Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Physiol ; 14: 1278632, 2023.
Article in English | MEDLINE | ID: mdl-37745235
3.
Front Physiol ; 13: 1033216, 2022.
Article in English | MEDLINE | ID: mdl-36589427

ABSTRACT

There is strong evidence that the omega-3 polyunsaturated fatty acids (n-3 PUFAs) docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) have cardioprotective effects. n-3 PUFAs cause vasodilation in hypertensive patients, in part controlled by increased membrane conductance to potassium. As KATP channels play a major role in vascular tone regulation and are involved in hypertension, we aimed to verify whether n-3 PUFA-mediated vasodilation involved the opening of KATP channels. We used a murine model in which the KATP channel pore subunit, Kir6.1, is deleted in vascular smooth muscle. The vasomotor response of preconstricted arteries to physiologically relevant concentrations of DHA and EPA was measured using wire myography, using the channel blocker PNU-37883A. The effect of n-3 PUFAs on potassium currents in wild-type native smooth muscle cells was investigated using whole-cell patch clamping. DHA and EPA induced vasodilation in mouse aorta and mesenteric arteries; relaxations in the aorta were sensitive to KATP blockade with PNU-37883A. Endothelium removal didn't affect relaxation to EPA and caused a small but significant inhibition of relaxation to DHA. In the knock-out model, relaxations to DHA and EPA were unaffected by channel knockdown but were still inhibited by PNU-37883A, indicating that the action of PNU-37883A on relaxation may not reflect inhibition of KATP. In native aortic smooth muscle cells DHA failed to activate KATP currents. We conclude that DHA and EPA cause vasodilation in mouse aorta and mesenteric arteries. Relaxations in blocker-treated arteries from knock-out mice demonstrate that KATP channels are not involved in the n-3 PUFA-induced relaxation.

4.
Br J Pharmacol ; 178(4): 860-877, 2021 02.
Article in English | MEDLINE | ID: mdl-33283269

ABSTRACT

Hypertension is often characterised by impaired vasodilation involving dysfunction of multiple vasodilatory mechanisms. ω-3 polyunsaturated fatty acids (PUFAs), docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) can reduce blood pressure and vasodilation. In the endothelium, DHA and EPA improve function including increased NO bioavailability. However, animal studies show that DHA- and EPA-mediated vasodilation persists after endothelial removal, indicating a role for vascular smooth muscle cells (VSMCs). The vasodilatory effects of ω-3 PUFAs on VSMCs are mediated via opening of large conductance calcium-activated potassium channels (BKCa ), ATP-sensitive potassium channels (KATP ) and possibly members of the Kv 7 family of voltage-activated potassium channels, resulting in hyperpolarisation and relaxation. ω-3 PUFA actions on BKCa and voltage-gated ion channels involve electrostatic interactions that are dependent on the polyunsaturated acyl tail, cis-geometry of these double bonds and negative charge of the carboxyl headgroup. This suggests structural manipulation of ω-3 PUFA could generate novel, targeted, therapeutic leads.


Subject(s)
Fatty Acids, Omega-3 , Hypertension , Animals , Docosahexaenoic Acids , Eicosapentaenoic Acid , Vasodilation
5.
Br J Pharmacol ; 177(4): 912-928, 2020 02.
Article in English | MEDLINE | ID: mdl-31693171

ABSTRACT

BACKGROUND AND PURPOSE: Epidiolex™, a form of highly purified cannabidiol (CBD) derived from Cannabis plants, has demonstrated seizure control activity in patients with Dravet syndrome, without a fully elucidated mechanism of action. We have employed an unbiased approach to investigate this mechanism at a cellular level. EXPERIMENTAL APPROACH: We use a tractable biomedical model organism, Dictyostelium, to identify a protein controlling the effect of CBD and characterize this mechanism. We then translate these results to a Dravet syndrome mouse model and an acute in vitro seizure model. KEY RESULTS: CBD activity is partially dependent upon the mitochondrial glycine cleavage system component, GcvH1 in Dictyostelium, orthologous to the human glycine cleavage system component H protein, which is functionally linked to folate one-carbon metabolism (FOCM). Analysis of FOCM components identified a mechanism for CBD in directly inhibiting methionine synthesis. Analysis of brain tissue from a Dravet syndrome mouse model also showed drastically altered levels of one-carbon components including methionine, and an in vitro rat seizure model showed an elevated level of methionine that is attenuated following CBD treatment. CONCLUSIONS AND IMPLICATIONS: Our results suggest a novel mechanism for CBD in the regulating methionine levels and identify altered one-carbon metabolism in Dravet syndrome and seizure activity.


