Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Acta Physiol (Oxf) ; 224(1): e13059, 2018 09.
Article in English | MEDLINE | ID: mdl-29480968

ABSTRACT

AIMS: The Na,K-ATPase is involved in a large number of regulatory activities including cSrc-dependent signalling. Upon inhibition of the Na,K-ATPase with ouabain, cSrc activation is shown to occur in many cell types. This study tests the hypothesis that acute potentiation of agonist-induced contraction by ouabain is mediated through Na,K-ATPase-cSrc signalling-dependent sensitization of vascular smooth muscle cells to Ca2+ . METHODS: Agonist-induced rat mesenteric small artery contraction was examined in vitro under isometric conditions and in vivo in anaesthetized rats. Arterial wall tension and [Ca2+ ]i in vascular smooth muscle cells were measured simultaneously. Changes in cSrc and myosin phosphatase targeting protein 1 (MYPT1) phosphorylation were analysed by Western blot. Protein expression was examined with immunohistochemistry. The α1 and α2 isoforms of the Na,K-ATPase were transiently downregulated by siRNA transfection in vivo. RESULTS: Ten micromolar ouabain, but not digoxin, potentiated contraction to noradrenaline. This effect was not endothelium-dependent. Ouabain sensitized smooth muscle cells to Ca2+ , and this was associated with increased phosphorylation of cSrc and MYPT1. Inhibition of tyrosine kinase by genistein, PP2 or pNaKtide abolished the potentiating effect of ouabain on arterial contraction and Ca2+ sensitization. Downregulation of the Na,K-ATPase α2 isoform made arterial contraction insensitive to ouabain and tyrosine kinase inhibition. CONCLUSION: Data suggest that micromolar ouabain potentiates agonist-induced contraction of rat mesenteric small artery via Na,K-ATPase-dependent cSrc activation, which increases Ca2+ sensitization of vascular smooth muscle cells by MYPT1 phosphorylation. This mechanism may be critical for acute control of vascular tone.


Subject(s)
Calcium Signaling , Mesenteric Arteries/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Vasoconstriction , src-Family Kinases/metabolism , Animals , Calcium Signaling/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Inhibitors/pharmacology , Male , Mesenteric Arteries/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Phosphorylation , Protein Phosphatase 1/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/genetics , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , src-Family Kinases/antagonists & inhibitors
3.
Am J Physiol Cell Physiol ; 310(3): C193-204, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26538090

ABSTRACT

The extracellular calcium-sensing receptor CaSR is expressed in blood vessels where its role is not completely understood. In this study, we tested the hypothesis that the CaSR expressed in vascular smooth muscle cells (VSMC) is directly involved in regulation of blood pressure and blood vessel tone. Mice with targeted CaSR gene ablation from vascular smooth muscle cells (VSMC) were generated by breeding exon 7 LoxP-CaSR mice with animals in which Cre recombinase is driven by a SM22α promoter (SM22α-Cre). Wire myography performed on Cre-negative [wild-type (WT)] and Cre-positive (SM22α)CaSR(Δflox/Δflox) [knockout (KO)] mice showed an endothelium-independent reduction in aorta and mesenteric artery contractility of KO compared with WT mice in response to KCl and to phenylephrine. Increasing extracellular calcium ion (Ca(2+)) concentrations (1-5 mM) evoked contraction in WT but only relaxation in KO aortas. Accordingly, diastolic and mean arterial blood pressures of KO animals were significantly reduced compared with WT, as measured by both tail cuff and radiotelemetry. This hypotension was mostly pronounced during the animals' active phase and was not rescued by either nitric oxide-synthase inhibition with nitro-l-arginine methyl ester or by a high-salt-supplemented diet. KO animals also exhibited cardiac remodeling, bradycardia, and reduced spontaneous activity in isolated hearts and cardiomyocyte-like cells. Our findings demonstrate a role for CaSR in the cardiovascular system and suggest that physiologically relevant changes in extracellular Ca(2+) concentrations could contribute to setting blood vessel tone levels and heart rate by directly acting on the cardiovascular CaSR.


