Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Exp Physiol ; 109(5): 791-803, 2024 May.
Article in English | MEDLINE | ID: mdl-38460127

ABSTRACT

The mechanisms behind renal vasodilatation elicited by stimulation of ß-adrenergic receptors are not clarified. As several classes of K channels are potentially activated, we tested the hypothesis that KV7 and BKCa channels contribute to the decreased renal vascular tone in vivo and in vitro. Changes in renal blood flow (RBF) during ß-adrenergic stimulation were measured in anaesthetized rats using an ultrasonic flow probe. The isometric tension of segmental arteries from normo- and hypertensive rats and segmental arteries from wild-type mice and mice lacking functional KV7.1 channels was examined in a wire-myograph. The ß-adrenergic agonist isoprenaline increased RBF significantly in vivo. Neither activation nor inhibition of KV7 and BKCa channels affected the ß-adrenergic RBF response. In segmental arteries from normo- and hypertensive rats, inhibition of KV7 channels significantly decreased the ß-adrenergic vasorelaxation. However, inhibiting BKCa channels was equally effective in reducing the ß-adrenergic vasorelaxation. The ß-adrenergic vasorelaxation was not different between segmental arteries from wild-type mice and mice lacking KV7.1 channels. As opposed to rats, inhibition of KV7 channels did not affect the murine ß-adrenergic vasorelaxation. Although inhibition and activation of KV7 channels or BKCa channels significantly changed baseline RBF in vivo, none of the treatments affected ß-adrenergic vasodilatation. In isolated segmental arteries, however, inhibition of KV7 and BKCa channels significantly reduced the ß-adrenergic vasorelaxation, indicating that the regulation of RBF in vivo is driven by several actors in order to maintain an adequate RBF. Our data illustrates the challenge in extrapolating results from in vitro to in vivo conditions.


Subject(s)
Kidney , Vasodilation , Animals , Vasodilation/drug effects , Vasodilation/physiology , Male , Rats , Mice , Kidney/metabolism , Kidney/blood supply , KCNQ1 Potassium Channel/metabolism , Isoproterenol/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Adrenergic beta-Agonists/pharmacology , Mice, Knockout , Receptors, Adrenergic, beta/metabolism , Renal Circulation/drug effects , Renal Circulation/physiology , Mice, Inbred C57BL , Rats, Wistar , Hypertension/physiopathology , Hypertension/metabolism
2.
Microvasc Res ; 141: 104333, 2022 05.
Article in English | MEDLINE | ID: mdl-35151723

ABSTRACT

The mechanisms behind development of diet-induced hypertension remain unclear. The kidneys play a paramount role in blood volume and blood pressure regulation. Increases in renal vascular resistance lead to increased mean arterial blood pressure (MAP) due to reduced glomerular filtration rate and Na+ excretion. Renal vascular resistance may be increased by several factors, e.g. sympathetic output, increased activity in the renin-angiotensin system or endothelial dysfunction. We examined if a 14-week diet rich in fat, fructose or both led to increased renal vascular resistance and blood pressure. Sixty male Sprague-Dawley rats received normal chow (Control), high-fat chow (High Fat), high-fructose in drinking water (High Fructose), or a combination of high-fat and high-fructose diet (High Fat + Fruc) for 14 weeks from age 4-weeks. Measurements included body weight (BW), telemetry blood pressures, renal blood flow in anesthetized rats, plasma concentrations of atrial natriuretic peptide and glucose, as well as vessel myography in renal segmental arteries. Body weight increased in both groups receiving high fat, whereas MAP increased only in the High Fat + Fruc group. Renal blood flow did not differ between groups showing that renal vascular resistance was not increased by the diets. After inhibiting nitric oxide and prostacyclin production, renal blood flow reductions to Angiotensin II infusions were exaggerated in the groups receiving high fructose. MAP correlated positively with heart rate in all rats tested. Our data suggest that diet-induced hypertension is not caused by an increase in renal vascular resistance. The pathophysiological mechanisms may include altered signaling in the renin-angiotensin system and increases in central sympathetic output in combination with reduced baroreceptor sensitivity leading to increased renal vasoconstrictor responses.


Subject(s)
Angiotensin II , Hypertension , Angiotensin II/pharmacology , Animals , Blood Pressure , Body Weight , Diet , Fructose/adverse effects , Hypertension/chemically induced , Kidney , Male , Rats , Rats, Sprague-Dawley , Vasoconstrictor Agents/pharmacology
3.
Physiol Rep ; 7(15): e14168, 2019 08.
Article in English | MEDLINE | ID: mdl-31368238

ABSTRACT

Endothelium derived signaling mechanisms play an important role in regulating vascular tone and endothelial dysfunction is often found in hypertension. Endothelium-derived hyperpolarization (EDH) plays a significant role in smaller renal arteries and arterioles, but its significance in vivo in hypertension is unresolved. The aim of this study was to characterize the EDH-induced renal vasodilation in normotensive and hypertensive rats during acute intrarenal infusion of ACh. Our hypothesis was that the increased renal vascular resistance (RVR) found early in hypertension would significantly correlate with reduced EDH-induced vasodilation. In isoflurane-anesthetized 12-week-old normo- and hypertensive rats blood pressure and renal blood flow (RBF) was measured continuously. RBF responses to acute intrarenal ACh infusions were measured before and after inhibition of NO and prostacyclin. Additionally, RVR was decreased or increased using inhibition or activation of adrenergic receptors or by use of papaverine and angiotensin II. Intrarenal infusion of ACh elicited a larger increase in RBF in hypertensive rats compared to normotensive rats suggesting that endothelial dysfunction is not present in 12-week-old hypertensive rats. The EDH-induced renal vasodilation (after inhibition of NO and prostacyclin) was similar between normo- and hypertensive rats. Reducing RVR by inhibition of α1 -adrenergic receptors significantly increased the renal EDH response in hypertensive rats, but a similar increase was found after activating α-adrenergic receptors using norepinephrine. The results show that renal EDH is present and functional in 12-week-old normo- and hypertensive rats. Interestingly, both activation and inactivation of α1 -adrenergic receptors elicited an increase in the renal EDH-induced vasodilation.


