Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 169(7): 1500-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23488724

ABSTRACT

BACKGROUND AND PURPOSE: Perivascular adipose tissue (PVAT) releases adipocyte-derived hyperpolarizing factors (ADHFs) that may partly act by opening myocyte K(+) channels. The present study in rat and mouse mesenteric arteries aimed to identify the myocyte K(+) channel activated by PVAT and to determine whether adiponectin contributed to the hyperpolarizing effects of PVAT. EXPERIMENTAL APPROACH: Myocyte membrane potential was recorded from de-endothelialized, non-contracted rat and mouse mesenteric arteries in the presence and absence of PVAT. KEY RESULTS: The ß3 -adrenoceptor agonist, CL-316,243 (10 µM), generated PVAT-dependent, iberiotoxin-sensitive myocyte hyperpolarizations resulting from BKCa channel opening and which were partially blocked by L-NMMA (100 µM). Adiponectin (5 µg·mL(-1) ) also produced iberiotoxin-sensitive hyperpolarizations in PVAT-denuded arterioles. Activation of myocyte AMP-activated protein kinase (AMPK) using 5 µM A-769662 also induced BKCa -mediated hyperpolarizations. Dorsomorphin abolished hyperpolarizations to CL-316,243, adiponectin and A-769662. In vessels from Adipo(-/-) mice, hyperpolarizations to CL-316,243 were absent whereas those to A-769662 and adiponectin were normal. In rat vessels, adipocyte-dependent hyperpolarizations were blocked by glibenclamide and clotrimazole but those to NS1619 (33 µM) were unaltered. CONCLUSIONS AND IMPLICATIONS: Under basal, non-contracted conditions, ß3 -adrenoceptor stimulation of PVAT releases an ADHF, which is probably adiponectin. This activates AMPK to open myocyte BKCa channels indirectly and additionally liberates NO, which also contributes to the observed PVAT-dependent myocyte hyperpolarizations. Clotrimazole and glibenclamide each reversed hyperpolarizations to adiponectin and A-769662, suggesting the involvement of myocyte TRPM4 channels in the ADHF-induced myocyte electrical changes mediated via the opening of BKCa channels.


Subject(s)
Adiponectin/metabolism , Adrenergic beta-3 Receptor Agonists/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Mesenteric Arteries/physiology , Muscle Cells/physiology , Pyrones/pharmacology , TRPM Cation Channels/metabolism , Thiophenes/pharmacology , Animals , Biphenyl Compounds , Dioxoles/pharmacology , Gene Expression Regulation , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-3/metabolism
3.
Br J Pharmacol ; 164(3): 922-33, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21506954

ABSTRACT

BACKGROUND AND PURPOSE: Controlling vascular tone involves K(+) efflux through endothelial cell small- and intermediate-conductance calcium-activated potassium channels (K(Ca)2.3 and K(Ca)3.1, respectively). We investigated the expression of these channels in astrocytes and the possibility that, by a similar mechanism, they might contribute to neurovascular coupling. EXPERIMENTAL APPROACH: Transgenic mice expressing enhanced green fluorescent protein (eGFP) in astrocytes were used to assess K(Ca)2.3 and K(Ca)3.1 expression by immunohistochemistry and RT-PCR. K(Ca) currents in eGFP-positive astrocytes were determined in situ using whole-cell patch clamp electrophysiology. The contribution of K(Ca)3.1 to neurovascular coupling was investigated in pharmacological experiments using electrical field stimulation (EFS) to evoke parenchymal arteriole dilatation in FVB/NJ mouse brain slices and whisker stimulation to evoke changes in cerebral blood flow in vivo, measured by laser Doppler flowmetry. KEY RESULTS: K(Ca)3.1 immunoreactivity was restricted to astrocyte processes and endfeet and RT-PCR confirmed astrocytic K(Ca)2.3 and K(Ca)3.1 mRNA expression. With 200 nM [Ca(2+)](i) , the K(Ca)2.1-2.3/K(Ca)3.1 opener NS309 increased whole-cell currents. CyPPA, a K(Ca)2.2/K(Ca)2.3 opener, was without effect. With 1 µM [Ca(2+)](i) , the K(Ca)3.1 inhibitor TRAM-34 reduced currents whereas apamin (K(Ca)2.1-2.3 blocker) had no effect. CyPPA also inhibited currents evoked by NS309 in HEK293 cells expressing K(Ca)3.1. EFS-evoked Fluo-4 fluorescence confirmed astrocyte endfoot recruitment into neurovascular coupling. TRAM-34 inhibited EFS-evoked arteriolar dilatation by 50% whereas charybdotoxin, a blocker of K(Ca)3.1 and the large-conductance K(Ca) channel, K(Ca)1.1, inhibited dilatation by 82%. TRAM-34 reduced the cortical hyperaemic response to whisker stimulation by 40%. CONCLUSION AND IMPLICATIONS: Astrocytes express functional K(Ca)3.1 channels, and these contribute to neurovascular coupling.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Arterioles/drug effects , Arterioles/metabolism , Astrocytes/drug effects , Brain/drug effects , Calcium/metabolism , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Female , HEK293 Cells , Humans , Immunohistochemistry/methods , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/genetics , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Vasodilation/drug effects
4.
Acta Physiol (Oxf) ; 203(1): 127-37, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21199402