Subject(s)
Blood Pressure , Calcium Signaling , Calcium/metabolism , Hypotension/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, G-Protein-Coupled/metabolism , Vasoconstriction , Vasodilation , Animals , Aorta/metabolism , Blood Pressure/drug effects , Blood Pressure/genetics , Bradycardia/genetics , Bradycardia/metabolism , Bradycardia/physiopathology , Calcium Signaling/drug effects , Calcium Signaling/genetics , Dose-Response Relationship, Drug , Genetic Predisposition to Disease , Heart Rate , Hypotension/genetics , Hypotension/physiopathology , Mesenteric Arteries/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Myocytes, Cardiac/metabolism , Phenotype , Receptors, Calcium-Sensing , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Vasoconstriction/drug effects , Vasoconstriction/genetics , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilation/genetics , Vasodilator Agents/pharmacology , Ventricular Remodeling
4.
Br J Pharmacol ; 172(16): 4158-72, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26013995

ABSTRACT

BACKGROUND AND PURPOSE: T16A(inh)-A01, CaCC(inh)-A01 and MONNA are identified as selective inhibitors of the TMEM16A calcium-activated chloride channel (CaCC). The aim of this study was to examine the chloride-specificity of these compounds on isolated resistance arteries in the presence and absence (±) of extracellular chloride. EXPERIMENTAL APPROACH: Isolated resistance arteries were maintained in a myograph and tension recorded, in some instances combined with microelectrode impalement for membrane potential measurements or intracellular calcium monitoring using fura-2. Voltage-dependent calcium currents (VDCC) were measured in A7r5 cells with voltage-clamp electrophysiology using barium as a charge carrier. KEY RESULTS: Rodent arteries preconstricted with noradrenaline or U46619 were concentration-dependently relaxed by T16A(inh) -A01 (0.1-10 µM): IC50 and maximum relaxation were equivalent in ±chloride (30 min aspartate substitution) and the T16A(inh) -A01-induced vasorelaxation ±chloride were accompanied by membrane hyperpolarization and lowering of intracellular calcium. However, agonist concentration-response curves ±chloride, with 10 µM T16A(inh) -A01 present, achieved similar maximum constrictions although agonist-sensitivity decreased. Contractions induced by elevated extracellular potassium were concentration-dependently relaxed by T16A(inh)-A01 ±chloride. Moreover, T16A(inh) -A01 inhibited VDCCs in A7r5 cells in a concentration-dependent manner. CaCC(inh) -A01 and MONNA (0.1-10 µM) induced vasorelaxation ±chloride and both compounds lowered maximum contractility. MONNA, 10 µM, induced substantial membrane hyperpolarization under resting conditions. CONCLUSIONS AND IMPLICATIONS: T16A(inh) -A01, CaCC(inh) -A01 and MONNA concentration-dependently relax rodent resistance arteries, but an equivalent vasorelaxation occurs when the transmembrane chloride gradient is abolished with an impermeant anion. These compounds therefore display poor selectivity for TMEM16A and inhibition of CaCC in vascular tissue in the concentration range that inhibits the isolated conductance.


Subject(s)
Chloride Channels/antagonists & inhibitors , Pyrimidines/pharmacology , Thiazoles/pharmacology , Thiophenes/pharmacology , Vasodilator Agents/pharmacology , ortho-Aminobenzoates/pharmacology , Animals , Cell Line , Cerebral Arteries/drug effects , Cerebral Arteries/physiology , Chloride Channels/physiology , Female , In Vitro Techniques , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Mice, Inbred C57BL , Rats, Wistar
5.
Br J Pharmacol ; 171(1): 69-82, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24111896

ABSTRACT

BACKGROUND AND PURPOSE: Hypoxia causes vasodilatation of coronary arteries, but the underlying mechanisms are poorly understood. We hypothesized that hypoxia reduces intracellular Ca(2+) concentration ([Ca(2+)](i)) by opening of K channels and release of H2S. EXPERIMENTAL APPROACH: Porcine coronary arteries without endothelium were mounted for measurement of isometric tension and [Ca(2+)](i), and the expression of voltage-gated K channels K(V)7 channels (encoded by KCNQ genes) and large-conductance calcium-activated K channels (K(Ca)1.1) was examined. Voltage clamp assessed the role of K(V)7 channels in hypoxia. KEY RESULTS: Gradual reduction of oxygen concentration from 95 to 1% dilated the precontracted coronary arteries and this was associated with reduced [Ca(2+)](i) in PGF(2α) (10 µM)-contracted arteries whereas no fall in [Ca(2+)](i) was observed in 30 mM K-contracted arteries. Blockers of ATP-sensitive voltage-gated potassium channels and K(Ca)1.1 inhibited hypoxia-induced dilatation in PGF2α -contracted arteries; this inhibition was more marked in the presence of the K(v)7 channel blockers, XE991 and linopirdine, while a K(V)7.1 blocker, failed to change hypoxic vasodilatation. XE991 also inhibited H2S- and adenosine-induced vasodilatation. PCR revealed the expression of K(V)7.1, K(V)7.4, K(V)7.5 and K(Ca)1.1 channels, and K(Ca)1.1, K(V)7.4 and K(V)7.5 were also identified by immunoblotting. Voltage clamp studies showed the XE991-sensitive current was more marked in hypoxic conditions. CONCLUSION: The K(V)7.4 and K(V)7.5 channels, which we identified in the coronary arteries, appear to have a major role in hypoxia-induced vasodilatation. The voltage clamp results further support the involvement of K(V)7 channels in this vasodilatation. Activation of these K(V)7 channels may be induced by H2S and adenosine.


