Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters










Publication year range
2.
Acta Physiol (Oxf) ; 219(1): 22-96, 2017 01.
Article in English | MEDLINE | ID: mdl-26706498

ABSTRACT

The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.


Subject(s)
Endothelium, Vascular/physiopathology , Muscle, Smooth, Vascular/physiopathology , Vascular Diseases/physiopathology , Animals , Cyclic GMP/metabolism , Endothelin-1/metabolism , Endothelium, Vascular/metabolism , Humans , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/metabolism , Vascular Diseases/metabolism , Vasoconstriction/physiology , Vasodilation/physiology
3.
Acta Physiol (Oxf) ; 196(2): 193-222, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19220204

ABSTRACT

The endothelium can evoke relaxations (dilatations) of the underlying vascular smooth muscle, by releasing vasodilator substances. The best characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO). The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDHF-mediated responses). Endothelium-dependent relaxations involve both pertussis toxin-sensitive G(i) (e.g. responses to serotonin and thrombin) and pertussis toxin-insensitive G(q) (e.g. adenosine diphosphate and bradykinin) coupling proteins. The release of NO by the endothelial cell can be up-regulated (e.g. by oestrogens, exercise and dietary factors) and down-regulated (e.g. oxidative stress, smoking and oxidized low-density lipoproteins). It is reduced in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively loose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and causing endothelium-dependent hyperpolarizations), endothelial cells also can evoke contraction (constriction) of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factor (EDCF). Most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells. EDCF-mediated responses are exacerbated when the production of NO is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive patients.


Subject(s)
Endothelium, Vascular/physiopathology , Vascular Diseases/physiopathology , Animals , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Humans , Models, Biological , Vascular Diseases/metabolism , Vasoconstriction/physiology , Vasodilation/physiology
4.
Naunyn Schmiedebergs Arch Pharmacol ; 377(2): 101-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18264694

ABSTRACT

This study was designed to determine whether putative openers of calcium-activated potassium channels of small and/or intermediate conductance (SK(Ca) and IK(Ca)) induce vascular smooth muscle hyperpolarizations and to identify the underlying mechanisms. The membrane potential of guinea pig carotid artery smooth muscle cells was recorded with intracellular microelectrodes in the presence of N(omega)-nitro-L-arginine and indomethacin. Acetylcholine and NS-309 produced endothelium-dependent hyperpolarizations. The effects of acetylcholine were partially and significantly inhibited by apamin. The combinations of charybdotoxin plus apamin and TRAM-34 plus apamin markedly and significantly reduced these hyperpolarizations. 1-ethyl-2-benzimidazolinone (1-EBIO) induced hyperpolarizations that were unaffected by TRAM-34 but partially inhibited by charybdotoxin, apamin, TRAM-34 plus apamin, and charybdotoxin plus apamin. Riluzole produced only marginal hyperpolarizations. Therefore, in the guinea pig carotid artery, endothelium-dependent hyperpolarization to acetylcholine involves the activation of both SK(Ca) and IK(Ca), with a predominant role for the former channel. 1-EBIO is a non-selective and weak opener of SK(Ca), while riluzole is virtually ineffective. By contrast, NS-309 is a reasonably potent and selective opener of both SK(Ca) and IK(Ca), and this compound mimics the endothelium-dependent hyperpolarizations to acetylcholine.


Subject(s)
Biological Factors/physiology , Carotid Arteries/physiology , Endothelium, Vascular/physiology , Potassium Channels, Calcium-Activated/drug effects , Acetylcholine/pharmacology , Animals , Benzimidazoles/pharmacology , Guinea Pigs , Indoles/pharmacology , Male , Membrane Potentials/drug effects , Oximes/pharmacology , Pyrazoles/pharmacology , Riluzole/pharmacology
5.
Br J Pharmacol ; 153(1): 57-65, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17906681

