Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nature ; 566(7745): 548-552, 2019 02.
Article in English | MEDLINE | ID: mdl-30760924

ABSTRACT

Singlet molecular oxygen (1O2) has well-established roles in photosynthetic plants, bacteria and fungi1-3, but not in mammals. Chemically generated 1O2 oxidizes the amino acid tryptophan to precursors of a key metabolite called N-formylkynurenine4, whereas enzymatic oxidation of tryptophan to N-formylkynurenine is catalysed by a family of dioxygenases, including indoleamine 2,3-dioxygenase 15. Under inflammatory conditions, this haem-containing enzyme is expressed in arterial endothelial cells, where it contributes to the regulation of blood pressure6. However, whether indoleamine 2,3-dioxygenase 1 forms 1O2 and whether this contributes to blood pressure control have remained unknown. Here we show that arterial indoleamine 2,3-dioxygenase 1 regulates blood pressure via formation of 1O2. We observed that in the presence of hydrogen peroxide, the enzyme generates 1O2 and that this is associated with the stereoselective oxidation of L-tryptophan to a tricyclic hydroperoxide via a previously unrecognized oxidative activation of the dioxygenase activity. The tryptophan-derived hydroperoxide acts in vivo as a signalling molecule, inducing arterial relaxation and decreasing blood pressure; this activity is dependent on Cys42 of protein kinase G1α. Our findings demonstrate a pathophysiological role for 1O2 in mammals through formation of an amino acid-derived hydroperoxide that regulates vascular tone and blood pressure under inflammatory conditions.


Subject(s)
Blood Pressure/physiology , Inflammation/blood , Inflammation/physiopathology , Singlet Oxygen/metabolism , Vasodilator Agents/metabolism , Animals , Cell Line , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/chemistry , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cysteine/metabolism , Enzyme Activation/drug effects , Female , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation/enzymology , Male , Oxidation-Reduction/drug effects , Rats , Signal Transduction , Singlet Oxygen/chemistry , Tryptophan/chemistry , Tryptophan/metabolism
2.
Arterioscler Thromb Vasc Biol ; 34(4): 887-93, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24558103

ABSTRACT

OBJECTIVE: Middle cerebral artery (MCA) diameter is regulated by inherent myogenic activity and the effect of potent vasodilators such as calcitonin gene-related peptide (CGRP). Previous studies showed that MCAs express KCNQ1, 4, and 5 potassium channel genes, and the expression products (Kv7 channels) participate in the myogenic control of MCA diameter. The present study investigated the contribution of Kv7.4 and Kv7.5 isoforms to myogenic and CGRP regulation of MCA diameter and determined whether they were affected in hypertensive animals. APPROACH AND RESULTS: Isometric tension recordings performed on MCA from normotensive rats produced CGRP vasodilations that were inhibited by the pan-Kv7 channel blocker linopirdine (P<0.01) and after transfection of arteries with siRNA against KCNQ4 (P<0.01) but not KCNQ5. However, isobaric myography revealed that myogenic constriction in response to increases in intravascular pressure (20-80 mm Hg) was affected by both KCNQ4 and KCNQ5 siRNA. Proximity ligation assay signals were equally abundant for Kv7.4/Kv7.4 or Kv7.4/Kv7.5 antibody combinations but minimal for Kv7.5/Kv7.5 antibodies or Kv7.4/7.1 combinations. In contrast to systemic arteries, Kv7 function and Kv7.4 abundance in MCA were not altered in hypertensive rats. CONCLUSIONS: This study reveals, for the first time to our knowledge, that in cerebral arteries, Kv7.4 and Kv7.5 proteins exist predominantly as a functional heterotetramer, which regulates intrinsic myogenicity and vasodilation attributed to CGRP. Surprisingly, unlike systemic arteries, Kv7 activity in MCAs is not affected by the development of hypertension, and CGRP-mediated vasodilation is well maintained. As such, cerebrovascular Kv7 channels could be amenable for therapeutic targeting in conditions such as cerebral vasospasm.