Subject(s)
Hypoxia/metabolism , KCNQ Potassium Channels/metabolism , Muscle, Smooth, Vascular/metabolism , Oxygen/metabolism , Vasodilation , Adenosine/pharmacology , Animals , Calcium Signaling , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Dose-Response Relationship, Drug , Hydrogen Sulfide/pharmacology , Hypoxia/genetics , Hypoxia/physiopathology , KCNQ Potassium Channels/drug effects , KCNQ Potassium Channels/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Membrane Potentials , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Potassium Channel Blockers/pharmacology , Signal Transduction , Swine , Time Factors , Vasodilation/drug effects , Vasodilator Agents/pharmacology
6.
Acta Physiol (Oxf) ; 202(3): 253-69, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21518271

ABSTRACT

This minireview discusses vasomotion, which is the oscillation in tone of blood vessels leading to flowmotion. We will briefly discuss the prevalence of vasomotion and its potential physiological and pathophysiological relevance. We will also discuss the models that have been suggested to explain how a coordinated oscillatory activity of the smooth muscle tone can occur and emphasize the role of the endothelium, the handling of intracellular Ca(2+) and the role of smooth muscle cell ion conductances. It is concluded that vasomotion is likely to enhance tissue dialysis, although this concept still requires more experimental verification, and that an understanding at the molecular level for the pathways leading to vasomotion is beginning to emerge.


Subject(s)
Endothelium, Vascular/physiology , Muscle, Smooth, Vascular/physiology , Vasomotor System/physiology , Animals , Calcium/metabolism , Calcium Signaling/physiology , Cell Communication/physiology , Chlorides/metabolism , Gap Junctions/metabolism , Humans , Membrane Potentials/physiology , Mesenteric Arteries/metabolism , Models, Biological
7.
Br J Pharmacol ; 158(6): 1465-76, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19845682

ABSTRACT

BACKGROUND AND PURPOSE: Large-conductance Ca(2+)-activated K(+) channels (BK(Ca)), located on the arterial and corporal smooth muscle, are potential targets for treatment of erectile dysfunction (ED). This study investigated whether NS11021 (1-(3,5-Bis-trifluoromethyl-phenyl)-3-[4-bromo-2-(1H-tetrazol-5-yl)-phenyl]-thiourea), a novel opener of BK(Ca) channels, relaxes erectile tissue in vitro and enhances erectile responses in intact rats. The effects were compared with sildenafil, an inhibitor of phosphodiesterase type 5. EXPERIMENTAL APPROACH: Patch clamp was used to record whole cell current in rat isolated corpus cavernosum smooth muscle cells (SMCs) and human umbilical vein endothelial cells (HUVECs). Isometric tension was measured in intracavernous arterial rings and corpus cavernosum strips isolated from rats and men, and simultaneous measurements of intracellular Ca(2+) concentration ([Ca(2+)](i)) and tension were performed in intracavernous arteries. Erectile response was measured in anaesthetized rats. KEY RESULTS: In patch clamp recordings, NS11021 increased currents sensitive to the selective BK(Ca) channel blocker, iberiotoxin (IbTX) in SMCs, but did not modulate K(+) current in HUVECs. NS11021 reduced [Ca(2+)](i) and tension in penile arteries. IbTX inhibited the vasorelaxation induced by NS11021 and sildenafil in human erectile tissue. NS11021 and sildenafil but not vehicle increased erectile responses in anaesthetized rats, an effect which was abolished after pretreatment with tetraethylammonium. CONCLUSIONS AND IMPLICATIONS: NS11021 leads to relaxation of both intracavernous arteries and corpus cavernosum strips primarily through opening of BK(Ca) channels. It is also effective in facilitating erectile responses in anaesthetized rats. These results suggest a potential for use of BK(Ca) openers in the treatment of ED.