ABSTRACT

The extracellular calcium-sensing receptor (CaR) was first described in the parathyroid gland. Recent studies have shown that the CaR is also expressed in blood vessels, especially in the endothelial and adventitial layers but its physiological function is still not clear. However, an understanding of its possible role(s) in the vasculature (perivascular-neurones, heart and blood vessels) is important because of the use of synthetic positive allosteric CaR modulators in hyperparathyroidism and the potential importance of negative modulators in the treatment of osteoporosis. In this review, the effects of CaR activation and inhibition are detailed and the possible role of the CaR as both an amplifier and attenuator of myo-endothelial coupling in the vasculature is described.


Subject(s)
Cardiovascular System/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Humans
5.
Br J Pharmacol ; 160(4): 836-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20233221

ABSTRACT

BACKGROUND AND PURPOSE: Previous studies have shown that endothelium-dependent hyperpolarization of myocytes is reduced in resistance arteries from spontaneously hypertensive rats (SHRs). The aim of the present study was to determine whether this reflects down-regulation of endothelial K(+) channels or their associated pathways. EXPERIMENTAL APPROACH: Changes in vascular K(+) channel responses and expression were determined by a combination of membrane potential recordings and Western blotting. KEY RESULTS: Endothelium-dependent myocyte hyperpolarizations induced by acetylcholine, 6,7-dichloro-1H-indole-2,3-dione 3-oxime (NS309) (opens small- and intermediate-conductance calcium-sensitive K(+) channels, SK(Ca) and IK(Ca), respectively) or cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine (SK(Ca) opener) were reduced in mesenteric arteries from SHRs. After blocking SK(Ca) channels with apamin, hyperpolarizations to acetylcholine and NS309 in SHR arteries were similar to those of controls. Hyperpolarization to 5 mM KCl was reduced in SHR arteries due to loss of the Ba(2+)-sensitive, inward-rectifier channel (K(IR)) component; the contribution of ouabain-sensitive, Na(+)/K(+)-ATPases was unaffected. Protein expression of both SK(Ca) and K(IR) channels was reduced in SHR arteries; the caveolin-1 monomer/dimer ratio was increased. CONCLUSIONS AND IMPLICATIONS: In SHRs, the distinct pathway that generates endothelium-dependent hyperpolarization in vascular myocyte by activation of IK(Ca) channels and Na(+)/K(+)-ATPases remains intact. The second pathway, initiated by endothelial SK(Ca) channel activation and amplified by K(IR) opening on both endothelial cells and myocytes is compromised in SHRs due to down-regulation of both SK(Ca) and K(IR) and to changes in caveolin-1 oligomers. These impairments in the SK(Ca)-K(IR) pathway shed new light on vascular control mechanisms and on the underlying vascular changes in hypertension.


Subject(s)
Down-Regulation/physiology , Hypertension/physiopathology , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Signal Transduction/physiology , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Blotting, Western , Caveolin 1/chemistry , Caveolin 1/metabolism , Dimerization , Hypertension/metabolism , In Vitro Techniques , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Membrane Potentials/drug effects , Membrane Transport Modulators/pharmacology , Mesenteric Arteries/drug effects , Muscle, Smooth, Vascular/drug effects , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism , Protein Subunits/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction/drug effects , Small-Conductance Calcium-Activated Potassium Channels/agonists , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism
6.
Br J Pharmacol ; 154(3): 652-62, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18414396

ABSTRACT

BACKGROUND AND PURPOSE: The extracellular calcium-sensing receptor (CaR) in vascular endothelial cells activates endothelial intermediate-conductance, calcium-sensitive K(+) channels (IK(Ca)) indirectly leading to myocyte hyperpolarization. We determined whether CaR expression and function was modified in a rat model of type II diabetes. EXPERIMENTAL APPROACH: Pressure myography, western blotting, sharp microelectrode and K(+)-selective electrode recordings were used to investigate the functional expression of the CaR and IK(Ca) in rat mesenteric arteries. KEY RESULTS: Myocyte hyperpolarization to the CaR activator calindol was inhibited by Calhex 231. U46619-induced vessel contraction elevated the extracellular [K(+)] around the myocytes, and inhibition of this 'K(+) cloud' by iberiotoxin was needed to reveal calindol-induced vasodilatations. These were antagonized by Calhex 231 and significantly smaller in Zucker diabetic fatty rat (ZDF) vessels than in Zucker lean (ZL) controls. Myocyte hyperpolarizations to calindol were also smaller in ZDF than in ZL arteries. In ZDF vessels, endothelial cell CaR protein expression was reduced; IK(Ca) expression was also diminished, but IK(Ca)-generated hyperpolarizations mediated by 1-EBIO were unaffected. CONCLUSIONS AND IMPLICATIONS: The reduced CaR-mediated hyperpolarizing and vasodilator responses in ZDF arteries result from a decrease in CaR expression, rather than from a modification of IK(Ca) channels. Detection of CaR-mediated vasodilatation required the presence of iberiotoxin, suggesting a CaR contribution to vascular diameter, that is, inversely related to the degree of vasoconstriction. Compromise of the CaR pathway would favour the long-term development of a higher basal vascular tone and could contribute to the vascular complications associated with type II diabetes.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Mesenteric Arteries/metabolism , Potassium Channels, Calcium-Activated/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Blotting, Western , Electrophysiology , Gene Expression Regulation , Male , Mesenteric Arteries/pathology , Microelectrodes , Muscle Cells/metabolism , Myography , Rats , Rats, Wistar , Rats, Zucker , Vasodilation/physiology
7.
Br J Pharmacol ; 151(3): 332-40, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17450174