ABSTRACT

BACKGROUND AND PURPOSE: C-type natriuretic peptide (CNP) has been proposed to make a fundamental contribution in arterial endothelium-dependent hyperpolarization to acetylcholine. The present study was designed to address this hypothesis in the guinea-pig carotid artery. EXPERIMENTAL APPROACH: The membrane potential of vascular smooth muscle cells was recorded in isolated arteries with intracellular microelectrodes. KEY RESULTS: Acetylcholine induced endothelium-dependent hyperpolarizations in the presence or absence of N (G)-nitro-L-arginine, indomethacin and/or thiorphan, inhibitors of NO-synthases, cyclooxygenases or neutral endopeptidase, respectively. Acetycholine hyperpolarized smooth muscle cells in resting arteries and produced repolarizations in phenylephrine-stimulated arteries. CNP produced hyperpolarizations with variable amplitude. They were observed only in the presence of inhibitors of NO-synthases and cyclooxygenases and were endothelium-independent, maintained in phenylephrine-depolarized carotid arteries, and not affected by the additional presence of thiorphan. In arteries with endothelium, the hyperpolarizations produced by CNP were always significantly smaller than those induced by acetylcholine. Upon repeated administration, a significant tachyphylaxis of the hyperpolarizing effect of CNP was observed, while consecutive administration of acetycholine produced sustained responses. The hyperpolarizations evoked by acetylcholine were abolished by the combination of apamin plus charybdotoxin, but unaffected by glibenclamide or tertiapin. In contrast, CNP-induced hyperpolarizations were abolished by glibenclamide and unaffected by the combination of apamin plus charybdotoxin. CONCLUSIONS AND IMPLICATIONS: In the isolated carotid artery of the guinea-pig, CNP activates K(ATP) and is a weak hyperpolarizing agent. In this artery, the contribution of CNP to EDHF-mediated responses is unlikely.


Subject(s)
Biological Factors/physiology , Carotid Arteries/drug effects , Endothelium, Vascular/physiology , Natriuretic Peptide, C-Type/pharmacology , Vasodilation/drug effects , Acetylcholine/pharmacology , Animals , Bee Venoms/pharmacology , Carotid Arteries/physiology , Glyburide/pharmacology , Guinea Pigs , In Vitro Techniques , Indomethacin/pharmacology , Male , Membrane Potentials/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Nitroarginine/pharmacology
6.
Br J Pharmacol ; 151(1): 15-23, 2007 May.
Article in English | MEDLINE | ID: mdl-17351662

ABSTRACT

BACKGROUND AND PURPOSE: Experiments were designed to assess whether or not the intracellular concentration of calcium and reactive oxygen species (ROS) increase in endothelial cells of the rat thoracic aorta in response to releasers of endothelium-derived contracting factor (EDCF) and if so, whether or not a difference exists between spontaneously hypertensive (SHR) and normotensive (WKY) rats. EXPERIMENTAL APPROACH: Calcium and ROS were measured by confocal microscopy, using Fura-red in combination with Fluo-4 and dichlorodihydrofluorescein diacetate, respectively. KEY RESULTS: Acetylcholine caused a rapid increase in cytosolic calcium concentration in endothelial cells of both SHR and WKY, which was significantly more pronounced in aortae of the former strain. This rise of calcium was not affected by indomethacin (an inhibitor of cyclooxygenase) or Tiron plus diethyldithiocarbamate acid (DETCA) (membrane permeable antioxidants). In the presence of a nitric oxide synthase blocker, acetylcholine also caused a rapid increase in ROS in endothelial cells of SHR but not in those of WKY. The burst of ROS was prevented by indomethacin or Tiron plus DETCA. CONCLUSIONS AND IMPLICATIONS: These experiments show that endothelial cells of SHR are more prone to calcium and ROS overload upon stimulation with acetylcholine. The abnormal accumulation of calcium is a prerequisite to initiate the release of EDCF and can be mimicked using the calcium ionophore A23187. The sequence of events occurring during endothelium-dependent contractions firstly requires the accumulation of calcium, which then activates cyclooxygenase and produces ROS along with EDCF that in turn stimulates TP-receptors, resulting in EDCF-mediated contractions.