Subject(s)
Erectile Dysfunction/drug therapy , Large-Conductance Calcium-Activated Potassium Channels/drug effects , Tetrazoles/pharmacology , Thiourea/analogs & derivatives , Vasodilator Agents/pharmacology , Aged , Animals , Calcium/metabolism , Erectile Dysfunction/physiopathology , Humans , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Middle Aged , Patch-Clamp Techniques , Penis/drug effects , Penis/metabolism , Penis/physiopathology , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Purines/pharmacology , Rats , Rats, Wistar , Sildenafil Citrate , Sulfones/pharmacology , Thiourea/pharmacology , Umbilical Veins/drug effects , Umbilical Veins/metabolism
8.
Pflugers Arch ; 457(2): 389-404, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18536933

ABSTRACT

The possibility that Ca(2+)-activated Cl(-) conductances (CaCCs) contribute to oscillations in vascular tone (vasomotion) is tested in isolated mesenteric small arteries from rats where cGMP independent (I (Cl(Ca))) and cGMP-dependent (I (Cl(Ca,cGMP))) chloride conductances are important. The effect of anion substitution and Cl(-) channel blockers on noradrenaline (NA)-stimulated tension in isometrically mounted mesenteric arteries and for chloride conductance of smooth muscle cells isolated from these arteries were assessed electrophysiologically. Cl(-) (o) replacement with aspartate blocked vasomotion while 36mM SCN(-) (o) (substituted for Cl(-)) was sufficient to inhibit vasomotion. Oscillations in tone, membrane potential, and [Ca(2+)](i) disappeared with 36mM SCN(-). DIDS and Zn(2+) blocked I (Cl(Ca,cGMP)) but not I (Cl(Ca)). Vasomotion was not sensitive to DIDS and Zn(2+), and DIDS and Zn(2+) induce vasomotion in arteries without endothelium. The vasomotion in the presence of DIDS and Zn(2+) was sensitive to 36mM SCN(-) (o). The anion substitution data indicate that Cl(-) is crucial for the V (m) and [Ca(2+)](i) oscillations underlying vasomotion. The Cl(-) channel blocker data are consistent with both CaCCs being important.


Subject(s)
Chloride Channels/metabolism , Chlorides/metabolism , Mesenteric Arteries/metabolism , Vasoconstriction , Vasodilation , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Aspartic Acid/metabolism , Calcium Signaling , Chloride Channels/drug effects , Dose-Response Relationship, Drug , Glycolates/pharmacology , Hydrogen-Ion Concentration , Male , Membrane Potentials , Mesenteric Arteries/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Niflumic Acid/pharmacology , Norepinephrine/pharmacology , Rats , Rats, Wistar , Sarcoplasmic Reticulum/metabolism , Thiocyanates/metabolism , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Zinc/metabolism
9.
Diabetes Obes Metab ; 10(11): 1074-85, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18435771

ABSTRACT

Recently, we showed that rebaudioside A potently stimulates the insulin secretion from isolated mouse islets in a dose-, glucose- and Ca(2+)-dependent manner. Little is known about the mechanisms underlying the insulinotropic action of rebaudioside A. The aim of this study was to define the signalling system by which, rebaudioside A acts. Isolated mouse islets were used in the cAMP[(125)I] scintillation proximity assay to measure total cAMP level, and in a luminometric method to measure intracellular ATP and ADP concentrations. Conventional and permeabilized whole-cell configuration of the patch-clamp technique was used to verify the effect of rebaudioside A on ATP-sensitive K(+)-channels from dispersed single beta cells from isolated mouse islets. Insulin was measured by radioimmunoassay from insulinoma MIN6 cells. In the presence of 16.7 mM glucose, the addition of the maximally effective concentration of rebaudioside A (10(-9) M) increased the ATP/ADP ratio significantly, while it did not change the intracellular cAMP level. Rebaudioside A (10(-9) M) and stevioside (10(-6) M) reduced the ATP-sensitive potassium channel (K(ATP)) conductance in a glucose-dependent manner. Moreover, rebaudioside A stimulated the insulin secretion from MIN6 cells in a dose- and glucose-dependent manner. In conclusion, the insulinotropic effect of rebaudioside A is mediated via inhibition of ATP-sensitive K(+)-channels and requires the presence of high glucose. The inhibition of ATP-sensitive K(+)-channels is probably induced by changes in the ATP/ADP ratio. The results indicate that rebaudioside A may offer a distinct therapeutic advantage over sulphonylureas because of less risk of causing hypoglycaemia.