ABSTRACT

BACKGROUND AND PURPOSE: The small and intermediate conductance, Ca2+-sensitive K+ channels (SK(Ca) and IK(Ca), respectively) which are pivotal in the EDHF pathway may be differentially activated. The importance of caveolae in the functioning of IK(Ca) and SK(Ca) channels was investigated. EXPERIMENTAL APPROACH: The effect of the caveolae-disrupting agent methyl-beta-cyclodextrin (MbetaCD) on IK(Ca) and SK(Ca) localization and function was determined. KEY RESULTS: EDHF-mediated, SK(Ca)-dependent myocyte hyperpolarizations evoked by acetylcholine in rat mesenteric arteries (following blockade of IK(Ca) with TRAM-34) were inhibited by MbetaCD. Hyperpolarizations evoked by direct SK(Ca) channel activation (using NS309 in the presence of TRAM-34) were also inhibited by MbetaCD, an effect reversed by cholesterol. In contrast, IK(Ca)-dependent hyperpolarizations (in the presence of apamin) were unaffected by MbetaCD. Similarly, in porcine coronary arteries, EDHF-mediated, SK(Ca)-dependent (but not IK(Ca)-dependent) endothelial cell hyperpolarizations evoked by substance P were inhibited by MbetaCD. In mesenteric artery homogenates subjected to sucrose-density centrifugation, caveolin-1 and SK3 (SK(Ca)) proteins but not IK1 (IK(Ca)) protein migrated to the buoyant, caveolin-rich fraction. MbetaCD pretreatment redistributed caveolin-1 and SK3 proteins into more dense fractions. In immunofluorescence images of porcine coronary artery endothelium, SK3 (but not IK1) and caveolin-1 were co-localized. Furthermore, caveolin-1 immunoprecipitates prepared from native porcine coronary artery endothelium contained SK3 but not IK1 protein. CONCLUSIONS AND IMPLICATIONS: These data provide strong evidence that endothelial cell SK(Ca) channels are located in caveolae while the IK(Ca) channels reside in a different membrane compartment. These studies reveal cellular organisation as a further complexity in the EDHF pathway signalling cascade.


Subject(s)
Arteries/drug effects , Biological Factors/physiology , Caveolae/physiology , Potassium Channels, Calcium-Activated/physiology , beta-Cyclodextrins/pharmacology , Acetylcholine/pharmacology , Animals , Arteries/cytology , Arteries/physiology , Blotting, Western , Caveolae/metabolism , Caveolins/metabolism , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/physiology , Dose-Response Relationship, Drug , Endothelium, Vascular/physiology , In Vitro Techniques , Indoles/pharmacology , Male , Membrane Potentials/drug effects , Mesenteric Arteries/cytology , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Oximes/pharmacology , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Potassium Channels, Calcium-Activated/metabolism , Pyrazoles/pharmacology , Rats , Rats, Wistar , Swine , Vasodilator Agents/pharmacology
8.
Br J Pharmacol ; 142(1): 192-202, 2004 May.
Article in English | MEDLINE | ID: mdl-15066906

ABSTRACT

1. Experiments were performed to elucidate the mechanism by which alterations of extracellular pH (pH(o)) change membrane potential (E(M)) in rat mesenteric and pulmonary arteries. 2. Changing pH(o) from 7.4 to 6.4 or 8.4 produced a depolarisation or hyperpolarisation, respectively, in mesenteric and pulmonary arteries. Anandamide (10 microm) or bupivacaine (100 microm) reversed the hyperpolarisation associated with alkaline pH(o), shifting the E(M) of both vessels to levels comparable to that at pH 6.4. In pulmonary arteries, clofilium (100 microm) caused a significant reversal of hyperpolarisation seen at pH 8.4 but was without effect at pH 7.4. 3. K(+) channel blockade by 4-aminopyridine (4-AP) (5 mm), tetraethylammonium (TEA) (10 mm), Ba(2+) (30 microm) and glibenclamide (10 microm) depolarised the pulmonary artery. However, shifts in E(M) with changes in pH(o) remained and were sensitive to anandamide (10 microm), bupivacaine (100 microm) or Zn(2+) (200 microm). 4. Anandamide (0.3-60 microm) or bupivacaine (0.3-300 microm) caused a concentration-dependent increase in basal tone in pulmonary arteries. 5. RT-PCR demonstrated the expression of TASK-1, TASK-2, THIK-1, TRAAK, TREK-1, TWIK-1 and TWIK-2 in mesenteric arteries and TASK-1, TASK-2, THIK-1, TREK-2 and TWIK-2 in pulmonary arteries. TASK-1, TASK-2, TREK-1 and TWIK-2 protein was demonstrated in both arteries by immunostaining. 6. These experiments provide evidence for the presence of two-pore domain K(+) channels in rat mesenteric and pulmonary arteries. Collectively, they strongly suggest that modulation of TASK-1 channels is most likely to have mediated the pH-induced changes in membrane potential observed in these vessels, and that blockade of these channels by anandamide or bupivacaine generates a small increase in pulmonary artery tone.