Subject(s)
Calcitonin Gene-Related Peptide/pharmacology , KCNQ Potassium Channels/metabolism , Middle Cerebral Artery/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Hypertension/metabolism , Hypertension/physiopathology , KATP Channels/metabolism , KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/genetics , Male , Middle Cerebral Artery/metabolism , Middle Cerebral Artery/physiopathology , Potassium Channel Blockers/pharmacology , Protein Isoforms , RNA Interference , Rats , Rats, Inbred SHR , Rats, Wistar , Transfection
3.
Am J Physiol Cell Physiol ; 304(8): C739-47, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23364266

ABSTRACT

This study investigated the molecular identity and impact of enhancing voltage-gated Na(+) (Na(V)) channels in the control of vascular tone. In rat isolated mesenteric and femoral arteries mounted for isometric tension recording, the vascular actions of the Na(V) channel activator veratridine were examined. Na(V) channel expression was probed by molecular techniques and immunocytochemistry. In mesenteric arteries, veratridine induced potent contractions (pEC(50) = 5.19 ± 0.20, E(max) = 12.0 ± 2.7 mN), which were inhibited by 1 µM TTX (a blocker of all Na(V) channel isoforms, except Na(V)1.5, Na(V)1.8, and Na(V)1.9), but not by selective blockers of Na(V)1.7 (ProTx-II, 10 nM) or Na(V)1.8 (A-80347, 1 µM) channels. The responses were insensitive to endothelium removal but were partly (~60%) reduced by chemical destruction of sympathetic nerves by 6-hydroxydopamine (2 mM) or antagonism at the α1-adrenoceptor by prazosin (1 µM). KB-R7943, a blocker of the reverse mode of the Na(+)/Ca(2+) exchanger (3 µM), inhibited veratridine contractions in the absence or presence of prazosin. T16A(inh)-A01, a Ca(2+)-activated Cl(-) channel blocker (10 µM), also inhibited the prazosin-resistant contraction to veratridine. Na(V) channel immunoreactivity was detected in freshly isolated mesenteric myocytes, with apparent colocalization with the Na(+)/Ca(2+) exchanger. Veratridine induced similar contractile effects in the femoral artery, and mRNA transcripts for Na(V)1.2 and Na(V)1.3 channels were evident in both vessel types. We conclude that, in addition to sympathetic nerves, NaV channels are expressed in vascular myocytes, where they are functionally coupled to the reverse mode of Na(+)/Ca(2+) exchanger and subsequent activation of Ca(2+)-activated Cl(-) channels, causing contraction. The TTX-sensitive Na(V)1.2 and Na(V)1.3 channels are likely involved in vascular control.


Subject(s)
NAV1.2 Voltage-Gated Sodium Channel/metabolism , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Vasoconstriction/physiology , Animals , Male , Mesenteric Arteries/physiology , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Wistar , Sodium Channel Blockers/pharmacology , Veratridine/pharmacology
4.
Histochem Cell Biol ; 139(2): 309-21, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23007290

ABSTRACT

Diet-induced obesity induces changes in mechanisms that are essential for the regulation of normal artery function, and in particular the function of the vascular endothelium. Using a rodent model that reflects the characteristics of human dietary obesity, in the rat saphenous artery we have previously demonstrated that endothelium-dependent vasodilation shifts from an entirely nitric oxide (NO)-mediated mechanism to one involving upregulation of myoendothelial gap junctions and intermediate conductance calcium-activated potassium channel activity and expression. This study investigates the changes in NO-mediated mechanisms that accompany this shift. In saphenous arteries from controls fed a normal chow diet, acetylcholine-mediated endothelium-dependent vasodilation was blocked by NO synthase and soluble guanylyl cyclase inhibitors, but in equivalent arteries from obese animals sensitivity to these agents was reduced. The expression of endothelial NO synthase (eNOS) and caveolin-3 in rat saphenous arteries was unaffected by obesity, whilst that of caveolin-1 monomer and large oligomeric complexes of caveolins-1 and -2 were increased in membrane-enriched samples. The density of caveolae was increased at the membrane and cytoplasm of endothelial and smooth muscle cells of saphenous arteries from obese rats. Dissociation of eNOS from caveolin-1, as a prerequisite for activation of the enzyme, may be compromised and thereby impair NO-mediated vasodilation in the saphenous artery from diet-induced obese rats. Such altered signaling mechanisms in obesity-related vascular disease represent significant potential targets for therapeutic intervention.