Subject(s)
Calcium/metabolism , Endothelial Cells/metabolism , Endothelins/physiology , Reactive Oxygen Species/metabolism , Animals , Calcimycin/pharmacology , Male , Oxidative Stress , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Vasoconstriction
7.
Br J Pharmacol ; 150(5): 624-32, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17245370

ABSTRACT

BACKGROUND AND PURPOSE: To study the importance of endothelium-derived contracting factors (EDCFs) in arteries of rats with type I diabetes. EXPERIMENTAL APPROACH: Rat femoral arteries were collected four or twelve weeks after induction of diabetes with streptozotocin. Rings, with or without endothelium, were suspended in organ chambers for isometric tension measurement. COX protein levels were determined by Western blotting. KEY RESULTS: Four weeks after the injection of streptozotocin, the endothelium-dependent relaxations (during contractions to phenylephrine) to A23817 were attenuated, but the endothelium-dependent contractions (quiescent preparations) to the ionophore were augmented. Indomethacin and S18886 prevented the endothelium-dependent contractions, while dazoxiben reduced them in rings from streptozotocin-treated rats, suggesting that thromboxane A2, activating TP- receptors, is involved. Twelve weeks after the injection of streptozotocin, the changes in endothelium-dependent relaxations and contractions to A23187 were even more noticeable. The protein expression of COX-1 was increased in femoral arteries of the diabetic rats. Valeryl salicylate and SC560 inhibited the contractions, suggesting that the EDCFs are produced by COX-1. At that time, a combination of S18886 with EP1-blockers was required to abolish the contractions, suggesting that the EDCFs involved act at both TP- and EP-receptors. Rings without endothelium from streptozotocin-treated rats exhibited a reduced maximal contraction to potassium chloride and U46619, combined with hyper-responsiveness to the latter, suggesting that more prolonged diabetes also alters the responsiveness of vascular smooth muscle. CONCLUSION AND IMPLICATIONS: The production of EDCFs is progressively increased in the course of type I diabetes. Eventually, the disease also damages vascular smooth muscle.


Subject(s)
Calcimycin/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Endothelium, Vascular/drug effects , Ionophores/pharmacology , Vasoconstriction/drug effects , Animals , Blotting, Western , Calcium/metabolism , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 2/biosynthesis , Cyclooxygenase Inhibitors/pharmacology , Diabetes Mellitus, Experimental/metabolism , Dose-Response Relationship, Drug , Endothelins/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Femoral Artery/drug effects , Femoral Artery/physiopathology , Membrane Proteins/biosynthesis , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Rats , Receptors, Prostaglandin E/metabolism , Receptors, Thromboxane/metabolism , Thromboxane A2/metabolism , Time Factors , Vasodilation/drug effects
8.
Int J Obes Relat Metab Disord ; 28(4): 628-39, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14758341

ABSTRACT

AIM: These studies were performed to test the hypothesis that endogenous neuropeptide Y (NPY) acting on the NPY Y(5) receptor subtype contributes to the control of food intake. The hypothesis was tested using S 25585-a newly synthesized NPY Y(5) receptor antagonist. METHODS AND RESULTS: S 25585 was shown to be a high-affinity antagonist of the NPY Y(5) receptor subtype (IC(50) 5 nM) with no significant affinity toward other NPY receptor subtypes and over 40 other receptors, channels or uptake systems. S 25585 (7.5 mg/kg, i.p.) did not induce a conditioned taste aversion, significantly alter need-induced sodium appetite or induce pica, suggesting that at this dose the compound did not induce illness or malaise. In satiated rats, S 25585 (5.0 and 7.5 mg/kg, i.p.) significantly decreased the overfeeding induced by i.c.v. injection of NPY (1 microg) and the highly selective NPY Y(5) receptor agonist [hPP(1-17), Ala(31), Aib(32)]NPY (0.7 microg). In rats fasted for 4 h immediately before the dark phase, analysis of the microstructure of feeding behavior revealed that S 25585 significantly increased latency to eat and significantly decreased the duration and size of the meals without altering the meal number or eating rate. Analysis of the behavioral satiety sequence at this time revealed that the animals passed through the normal pattern of feeding, grooming and resting. Although S 25585 appeared to be influencing a physiological system controlling appetite, this does not involve the NPY Y(5) receptor since the antagonist also markedly reduced food intake in the NPY Y(5) knockout mouse. CONCLUSIONS: The results presented do not support a role for the NPY Y(5) receptor in the control of food intake. The results further illustrate that it is imperative that the activity of any new NPY Y(5) antagonist be assessed in the NPY Y(5) knockout mouse before assuming that its effect on food intake is due to blockade of this receptor.