Subject(s)
Mesenteric Arteries/physiology , Potassium Channels, Tandem Pore Domain/physiology , Pulmonary Artery/physiology , Animals , Arachidonic Acids/pharmacology , Bupivacaine/pharmacology , Dose-Response Relationship, Drug , Endocannabinoids , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mesenteric Arteries/drug effects , Polyunsaturated Alkamides , Potassium Channels/physiology , Pulmonary Artery/drug effects , Rats , Rats, Sprague-Dawley
9.
Br J Pharmacol ; 136(6): 918-26, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12110616

ABSTRACT

1. Mechanisms underlying K(+)-induced hyperpolarizations in the presence and absence of phenylephrine were investigated in endothelium-denuded rat mesenteric arteries (for all mean values, n=4). 2. Myocyte resting membrane potential (m.p.) was -58.8+/-0.8 mV. Application of 5 mM KCl produced similar hyperpolarizations in the absence (17.6+/-0.7 mV) or presence (15.8+/-1.0 mV) of 500 nM ouabain. In the presence of ouabain +30 microM barium, hyperpolarization to 5 mM KCl was essentially abolished. 3. In the presence of 10 microM phenylephrine (m.p. -33.7+/-3 mV), repolarization to 5 mM KCl did not occur in the presence or absence of 4-aminopyridine but was restored (-26.9+/-1.8 mV) on addition of iberiotoxin (100 nM). Under these conditions the K+-induced repolarization was insensitive to barium (30 microM) but abolished by 500 nM ouabain alone. 4. In the presence of phenylephrine + iberiotoxin the hyperpolarization to 5 mM K(+) was inhibited in the additional presence of 300 nM levcromakalim, an action which was reversed by 10 microM glibenclamide. 5. RT-PCR, Western blotting and immunohistochemical techniques collectively showed the presence of alpha(1)-, alpha(2)- and alpha(3)-subunits of Na(+)/K(+)-ATPase in the myocytes. 6. In K(+)-free solution, re-introduction of K(+) (to 4.6 mM) hyperpolarized myocytes by 20.9+/-0.5 mV, an effect unchanged by 500 nM ouabain but abolished by 500 microM ouabain. 7. We conclude that under basal conditions, Na(+)/K(+)-ATPases containing alpha(2)- and/or alpha(3)-subunits are partially responsible for the observed K(+)-induced effects. The opening of myocyte K(+) channels (by levcromakalim or phenylephrine) creates a 'K(+) cloud' around the cells which fully activates Na(+)/K(+)-ATPase and thereby abolishes further responses to [K(+)](o) elevation.


Subject(s)
Mesenteric Arteries/physiology , Muscle, Smooth, Vascular/physiology , Potassium/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Blotting, Western , Endothelium, Vascular/physiology , Fluorescent Antibody Technique , In Vitro Techniques , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/enzymology , Microelectrodes , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Phenylephrine/pharmacology , Protein Isoforms , Protein Subunits , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Vasoconstrictor Agents/pharmacology
10.
Br J Pharmacol ; 135(5): 1133-43, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11877319

ABSTRACT

1. The apamin-sensitive small-conductance Ca(2+)-activated K(+) channel (SK(Ca)) was characterized in porcine coronary arteries. 2. In intact arteries, 100 nM substance P and 600 microM 1-ethyl-2-benzimidazolinone (1-EBIO) produced endothelial cell hyperpolarizations (27.8 +/- 0.8 mV and 24.1 +/- 1.0 mV, respectively). Charybdotoxin (100 nM) abolished the 1-EBIO response but substance P continued to induce a hyperpolarization (25.8 +/- 0.3 mV). 3. In freshly-isolated endothelial cells, outside-out patch recordings revealed a unitary K(+) conductance of 6.8 +/- 0.04 pS. The open-probability was increased by Ca(2+) and reduced by apamin (100 nM). Substance P activated an outward current under whole-cell perforated-patch conditions and a component of this current (38%) was inhibited by apamin. A second conductance of 2.7 +/- 0.03 pS inhibited by d-tubocurarine was observed infrequently. 4. Messenger RNA encoding the SK2 and SK3, but not the SK1, subunits of SK(Ca) was detected by RT - PCR in samples of endothelium. Western blotting indicated that SK3 protein was abundant in samples of endothelium compared to whole arteries. SK2 protein was present in whole artery nuclear fractions. 5. Immunofluorescent labelling confirmed that SK3 was highly expressed at the plasmalemma of endothelial cells and was not expressed in smooth muscle. SK2 was restricted to the peri-nuclear regions of both endothelial and smooth muscle cells. 6. In conclusion, the porcine coronary artery endothelium expresses an apamin-sensitive SK(Ca) containing the SK3 subunit. These channels are likely to confer all or part of the apamin-sensitive component of the endothelium-derived hyperpolarizing factor (EDHF) response.