Subject(s)
Caveolae/metabolism , Caveolin 1/biosynthesis , Diet, High-Fat/adverse effects , Nitric Oxide/metabolism , Obesity/metabolism , Vasodilation , Animals , Caveolin 1/metabolism , Male , Rats , Rats, Sprague-Dawley
5.
Circulation ; 124(5): 602-11, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21747056

ABSTRACT

BACKGROUND: Voltage-gated potassium (K(+)) channels encoded by KCNQ genes (Kv7 channels) have been identified in various rodent and human blood vessels as key regulators of vascular tone; however, nothing is known about the functional impact of these channels in vascular disease. We ascertained the effect of 3 structurally different activators of Kv7.2 through Kv7.5 channels (BMS-204352, S-1, and retigabine) on blood vessels from normotensive and hypertensive animals. METHODS AND RESULTS: Precontracted thoracic aorta and mesenteric artery segments from normotensive rats were relaxed by all 3 Kv7 activators, with potencies of BMS-204352=S-1>retigabine. We also tested these agents in the coronary circulation using the Langendorff heart preparation. BMS-204352 and S-1 dose dependently increased coronary perfusion at concentrations between 0.1 and 10 µmol/L, whereas retigabine was effective at 1 to 10 µmol/L. In addition, S-1 increased K(+) currents in isolated mesenteric artery myocytes. The ability of these agents to relax precontracted vessels, increase coronary flow, or augment K(+) currents was impaired considerably in tissues isolated from spontaneously hypertensive rats (SHRs). Of the 5 KCNQ genes, only the expression of KCNQ4 was reduced (≈3.7 fold) in SHRs aorta. Kv7.4 protein levels were ≈50% lower in aortas and mesenteric arteries from spontaneously hypertensive rats compared with normotensive vessels. A similar attenuated response to S-1 and decreased Kv7.4 were observed in mesenteric arteries from mice made hypertensive by angiotensin II infusion compared with normotensive controls. CONCLUSIONS: In 2 different rat and mouse models of hypertension, the functional impact of Kv7 channels was dramatically downregulated.


Subject(s)
Hypertension/physiopathology , KCNQ Potassium Channels/physiology , Angiotensin II/pharmacology , Animals , Aorta, Thoracic/physiology , Blood Pressure/physiology , Carbamates/pharmacology , Down-Regulation/physiology , Hypertension/chemically induced , In Vitro Techniques , Indoles/pharmacology , KCNQ Potassium Channels/agonists , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mesenteric Arteries/physiology , Mice , Phenylenediamines/pharmacology , Rats , Rats, Inbred SHR , Rats, Wistar , Vasoconstrictor Agents/pharmacology
6.
J Pharmacol Exp Ther ; 336(3): 701-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21172909

ABSTRACT

Myoendothelial microdomain signaling via localized calcium-activated potassium channel (K(Ca)) and gap junction connexins (Cx) is critical for endothelium-dependent vasodilation in rat mesenteric artery. The present study determines the relative contribution of NO and gap junction-K(Ca) mediated microdomain signaling to endothelium-dependent vasodilation in human mesenteric artery. The hypothesis tested was that such activity is due to NO and localized K(Ca) and Cx activity. In mesenteric arteries from intestinal surgery patients, endothelium-dependent vasodilation was characterized using pressure myography with pharmacological intervention. Vessel morphology was examined using immunohistochemical and ultrastructural techniques. In vessel segments at 80 mm Hg, the intermediate (I)K(Ca) blocker 1-[(2-chlorophenyl)diphenyl-methyl]-1H-pyrazole (TRAM-34; 1 µM) inhibited bradykinin (0.1 nM-3 µM)-induced vasodilation, whereas the small (S) K(Ca) blocker apamin (50 and 100 nM) had no effect. Direct IK(Ca) activation with 1-ethyl-2-benzimidazolinone (1-EBIO; 10-300 µM) induced vasodilation, whereas cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine (1-30 µM), the SK(Ca) activator, failed to dilate arteries, whereas dilation induced by 1-EBIO (10-100 µM) was blocked by TRAM-34. Bradykinin-mediated vasodilation was attenuated by putative gap junction block with carbenoxolone (100 µM), with remaining dilation blocked by N-nitro l-arginine methyl ester (100 µM) and [1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one] (10 µM), NO synthase and soluble guanylate cyclase blockers, respectively. In human mesenteric artery, myoendothelial gap junction and IK(Ca) activity are consistent with Cx37 and IK(Ca) microdomain expression and distribution. Data suggest that endothelium-dependent vasodilation is primarily mediated by NO, IK(Ca), and gap junction Cx37 in this vessel. Myoendothelial microdomain signaling sites are present in human mesenteric artery and are likely to contribute to endothelium-dependent vasodilation via a mechanism that is conserved between species.