Subject(s)
Cannabidiol , Dictyostelium , Epilepsy , Lennox Gastaut Syndrome , Animals , Anticonvulsants/therapeutic use , Cannabidiol/therapeutic use , Carbon Cycle , Epilepsy/drug therapy , Humans , Lennox Gastaut Syndrome/drug therapy , Methionine/therapeutic use , Rats
6.
Hypertension ; 71(6): 1091-1100, 2018 06.
Article in English | MEDLINE | ID: mdl-29686000

ABSTRACT

Voltage-gated Kv7.4 channels have been implicated in vascular smooth muscle cells' activity because they modulate basal arterial contractility, mediate responses to endogenous vasorelaxants, and are downregulated in several arterial beds in different models of hypertension. Angiotensin II (Ang II) is a key player in hypertension that affects the expression of several classes of ion channels. In this study, we evaluated the effects of Ang II on the expression and function of vascular Kv7.4. Western blot and quantitative polymerase chain reaction revealed that in whole rat mesenteric artery, Ang II incubation for 1 to 7 hours decreased Kv7.4 protein expression without reducing transcript levels. Moreover, Ang II decreased XE991 (Kv7)-sensitive currents and attenuated membrane potential hyperpolarization and relaxation induced by the Kv7 activator ML213. Ang II also reduced Kv7.4 staining at the plasma membrane of vascular smooth muscle cells. Proteasome inhibition with MG132 prevented Ang II-induced decrease of Kv7.4 levels and counteracted the functional impairment of ML213-induced relaxation in myography experiments. Proximity ligation assays showed that Ang II impaired the interaction of Kv7.4 with the molecular chaperone HSP90 (heat shock protein 90), enhanced the interaction of Kv7.4 with the E3 ubiquitin ligase CHIP (C terminus of Hsp70-interacting protein), and increased Kv7.4 ubiquitination. Similar alterations were found in mesenteric vascular smooth muscle cells isolated from Ang II-infused mice. The effect of Ang II was emulated by 17-AAG (17-demethoxy-17-(2-propenylamino) geldanamycin) that inhibits HSP90 interactions with client proteins. These results show that Ang II downregulates Kv7.4 by altering protein stability through a decrease of its interaction with HSP90. This leads to the recruitment of CHIP and Kv7.4 ubiquitination and degradation via the proteasome.


Subject(s)
Angiotensin II/pharmacology , Down-Regulation , HSP90 Heat-Shock Proteins/metabolism , Hypertension/genetics , KCNQ Potassium Channels/genetics , Muscle, Smooth, Vascular/metabolism , Vasodilation/physiology , Animals , Blotting, Western , Disease Models, Animal , Gene Expression Regulation , Hypertension/metabolism , Hypertension/physiopathology , KCNQ Potassium Channels/biosynthesis , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Muscle, Smooth, Vascular/physiopathology , Oxidative Stress , Rats , Rats, Wistar
7.
PLoS One ; 13(2): e0192484, 2018.
Article in English | MEDLINE | ID: mdl-29394279