Subject(s)
Eating/drug effects , Eating/physiology , Receptors, Neuropeptide Y/antagonists & inhibitors , Animals , Appetite/physiology , Conditioning, Psychological/physiology , Male , Mice , Mice, Knockout , Pica/physiopathology , Rats , Rats, Long-Evans , Rats, Wistar , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/physiology , Satiety Response/physiology , Sodium Chloride, Dietary/administration & dosage , Taste/physiology
10.
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
11.
Int J Obes Relat Metab Disord ; 26(3): 281-98, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11896483

ABSTRACT

AIM: The aim of this review is to critically assess available evidence that blockade of the actions of NPY at one of the five NPY receptor subtypes represents an attractive new drug discovery target for the development of an appetite suppressant drug. RESULTS: Blockade of the central actions of NPY using anti-NPY antibodies, antisense oligodeoxynucleotides against NPY and NPY receptor antagonists results in a decrease in food intake in energy-deprived animals. These results appear to show that endogenous NPY plays a role in the control of appetite. The fact that NPY receptors exist as at least five different subtypes raises the possibility that the actions of endogenous NPY on food intake can be adequately dissociated from other effects of the peptide. Current drug discovery has produced a number of highly selective NPY receptor antagonists which have been used to establish the NPY Y(1) receptor subtype as the most critical in regulating short-term food intake. However, additional studies are now needed to more clearly define the relative contribution of NPY acting through the NPY Y2 and NPY Y5 receptors in the complex sequence of physiological and behavioral events that underlie the long-term control of appetite. CONCLUSIONS: Blockade of the NPY receptor may produce appetite-suppressing drugs. However, it is too early to state with certainty whether a single subtype selective drug used alone or a combination of NPY receptor selective antagonists used in combination will be necessary to adequately influence appetite regulation.


Subject(s)
Appetite Depressants , Receptors, Neuropeptide Y/antagonists & inhibitors , Animals , Antibodies/pharmacology , Brain/drug effects , Brain/metabolism , Drug Design , Eating/drug effects , Energy Metabolism , Humans , Mice , Mice, Knockout , Naphthalenes/pharmacology , Neuropeptide Y/antagonists & inhibitors , Neuropeptide Y/genetics , Neuropeptide Y/immunology , Neuropeptide Y/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Pyrimidines/pharmacology , Receptors, Neuropeptide Y/deficiency , Receptors, Neuropeptide Y/metabolism
12.
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
13.
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
14.
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
15.
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
16.
Am J Physiol Lung Cell Mol Physiol ; 281(1): L258-67, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11404269

ABSTRACT

Nitric oxide (NO) is a potent vasodilator, but it can also modulate contractile responses of the airway smooth muscle. Whether or not endothelial (e) NO synthase (NOS) contributes to the regulation of bronchial tone is unknown at present. Experiments were designed to investigate the isoforms of NOS that are expressed in murine airways and to determine whether or not the endogenous release of NO modulates bronchial tone in wild-type mice and in mice with targeted deletion of eNOS [eNOS(-/-)]. The presence of neuronal NOS (nNOS), inducible NOS (iNOS), and eNOS in murine trachea and lung parenchyma was assessed by RT-PCR, immunoblotting, and immunohistochemistry. Airway resistance was measured in conscious unrestrained mice by means of a whole body plethysmography chamber. The three isoforms of NOS were constitutively present in lungs of wild-type mice, whereas only iNOS and nNOS were present in eNOS(-/-) mice. Labeling of nNOS was localized in submucosal airway nerves but was not consistently detected, and iNOS immunoreactivity was observed in tracheal and bronchiolar epithelial cells, whereas eNOS was expressed in endothelial cells. In wild-type mice, treatment with N-nitro-L-arginine methyl ester, but not with aminoguanidine, potentiated the increase in airway resistance produced by inhalation of methacholine. eNOS(-/-) mice were hyperresponsive to inhaled methacholine and markedly less sensitive to N-nitro-L-arginine methyl ester. These results demonstrate that the three NOS isoforms are expressed constitutively in murine lung and that NO derived from eNOS plays a physiological role in controlling bronchial airway reactivity.