Subject(s)
Endothelium-Dependent Relaxing Factors/physiology , Gap Junctions/physiology , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Mesenteric Arteries/physiology , Nitric Oxide/physiology , Connexins/physiology , Female , Humans , Male , Middle Aged , Vasodilation/physiology , Gap Junction alpha-4 Protein
7.
J Pharmacol Exp Ther ; 335(2): 284-93, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20671071

ABSTRACT

The mechanisms involved in altered endothelial function in obesity-related cardiovascular disease are poorly understood. This study investigates the effect of chronic obesity on endothelium-dependent vasodilation and the relative contribution of nitric oxide (NO), calcium-activated potassium channels (K(Ca)), and myoendothelial gap junctions (MEGJs) in the rat saphenous artery. Obesity was induced by feeding rats a cafeteria-style diet (∼30 kJ as fat) for 16 to 20 weeks, with this model reflecting human dietary obesity etiology. Age- and sex-matched controls received standard chow (∼12 kJ as fat). Endothelium-dependent vasodilation was characterized in saphenous arteries by using pressure myography with pharmacological intervention, Western blotting, immunohistochemistry, and ultrastructural techniques. In saphenous artery from control, acetylcholine (ACh)-mediated endothelium-dependent vasodilation was blocked by NO synthase and soluble guanylate cyclase inhibition, whereas in obese rats, the ACh response was less sensitive to such inhibition. Conversely, the intermediate conductance K(Ca) (IK(Ca)) blocker 1-[(2-chlorophenyl)diphenyl-methyl]-1H pyrazole attenuates ACh-mediated dilation in obese, but not control, vessels. In a similar manner, putative gap junction block with carbenoxolone increased the pEC(50) for ACh in arteries from obese, but not control, rats. IK1 protein and MEGJ expression was up-regulated in the arteries of obese rats, an observation absent in control. Addition of the small conductance K(Ca) blocker apamin had no effect on ACh-mediated dilation in either control or obese rat vessels, consistent with unaltered SK3 expression. Up-regulation of distinct IK(Ca)- and gap junction-mediated pathways at myoendothelial microdomain sites, key mechanisms for endothelial-derived hyperpolarization-type activity, maintains endothelium-dependent vasodilation in diet-induced obese rat saphenous artery. Plasticity of myoendothelial coupling mechanisms represents a significant potential target for therapeutic intervention.


Subject(s)
Endothelium, Vascular/physiology , Gap Junctions/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/biosynthesis , Muscle, Smooth, Vascular/metabolism , Obesity/physiopathology , Vasodilation/physiology , Animals , Blotting, Western , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/ultrastructure , Gap Junctions/physiology , Gap Junctions/ultrastructure , Immunohistochemistry , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Male , Microscopy, Electron , Muscle, Smooth, Vascular/physiology , Muscle, Smooth, Vascular/ultrastructure , Myography , Obesity/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects
8.
Clin Exp Pharmacol Physiol ; 36(1): 67-76, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19018806