ABSTRACT

BACKGROUND AND PURPOSE: Increasing evidence suggests that the omega-3 polyunsaturated acids (n-3 PUFA), docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), are beneficial to cardiovascular health, promoting relaxation of vascular smooth muscle cells and vasodilation. Numerous studies have attempted to study these responses, but to date there has not been a systematic characterisation of both DHA and EPA mediated vasodilation in conduit and resistance arteries. Therefore, we aimed to fully characterise the n-3 PUFA-induced vasodilation pathways in rat aorta and mesenteric artery. METHODS: Wire myography was used to measure the vasomotor responses of freshly dissected rat mesenteric artery and aorta. Arteries were pre-constricted with U46619 and cumulative concentrations of either DHA or EPA (10 nM-30 µM) were added. The mechanisms by which n-3 PUFA relaxed arteries were investigated using inhibitors of vasodilator pathways, which include: nitric oxide synthase (NOS; L-NAME), cycloxygenase (COX; indomethacin), cytochrome P450 epoxygenase (CYP450; clotrimazole); and calcium-activated potassium channels (KCa), SKCa (apamin), IKCa (TRAM-34) and BKCa (paxilline). RESULTS: Both DHA- and EPA-induced relaxations were partially inhibited following endothelium removal in rat mesenteric arteries. Similarly, in aorta EPA-induced relaxation was partially suppressed due to endothelium removal. CYP450 also contributed to EPA-induced relaxation in mesenteric artery. Inhibition of IKCa partially attenuated DHA-induced relaxation in aorta and mesenteric artery along with EPA-induced relaxation in mesenteric artery. Furthermore, this inhibition of DHA- and EPA-induced relaxation was increased following the additional blockade of BKCa in these arteries. CONCLUSIONS: This study provides evidence of heterogeneity in the vasodilation mechanisms of DHA and EPA in different vascular beds. Our data also demonstrates that endothelium removal has little effect on relaxations produced by either PUFA. We demonstrate IKCa and BKCa are involved in DHA-induced relaxation in rat aorta and mesenteric artery; and EPA-induced relaxation in rat mesenteric artery only. CYP450 derived metabolites of EPA may also be involved in BKCa dependent relaxation. To our knowledge this is the first study indicating the involvement of IKCa in n-3 PUFA mediated relaxation.


Subject(s)
Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Omega-3/pharmacology , Vasodilation/drug effects , Animals , Cytochrome P-450 Enzyme System/metabolism , Male , Nitric Oxide Synthase Type III/pharmacology , Rats
8.
Sci Rep ; 7(1): 12389, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28959041

ABSTRACT

Calcitonin receptor-like receptor (CLR) and the receptor activity-modifying protein 2 (RAMP2) comprise a receptor for adrenomedullin (AM). Although it is known that AM induces internalization of CLR•RAMP2, little is known about the molecular mechanisms that regulate the trafficking of CLR•RAMP2. Using HEK and HMEC-1 cells, we observed that AM-induced activation of CLR•RAMP2 promoted ubiquitination of CLR. A mutant (CLRΔ9KR), lacking all intracellular lysine residues was functional and trafficked similar to the wild-type receptor, but was not ubiquitinated. Degradation of CLR•RAMP2 and CLRΔ9KR•RAMP2 was not dependent on the duration of AM stimulation or ubiquitination and occurred via a mechanism that was partially prevented by peptidase inhibitors. Degradation of CLR•RAMP2 was sensitive to overexpression of hepatocyte growth factor-regulated tyrosine kinase substrate (HRS), but not to HRS knockdown, whereas CLRΔ9KR•RAMP2 degradation was unaffected. Overexpression, but not knockdown of HRS, promoted hyperubiquitination of CLR under basal conditions. Thus, we propose a role for ubiquitin and HRS in the regulation of AM-induced degradation of CLR•RAMP2.


Subject(s)
Adrenomedullin/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Phosphoproteins/metabolism , Receptors, Adrenomedullin/metabolism , Ubiquitination/physiology , Calcitonin Receptor-Like Protein/genetics , Calcitonin Receptor-Like Protein/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Phosphoproteins/genetics , Protein Transport , Proteolysis , RNA, Small Interfering/metabolism , Receptor Activity-Modifying Protein 2/genetics , Receptor Activity-Modifying Protein 2/metabolism , Ubiquitin/metabolism
9.
Front Physiol ; 8: 500, 2017.
Article in English | MEDLINE | ID: mdl-28747891