Subject(s)
Apamin/pharmacology , Biological Factors/physiology , Endothelium, Vascular/physiology , Muscle, Smooth, Vascular/physiology , Potassium Channels, Calcium-Activated , Potassium Channels/physiology , Amino Acid Sequence , Animals , Coronary Vessels/drug effects , Coronary Vessels/physiology , DNA, Complementary/analysis , Endothelium, Vascular/drug effects , Female , Male , Membrane Potentials/drug effects , Microelectrodes , Molecular Sequence Data , Muscle, Smooth, Vascular/drug effects , Patch-Clamp Techniques , Potassium Channels/drug effects , Potassium Channels/genetics , Reverse Transcriptase Polymerase Chain Reaction , Small-Conductance Calcium-Activated Potassium Channels , Substance P/pharmacology , Swine
11.
Br J Pharmacol ; 133(7): 1145-53, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11487526

ABSTRACT

In porcine coronary arteries, smooth muscle hyperpolarizations produced by the nitric oxide donor, NOR-1, and the prostacyclin analogue, iloprost, were compared with those induced by substance P and bradykinin and attributed to the endothelium-derived hyperpolarizing factor (EDHF). In the presence of 300 microM L-nitroarginine and 10 microM indomethacin, iloprost-induced hyperpolarizations were partially inhibited by 10 microM glibenclamide whereas those to NOR-1, substance P and bradykinin were unaffected. Hyperpolarizations produced by maximally-effective concentrations of NOR-1 and NS1619 were identical (to -65 mV). They were significantly less than those generated by either substance P or bradykinin (to approximately -80 mV) and were abolished by iberiotoxin 100 nM, a concentration which had essentially no effect on responses to substance P or bradykinin. Incubation of segments of intact arteries for 16 - 22 h in bicarbonate-buffered Krebs solution had little effect on EDHF responses to substance P or bradykinin. In contrast, after incubation for this period of time in HEPES-buffered Tyrode solution or Krebs containing 10 mM HEPES the EDHF response to substance P was abolished and that to bradykinin was markedly reduced. The residual bradykinin-induced hyperpolarization following incubation in Tyrode solution was inhibited by iberiotoxin and by 10 microM 17-octadecynoic acid. We conclude that substance P activates only the EDHF pathway in the presence of nitric oxide synthase and cyclo-oxygenase inhibitors. Incubation in HEPES-buffered Tyrode solution abolishes the EDHF responses to substance P and bradykinin to reveal an additional hyperpolarizing mechanism, associated with the opening of K(+) channels, activated only by bradykinin.


Subject(s)
Bradykinin/pharmacology , Coronary Vessels/drug effects , Endothelium, Vascular/physiology , Membrane Potentials/drug effects , Substance P/pharmacology , Animals , Bicarbonates/pharmacology , Biological Factors/physiology , Buffers , Coronary Vessels/physiology , Cromakalim/pharmacology , Epoprostenol/pharmacology , Glyburide/pharmacology , HEPES/pharmacology , In Vitro Techniques , Indomethacin/pharmacology , Isotonic Solutions/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Nitric Oxide Donors/pharmacology , Nitroarginine/pharmacology , Peptides/pharmacology , Sodium Chloride/pharmacology , Swine , Time Factors , Vasodilator Agents/pharmacology
12.
Br J Pharmacol ; 134(1): 1-5, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11522590

ABSTRACT

In intact mesenteric arteries, increasing [K(+)]o by 5 mM hyperpolarized both endothelial and smooth muscle cells. Subsequent exposure to 10 microM phenylephrine depolarized both cell types which were then repolarized by a 5 mM increase in [K(+)]o. In endothelium-denuded vessels, increasing [K(+)]o by 5 mM hyperpolarized the smooth muscle but K(+) had no effect after depolarization by 10 microM phenylephrine. On subsequent exposure to iberiotoxin plus 4-aminopyridine, the repolarizing action of 5 mM K(+) was restored. In endothelium-intact vessels exposed to phenylephrine, pretreatment with a gap junction inhibitor (gap 27) reduced K(+)-mediated smooth muscle repolarization without affecting the endothelial cell response. It is concluded that phenylephrine-induced efflux of K(+) via smooth muscle K(+) channels produces a local increase in [K(+)]o which impairs repolarization to added K(+). Thus, studies involving vessels precontracted with agonists which increase [K(+)]o maximize the role of gap junctions and minimize any contribution to the EDHF pathway from endothelium-derived K(+).