ABSTRACT

1. Modulation of vascular cell calcium is critical for the control of vascular tone, blood flow and pressure. 2. Specialized microdomain signalling sites associated with calcium modulation are present in vascular smooth muscle cells, where spatially localized channels and calcium store receptors interact functionally. Anatomical studies suggest that such sites are also present in endothelial cells. 3. The characteristics of these sites near heterocellular myoendothelial gap junctions (MEGJs) are described, focusing on rat mesenteric artery. The MEGJs enable current and small molecule transfer to coordinate arterial function and are thus critical for endothelium-derived hyperpolarization, regulation of smooth muscle cell diameter in response to contractile stimuli and vasomotor conduction over distance. 4. Although MEGJs occur on endothelial cell projections within internal elastic lamina (IEL) holes, not all IEL holes have MEGJ-related projections (approximately 0-50% of such holes have MEGJ-related projections, with variations occurring within and between vessels, species, strains and disease). 5. In rat mesenteric, saphenous and caudal cerebellar artery and hamster cheek pouch arteriole, but not rat middle cerebral artery or cremaster arteriole, intermediate conductance calcium-activated potassium channels (IK(Ca)) localize to endothelial cell projections. 6. Rat mesenteric artery MEGJ connexins and IK(Ca) are in close spatial association with endothelial cell inositol 1,4,5-trisphosphate receptors and endoplasmic reticulum. 7. Data suggest a relationship between spatially associated endothelial cell ion channels and calcium stores in modulation of calcium release and action. Differences in spatial relationships between ion channels and calcium stores in different vessels reflect heterogeneity in vasomotor function, representing a selective target for the control of endothelial and vascular function.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/physiology , Membrane Microdomains/physiology , Signal Transduction/physiology , Animals , Cricetinae , Gap Junctions/physiology , Mice , Rats
9.
Hypertension ; 62(6): 1090-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24082059

ABSTRACT

The goal of the present study was to determine the role of KCNQ-encoded Kv channels (Kv7 channels) in the passive and active regulation of coronary flow in normotensive and hypertensive rats. In left anterior descending coronary arteries from normotensive rats, structurally different Kv7.2 to 7.5 activators produced relaxations, which were considerably less in arteries from hypertensive rats and were not mimicked by the Kv7.1-specific activator R-L3. In isolated, perfused heart preparations, coronary flow rate increased in response to the Kv7.2 to 7.5 activator (S)-1 and was diminished in the presence of a Kv7 inhibitor. The expression levels of KCNQ1-5 and their known accessory KCNE1-5 subunits in coronary arteries were similar in normotensive and hypertensive rats as measured by quantitative polymerase chain reaction. However, Kv7.4 protein expression was reduced in hypertensive rats. Application of adenosine or A2A receptor agonist CGS-21680 produced concentration-dependent relaxations of coronary arteries from normotensive rats, which were attenuated by application of Kv7 inhibitors. Kv7 blockers also attenuated the ischemia-induced increase in coronary perfusion in Langendorff studies. Overall, these data establish Kv7 channels as crucial regulators of coronary flow at resting and after hypoxic insult.


Subject(s)
Blood Pressure/physiology , Coronary Vessels/metabolism , Hypertension/metabolism , KCNQ Potassium Channels/metabolism , Regional Blood Flow/physiology , Adenosine/pharmacology , Animals , Blood Pressure/drug effects , Coronary Vessels/drug effects , Coronary Vessels/physiopathology , Dose-Response Relationship, Drug , Hypertension/physiopathology , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/antagonists & inhibitors , Male , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/drug effects , Vasodilation/physiology
10.
Br J Pharmacol ; 168(3): 773-84, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22946562