ABSTRACT

K+ channels encoded by the ether-a-go-go related gene (ERG1 or KCNH2) are important determinants of the cardiac action potential. Expression of both cardiac isoforms (ERG1a and ERG1b) were identified in murine portal vein and distinctive voltage-gated K+ currents were recorded from single myocytes. The aim of the present study was to ascertain the expression and functional impact of ERG channels in murine arteries. Methods: Quantitative RT-PCR was undertaken on RNA extracted from a number of murine arteries. Immunofluorescence was performed on single vascular smooth muscle cells using antibodies against the ERG1 expression product (Kv11.1). Single cell electrophysiology was performed on myocytes from portal vein and several different arteries, complimented by isometric tension recordings. Proliferation assays were undertaken on smooth muscle cells isolated from femoral arteries. Results: ERG1 transcripts were detected in all murine blood vessels, and Kv11.1 immunofluorescence was observed in all smooth muscle cells. However, K+ currents with properties consistent with ERG channels were only recorded in portal vein myocytes. Moreover, ERG channel blockers (E4031 or dofetilide, 1 µM) failed to depolarize carotid arteries or produce contraction. Proliferation of arterial smooth muscle cells was associated with a marked increase in ERG1 expression and ERG blockers suppressed proliferation significantly. Conclusions: These data reveal that arterial blood vessels express ERG channels that appear to be functional silent in contractile smooth muscle but contribute to proliferative response.

10.
PeerJ ; 2: e414, 2014.
Article in English | MEDLINE | ID: mdl-24949235

ABSTRACT

Background and Purpose. In rat middle cerebral arteries, endothelium-dependent hyperpolarization (EDH) is mediated by activation of calcium-activated potassium (KCa) channels specifically KCa2.3 and KCa3.1. Lipoxygenase (LOX) products function as endothelium-derived hyperpolarizing factors (EDHFs) in rabbit arteries by stimulating KCa2.3. We investigated if LOX products contribute to EDH in rat cerebral arteries. Methods. Arachidonic acid (AA) metabolites produced in middle cerebral arteries were measured using HPLC and LC/MS. Vascular tension and membrane potential responses to SLIGRL were simultaneously recorded using wire myography and intracellular microelectrodes. Results. SLIGRL, an agonist at PAR2 receptors, caused EDH that was inhibited by a combination of KCa2.3 and KCa3.1 blockade. Non-selective LOX-inhibition reduced EDH, whereas inhibition of 12-LOX had no effect. Soluble epoxide hydrolase (sEH) inhibition enhanced the KCa2.3 component of EDH. Following NO synthase (NOS) inhibition, the KCa2.3 component of EDH was absent. Using HPLC, middle cerebral arteries metabolized (14)C-AA to 15- and 12-LOX products under control conditions. With NOS inhibition, there was little change in LOX metabolites, but increased F-type isoprostanes. 8-iso-PGF2α inhibited the KCa2.3 component of EDH. Conclusions. LOX metabolites mediate EDH in rat middle cerebral arteries. Inhibition of sEH increases the KCa2.3 component of EDH. Following NOS inhibition, loss of KCa2.3 function is independent of changes in LOX production or sEH inhibition but due to increased isoprostane production and subsequent stimulation of TP receptors. These findings have important implications in diseases associated with loss of NO signaling such as stroke; where inhibition of sEH and/or isoprostane formation may of benefit.

11.
PLoS One ; 7(10): e46735, 2012.
Article in English | MEDLINE | ID: mdl-23056429

ABSTRACT

BACKGROUND: In rat middle cerebral and mesenteric arteries the K(Ca)2.3 component of endothelium-dependent hyperpolarization (EDH) is lost following stimulation of thromboxane (TP) receptors, an effect that may contribute to the endothelial dysfunction associated with cardiovascular disease. In cerebral arteries, K(Ca)2.3 loss is associated with NO synthase inhibition, but is restored if TP receptors are blocked. The Rho/Rho kinase pathway is central for TP signalling and statins indirectly inhibit this pathway. The possibility that Rho kinase inhibition and statins sustain K(Ca)2.3 hyperpolarization was investigated in rat middle cerebral arteries (MCA). METHODS: MCAs were mounted in a wire myograph. The PAR2 agonist, SLIGRL was used to stimulate EDH responses, assessed by simultaneous measurement of smooth muscle membrane potential and tension. TP expression was assessed with rt-PCR and immunofluorescence. RESULTS: Immunofluorescence detected TP in the endothelial cell layer of MCA. Vasoconstriction to the TP agonist, U46619 was reduced by Rho kinase inhibition. TP receptor stimulation lead to loss of K(Ca)2.3 mediated hyperpolarization, an effect that was reversed by Rho kinase inhibitors or simvastatin. K(Ca)2.3 activity was lost in L-NAME-treated arteries, but was restored by Rho kinase inhibition or statin treatment. The restorative effect of simvastatin was blocked after incubation with geranylgeranyl-pyrophosphate to circumvent loss of isoprenylation. CONCLUSIONS: Rho/Rho kinase signalling following TP stimulation and L-NAME regulates endothelial cell K(Ca)2.3 function. The ability of statins to prevent isoprenylation and perhaps inhibit of Rho restores/protects the input of K(Ca)2.3 to EDH in the MCA, and represents a beneficial pleiotropic effect of statin treatment.