Subject(s)
Membrane Potentials/drug effects , Mesenteric Arteries/drug effects , Phenylephrine/pharmacology , Potassium/pharmacology , Vasoconstrictor Agents/pharmacology , 4-Aminopyridine/pharmacology , Animals , Connexins/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , In Vitro Techniques , Male , Mesenteric Arteries/cytology , Mesenteric Arteries/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Peptides/pharmacology , Rats , Rats, Sprague-Dawley
13.
Eur J Pharmacol ; 426(1-2): 89-94, 2001 Aug 24.
Article in English | MEDLINE | ID: mdl-11525776

ABSTRACT

The effects on membrane potential of prostacyclin and iloprost were compared in smooth muscle cells of the guinea pig carotid artery. Both prostacyclin and iloprost induced hyperpolarization of the smooth muscle cells. In the presence of (3R)-3-(4-fluorophenyl-sulfonamido)-1,2,3,4-tetrahydro-9-carbazolepropanoic acid (Bay U3405), an antagonist of TP receptors, the response to iloprost was unaffected while that to prostacyclin was increased. Iloprost-induced hyperpolarizations were abolished by glibenclamide while those to prostacyclin were either not affected, or converted to either depolarization or to rhythmic electrical activity. The latter effects of prostacyclin were abolished by Bay U3405. After removal of the endothelium, iloprost and prostacyclin produced hyperpolarizations similar to those observed in control blood vessels. However, in the presence of glibenclamide, prostacyclin produced only depolarizations inhibited by Bay U3405. These results suggest that iloprost activates IP receptors and K(ATP) channels in smooth muscle. In contrast, prostacyclin produces additional endothelium-dependent and -independent effects via activation of TP receptors.


Subject(s)
Carotid Arteries/drug effects , Epoprostenol/pharmacology , Iloprost/pharmacology , Animals , Carotid Arteries/cytology , Carotid Arteries/physiology , Dose-Response Relationship, Drug , Endothelium, Vascular/physiology , Glyburide/pharmacology , Guinea Pigs , In Vitro Techniques , Male , Membrane Potentials/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology
15.
Eur J Pharmacol ; 399(1): 9-16, 2000 Jun 30.
Article in English | MEDLINE | ID: mdl-10876017

ABSTRACT

Experiments were designed to determine the subtype of K(+) channels activated by the nitrovasodilator 3-morpholinosydnonimine (SIN-1) in smooth muscle cells of the rabbit and guinea pig carotid arteries. Membrane potential was recorded in isolated segments with intracellular microelectrode and K(+) currents in freshly dissociated smooth muscle cells, with the patch-clamp technique. In the guinea pig carotid artery, SIN-1 caused a glibenclamide-sensitive hyperpolarization. The nitrovasodilator did not affect the whole-cell K(+) current, but activated a glibenclamide-sensitive K(+) current. In the rabbit carotid artery, SIN-1 induced only an iberiotoxin-sensitive repolarization in phenylephrine-depolarized tissue and in isolated cells, enhanced the activity of an iberiotoxin-sensitive K(+) current. These findings demonstrate that the population of K(+) channels activated by nitric oxide (NO) is species-dependent and support the conclusion that, in the guinea pig carotid artery, in contrast to the rabbit carotid artery, the release of NO cannot account for the responses attributed to endothelium-derived hyperpolarizing factor (EDHF).


Subject(s)
Molsidomine/analogs & derivatives , Nitric Oxide Donors/pharmacology , Potassium Channels/drug effects , Animals , Calcium/pharmacology , Guinea Pigs , In Vitro Techniques , Male , Microelectrodes , Molsidomine/pharmacology , Nitric Oxide/physiology , Patch-Clamp Techniques , Rabbits , Species Specificity
16.
Br J Pharmacol ; 129(6): 1103-12, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10725258

ABSTRACT

1. Experiments were performed to identify the potassium channels involved in the acetylcholine-induced endothelium-dependent hyperpolarization of the guinea-pig internal carotid artery. Smooth muscle and endothelial cell membrane potentials were recorded in isolated arteries with intracellular microelectrodes. Potassium currents were recorded in freshly-dissociated smooth muscle cells using patch clamp techniques. 2. In single myocytes, iberiotoxin (0.1 microM)-, charybdotoxin (0.1 microM)-, apamin (0.5 microM)- and 4-aminopyridine (5 mM)-sensitive potassium currents were identified indicating the presence of large- and small-conductance calcium-sensitive potassium channels (BK(Ca) and SK(Ca)) as well as voltage-dependent potassium channels (K(V)). Charybdotoxin and iberiotoxin inhibited the same population of BK(Ca) but a conductance specifically sensitive to the combination of charybdotoxin plus apamin could not be detected. 4-aminopyridine (0. 1 - 25 mM) induced a concentration-dependent inhibition of K(V) without affecting the iberiotoxin- or the apamin-sensitive currents. 3. In isolated arteries, both the endothelium-dependent hyperpolarization of smooth muscle and the hyperpolarization of endothelial cells induced by acetylcholine or by substance P were inhibited by 5 mM 4-aminopyridine. 4. These results indicate that in the vascular smooth muscle cells of the guinea-pig carotid artery, a conductance specifically sensitive to the combination of charybdotoxin plus apamin could not be detected, comforting the hypothesis that the combination of these two toxins should act on the endothelial cells. Furthermore, the inhibition by 4-aminopyridine of both smooth muscle and endothelial hyperpolarizations, suggests that in order to observe an endothelium-dependent hyperpolarization of the vascular smooth muscle cells, the activation of endothelial potassium channels is likely to be required.