Subject(s)
Airway Resistance/physiology , Bronchi/physiology , Nitric Oxide Synthase/physiology , Airway Resistance/drug effects , Animals , Brain/metabolism , Bronchi/drug effects , Cholinergic Agonists/pharmacology , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout/genetics , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , RNA, Messenger/metabolism , Reference Values , Trachea/drug effects , Trachea/metabolism
17.
Br J Pharmacol ; 132(6): 1342-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11250886

ABSTRACT

1. The purpose of this study in the hamster cheek pouch was to determine whether or not vascular endothelial growth factor (VEGF) induced changes in plasma extravasation and if so, the mechanism(s) involved. 2. The cheek pouch microcirculatory bed of the anaesthetized hamster was directly observed under microscope and the number of vascular leakage sites, as shown by fluorescein isothiocyanate (FITC-dextran, 150 kD) extravasation, was counted. Drugs and VEGF were applied topically. VEGF from 0.05 to 0.5 microg ml(-1) (1.2 to 12 nM) produced a dose-dependent increase in the number of microvascular leakage sites from virtually none in basal conditions to up to 250 in some pouches. The effects of VEGF (0.1 microg ml(-1) or 2.4 nM) were blocked in a concentration-dependent manner by the non-specific heparin growth factor antagonist TBC-1635 (0.1, 1 and 3microM). The placenta growth factor (PlGF-1: 0.1 and 0.5 microg ml(-1) or 3.4 and 17 nM) did not increase plasma extravasation, per se, but abolished the effects of VEGF (2.4 nM). 3. The increases in microvascular leakage produced by VEGF (2.4 nM) were partially but significantly (P<0.05) inhibited by genistein (5 and 10 microM, up to 33% inhibition), LY 294002 (30 microM, 41%), bisindolylmaleimide (1 microM, 65%) and virtually abolished by indomethacin (3 microM, 88%) and L-nitro-arginine (10 microM, 95%), these drugs being inhibitors of tyrosine kinase, phosphatidylinositol-3-kinase, protein kinase C, cyclo-oxygenase and nitric oxide synthase respectively. None of these inhibitors, at the concentration tested, induced alone an increase in plasma extravasation. 4. These results indicate that the VEGF-induced plasma extravasation may involve the stimulation of VEGF-R2 (Flk-1/KDR) and the activation of phosphatidylinositol-3-kinase and protein kinase C. The production of both nitric oxide and prostaglandin is required to observe an increase in vascular leakage.


Subject(s)
Cheek/physiology , Endothelial Growth Factors/physiology , Extravasation of Diagnostic and Therapeutic Materials , Lymphokines/physiology , Animals , Cricetinae , Male , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
18.
Int J Obes Relat Metab Disord ; 25(1): 84-94, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11244462

ABSTRACT

INTRODUCTION: The purpose of this study was to test the continuing validity of the hypothesis that neuropeptide Y (NPY) produced in the brain controls food intake through an interaction with the NPY Y(5) receptor subtype. METHODS: The hypothesis was tested using CGP 71683A a potent and highly selective non-peptide antagonist of the NPY Y(5) receptor which was administered into the right lateral ventricle of obese Zucker fa/fa rats. RESULTS: Intraventricular injection of 3.4 nmol/kg NPY increased food intake during a 2 h test period. Doses of CGP 71683A in excess of 15 nmol/kg (i.cv.) resulted in blockade of the increase in food intake produced by NPY. Repeated daily injection of CGP 71683A (30--300 nmol/kg, i.cv.) immediately before the dark phase produced a dose-dependent and slowly developing decrease in food intake. CGP 71683A has a low affinity for NPY Y(1), Y(2) and Y(4) receptors but a very high affinity for the NPY Y(5) receptor (Ki, 1.4 nM). Surprisingly, CGP 71683A had similarly high affinity for muscarinic receptors (Ki, 2.7 nM) and for the serotonin uptake recognition site (Ki, 6.2 nM) in rat brain. Anatomic analysis of the brain after treatment with CGP 71683A demonstrated an inflammatory response associated with the fall in food intake. CONCLUSIONS: While the fall in food intake in response to CGP 71683A may have a Y(5) component, interactions with other receptors or inflammatory mediators may also play a role. It is concluded that CGP 71683A is an imprecise tool for investigating the role of the NPY Y(5) receptor in the control of physiological processes including food intake. International Journal of Obesity (2001) 25, 84-94


