa protein corresponding to the rabbit sEH. In preconstricted aortic rings, AA (10(-7)-10(-4) M) and acetylcholine (10(-9)-10(-6) M) caused concentration-related relaxations that were enhanced by pretreatment with AUDA. These enhanced relaxations were inhibited by increasing extracellular [K(+)] from 4.8 to 20 mM. AA (3 x 10(-6) M) induced cell membrane hyperpolarization (from -31.0 +/- 1 to -46.8 +/- 2 mV) in aortic strips with an intact endothelium, which was enhanced by AUDA. These results indicate that 15-H-11,12-EETA is produced by the aorta, hydrolyzed by sEH to 11,12,15-THETA, and mediates relaxations by membrane hyperpolarization. 15-H-11,12-EETA represents an endothelium-derived hyperpolarizing factor.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Aorta, Thoracic/metabolism , Arachidonate 15-Lipoxygenase/metabolism , 8,11,14-Eicosatrienoic Acid/chemistry , 8,11,14-Eicosatrienoic Acid/metabolism , Acetylcholine/metabolism , Animals , Arachidonic Acids/metabolism , Blotting, Western , Chromatography, High Pressure Liquid , Epoxide Hydrolases/metabolism , Gas Chromatography-Mass Spectrometry , Membrane Potentials/physiology , Rabbits
12.
Hypertension ; 51(2): 246-51, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18180398

ABSTRACT

Arachidonic acid is metabolized by the 15-lipoxygenase-1 pathway to the vasodilatory eicosanoids hydroxy-epoxyeicosatrienoic acid and trihydroxyeicosatrienoic acid. We determined the in vitro and in vivo effects of the 15-lipoxygenase-1 metabolites in rabbits infected with adenovirus containing cDNA for human 15-lipoxygenase-1 (Ad-15-LO-1). Forty hours after intravenous adenoviral injection, 15-lipoxygenase-1 expression increased in liver and mesenteric arteries 10-fold and 3-fold, respectively. Expression of 15-LO-1 was limited to the endothelium of mesenteric arteries. Overexpression did not occur in tissues from rabbits infected with a beta-galactosidase containing adenovirus. Trihydroxyeicosatrienoic acid and hydroxy-epoxyeicosatrienoic acid synthesis per milligram of tissue increased by 2.1- and 1.5-fold, respectively, in mesenteric arteries from Ad-15-LO-1-infected rabbits compared with normal rabbits. Pretreatment with a 15-lipoxygenase inhibitor BW755C inhibited the synthesis. NO and prostaglandin-independent, maximal relaxations to acetylcholine were greater in mesenteric arteries from Ad-15-LO-1-infected rabbits (98.3+/-1.7%) compared with normal (60.93+/-10.5%) and beta-galactosidase containing adenovirus-infected rabbits (68.3+/-7.7%). Pretreatment with BW755C decreased these relaxations. Mean arterial pressure and heart rate did not differ in Ad-15-LO-1-infected rabbits compared with normal or beta-galactosidase containing adenovirus-infected rabbits. The hypotensive responses to acetylcholine in the presence and absence of inhibition of NO and prostaglandins were greater in Ad-15-LO-1-infected rabbits (-52+/-2% and -47+/-2%) compared with normal (-31+/-3% and -25+/-5%) or beta-galactosidase containing adenovirus-infected rabbits (-38+/-2% and -30+/-3%). Therefore, increased expression of 15-LO-1 increases acetylcholine relaxation in arteries and hypotensive responses in rabbits because of increased hydroxy-epoxyeicosatrienoic acid and trihydroxyeicosatrienoic acid synthesis.


Subject(s)
Acetylcholine , Arachidonate 15-Lipoxygenase/metabolism , Arteries/physiopathology , Endothelium, Vascular/enzymology , Hypotension/chemically induced , Vasodilation , Vasodilator Agents , Animals , Arachidonate 15-Lipoxygenase/genetics , Arachidonic Acid/metabolism , Arteries/enzymology , Blotting, Western , Gene Transfer Techniques , Hemodynamics , Humans , Hypotension/physiopathology , Immunohistochemistry , In Vitro Techniques , Isoenzymes/genetics , Isoenzymes/metabolism , Isometric Contraction , Liver/blood supply , Mesenteric Arteries/physiopathology , Rabbits , Up-Regulation
13.
Am J Physiol Heart Circ Physiol ; 292(2): H1033-41, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17040969

ABSTRACT

Endothelium-dependent vasorelaxation of the rabbit aorta is mediated by either nitric oxide (NO) or arachidonic acid (AA) metabolites from cyclooxygenase (COX) and 15-lipoxygenase (15-LO) pathways. 15-LO-1 metabolites of AA, 11,12,15-trihydroxyeicosatrienoic acid (THETA), and 15-hydroxy-11,12-epoxyeicosatrienoic acid (HEETA) cause concentration-dependent relaxation. We tested the hypothesis that in the 15-LO pathway of AA metabolism, 15-LO-1 is sufficient and is the rate-limiting step in inducing relaxations in rabbit aorta. Aorta and rabbit aortic endothelial cells were treated with adenoviruses containing human 15-LO-1 cDNA (Ad-15-LO-1) or beta-galactosidase (Ad-beta-Gal). Ad-15-LO-1-transduction increased the expression of a 75-kDa protein corresponding to 15-LO-1, detected by immunoblotting with an anti-human15-LO-1 antibody, and increased the production of HEETA and THETA from [(14)C]AA. Immunohistochemical studies on Ad-15-LO-1-transduced rabbit aorta showed the presence of 15-LO-1 in endothelial cells. Ad-15-LO-1-treated aortic rings showed enhanced relaxation to AA (max 31.7 +/- 3.2%) compared with Ad-beta-Gal-treated (max 12.7 +/- 3.2%) or control nontreated rings (max 13.1 +/- 1.6%) (P < 0.01). The relaxations in Ad-15-LO-1-treated aorta were blocked by the 15-LO inhibitor cinnamyl-3,4-dihydroxy-a-cyanocinnamate. Overexpression of 15-LO-1 in the rabbit aortic endothelium is sufficient to increase the production of the vasodilatory HEETA and THETA and enhance the relaxations to AA. This confirms the role of HEETA and THETA as endothelium-derived relaxing factors.


Subject(s)
Adenoviridae/genetics , Aorta, Thoracic/metabolism , Arachidonate 15-Lipoxygenase/biosynthesis , Arachidonic Acid/metabolism , Genetic Vectors , Vasodilation , Vasodilator Agents/metabolism , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/metabolism , Animals , Aorta, Thoracic/drug effects , Arachidonate 15-Lipoxygenase/genetics , Arachidonic Acid/pharmacology , Cells, Cultured , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Immunohistochemistry , In Vitro Techniques , Lipoxygenase Inhibitors/pharmacology , Molecular Structure , Rabbits , Tandem Mass Spectrometry , Transduction, Genetic , Vasodilation/drug effects , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...