Subject(s)
Diterpenes, Kaurane/pharmacology , Glucose/pharmacology , Insulin-Secreting Cells/drug effects , Insulin/metabolism , KATP Channels/metabolism , Potassium Channel Blockers/pharmacology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Cell Line , Cyclic AMP/metabolism , Female , Glucosides/pharmacology , Glyburide/pharmacology , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Patch-Clamp Techniques , Stimulation, Chemical
10.
Circ Res ; 88(8): 810-5, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11325873

ABSTRACT

Vasomotion is the regular variation in tone of arteries. In our study, we suggest a model for the initiation of vasomotion. We suggest that intermittent release of Ca(2+) from the sarcoplasmic reticulum (SR, cytosolic oscillator), which is initially unsynchronized between the vascular smooth muscle cells, becomes synchronized to initiate vasomotion. The synchronization is achieved by an ion current over the cell membrane, which is activated by the oscillating Ca(2+) release. This current results in an oscillating membrane potential, which synchronizes the SR in the vessel wall and starts vasomotion. Therefore, the pacemaker of the vascular wall can be envisaged as a diffuse array of individual cytosolic oscillators that become entrained by a reciprocal interaction with the cell membrane. The model is supported by experimental data. Confocal [Ca(2+)](i) imaging and isometric force development in isolated rat resistance arteries showed that low norepinephrine concentrations induced SR-dependent unsynchronized waves of Ca(2+) in the vascular smooth muscle. In the presence of the endothelium, the waves converted to global synchronized oscillations of [Ca(2+)](i) after some time, and vasomotion appeared. Synchronization was also seen in the absence of endothelium if 8-bromo-cGMP was added to the bath. Using the patch-clamp technique and microelectrodes, we showed that Ca(2+) release can activate an inward current in isolated smooth muscle cells from the arteries and cause depolarization. These electrophysiological effects of Ca(2+) release were cGMP dependent, which is consistent with the possibility that they are important for the cGMP-dependent synchronization. Further support for the model is the observation that a short-lasting current pulse can initiate vasomotion in an unsynchronized artery as expected from the model.


Subject(s)
Models, Cardiovascular , Vasoconstriction/physiology , Vasomotor System/physiology , Animals , Biological Clocks/drug effects , Biological Clocks/physiology , Caffeine/pharmacology , Calcium Signaling , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Fluorescent Dyes , In Vitro Techniques , Isometric Contraction/drug effects , Isometric Contraction/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Microelectrodes , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Norepinephrine/pharmacology , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Rats , Rats, Wistar , Vascular Resistance/physiology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasomotor System/drug effects
11.
Environ Med ; 43(1): 1-9, 1999 Dec.
Article in English | MEDLINE | ID: mdl-12227374

ABSTRACT

Adrenoreactivity of rat hindlimb vessels was studied in experiments with constant-pressure saline perfusion. An original mathematical model was applied to evaluate the mechanism of changes in vascular tone regulation. A 3-week suspension resulted in decreased responses to sympathetic nerve stimulation, as well as to exogenous noradrenaline, the latter effect being registered when the pressure level was close to normal. Mathematical simulation indicated that long-term suspension induces both structural and functional changes in the vascular bed of the hind limbs, one of which is a disorder in the myogenic mechanisms of vascular tone regulation. In suspended rats, suppression of the myogenic response can be one of the reasons for decreased vessel reactivity to constrictor stimuli and, consequently, for disturbances in blood flow regulation in skeletal muscles of the hind limbs.


Subject(s)
Femoral Artery/drug effects , Hindlimb Suspension , Models, Biological , Norepinephrine/pharmacology , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology , Animals , Electric Stimulation , Femoral Artery/physiology , Femoral Artery/physiopathology , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Muscle, Smooth, Vascular/physiopathology , Perfusion , Rats , Rats, Wistar , Sodium Chloride , Vasoconstriction/drug effects , Vasomotor System/drug effects , Vasomotor System/physiology , Vasomotor System/physiopathology , Weightlessness Simulation
SELECTION OF CITATIONS
SEARCH DETAIL
...