Subject(s)
Biological Factors/pharmacology , Carotid Arteries/drug effects , Endothelium, Vascular/physiology , 4-Aminopyridine/pharmacology , Acetylcholine/pharmacology , Animals , Biological Factors/antagonists & inhibitors , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/physiology , Electrophysiology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Guinea Pigs , In Vitro Techniques , Male , Membrane Potentials/physiology , Microelectrodes , Muscle, Smooth, Vascular/drug effects , Patch-Clamp Techniques , Peptides/pharmacology , Potassium Channel Blockers , Potassium Channels/drug effects , Substance P/pharmacology
17.
Br J Pharmacol ; 129(6): 1145-54, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10725263

ABSTRACT

1. The effects of endothelium-derived hyperpolarizing factor (EDHF: elicited using substance P or bradykinin) were compared with those of 11,12-EET in pig coronary artery. Smooth muscle cells were usually impaled with microelectrodes through the adventitial surface. 2. Substance P (100 nM) and 11,12-EET (11,12-epoxyeicosatrienoic acid; 3 microM) hyperpolarized endothelial cells in intact arteries. These actions were unaffected by 100 nM iberiotoxin but were abolished by charybdotoxin plus apamin (each 100 nM). 3. Substance P (100 nM) and bradykinin (30 nM) hyperpolarized intact artery smooth muscle; Substance P had no effect after endothelium removal. 11,12-EET hyperpolarized de-endothelialized vessels by 12.6+/-0.3 mV, an effect abolished by 100 nM iberiotoxin. 4. 11,12-EET hyperpolarized intact arteries by 18.6+/-0.8 mV, an action reduced by iberiotoxin, which was ineffective against substance P. Hyperpolarizations to 11, 12-EET and substance P were partially inhibited by 100 nM charybdotoxin and abolished by further addition of 100 nM apamin. 5. 30 microM barium plus 500 nM ouabain depolarized intact artery smooth muscle but responses to substance P and bradykinin were unchanged. 500 microM gap 27 markedly reduced hyperpolarizations to substance P and bradykinin which were abolished in the additional presence of barium plus ouabain. 6. Substance P-induced hyperpolarizations of smooth muscle cells immediately below the internal elastic lamina were unaffected by gap 27, even in the presence of barium plus ouabain. 7. In pig coronary artery, 11,12-EET is not EDHF. Smooth muscle hyperpolarizations attributed to 'EDHF' are initiated by endothelial cell hyperpolarization involving charybdotoxin- (but not iberiotoxin) and apamin-sensitive K(+) channels. This may spread electrotonically via myoendothelial gap junctions but the involvement of an unknown endothelial factor cannot be excluded.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Biological Factors/pharmacology , Coronary Vessels/drug effects , Endothelium, Vascular/physiology , Gap Junctions/drug effects , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Barium/pharmacology , Charybdotoxin/pharmacology , Electrophysiology , Endothelium, Vascular/drug effects , Female , In Vitro Techniques , Male , Membrane Potentials/drug effects , Microelectrodes , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/innervation , Ouabain/pharmacology , Peptides/pharmacology , Substance P/pharmacology , Swine
18.
Br J Pharmacol ; 128(8): 1788-94, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10588935

ABSTRACT

1. In guinea-pig internal carotid arteries with an intact endothelium, acetylcholine (10 microM) and levcromakalim (10 microM) each hyperpolarized the smooth muscle whereas a 5 mM elevation of extracellular K(+) was without effect. 2. Incubation of the carotid artery with the gap junction inhibitors carbenoxolone (100 microM) or gap 27 (500 microM) essentially abolished the hyperpolarization to acetylcholine but it was without effect on that to levcromakalim. Carbenoxolone had no effect on the acetylcholine-induced endothelial cell hyperpolarization but inhibited the smooth muscle hyperpolarization induced by the endothelial cell K(+) channel opener, 1-ethyl-2-benzimidazolinone (600 microM). 3. In rat hepatic and mesenteric arteries with endothelium, carbenoxolone (100 or 500 microM) depolarized the smooth muscle but did not modify hyperpolarizations induced by KCl or levcromakalim. In the mesenteric (but not the hepatic) artery, the acetylcholine-induced hyperpolarization was inhibited by carbenoxolone. 4. Phenylephrine (1 microM) depolarized the smooth muscle cells of intact hepatic and mesenteric arteries, an effect enhanced by carbenoxolone. Gap 27 did not have a depolarizing action. In the presence of phenylephrine, acetylcholine-induced hyperpolarization of both hepatic and mesenteric artery myocytes was partially inhibited by each of the gap junction inhibitors. 5. Collectively, the data suggest that gap junctions play some role in the EDHF (endothelium-derived hyperpolarizing factor) response in rat hepatic and mesenteric arteries. However, in the guinea-pig internal carotid artery, electrotonic propagation of endothelial cell hyperpolarizations via gap junctions may be the sole mechanism underlying the response previously attributed to EDHF.