ABSTRACT

BACKGROUND AND PURPOSE: T16A(inh) -A01 is a recently identified inhibitor of the calcium-activated chloride channel TMEM16A. The aim of this study was to test the efficacy of T16A(inh) -A01 for inhibition of calcium-activated chloride channels in vascular smooth muscle and consequent effects on vascular tone. EXPERIMENTAL APPROACH: Single channel and whole cell patch clamp was performed on single smooth muscle cells from rabbit pulmonary artery and mouse thoracic aorta. Isometric tension studies were performed on mouse thoracic aorta and mesenteric artery as well as human abdominal visceral adipose artery. KEY RESULTS: In rabbit pulmonary artery myocytes T16A(inh) -A01 (1-30 µM) inhibited single calcium (Ca(2+) )-activated chloride (Cl(-) ) channels and whole cell currents activated by 500 nM free Ca(2+) . Similar effects were observed for single Ca(2+) -activated Cl(-) channels in mouse thoracic aorta, and in both cell types, channel activity was abolished by two antisera raised against TMEM16A but not by a bestrophin antibody. The TMEM16A potentiator, F(act) (10 µM), increased single channel and whole cell Ca(2+) -activated Cl(-) currents in rabbit pulmonary arteries. In isometric tension studies, T16A(inh) -A01 relaxed mouse thoracic aorta pre-contracted with methoxamine with an IC(50) of 1.6 µM and suppressed the methoxamine concentration-effect curve. T16A(inh) -A01 did not affect the maximal contraction produced by 60 mM KCl and the relaxant effect of 10 µM T16A(inh) -A01 was not altered by incubation of mouse thoracic aorta in a cocktail of potassium (K(+) ) channel blockers. T16A(inh) -A01 (10 µM) also relaxed human visceral adipose arteries by 88 ± 3%. CONCLUSIONS AND IMPLICATIONS: T16A(inh) -A01 blocks calcium-activated chloride channels in vascular smooth muscle cells and relaxes murine and human blood vessels.


Subject(s)
Chloride Channels/antagonists & inhibitors , Myocytes, Smooth Muscle/drug effects , Neoplasm Proteins/antagonists & inhibitors , Pyrimidines/pharmacology , Thiazoles/pharmacology , Vasodilator Agents/pharmacology , Adipose Tissue/blood supply , Adipose Tissue/physiology , Animals , Anoctamin-1 , Aorta, Thoracic/cytology , Aorta, Thoracic/physiology , Chloride Channels/physiology , Humans , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Mice , Mice, Inbred BALB C , Myocytes, Smooth Muscle/physiology , Neoplasm Proteins/physiology , Pulmonary Artery/cytology , Pulmonary Artery/physiology , Rabbits
11.
Br J Pharmacol ; 166(4): 1377-87, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22251082

ABSTRACT

BACKGROUND AND PURPOSE: The aim of this study was to characterize the functional impact of KCNQ1-encoded voltage-dependent potassium channels (K(v)7.1) in the vasculature. EXPERIMENTAL APPROACH: Mesenteric arteries, intrapulmonary arteries and thoracic aortae were isolated from adult rats. K(v)7.1 channel expression was established by fluorescence immunocytochemistry. Wire myography determined functionality of these channels in response to selective blockers and activators. Xenopus oocytes expressing K(v)7.1 channels were used to assess the effectiveness of selective K(v)7.1 channel blockers. KEY RESULTS: K(v)7.1 channels were identified in arterial myocytes by immunocytochemistry. K(v)7.1 blockers HMR1556, L-768,673 (10 µM) and JNJ39490282 (JNJ282; 1 µM) had no contractile effects in arteries, whereas the pan-K(v)7 channel blocker linopirdine (10 µM) evoked robust contractions. Application of two compounds purported to activate K(v)7.1 channels, L-364 373 (R-L3) and mefenamic acid, relaxed mesenteric arteries preconstricted by methoxamine. These responses were reversed by HMR1556 or L-768,673 but not JNJ282. Similar effects were observed in the thoracic aorta and intrapulmonary arteries. CONCLUSIONS AND IMPLICATIONS: In contrast to previous assumptions, K(v)7.1 channels expressed in arterial myocytes are functional ion channels. Although these channels do not appear to contribute to resting vascular tone, K(v)7.1 activators were effective vasorelaxants.