Subject(s)
Cerebral Arteries/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Receptors, Thromboxane/metabolism , rho-Associated Kinases/metabolism , Animals , Fluorescent Antibody Technique , In Vitro Techniques , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Male , Membrane Potentials/drug effects , Oligopeptides/pharmacology , Rats , Rats, Wistar , Receptors, Thromboxane/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vasodilation/drug effects , rho-Associated Kinases/antagonists & inhibitors
12.
J Vasc Res ; 47(2): 93-107, 2010.
Article in English | MEDLINE | ID: mdl-19729956

ABSTRACT

BACKGROUND/AIMS: In cerebral arteries, nitric oxide (NO) release plays a key role in suppressing vasomotion. Our aim was to establish the pathways affected by NO in rat middle cerebral arteries. METHODS: In isolated segments of artery, isometric tension and simultaneous measurements of either smooth muscle membrane potential or intracellular [Ca(2+)] ([Ca(2+)](SMC)) changes were recorded. RESULTS: In the absence of L-NAME, asynchronous propagating Ca(2+) waves were recorded that were sensitive to block with ryanodine, but not nifedipine. L-NAME stimulated pronounced vasomotion and synchronous Ca(2+) oscillations with close temporal coupling between membrane potential, tone and [Ca(2+)](SMC). If nifedipine was applied together with L-NAME, [Ca(2+)](SMC) decreased and synchronous Ca(2+) oscillations were lost, but asynchronous propagating Ca(2+) waves persisted. Vasomotion was similarly evoked by either iberiotoxin, or by ryanodine, and to a lesser extent by ODQ. Exogenous application of NONOate stimulated endothelium-independent hyperpolarization and relaxation of either L-NAME-induced or spontaneous arterial tone. NO-evoked hyperpolarization involved activation of BK(Ca) channels via ryanodine receptors (RYRs), with little involvement of sGC. Further, in whole cell mode, NO inhibited current through L-type voltage-gated Ca(2+) channels (VGCC), which was independent of both voltage and sGC. CONCLUSION: NO exerts sGC-independent actions at RYRs and at VGCC, both of which normally suppress cerebral artery myogenic tone.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling , Guanylate Cyclase/metabolism , Muscle, Smooth, Vascular/enzymology , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Vasoconstriction , Vasodilation , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , In Vitro Techniques , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Membrane Potentials , Middle Cerebral Artery/enzymology , Muscle, Smooth, Vascular/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Potassium Channel Blockers/pharmacology , Rats , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Ryanodine Receptor Calcium Release Channel/drug effects , Soluble Guanylyl Cyclase , Time Factors , Vasoconstriction/drug effects , Vasodilation/drug effects
13.
Stroke ; 37(5): 1277-82, 2006 May.
Article in English | MEDLINE | ID: mdl-16556879