Subject(s)
Eating/physiology , Energy Intake/drug effects , Naphthalenes/pharmacology , Neuropeptide Y/metabolism , Pyrimidines/pharmacology , Receptors, Neuropeptide Y/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Eating/drug effects , Encephalitis , Energy Intake/genetics , Injections, Intraventricular , Male , Naphthalenes/administration & dosage , Obesity/metabolism , Pyrimidines/administration & dosage , Rats , Rats, Wistar , Rats, Zucker , Receptors, Muscarinic/metabolism , Receptors, Serotonin/metabolism , Time Factors
19.
Proc Natl Acad Sci U S A ; 97(17): 9747-52, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10944233

ABSTRACT

In addition to nitric oxide (NO) and prostacyclin (PGI(2)), the endothelium generates the endothelium-derived hyperpolarizing factor (EDHF). We set out to determine whether an EDHF-like response can be detected in wild-type (WT) and endothelial NO synthase knockout mice (eNOS -/-) mice. Vasodilator responses to endothelium-dependent agonists were determined in vivo and in vitro. In vivo, bradykinin induced a pronounced, dose-dependent decrease in mean arterial pressure (MAP) which did not differ between WT and eNOS -/- mice and was unaffected by treatment with N(omega)-nitro-l-arginine methyl ester and diclofenac. In the saline-perfused hindlimb of WT and eNOS -/- mice, marked N(omega)-nitro-l-arginine (l-NA, 300 micromol/liter)- and diclofenac-insensitive vasodilations in response to both bradykinin and acetylcholine (ACh) were observed, which were more pronounced than the agonist-induced vasodilation in the hindlimb of WT in the absence of l-NA. This endothelium-dependent, NO/PGI(2)-independent vasodilatation was sensitive to KCl (40 mM) and to the combination of apamin and charybdotoxin. Gap junction inhibitors (18alpha-glycyrrhetinic acid, octanol, heptanol) and CB-1 cannabinoid-receptor agonists (Delta(9)-tetrahydrocannabinol, HU210) impaired EDHF-mediated vasodilation, whereas inhibition of cytochrome P450 enzymes, soluble guanylyl cyclase, or adenosine receptors had no effect on EDHF-mediated responses. These results demonstrate that in murine resistance vessels the predominant agonist-induced endothelium-dependent vasodilation in vivo and in vitro is not mediated by NO, PGI(2), or a cytochrome P450 metabolite, but by an EDHF-like principle that requires functional gap junctions.


Subject(s)
Biological Factors/metabolism , Endothelium, Vascular/metabolism , Epoprostenol/metabolism , Nitric Oxide Synthase/deficiency , Nitric Oxide/metabolism , Vasodilator Agents/metabolism , Acetylcholine/pharmacology , Animals , Arteries/drug effects , Arteries/physiology , Blood Pressure/drug effects , Bradykinin/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Diclofenac/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Gap Junctions/drug effects , Gap Junctions/physiology , Gene Deletion , Hindlimb/blood supply , Hindlimb/drug effects , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitroarginine/pharmacology , Nitroprusside/pharmacology , Perfusion , Potassium Channel Blockers , Potassium Channels/metabolism , Receptors, Cannabinoid , Receptors, Drug/antagonists & inhibitors , Receptors, Drug/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...