Subject(s)
Endothelium, Vascular/drug effects , Hypertension/physiopathology , Renal Circulation/drug effects , Vasodilator Agents/pharmacology , Acetylcholine/pharmacology , Animals , Blood Pressure/drug effects , Hypertension/drug therapy , Male , Rats, Sprague-Dawley , Signal Transduction/drug effects , Vasodilation/drug effects
4.
Physiol Rep ; 6(17): e13863, 2018 09.
Article in English | MEDLINE | ID: mdl-30198176

ABSTRACT

The myogenic response (MR) and myogenic tone (MT) in resistance vessels is crucial for maintaining peripheral vascular resistance and blood flow autoregulation. Development of MT involves G protein-coupled receptors, and may be affected by aging. AIMS: (1) to estimate the mesenteric blood flow in myogenically active small mesenteric arteries; (2) to investigate the signaling from Gαq/11 and/or Gα12 activation to MT development; (3) to investigate the role of Rho-kinase 2 and aging on MT in mesenteric resistance arteries. METHODS: we used pressure myography, quantitative real-time PCR, and immunolocalization to study small (<200 µm) mesenteric arteries (SMA) from young, mature adult, and middle aged mice. RESULTS: Poiseuille flow calculations indicated autoregulation of blood flow at 60-120 mm Hg arterial pressure. Gαq/11 and Gα12 were abundantly expressed at the mRNA and protein levels in SMA. The Gαq/11 inhibitor YM-254890 suppressed MT development, and the Phosholipase C inhibitors U73122 and ET-18-OCH3 robustly inhibited it. We found an age-dependent increase in ROCK2 mRNA expression, and in basal MT. The specific ROCK2 inhibitor KD025 robustly inhibited MT in SMAs in all mice with an age-dependent variation in KD025 sensitivity. The inhibitory effect of KD025 was not prevented by the L-type Ca2+ channel activator BayK 8644. KD025 reversibly inhibited MT and endothelin-1 vasoconstriction in small pial arteries from Göttingen minipigs. CONCLUSIONS: MT development in SMAs occurs through a Gαq/11 /PLC/Ca2+ -dependent pathway, and is maintained via ROCK2-mediated Ca2+ sensitization. Increased MT at mature adulthood can be explained by increased ROCK2 expression/activity.


Subject(s)
Aging/physiology , GTP-Binding Protein alpha Subunits/metabolism , Mesenteric Arteries/metabolism , Muscle, Smooth, Vascular/physiology , Signal Transduction , rho-Associated Kinases/metabolism , Aging/metabolism , Animals , Calcium Channels, L-Type/metabolism , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , GTP-Binding Protein alpha Subunits/genetics , Male , Mesenteric Arteries/growth & development , Mesenteric Arteries/physiology , Mice , Mice, Inbred C57BL , Muscle Tonus , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/metabolism , Swine , Swine, Miniature , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics
5.
Clin Sci (Lond) ; 132(4): 461-474, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29436481

ABSTRACT

Abdominal obesity and/or a high intake of fructose may cause hypertension. K+ channels, Na/K-ATPase, and voltage-gated Ca2+ channels are crucial determinants of resistance artery tone and thus the control of blood pressure. Limited information is available on the role of K+ transporters in long-term diet-induced hypertension in rats. We hypothesized that a 28-week diet rich in fat, fructose, or both, will lead to changes in K+ transporter expression and function, which is associated with increased blood pressure and decreased arterial function. Male Sprague-Dawley (SD) rats received a diet containing normal chow (Control), high-fat chow (High Fat), high-fructose in drinking water (High Fructose), or a combination of high-fat and high-fructose diet (High Fat/Fruc) for 28 weeks from the age of 4 weeks. Measurements included body weight (BW), systolic blood pressure (SBP), mRNA expression of vascular K+ transporters, and vessel myography in small mesenteric arteries (SMAs). BW was increased in the High Fat and High Fat/Fruc groups, and SBP was increased in the High Fat/Fruc group. mRNA expression of small conductance calcium-activated K+ channel (SKCa), intermediate conductance calcium-activated K+ (IKCa), and Kir2.1 inward rectifier K+ channels were reduced in the High Fat/Fruc group. Reduced endothelium-derived hyperpolarization (EDH)-type relaxation to acetylcholine (ACh) was seen in the High Fat and High Fat/Fruc groups. Ba2+-sensitive dilatation to extracellular K+ was impaired in all the experimental diet groups. In conclusion, reduced expression and function of SKCa, IKCa, and Kir2.1 channels are associated with elevated blood pressure in rats fed a long-term High Fat/Fruc. Rats fed a 28-week High Fat/Fruc provide a relevant model of diet-induced hypertension.