Subject(s)
Anti-Ulcer Agents/pharmacology , Arteries/drug effects , Biological Factors/pharmacology , Carbenoxolone/pharmacology , Connexins/pharmacology , Gap Junctions/drug effects , Potassium/pharmacology , Acetylcholine/pharmacology , Animals , Arteries/physiology , Gap Junctions/physiology , Guinea Pigs , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Muscle, Smooth, Vascular/drug effects , Potassium Chloride/pharmacology , Rats , Vasodilator Agents/pharmacology
19.
Br J Pharmacol ; 128(5): 1064-70, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10556944

ABSTRACT

1. The characteristics of endothelium-dependent hyperpolarization in rat hepatic artery have been further investigated in the presence of inhibitors of cyclo-oxygenase and nitric oxide synthase. 2. Using sharp micro-electrodes, the smooth muscle hyperpolarization induced by acetylcholine, KCl or 1-ethyl-2-benzimidazolinone (1-EBIO) in intact hepatic arteries was abolished by 30 micronM barium plus 500 nM ouabain. 3. In vessels without endothelium, the smooth muscle hyperpolarization induced by KCl was not reduced by 30 micronM barium alone. However, in the presence of barium the effects of KCl were partially inhibited by 100 nM ouabain and essentially abolished by 500 nM ouabain. 4. Using sharp micro-electrodes, the hyperpolarization of both the smooth muscle and the endothelium induced by 1-EBIO or by acetylcholine was unaffected by 100 nM iberiotoxin. However, in the presence of 100 nM charybdotoxin, the effects of 1-EBIO were abolished whereas those of acetylcholine were only partially reduced. The hyperpolarization induced by levcromakalim was unaffected by either charybdotoxin or iberiotoxin. 5 Under whole-cell patch-clamp recording conditions, 1-EBIO induced a voltage-insensitive, charybdotoxin-sensitive K+ current in cultured endothelial cells but was without effect on K+ currents in smooth muscle cells isolated from hepatic arteries. 6 It is concluded that the endothelium-dependent hyperpolarization of smooth muscle induced by either acetylcholine or by 1-EBIO in rat hepatic artery is initially associated with the opening of endothelial calcium-sensitive K+-channels insensitive to iberiotoxin. The resulting accumulation of K+ in the myoendothelial space activates an isoform of Na+/K+-ATPase which is sensitive to low concentrations of ouabain.


Subject(s)
Benzimidazoles/pharmacology , Biological Factors/pharmacology , Calcium Channel Agonists/pharmacology , Enzyme Inhibitors/pharmacology , Hepatic Artery/drug effects , Ouabain/pharmacology , Acetylcholine/pharmacology , Animals , Cromakalim/pharmacology , Electrophysiology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Hepatic Artery/cytology , In Vitro Techniques , Male , Microelectrodes , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Patch-Clamp Techniques , Potassium/pharmacology , Potassium Channels/drug effects , Potassium Channels/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Exchanging ATPase/metabolism , Vasodilator Agents/pharmacology
20.
Nature ; 396(6708): 269-72, 1998 Nov 19.
Article in English | MEDLINE | ID: mdl-9834033

ABSTRACT

In arteries, muscarinic agonists such as acetylcholine release an unidentified, endothelium-derived hyperpolarizing factor (EDHF) which is neither prostacyclin nor nitric oxide. Here we show that EDHF-induced hyperpolarization of smooth muscle and relaxation of small resistance arteries are inhibited by ouabain plus Ba2+; ouabain is a blocker of Na+/K+ ATPase and Ba2+ blocks inwardly rectifying K+ channels. Small increases in the amount of extracellular K+ mimic these effects of EDHF in a ouabain- and Ba2+-sensitive, but endothelium-independent, manner. Acetylcholine hyperpolarizes endothelial cells and increases the K+ concentration in the myoendothelial space; these effects are abolished by charbdotoxin plus apamin. Hyperpolarization of smooth muscle by EDHF is also abolished by this toxin combination, but these toxins do not affect the hyperpolarizaiton of smooth muscle by added K+. These data show that EDHF is K+ that effluxes through charybdotoxin- and apamin-sensitive K+ channels on endothelial cells. The resulting increase in myoendothelial K+ concentration hyperpolarizes and relaxes adjacent smooth-muscle cells by activating Ba2+-sensitive K+ channels and Na+/K+ ATPase. These results show that fluctuations in K+ levels originating within the blood vessel itself are important in regulating mammalian blood pressure and flow.


Subject(s)
Arteries/physiology , Biological Factors/physiology , Potassium/physiology , Acetylcholine/pharmacology , Animals , Apamin/pharmacology , Arteries/drug effects , Barium/pharmacology , Biological Factors/metabolism , Charybdotoxin/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Enzyme Inhibitors/pharmacology , Hepatic Artery , In Vitro Techniques , Male , Membrane Potentials , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Ouabain/pharmacology , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channel Blockers , Potassium Channels/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism , Vasodilation/drug effects , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...