ABSTRACT

BACKGROUND AND PURPOSE: Endothelium-derived hyperpolarizing factor responses in the rat middle cerebral artery are blocked by inhibiting IKCa channels alone, contrasting with peripheral vessels where block of both IKCa and SKCa is required. As the contribution of IKCa and SKCa to endothelium-dependent hyperpolarization differs in peripheral arteries, depending on the level of arterial constriction, we investigated the possibility that SKCa might contribute to equivalent hyperpolarization in cerebral arteries under certain conditions. METHODS: Rat middle cerebral arteries (approximately 175 microm) were mounted in a wire myograph. The effect of KCa channel blockers on endothelium-dependent responses to the protease-activated receptor 2 agonist, SLIGRL (20 micromol/L), were then assessed as simultaneous changes in tension and membrane potential. These data were correlated with the distribution of arterial KCa channels revealed with immunohistochemistry. RESULTS: SLIGRL hyperpolarized and relaxed cerebral arteries undergoing variable levels of stretch-induced tone. The relaxation was unaffected by specific inhibitors of IKCa (TRAM-34, 1 micromol/L) or SKCa (apamin, 50 nmol/L) alone or in combination. In contrast, the associated smooth-muscle hyperpolarization was inhibited, but only with these blockers in combination. Blocking nitric oxide synthase (NOS) or guanylyl cyclase evoked smooth-muscle depolarization and constriction, with both hyperpolarization and relaxation to SLIGRL being abolished by TRAM-34 alone, whereas apamin had no effect. Immunolabeling showed SKCa and IKCa within the endothelium. CONCLUSIONS: In the absence of NO, IKCa underpins endothelium-dependent hyperpolarization and relaxation in cerebral arteries. However, when NOS is active SKCa contributes to hyperpolarization, whatever the extent of background contraction. These changes may have relevance in vascular disease states where NO release is compromised and when the levels of SKCa expression may be altered.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Middle Cerebral Artery/physiology , Small-Conductance Calcium-Activated Potassium Channels/physiology , Animals , Apamin/pharmacology , In Vitro Techniques , Male , Membrane Potentials/drug effects , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Oligopeptides/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptor, PAR-2/agonists , Receptors, Thrombin/agonists
14.
Stroke ; 36(7): 1526-32, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15933259

ABSTRACT

BACKGROUND AND PURPOSE: Endothelium-derived hyperpolarizing factor (EDHF) and K+ are vasodilators in the cerebral circulation. Recently, K+ has been suggested to contribute to EDHF-mediated responses in peripheral vessels. The EDHF response to the protease-activated receptor 2 ligand SLIGRL was characterized in cerebral arteries and used to assess whether K+ contributes as an EDHF. METHODS: Rat middle cerebral arteries were mounted in either a wire or pressure myograph. Concentration-response curves to SLIGRL and K+ were constructed in the presence and absence of a variety of blocking agents. In some experiments, changes in tension and smooth muscle cell membrane potential were recorded simultaneously. RESULTS: SLIGRL (0.02 to 20 micromol/L) stimulated concentration and endothelium-dependent relaxation. In the presence of NG-nitro-L-arginine methyl ester, relaxation to SLIGRL was associated with hyperpolarization and sensitivity to a specific inhibitor of IKCa, 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole (1 micromol/L), reflecting activation of EDHF. Combined inhibition of KIR with Ba2+ (30 micromol/L) and Na+/K+-ATPase with ouabain (1 micromol/L) markedly attenuated the relaxation to EDHF. Raising extracellular [K+] to 15 mmol/L also stimulated smooth muscle relaxation and hyperpolarization, which was also attenuated by combined application of Ba2+ and ouabain. CONCLUSIONS: SLIGRL evokes EDHF-mediated relaxation in the rat middle cerebral artery, underpinned by hyperpolarization of the smooth muscle. The profile of blockade of EDHF-mediated hyperpolarization and relaxation supports a pivotal role for IKCa channels. Furthermore, similar inhibition of responses to EDHF and exogenous K+ with Ba2+ and ouabain suggests that K+ may contribute as an EDHF in the middle cerebral artery.