Subject(s)
Acetylcholine/pharmacology , Diet , Hypertension/etiology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Animals , Calcium/metabolism , Endothelium, Vascular/metabolism , Male , Rats, Sprague-Dawley , Time
6.
Channels (Austin) ; 11(3): 183-195, 2017 May 04.
Article in English | MEDLINE | ID: mdl-28055302

ABSTRACT

L-type voltage gated Ca2+ channels are considered to be the primary source of calcium influx during the myogenic response. However, many vascular beds also express T-type voltage gated Ca2+ channels. Recent studies suggest that these channels may also play a role in autoregulation. At low pressures (40-80 mmHg) T-type channels affect myogenic responses in cerebral and mesenteric vascular beds. T-type channels also seem to be involved in skeletal muscle autoregulation. This review discusses the expression and role of T-type voltage gated Ca2+ channels in the autoregulation of several different vascular beds. Lack of specific pharmacological inhibitors has been a huge challenge in the field. Now the research has been strengthened by genetically modified models such as mice lacking expression of T-type voltage gated Ca2+ channels (CaV3.1 and CaV3.2). Hopefully, these new tools will help further elucidate the role of voltage gated T-type Ca2+ channels in autoregulation and vascular function.


Subject(s)
Calcium Channels, T-Type/metabolism , Homeostasis , Regional Blood Flow , Animals , Humans , Membrane Potentials , Muscle Contraction
7.
Am J Physiol Renal Physiol ; 312(3): F543-F548, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27881397

ABSTRACT

Intrarenal drug infusion plays an important role in renal experimental research. Laminar flow of the blood can cause streaming and inhomogeneous intrarenal distribution of infused drugs. We suggest a simple method to achieve a homogeneous intravascular distribution of drugs infused into the renal artery of anesthetized rats. The method employs a multiple sidehole catheter inserted into the renal artery, which enables an efficient drug mixing with the arterial blood. To verify the efficiency of this method, we use laser speckle imaging and renal artery flowmetry. The results show that, compared with the conventional single-hole catheter, the multiple sidehole catheter provides a more uniform drug distribution and a homogenous vascular response on the surface of the kidney.


Subject(s)
Angiotensin II/administration & dosage , Catheterization, Peripheral/methods , Kidney/blood supply , Renal Artery/drug effects , Renal Circulation/drug effects , Vasoconstriction/drug effects , Vasoconstrictor Agents/administration & dosage , Angiotensin II/blood , Animals , Blood Flow Velocity , Catheterization, Peripheral/instrumentation , Equipment Design , Infusions, Intra-Arterial , Laser-Doppler Flowmetry , Male , Models, Cardiovascular , Perfusion Imaging/methods , Rats, Sprague-Dawley , Renal Artery/physiology , Time Factors , Vascular Access Devices , Vasoconstrictor Agents/blood
8.
Pflugers Arch ; 468(7): 1139-1149, 2016 07.
Article in English | MEDLINE | ID: mdl-26965146

ABSTRACT

We investigated the mechanisms behind the endothelial-derived hyperpolarization (EDH)-induced renal vasodilation in vivo and in vitro in rats. We assessed the role of Ca(2+)-activated K(+) channels and whether K(+) released from the endothelial cells activates inward rectifier K(+) (Kir) channels and/or the Na(+)/K(+)-ATPase. Also, involvement of renal myoendothelial gap junctions was evaluated in vitro. Isometric tension in rat renal interlobar arteries was measured using a wire myograph. Renal blood flow was measured in isoflurane anesthetized rats. The EDH response was defined as the ACh-induced vasodilation assessed after inhibition of nitric oxide synthase and cyclooxygenase using L-NAME and indomethacin, respectively. After inhibition of small conductance Ca(2+)-activated K(+) channels (SKCa) and intermediate conductance Ca(2+)-activated K(+) channels (IKCa) (by apamin and TRAM-34, respectively), the EDH response in vitro was strongly attenuated whereas the EDH response in vivo was not significantly reduced. Inhibition of Kir channels and Na(+)/K(+)-ATPases (by ouabain and Ba(2+), respectively) significantly attenuated renal vasorelaxation in vitro but did not affect the response in vivo. Inhibition of gap junctions in vitro using carbenoxolone or 18α-glycyrrhetinic acid significantly reduced the endothelial-derived hyperpolarization-induced vasorelaxation. We conclude that SKCa and IKCa channels are important for EDH-induced renal vasorelaxation in vitro. Activation of Kir channels and Na(+)/K(+)-ATPases plays a significant role in the renal vascular EDH response in vitro but not in vivo. The renal EDH response in vivo is complex and may consist of several overlapping mechanisms some of which remain obscure.


Subject(s)
Endothelium, Vascular/metabolism , Potassium Channels, Calcium-Activated/metabolism , Vasodilation/physiology , Acetylcholine/pharmacology , Animals , Carbenoxolone/pharmacology , Endothelium, Vascular/drug effects , Gap Junctions/drug effects , Gap Junctions/metabolism , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , Kidney/drug effects , Kidney/metabolism , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Renal Circulation/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Vasodilation/drug effects
9.
J Hypertens ; 34(3): 513-23; discussion 523, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26820478

ABSTRACT

OBJECTIVE: Despite its high prevalence among patients suffering myocardial infarction, the significance of left ventricle hypertrophy for infarct size is not known. We asked whether infarct size might be increased by this condition, and whether any such increase might be associated with an increased mitochondrial damage following coronary occlusion. METHODS: Occlusion of the left descending artery in isolated, perfused hearts of SHR-SP (spontaneously hypertensive rat stroke-prone) (left ventricular hypertrophy) or Wistar-Kyoto (WKY) (control) rats was used, followed by reperfusion with or without exendin-4 (Exe-4), a glucagon-like peptide-1 receptor agonist. Infarct size relative to area-at-risk was determined. Separately, mitochondria were isolated after global ischemia. Activities of complexes III and IV and amounts of selected complex subunits and cytochromes a, b, c, and c1 were determined. RESULTS: Infarct size (ischemia 35  min and 120  min reperfusion) was 65.8% (±3.3%) and 37.1% (±3.4%) in the SHR-SP and WKY hearts, respectively (P < 0.05). Exe-4 significantly decreased infarct size and hypercontracture in WKY, but not in SHR-SP, hearts. After ischemia 15  min in SHR-SP hearts, Exe-4 reduced the infarct (26.6%, ±3.8% to 9.3% ± 1.5%; P < 0.05). Mitochondria from postischemic SHR-SP hearts showed a reduction of complex III (368.1 ± 37.5 to 175.8 ± 23.0  nmoles/min × mg; P < 0.05) and complex IV (14.4 ± 0.22 to 5.8 ± 0.8 1/s × mg; P < 0.05) activities and decreased amounts of cytochromes a, b, and c. CONCLUSION: Hearts from hypertensive (SHR-SP) rats with left ventricle hypertrophy appeared more vulnerable to ischemia-reperfusion injury, as supported by a more profound infarct development and an earlier loss of postconditioning by Exe-4. Mitochondrial complexes III and IV were identified among possible loci of this increased, hypertrophy-associated vulnerability.