Subject(s)
Aorta, Thoracic/metabolism , KCNQ1 Potassium Channel/metabolism , Mesenteric Arteries/metabolism , Potassium Channel Blockers/pharmacology , Pulmonary Artery/metabolism , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Female , Humans , In Vitro Techniques , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/genetics , KCNQ Potassium Channels/metabolism , KCNQ1 Potassium Channel/agonists , KCNQ1 Potassium Channel/antagonists & inhibitors , KCNQ1 Potassium Channel/genetics , Male , Membrane Potentials/drug effects , Mesenteric Arteries/cytology , Mesenteric Arteries/drug effects , Oocytes/drug effects , Oocytes/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Rats , Rats, Wistar , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Xenopus laevis
12.
Hypertension ; 59(4): 877-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22353613

ABSTRACT

KCNQ4-encoded voltage-dependent potassium (Kv7.4) channels are important regulators of vascular tone that are severely compromised in models of hypertension. However, there is no information as to the role of these channels in responses to endogenous vasodilators. We used a molecular knockdown strategy, as well as pharmacological tools, to examine the hypothesis that Kv7.4 channels contribute to ß-adrenoceptor-mediated vasodilation in the renal vasculature and underlie the vascular deficit in spontaneously hypertensive rats. Quantitative PCR and immunohistochemistry confirmed gene and protein expression of KCNQ1, KCNQ3, KCNQ4, KCNQ5, and Kv7.1, Kv7.4, and Kv7.5 in rat renal artery. Isoproterenol produced concentration-dependent relaxation of precontracted renal arteries and increased Kv7 channel currents in isolated smooth muscle cells. Application of the Kv7 blocker linopirdine attenuated isoproterenol-induced relaxation and current. Isoproterenol-induced relaxations were also reduced in arteries incubated with small interference RNAs targeted to KCNQ4 that produced a ≈60% decrease in Kv7.4 protein level. Relaxation to isoproterenol and the Kv7 activator S-1 were abolished in arteries from spontaneously hypertensive rats, which was associated with ≈60% decrease in Kv7.4 abundance. This study provides the first evidence that Kv7 channels contribute to ß-adrenoceptor-mediated vasodilation in the renal vasculature and that abrogation of Kv7.4 channels is strongly implicated in the impaired ß-adrenoceptor pathway in spontaneously hypertensive rats. These findings may provide a novel pathogenic link between arterial dysfunction and hypertension.


Subject(s)
Hypertension/physiopathology , KCNQ Potassium Channels/deficiency , Receptors, Adrenergic, beta/physiology , Renal Artery/physiology , Vasodilation/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Isoproterenol/pharmacology , KCNQ Potassium Channels/drug effects , KCNQ Potassium Channels/genetics , Male , RNA, Small Interfering/pharmacology , Rats , Rats, Inbred SHR , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Vasodilation/drug effects
13.
Cardiovasc Res ; 95(4): 439-47, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22721989

ABSTRACT

AIMS: Microdomain signalling mechanisms underlie key aspects of artery function and the modulation of intracellular calcium, with transient receptor potential (TRP) channels playing an integral role. This study determines the distribution and role of TRP canonical type 3 (C3) channels in the control of endothelium-derived hyperpolarization (EDH)-mediated vasodilator tone in rat mesenteric artery. METHODS AND RESULTS: TRPC3 antibody specificity was verified using rat tissue, human embryonic kidney (HEK)-293 cells stably transfected with mouse TRPC3 cDNA, and TRPC3 knock-out (KO) mouse tissue using western blotting and confocal and ultrastructural immunohistochemistry. TRPC3-Pyr3 (ethyl-1-(4-(2,3,3-trichloroacrylamide)phenyl)-5-(trifluoromethyl)-1H-pyrazole-4-carboxylate) specificity was verified using patch clamp of mouse mesenteric artery endothelial and TRPC3-transfected HEK cells, and TRPC3 KO and wild-type mouse aortic endothelial cell calcium imaging and mesenteric artery pressure myography. TRPC3 distribution, expression, and role in EDH-mediated function were examined in rat mesenteric artery using immunohistochemistry and western blotting, and pressure myography and endothelial cell membrane potential recordings. In rat mesenteric artery, TRPC3 was diffusely distributed in the endothelium, with approximately five-fold higher expression at potential myoendothelial microdomain contact sites, and immunoelectron microscopy confirmed TRPC3 at these sites. Western blotting and endothelial damage confirmed primary endothelial TRPC3 expression. In rat mesenteric artery endothelial cells, Pyr3 inhibited hyperpolarization generation, and with individual SK(Ca) (apamin) or IK(Ca) (TRAM-34) block, Pyr3 abolished the residual respective IK(Ca)- and SK(Ca)-dependent EDH-mediated vasodilation. CONCLUSION: The spatial localization of TRPC3 and associated channels, receptors, and calcium stores are integral for myoendothelial microdomain function. TRPC3 facilitates endothelial SK(Ca) and IK(Ca) activation, as key components of EDH-mediated vasodilator activity and for regulating mesenteric artery tone.