Subject(s)
Biological Factors/physiology , Middle Cerebral Artery/pathology , Potassium/physiology , Animals , Barium/pharmacology , Biological Factors/metabolism , Bradykinin/pharmacology , Cerebrovascular Circulation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Inflammation , Male , Middle Cerebral Artery/metabolism , Myography/methods , NG-Nitroarginine Methyl Ester/pharmacology , Oligopeptides/metabolism , Ouabain/pharmacology , Potassium/chemistry , Potassium Channels/metabolism , Pressure , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptor, PAR-2/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
15.
Br J Pharmacol ; 138(6): 1172-80, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12684274

ABSTRACT

1. The ability of ascorbate to inhibit endothelium-derived hyperpolarizing factor (EDHF)-mediated vasodilatation was compared in the bovine perfused ciliary vascular bed and isolated rings of coronary artery. 2. Acetylcholine-induced, EDHF-mediated vasodilatation of the ciliary circulation was blocked following inclusion of ascorbate (50 micro M, 120 min) in the perfusion fluid. The blockade was highly selective since ascorbate had no effect on the vasodilator actions of the K(ATP) channel opener, levcromakalim, nor on the tonic vasodepressor action of basally released nitric oxide. 3. The possibility that concentration of ascorbate by the ciliary body was a prerequisite for blockade to occur was ruled out, since EDHF was still blocked when the anterior and posterior chambers were continuously flushed with Krebs solution or when both the aqueous and vitreous humour were drained. 4. Ascorbate at 50 micro M failed to affect bradykinin- or acetylcholine-induced, EDHF-mediated vasodilatation in rings of bovine coronary artery. Raising the concentration to 3 mM did produce blockade of EDHF, but this was nonselective, since vasodilator responses to endothelium-derived nitric oxide were also inhibited. 5. Thus, ascorbate (50 micro M) is not a universal blocker of EDHF. Whether its ability to block in the bovine ciliary circulation, but not in the coronary artery, is due to differences in the nature of EDHF at the two sites, differences in vessel size (resistance arterioles versus conduit artery), the presence or absence of flow, or to some other factor remains to be determined.


Subject(s)
Acetylcholine/pharmacology , Ascorbate Oxidase/pharmacology , Biological Factors/antagonists & inhibitors , Bradykinin/pharmacology , Ciliary Body/drug effects , Cromakalim/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Vasodilation/physiology , Animals , Aqueous Humor/chemistry , Aqueous Humor/drug effects , Biological Factors/physiology , Cattle , Ciliary Arteries/drug effects , Ciliary Body/blood supply , Collateral Circulation/drug effects , Collateral Circulation/physiology , Coronary Circulation/drug effects , Dose-Response Relationship, Drug , Potassium Channels, Calcium-Activated , Vasodilation/drug effects
16.
Br J Pharmacol ; 135(7): 1801-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11934822

ABSTRACT

1. The effects of ascorbate were assessed on vasodilatation mediated by endothelium-derived hyperpolarizing factor (EDHF) in the ciliary vascular bed of the bovine isolated perfused eye and in the rat isolated perfused mesenteric arterial bed. 2. In the bovine eye, EDHF-mediated vasodilator responses induced by acetylcholine or bradykinin were powerfully blocked when ascorbate (50 microM) was included in the perfusion medium for at least 120 min; with acetylcholine a normally-masked muscarinic vasoconstrictor response was also uncovered. 3. The blockade of EDHF-mediated vasodilatation by ascorbate was time-dependent (maximum blockade at 120 min) and concentration-dependent (10 - 150 microM). 4. Ascorbate (50 microM) also blocked acetylcholine-induced, EDHF-mediated vasodilator responses in the rat mesenteric arterial bed in a time-dependent manner (maximum blockade at 180 min). 5. The ability of ascorbate to block EDHF-mediated vasodilatation is likely to result from its reducing properties, since this action was mimicked in the bovine eye by two other reducing agents, namely, N-acetyl-L-cysteine (1 mM) and dithiothreitol (100 microM), but not by the redox-inactive analogue, dehydroascorbate (50 microM). 6. In conclusion, concentrations of ascorbate present in normal plasma block EDHF-mediated vasodilator responses in the bovine eye and rat mesentery. The mechanism and physiological consequences of this blockade remain to be determined.


Subject(s)
Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Biological Factors/physiology , Ciliary Body/drug effects , Mesenteric Arteries/drug effects , Vasodilation/drug effects , Acetylcholine , Animals , Bradykinin/pharmacology , Cattle , Ciliary Body/physiology , Mesenteric Arteries/physiology , Perfusion , Rats , Time Factors , Vasoconstriction
SELECTION OF CITATIONS
SEARCH DETAIL
...