Subject(s)
Hypertension/complications , Hypertrophy, Left Ventricular/complications , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Animals , Cytochromes/drug effects , Cytochromes/metabolism , Electron Transport Complex III/drug effects , Electron Transport Complex III/metabolism , Electron Transport Complex IV/drug effects , Electron Transport Complex IV/metabolism , Exenatide , Heart/drug effects , Incretins/pharmacology , Male , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Peptides/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Severity of Illness Index , Venoms/pharmacology
10.
Pflugers Arch ; 468(4): 541-50, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26658945

ABSTRACT

Renal autoregulation protects glomerular capillaries against increases in renal perfusion pressure (RPP). In the mesentery, both L- and T-type calcium channels are involved in autoregulation. L-type calcium channels participate in renal autoregulation, but the role of T-type channels is not fully elucidated due to lack of selective pharmacological inhibitors. The role of T- and L-type calcium channels in the response to acute increases in RPP in T-type channel knockout mice (CaV3.1) and normo- and hypertensive rats was examined. Changes in afferent arteriolar diameter in the kidneys from wild-type and CaV3.1 knockout mice were assessed. Autoregulation of renal blood flow was examined during acute increases in RPP in normo- and hypertensive rats under pharmacological blockade of T- and L-type calcium channels using mibefradil (0.1 µM) and nifedipine (1 µM). In contrast to the results from previous pharmacological studies, genetic deletion of T-type channels CaV3.1 did not affect renal autoregulation. Pharmacological blockade of T-type channels using concentrations of mibefradil which specifically blocks T-type channels also had no effect in wild-type or knockout mice. Blockade of L-type channels significantly attenuated renal autoregulation in both strains. These findings are supported by in vivo studies where blockade of T-type channels had no effect on changes in the renal vascular resistance after acute increases in RPP in normo- and hypertensive rats. These findings show that genetic deletion of T-type channels CaV3.1 or treatment with low concentrations of mibefradil does not affect renal autoregulation. Thus, T-type calcium channels are not involved in renal autoregulation in response to acute increases in RPP.


Subject(s)
Calcium Channels, T-Type/metabolism , Homeostasis , Kidney/metabolism , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/genetics , Gene Deletion , Kidney/blood supply , Kidney/physiology , Mibefradil/pharmacology , Mice , Mice, Inbred C57BL , Renal Circulation
11.
Am J Physiol Renal Physiol ; 303(6): F855-63, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22811484

ABSTRACT

Connexins in renal arterioles affect autoregulation of arteriolar tonus and renal blood flow and are believed to be involved in the transmission of the tubuloglomerular feedback (TGF) response across the cells of the juxtaglomerular apparatus. Connexin40 (Cx40) also plays a significant role in the regulation of renin secretion. We investigated the effect of deleting the Cx40 gene on autoregulation of afferent arteriolar diameter in response to acute changes in renal perfusion pressure. The experiments were performed using the isolated blood perfused juxtamedullary nephron preparation in kidneys obtained from wild-type or Cx40 knockout mice. Renal perfusion pressure was increased in steps from 75 to 155 mmHg, and the response in afferent arteriolar diameter was measured. Hereafter, a papillectomy was performed to inhibit TGF, and the pressure steps were repeated. Conduction of intercellular Ca(2+) changes in response to local electrical stimulation was examined in isolated interlobular arteries and afferent arterioles from wild-type or Cx40 knockout mice. Cx40 knockout mice had an impaired autoregulatory response to acute changes in renal perfusion pressure compared with wild-type mice. Inhibition of TGF by papillectomy significantly reduced autoregulation of afferent arteriolar diameter in wild-type mice. In Cx40 knockout mice, papillectomy did not affect the autoregulatory response, indicating that these mice have no functional TGF. Also, Cx40 knockout mice showed no conduction of intercellular Ca(2+) changes in response to local electrical stimulation of interlobular arteries, whereas the Ca(2+) response to norepinephrine was unaffected. These results suggest that Cx40 plays a significant role in the renal autoregulatory response of preglomerular resistance vessels.