Subject(s)
Biological Factors/metabolism , Endothelium, Vascular/metabolism , Mesenteric Arteries/metabolism , TRPC Cation Channels/metabolism , Vasodilation , Animals , Arterial Pressure , Blotting, Western , Calcium/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/ultrastructure , HEK293 Cells , Humans , Immunohistochemistry , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Membrane Potentials , Mesenteric Arteries/drug effects , Mesenteric Arteries/ultrastructure , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Immunoelectron , Myography , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/metabolism , TRPC Cation Channels/drug effects , TRPC Cation Channels/genetics , Transfection , Vasodilation/drug effects , Vasodilator Agents/pharmacology
14.
PLoS One ; 6(1): e16423, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21283658

ABSTRACT

BACKGROUND: The vascular endothelium plays a critical role in the control of blood flow. Altered endothelium-mediated vasodilator and vasoconstrictor mechanisms underlie key aspects of cardiovascular disease, including those in obesity. Whilst the mechanism of nitric oxide (NO)-mediated vasodilation has been extensively studied in obesity, little is known about the impact of obesity on vasodilation to the endothelium-derived hyperpolarization (EDH) mechanism; which predominates in smaller resistance vessels and is characterized in this study. METHODOLOGY/PRINCIPAL FINDINGS: Membrane potential, vessel diameter and luminal pressure were recorded in 4(th) order mesenteric arteries with pressure-induced myogenic tone, in control and diet-induced obese rats. Obesity, reflecting that of human dietary etiology, was induced with a cafeteria-style diet (∼30 kJ, fat) over 16-20 weeks. Age and sexed matched controls received standard chow (∼12 kJ, fat). Channel protein distribution, expression and vessel morphology were determined using immunohistochemistry, Western blotting and ultrastructural techniques. In control and obese rat vessels, acetylcholine-mediated EDH was abolished by small and intermediate conductance calcium-activated potassium channel (SK(Ca)/IK(Ca)) inhibition; with such activity being impaired in obesity. SK(Ca)-IK(Ca) activation with cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine (CyPPA) and 1-ethyl-2-benzimidazolinone (1-EBIO), respectively, hyperpolarized and relaxed vessels from control and obese rats. IK(Ca)-mediated EDH contribution was increased in obesity, and associated with altered IK(Ca) distribution and elevated expression. In contrast, the SK(Ca)-dependent-EDH component was reduced in obesity. Inward-rectifying potassium channel (K(ir)) and Na(+)/K(+)-ATPase inhibition by barium/ouabain, respectively, attenuated and abolished EDH in arteries from control and obese rats, respectively; reflecting differential K(ir) expression and distribution. Although changes in medial properties occurred, obesity had no effect on myoendothelial gap junction density. CONCLUSION/SIGNIFICANCE: In obese rats, vasodilation to EDH is impaired due to changes in the underlying potassium channel signaling mechanisms. Whilst myoendothelial gap junction density is unchanged in arteries of obese compared to control, increased IK(Ca) and Na(+)/K(+)-ATPase, and decreased K(ir) underlie changes in the EDH mechanism.


Subject(s)
Diet/adverse effects , Endothelium, Vascular/pathology , Membrane Potentials , Obesity/physiopathology , Potassium Channels/analysis , Signal Transduction/physiology , Animals , Gap Junctions , Obesity/etiology , Rats , Tissue Distribution , Vasodilation
SELECTION OF CITATIONS
SEARCH DETAIL