Subject(s)
Arterioles/physiology , Connexins/physiology , Kidney/physiology , Renal Circulation/physiology , Animals , Arterioles/drug effects , Calcium/physiology , Cells, Cultured , Connexins/genetics , Electric Stimulation , Female , Homeostasis/drug effects , Kidney/blood supply , Kidney/drug effects , Male , Mice , Mice, Knockout , Norepinephrine/pharmacology , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Transforming Growth Factors/physiology , Vasoconstrictor Agents/pharmacology , Gap Junction alpha-5 Protein
12.
Am J Physiol Renal Physiol ; 302(5): F505-18, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22169005

ABSTRACT

K(+) conductance is a major determinant of membrane potential (V(m)) in vascular smooth muscle (VSMC) and endothelial cells (EC). The vascular tone is controlled by V(m) through the action of voltage-operated Ca(2+) channels (VOCC) in VSMC. Increased K(+) conductance leads to hyperpolarization and vasodilation, while inactivation of K(+) channels causes depolarization and vasoconstriction. K(+) channels in EC indirectly participate in the control of vascular tone by several mechanisms, e.g., release of nitric oxide and endothelium-derived hyperpolarizing factor. In the kidney, a change in the activity of one or more classes of K(+) channels will lead to a change in hemodynamic resistance and therefore of renal blood flow and glomerular filtration pressure. Through these effects, the activity of renal vascular K(+) channels influences renal salt and water excretion, fluid homeostasis, and ultimately blood pressure. Four main classes of K(+) channels [calcium activated (K(Ca)), inward rectifier (K(ir)), voltage activated (K(V)), and ATP sensitive (K(ATP))] are found in the renal vasculature. Several in vitro experiments have suggested a role for individual classes of K(+) channels in the regulation of renal vascular function. Results from in vivo experiments are sparse. We discuss the role of the different classes of renal vascular K(+) channels and their possible role in the integrated function of the renal microvasculature. Since several pathological conditions, among them hypertension, are associated with alterations in K(+) channel function, the role of renal vascular K(+) channels in the control of salt and water excretion deserves attention.


Subject(s)
Endothelium, Vascular/physiology , Hemodynamics/physiology , Kidney/blood supply , Potassium Channels/physiology , Animals , Humans , Hypertension, Renovascular/physiopathology , Kidney/physiopathology
13.
Pflugers Arch ; 462(5): 655-67, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21874333

ABSTRACT

Inhibition of K(+) channels might mediate renal vasoconstriction. As inhibition of a single type of K(+) channel caused minor or no renal vasoconstriction in vivo in rats, we hypothesized that several classes of K(+) channels must be blocked to elicit renal vasoconstriction. We measured renal blood flow (RBF) in vivo in anesthetized Sprague-Dawley rats. Test agents were infused directly into the renal artery to avoid systemic effects. Inhibition of BK(Ca) and K(ir) channels (with TEA and Ba(2+), respectively) caused small and transient reductions in RBF (to 93 ± 2% and 95 ± 1% of baseline, respectively). K(ATP), SK(Ca) or K(v) channel blockade (with glibenclamide, apamin and 4-aminopyridine, respectively) was without effect. However, a cocktail of all blockers caused a massive reduction of RBF (to 15 ± 10% of baseline). Nifedipine and mibefradil abolished and reduced, respectively, this RBF reduction. The effect of the cocktail of K(+) channel blockers was confirmed in mice using the isolated blood-perfused juxtamedullary nephron preparation. A cocktail of K(+) channel openers (K(+), NS309, NS1619 and pinacidil) had only a minor effect on baseline RBF in vivo in rats, but reduced the vasoconstriction induced by bolus injections of norepinephrine or angiotensin II (by 33 ± 5% and 60 ± 5%, respectively). Our results indicate that closure of numerous types of K(+) channels could participate in the mediation of agonist-induced renal vasoconstriction. Our results also suggest that renal vasoconstriction elicited by K(+) channel blockade is mediated by nifedipine-sensitive Ca(2+) channels and partly by mibefradil-sensitive Ca(2+) channels.


Subject(s)
Calcium Channels/physiology , Renal Circulation/physiology , Vascular Resistance/drug effects , Animals , Arterioles/drug effects , Benzimidazoles/pharmacology , Calcium Channels/drug effects , Large-Conductance Calcium-Activated Potassium Channels/drug effects , Male , Membrane Potentials/drug effects , Mibefradil/pharmacology , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Nifedipine/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels/agonists , Potassium Channels, Inwardly Rectifying/drug effects , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Vasoconstriction/drug effects
14.
Circ Res ; 104(12): 1399-409, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19443836

ABSTRACT

TRPC6 is a non-voltage-gated Ca(2+) entry/depolarization channel associated with vascular tone regulation and remodeling. Expressed TRPC6 channel responds to both neurohormonal and mechanical stimuli, the mechanism for which remains controversial. In this study, we examined the possible interactions of receptor and mechanical stimulations in activating this channel using the patch clamp technique. In HEK293 cells expressing TRPC6, application of mechanical stimuli (hypotonicity, shear, 2,4,6-trinitrophenol) caused, albeit not effective by themselves, a prominent potentiation of cationic currents (I(TRPC6)) induced by a muscarinic receptor agonist carbachol. This effect was insensitive to a tarantula toxin GsMTx-4 (5 mumol/L). A similar extent of mechanical potentiation was observed after activation of I(TRPC6) by GTPgammaS or a diacylglycerol analog 1-oleoyl-2-acetyl-sn-glycerol (OAG). Single TRPC6 channel activity evoked by carbachol was also enhanced by a negative pressure added in the patch pipette. Mechanical potentiation of carbachol- or OAG-induced I(TRPC6) was abolished by small interfering RNA knockdown of cytosolic phospholipase A(2) or pharmacological inhibition of omega-hydroxylation of arachidonic acid into 20-HETE (20-hydroxyeicosatetraenoic acid). Conversely, direct application of 20-HETE enhanced both OAG-induced macroscopic and single channel TRPC6 currents. Essentially the same results were obtained for TRPC6-like cation channel in A7r5 myocytes, where its activation by noradrenaline or Arg8 vasopressin was greatly enhanced by mechanical stimuli via 20-HETE production. Furthermore, myogenic response of pressurized mesenteric artery was significantly enhanced by weak receptor stimulation dependently on 20-HETE production. These results collectively suggest that simultaneous operation of receptor and mechanical stimulations may synergistically amplify transmembrane Ca(2+) mobilization through TRPC6 activation, thereby enhancing the vascular tone via phospholipase C/diacylglycerol and phospholipase A(2)/omega-hydroxylase/20-HETE pathways.


Subject(s)
Cytochrome P-450 CYP4A/metabolism , Hydroxyeicosatetraenoic Acids/pharmacology , Mechanotransduction, Cellular/drug effects , Muscle Cells/metabolism , Phospholipases A2/metabolism , TRPC Cation Channels/metabolism , Type C Phospholipases/metabolism , Animals , Carbachol/pharmacology , Cell Line , Cholinergic Agonists/pharmacology , Cytochrome P-450 CYP4A/genetics , Dose-Response Relationship, Drug , Humans , Hydroxyeicosatetraenoic Acids/agonists , Hydroxylation/drug effects , Intercellular Signaling Peptides and Proteins , Male , Peptides/pharmacology , Phospholipases A2/genetics , Rats , Rats, Sprague-Dawley , Receptors, Muscarinic/metabolism , Spider Venoms/pharmacology , TRPC Cation Channels/agonists , TRPC Cation Channels/genetics , TRPC6 Cation Channel , Type C Phospholipases/genetics
15.
Am J Physiol Regul Integr Comp Physiol ; 295(3): R840-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18614765

ABSTRACT

Vascular conducted responses are believed to play a central role in controlling the microcirculatory blood flow. The responses most likely spread through gap junctions in the vascular wall. At present, four different connexins (Cx) have been detected in the renal vasculature, but their role in transmission of conducted vasoconstrictor signals in the preglomerular arterioles is unknown. Connexin mimetic peptides were previously reported to target and inhibit specific connexins. We, therefore, investigated whether conducted vasoconstriction in isolated renal arterioles could be blocked by the use of mimetic peptides directed against one or more connexins. Preglomerular resistance vessels were microdissected from kidneys of Sprague-Dawley rats and loaded with fura 2. The vessels were stimulated locally by applying electrical current through a micropipette, and the conducted calcium response was measured 500 mum from the site of stimulation. Application of connexin mimetic peptides directed against Cx40, 37/43, 45, or a cocktail with equimolar amounts of each, did not inhibit the propagated response, whereas the nonselective gap junction uncoupler carbenoxolone completely abolished the propagated response. However, the connexin mimetic peptides were able to reduce dye coupling between rat aorta endothelial cells shown to express primarily Cx40. In conclusion, we did not observe any attenuating effects on conducted calcium responses in isolated rat interlobular arteries when exposed to connexin mimetic peptides directed against Cx40, 37/43, or 45. Further studies are needed to determine whether conducted vasoconstriction is mediated via previously undescribed pathways.


Subject(s)
Calcium/metabolism , Connexins/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Kidney Glomerulus/blood supply , Renal Circulation/physiology , Animals , Aorta/cytology , Arterioles/cytology , Arterioles/physiology , Cell Communication/drug effects , Cell Communication/physiology , Cells, Cultured , Connexin 43/genetics , Connexin 43/pharmacology , Connexins/genetics , Electric Stimulation , Endothelial Cells/cytology , HeLa Cells , Humans , Molecular Mimicry , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Transfection , Vasoconstriction/drug effects , Vasoconstriction/physiology , Gap Junction alpha-5 Protein , Gap Junction alpha-4 Protein
16.
Am J Physiol Regul Integr Comp Physiol ; 292(1): R345-53, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16973937

ABSTRACT

We investigated the role of large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels for the basal renal vascular tone in vivo. Furthermore, the possible buffering by BK(Ca) of the vasoconstriction elicited by angiotensin II (ANG II) or norepinephrine (NE) was investigated. The possible activation of renal vascular BK(Ca) channels by cAMP was investigated by infusing forskolin. Renal blood flow (RBF) was measured in vivo using electromagnetic flowmetry or ultrasonic Doppler. Renal preinfusion of tetraethylammonium (TEA; 3.0 mumol/min) caused a small reduction of baseline RBF, but iberiotoxin (IBT; 0.3 nmol/min) did not have any effect. Renal injection of ANG II (1-4 ng) or NE (10-40 ng) produced a transient decrease in RBF. These responses were not affected by preinfusion of TEA or IBT. Renal infusion of the BK(Ca) opener NS-1619 (90.0 nmol/min) did not affect basal RBF or the response to NE, but it attenuated the response to ANG II. Coadministration of NS-1619 with TEA or IBT abolished this effect. Forskolin caused renal vasodilation that was not inhibited by IBT. The presence of BK(Ca) channels in the preglomerular vessels was confirmed by immunohistochemistry. Despite their presence, there is no indication for a major role for BK(Ca) channels in the control of basal renal tone in vivo. Furthermore, BK(Ca) channels do not have a buffering effect on the rat renal vascular responses to ANG II and NE. The fact that NS-1619 attenuates the ANG II response indicates that the renal vascular BK(Ca) channels can be activated under certain conditions.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/agonists , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle, Smooth, Vascular/physiology , Renal Circulation/physiology , Adenylyl Cyclase Inhibitors , Angiotensin II/pharmacology , Animals , Benzimidazoles/pharmacology , Blood Pressure/drug effects , Colforsin/pharmacology , Electromagnetic Fields , Immunohistochemistry , Male , Muscle Tonus/physiology , Norepinephrine/pharmacology , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Rheology , Stimulation, Chemical , Tetraethylammonium/pharmacology , Vasoconstrictor Agents/pharmacology
17.
Am J Physiol Renal Physiol ; 289(6): F1227-34, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16033921

ABSTRACT

We investigated whether tempol, a superoxide dismutase mimetic, affected renal hemodynamics and arterial pressure in spontaneously hypertensive rats (SHR) and Sprague-Dawley (SD) rats. We also examined whether tempol affected exaggerated renal vasoconstrictor responses to ANG II in SHR. To test whether the effects of tempol were due to a restored NO system, we used the NOS inhibitor N(w)-nitro-L-arginine methyl ester (L-NAME). Renal blood flow (RBF) and mean arterial pressure (MAP) were measured in vivo by electromagnetic flowmetry and arterial catheterization in 10- to 12-wk-old anesthetized SHR and SD rats. Systolic arterial pressure (SAP) was measured in conscious rats using the tail cuff method. Tempol (1 mM) was given in the drinking water to SD (SD-T) and SHR (SHR-T) for 5-7 days for RBF measurements and for 15 days for SAP measurements. Age-matched SD (SD-C) and SHR (SHR-C) were used as controls. ANG II (1-4 ng) was administered as a bolus via a renal artery catheter. L-NAME was administered intravenously for 15-20 min. Renal vascular resistance (RVR) was elevated in SHR-C compared with SD-C. In SHR-T, baseline RVR was not different from SD-C and SD-T rats. Tempol had no effect on RVR in SD. L-NAME elevated RVR to the same extent in all four groups. Arterial pressure was not affected by tempol. The RVR responses to ANG II were higher in SHR-C than in the SD-C group. ANG II responses were not different between SHR-T and SD-T. Overall, tempol reduced the renovascular responses to ANG II in SHR. L-NAME elevated the effects of ANG II in SD-C rats but had no effect on the ANG II responses in the other groups. Thus L-NAME treatment did not influence tempol's effects on baseline RVR or ANG II responses. We conclude that in SHR, tempol has a significant renal vasodilator effect and that it normalizes the increased renovascular ANG II sensitivity. As the effects of L-NAME are not greater in SHR-T rats, it is not likely that the elevated renal resistance and ANG II sensitivity in SHR are due to reactive oxygen species-induced quenching of nitric oxide.


Subject(s)
Cyclic N-Oxides/pharmacology , Kidney/blood supply , Nitric Oxide/physiology , Renal Circulation/drug effects , Vascular Resistance/drug effects , Vasodilation/drug effects , Angiotensin II/pharmacology , Animals , Kidney/drug effects , Male , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Rats, Inbred SHR , Spin Labels
18.
Br J Pharmacol ; 142(4): 709-18, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15172957

ABSTRACT

1. In this study, intracellular Ca(2+) was measured as the Fura-2 ratio (R) of fluorescence excited at 340 and 380 nm (F(340)/F(380)) in nonpressurized rat mesenteric small arterioles ( (lumen diameter) 10-25 microm). 2. The response to depolarization using 75 mm KCl was an increase in R from a baseline of 0.96+/-0.01 ([Ca(2+)](i) approximately 74 nm) to 1.04+/-0.01 ( approximately 128 nm) (n=80). The response to 75 mm K(+) was reversibly abolished in Ca(2+)-free physiological saline solution, whereas phentolamine (10 microm) or tetrodotoxin (1 microm) had no effects. LaCl(3) (200 microm) inhibited 61+/-9% of the response. 3. A [K(+)]-response curve indicated that the Ca(2+) response was activated between 15 and 25 mm K(+). The data suggest that the Ca(2+) response was caused by the activation of voltage-dependent Ca(2+) channels. 4. Mibefradil use dependently inhibited the Ca(2+) response to 75 mm K(+) by 29+/-2% (100 nm), 73+/-7% (1 microm) or 89+/-7% (10 microm). Pimozide (500 nm) use dependently inhibited the Ca(2+) response by 85+/-1%. 5. Nifedipine (1 microm) inhibited the Ca(2+) response to 75 mm K(+) by 41+/-12%. The response was not inhibited by calciseptine (500 nm), omega-agatoxin IVA (100 nm), omega-conotoxin MVIIA (500 nm), or SNX-482 (100 nm). 6. Using reverse transcriptase-polymerase chain reaction, it was shown that neither Ca(V)2.1a (P-type) nor Ca(V)2.1b (Q-type) voltage-dependent Ca(2+) channels were expressed in mesenteric arterioles, whereas the Ca(V)3.1 (T-type) channel was expressed. Furthermore, no amplification products were detected when using specific primers for the beta(1b), beta(2), or beta(3) auxiliary subunits of high-voltage-activated Ca(2+) channels. 7. The results suggest that the voltage-dependent Ca(2+) channel activated by sustained depolarization in mesenteric arterioles does not classify as any of the high-voltage-activated channels (L-, P/Q-, N-, or R-type), but is likely to be a T-type channel. The possibility that the sustained Ca(2+) influx observed was the result of a T-type window current is discussed.


Subject(s)
Calcium Channels/drug effects , Calcium/metabolism , Membrane Potentials/physiology , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Mibefradil/pharmacology , Animals , Arterioles/anatomy & histology , Arterioles/drug effects , Arterioles/ultrastructure , Blotting, Southern/methods , Calcium/chemistry , Calcium Channels/physiology , Denmark , Elapid Venoms/pharmacology , Fluorescence , Fura-2/pharmacology , Gene Expression/drug effects , Gene Expression/physiology , Lanthanum/pharmacology , Male , Mesenteric Arteries/anatomy & histology , Muscle, Smooth, Vascular/anatomy & histology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Nifedipine/pharmacology , Phentolamine/pharmacology , Pimozide/pharmacology , Potassium Chloride/pharmacology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods , Sodium-Calcium Exchanger/metabolism , Solutions/chemistry , Spider Venoms/pharmacology , Tetrodotoxin/pharmacology , omega-Agatoxin IVA/pharmacology , omega-Conotoxins/pharmacology
19.
Am J Physiol Renal Physiol ; 286(5): F866-74, 2004 May.
Article in English | MEDLINE | ID: mdl-15075182

ABSTRACT

We used genistein (Gen) and tyrphostin 23 (Tyr-23) to evaluate the importance of tyrosine phosphorylation in norepinephrine (NE)-induced changes in intracellular free calcium concentration ([Ca(2+)](i)) in rat afferent arterioles. [Ca(2+)](i) was measured in microdissected arterioles using ratiometric photometry of fura 2 fluorescence. The control [Ca(2+)](i) response to NE (1 microM) consisted of a rapid initial peak followed by a plateau phase sustained above baseline. Pretreatment with the tyrosine kinase inhibitor Tyr-23 (50 microM, 10 min) caused a slow 40% increase in baseline [Ca(2+)](i). Tyr-23 attenuated peak and plateau responses to NE, both by approximately 70%. In the absence of extracellular Ca(2+) (0 Ca), Tyr-23 reduced the immediate [Ca(2+)](i) response to NE by approximately 60%, indicative of mobilization of internal stores, and abolished the plateau phase. In other arterioles, the [Ca(2+)](i) response to depolarization induced by KCl (50 mM) was not attenuated by Tyr-23, indicating no direct effect on L-type Ca(+) channels activated by depolarization. The Ca(2+) channel blocker nifedipine (1 microM) inhibited the NE response by approximately 50%; the effects of nifedipine and Tyr-23 were not additive. Nifedipine had no inhibitory effect after Tyr-23 pretreatment, indicating Tyr-23 inhibition of Ca(2+) entry. Another tyrosine kinase inhibitor, Gen (5 and 50 microM), did not affect baseline [Ca(2+)](i). High-dose Gen inhibited the peak and plateau response to NE by 87 and 75%, respectively; low-dose Gen attenuated both responses by approximately 20%. In 0 Ca, Gen (50 microM) abolished the immediate [Ca(2+)](i) mobilization response. Combined nifedipine and Gen (50 microM) inhibited the rapid NE response by approximately 90% in the presence of extracellular Ca(2+). Gen (50 microM) also inhibited by 60% the [Ca(2+)](i) response to 50 mM KCl, indicating a direct interaction with voltage-sensitive, L-type Ca(2+) entry channels. These results indicate that tyrosine phosphorylation is an important link in the chain of events leading to alpha-adrenoceptor-induced Ca(2+) recruitment (both entry and release) in afferent arteriolar smooth muscle cells. Furthermore, different blockers of tyrosine kinase appear to have different modes of action in renal microvessels.


Subject(s)
Calcium/metabolism , Kidney Glomerulus/blood supply , Norepinephrine/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Vasoconstrictor Agents/pharmacology , Animals , Arterioles/drug effects , Arterioles/enzymology , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Fura-2 , Genistein/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Rats , Rats, Inbred WKY , Renal Circulation/drug effects , Renal Circulation/physiology , Tyrphostins/pharmacology
20.
Am J Physiol Regul Integr Comp Physiol ; 286(5): R865-73, 2004 May.
Article in English | MEDLINE | ID: mdl-14715487

ABSTRACT

Previous experiments from our laboratory showed that longer-lasting reductions in renal perfusion pressure (RPP) are associated with a gradual decrease in renal blood flow (RBF) that can be abolished by clamping plasma ANG II concentration ([ANG II]). The aim of the present study was to investigate the mechanisms behind the RBF downregulation in halothane-anesthetized Sprague-Dawley rats during a 30-min reduction in RPP to 88 mmHg. During the 30 min of reduced RPP we also measured glomerular filtration rate (GFR), proximal tubular pressure (P(prox)), and proximal tubular flow rate (Q(LP)). Early distal tubular fluid conductivity was measured as an estimate of early distal [NaCl] ([NaCl](ED)), and changes in plasma renin concentration (PRC) over time were measured. During 30 min of reduced RPP, RBF decreased gradually from 6.5 +/- 0.3 to 6.0 +/- 0.3 ml/min after 5 min (NS) to 5.2 +/- 0.2 ml/min after 30 min (P < 0.05). This decrease occurred in parallel with a gradual increase in PRC from 38.2 +/- 11.0 x 10(-5) to 87.1 +/- 25.1 x 10(-5) Goldblatt units (GU)/ml after 5 min (P < 0.05) to 158.5 +/- 42.9 x 10(-5) GU/ml after 30 min (P < 0.01). GFR, P(prox), and [NaCl](ED) all decreased significantly after 5 min and remained low. Estimates of pre- and postglomerular resistances showed that the autoregulatory mechanisms initially dilated preglomerular vessels to maintain RBF and GFR. However, after 30 min of reduced RPP, both pre- and postglomerular resistance had increased. We conclude that the decrease in RBF over time is caused by increases in both pre- and postglomerular resistance due to rising plasma renin and ANG II concentrations.


Subject(s)
Angiotensin II/pharmacology , Kidney Glomerulus/blood supply , Renal Circulation/drug effects , Vascular Resistance/physiology , Vasoconstrictor Agents/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Down-Regulation/drug effects , Down-Regulation/physiology , Glomerular Filtration Rate/physiology , Homeostasis/physiology , Kidney Glomerulus/physiology , Kidney Tubules, Proximal/physiology , Male , Models, Biological , Nephrons/physiology , Potassium/blood , Rats , Rats, Sprague-Dawley , Renal Circulation/physiology , Sodium Chloride/blood , Water/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL