Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters










Publication year range
1.
Acta Physiol Scand ; 181(4): 421-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15283754

ABSTRACT

The cytosolic intracellular calcium concentration ([Ca(2+)](i)) is a major determining factor in the vascular smooth muscle tone. In the afferent arteriole it has been shown that agonists utilizing G-protein coupled receptors recruit Ca(2+) via release from intracellular stores and entry via pathways in the plasma membrane. The relative importances of entry vs. mobilization seem to differ between different agonists, species and preparations. The entry pathway might include different types of voltage sensitive Ca(2+) channels located in the plasmalemma such as dihydropyridine sensitive L-type channels, T-type channels and P/Q channels. A role for non-voltage sensitive entry pathways has also been suggested. The importance of voltage sensitive Ca(2+) channels in the control of the tone of the afferent arteriole (and thus in the control of renal function and whole body control of extracellular fluid volume and blood pressure) sheds light on the control of the membrane potential of afferent arteriolar smooth muscle cells. Thus, K(+) and Cl(-) channels are of importance in their role as major determinants of membrane potential. Some studies suggest a role for calcium-activated chloride (Cl(Ca)) channels in the renal vasoconstriction elicited by agonists. Other investigators have found evidence for several types of K(+) channels in the regulation of the afferent arteriolar tone. The available literature in this field regarding afferent arterioles is, however, relatively sparse and not conclusive. This review is an attempt to summarize the results obtained by others and ourselves in the field of agonist induced afferent arteriolar Ca(2+) recruitment, with special emphasis on the control of voltage sensitive Ca(2+) entry. Outline of the Manuscript: This manuscript is structured as follows: it begins with an introduction where the general role for [Ca(2+)](i) as a key factor in the regulation of the tone of vascular smooth muscles (VSMC) is detailed. In this section there is an emphasis is on observations that could be attributed to afferent arteriolar function. We then investigate the literature and describe our results regarding the relative roles for Ca(2+) entry and intracellular release in afferent arterioles in response to vasoactive agents, with the focus on noradrenalin (NA) and angiotensin II (Ang II). Finally, we examine the role of ion channels (i.e. K(+) and Cl(-) channels) for the membrane potential, and thus activation of voltage sensitive Ca(2+) channels.


Subject(s)
Calcium/physiology , Kidney/blood supply , Animals , Arterioles/physiology , Chloride Channels/physiology , Humans , Membrane Potentials/physiology , Muscle, Smooth, Vascular/physiology , Potassium Channels/physiology
2.
Am J Physiol Renal Physiol ; 281(2): F264-72, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11457717

ABSTRACT

This study provides new information about the relative importance of calcium mobilization and entry in the renal vascular response to adrenoceptor activation in afferent arterioles isolated from 7- to 8-wk-old Wistar-Kyoto (WKY) and spontaneously hypertensive rats (SHR). Intracellular free calcium concentration ([Ca(2+)](i)) was measured in microdissected arterioles utilizing ratiometric photometry of fura 2 fluorescence. There was no significant strain difference in baseline [Ca(2+)](i). Norepinephrine (NE; 10(-6) and 10(-7) M) elicited immediate, sustained increases in [Ca(2+)](i). The general temporal pattern of response to 10(-6) M NE consisted of an initial peak and a maintained plateau phase. The response to NE was partially blocked by nifedipine (10(-6) M) or 8-(N,N-diethylamino) octyl-3,4,5-trimetoxybenzoate (TMB-8; 10(-5) M). A calcium-free external solution abolished the sustained [Ca(2+)](i) plateau response to NE, with less influence on the peak response. In the absence of calcium entry, TMB-8 (10(-5) M) completely blocked the calcium response to NE in WKY but not SHR, suggesting strain differences in mobilization. A higher concentration of TMB-8 (10(-4) M), however, blocked all discernible mobilization in both strains. We conclude that there are differences in Ca(2+) handling in renal resistance vessels between young WKY and SHR with respect to mobilization stimulated by alpha-adrenoceptors. Afferent arterioles of young SHR appear to have a larger inositol-1,4,5-trisphosphate-sensitive pool or release from a site less accessible to TMB-8.


Subject(s)
Arterioles/metabolism , Calcium Signaling , Calcium/metabolism , Hypertension/metabolism , Kidney/blood supply , Norepinephrine/pharmacology , Adrenergic alpha-Agonists/pharmacology , Animals , Arterioles/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels/metabolism , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , In Vitro Techniques , Kidney/anatomy & histology , Nifedipine/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Vasoconstrictor Agents/pharmacology
3.
Am J Physiol Renal Physiol ; 281(1): F172-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11399658

ABSTRACT

We utilized [3H]prazosin saturation and competition radioligand binding studies to characterize the expression of alpha1-adrenoceptors in preglomerular vessels. mRNA for adrenoceptor subtypes was assayed using RT-PCR. The vessels were isolated using an iron oxide-sieving method. [3H]prazosin bound to a single class of binding sites (Kd 0.087 +/- 0.012 nM, Bmax 326 +/- 56 fmol/mg protein). Phentolamine displaced [3H]prazosin (0.2 nM) with a pK(i) of 8.37 +/- 0.09. Competition with the selective alpha1A-adrenoceptor antagonist 5-methylurapidil fit a two-site model (pK(i) 9.38 +/- 0.21 and 7.04 +/- 0.15); 59 +/- 3% of the sites were high-affinity, and 41 +/- 3% were low-affinity binding sites. Competition with the alpha1D-adrenoceptor antagonist 8-(2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl)-8-azaspiro[4.5]decane-7,9-dione dihydrochloride (BMY-7378) fit a one-site model with low affinity (pK(i) 6.83 +/- 0.03). The relative contents of alpha1A-, alpha1B-, and alpha1D-adrenoceptor mRNAs were 64 +/- 5, 25 +/- 5, and 11 +/- 1%, respectively. Thus there was a very good correlation between mRNA and receptor binding for the subtypes. These data indicate a predominance of the alpha1A-adrenoceptor subtype in rat renal resistance vessels, with smaller densities of alpha1B- and alpha1D-adrenoceptors.


Subject(s)
Arterioles/metabolism , Kidney/blood supply , Receptors, Adrenergic, alpha-1/analysis , Animals , Binding, Competitive , Male , RNA, Messenger/analysis , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/classification , Receptors, Adrenergic, alpha-1/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Vascular Resistance
4.
Am J Physiol Heart Circ Physiol ; 280(1): H327-33, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11123248

ABSTRACT

The kidney plays a central role in long-term regulation of arterial blood pressure and salt and water homeostasis. This is achieved in part by the local actions of paracrine and autacoid mediators such as the arachidonic acid-prostanoid system. The present study tested the role of specific PGE(2) E-prostanoid (EP) receptors in the regulation of renal hemodynamics and vascular reactivity to PGE(2). Specifically, we determined the extent to which the EP(2) and EP(3) receptor subtypes mediate the actions of PGE(2) on renal vascular tone. Renal blood flow (RBF) was measured by ultrasonic flowmetry, whereas vasoactive agents were injected directly into the renal artery of male mice. Studies were performed on two independent mouse lines lacking either EP(2) or EP(3) (-/-) receptors and the results were compared with wild-type controls (+/+). Our results do not support a unique role of the EP(2) receptor in regulating overall renal hemodynamics. Baseline renal hemodynamics in EP(2)-/- mice [RBF EP(2)-/-: 5.3 +/- 0.8 ml. min(-1). 100 g kidney wt(-1); renal vascular resistance (RVR) 19.7 +/- 3.6 mmHg. ml(-1). min. g kidney wt] did not differ statistically from control mice (RBF +/+: 4.0 +/- 0.5 ml. min(-1). 100 g kidney wt(-1); RVR +/+: 25.4 +/- 4.9 mmHg. ml(-1). min. 100 g kidney wt(-1)). This was also the case for the peak RBF increase after local PGE(2) (500 ng) injection into the renal artery (EP(2)-/-: 116 +/- 4 vs. +/+: 112 +/- 2% baseline RBF). In contrast, we found that the absence of EP(3) receptors in EP(3)-/- mice caused a significant increase (43%) in basal RBF (7.9 +/- 0.8 ml. min(-1). g kidney wt(-1), P < 0.05 vs. +/+) and a significant decrease (41%) in resting RVR (11.6 +/- 1.4 mmHg. ml(-1). min. g kidney wt(-1), P < 0.05 vs. +/+). Local administration of 500 ng of PGE(2) into the renal artery caused more pronounced renal vasodilation in EP(3)-/- mice (128 +/- 2% of basal RBF, P < 0.05 vs. +/+). We conclude that EP(3 )receptors mediate vasoconstriction in the kidney of male mice and its actions are tonically active in the basal state. Furthermore, EP(3) receptors are capable of buffering PGE(2)-mediated renal vasodilation.


Subject(s)
Dinoprostone/pharmacology , Receptors, Prostaglandin E/physiology , Renal Circulation/physiology , Animals , Dinoprostone/metabolism , Genotype , Kidney/blood supply , Kidney/diagnostic imaging , Male , Mice , Mice, Knockout , Microcirculation , Mutation , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP3 Subtype , Renal Circulation/drug effects , Ultrasonography
5.
Am J Physiol Renal Physiol ; 279(4): F755-64, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10997926

ABSTRACT

Vasodilator prostaglandin PGE(2) protects the kidney from excessive vasoconstriction during contraction of extracellular fluid volume and pathophysiological states. However, it is not yet clear which of the four known E-prostanoid (EP) receptors is localized to resistance vessels and mediates net vasodilation. In the present study, we assessed the presence, signal transduction, and actions of EP receptor subtypes in preglomerular arterioles of Sprague-Dawley rat kidneys. RNA encoding EP(1), an EP(1)-variant, and EP(4) receptors was identified by RT-PCR in freshly isolated preglomerular microvessels; cultured preglomerular vascular smooth muscle cells (VSMC) had EP(1)-variant and EP(4) RNA but lacked EP(1). EP(2) and EP(3) receptors were undetectable in both vascular preparations. In studies of cell signaling, stimulation of cAMP by various receptor agonists is consistent with primary actions of PGE(2) on the EP(4) receptor, with no inhibition of cAMP by EP(1) receptors. Studies of cytosolic calcium concentration in cultured renal VSMC support an inhibitory role of EP(4) during ANG II stimulation. In vivo renal blood flow (RBF) studies indicate that the EP(4) receptor is the primary receptor mediating sustained renal vasodilation produced by PGE(2), whereas the EP(1) receptor elicits transient vasoconstriction. The EP(1)-variant receptor does not appear to possess any cAMP or cytosolic calcium signaling capable of affecting RBF. Collectively, these studies demonstrate that the EP(4) receptor is the major receptor in preglomerular VSMC. EP(4) mediates PGE(2)-induced vasodilation in the rat kidney and signals through G(s) proteins to stimulate cAMP and inhibit cytosolic calcium concentration.


Subject(s)
Receptors, Prostaglandin E/physiology , Renal Circulation/physiology , Animals , Arterioles/physiology , Calcium/metabolism , Cyclic AMP/biosynthesis , Cytosol/physiology , Dinoprostone/pharmacology , GTP-Binding Protein alpha Subunits, Gs/physiology , In Vitro Techniques , Male , Microcirculation/physiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Renal Artery/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Vascular Resistance/physiology
6.
Kidney Int ; 58(4): 1686-94, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11012902

ABSTRACT

BACKGROUND: A multiplicity of hormonal, neural, and paracrine factors regulates preglomerular arterial tone by stimulating calcium entry or mobilization. We have previously provided evidence for capacitative (store-operated) Ca2+ entry in fresh renal vascular smooth muscle cells (VSMCs). Ryanodine-sensitive receptors (RyRs) have recently been identified in a variety of nonrenal vascular beds. METHODS: We isolated fresh rat preglomerular VSMCs with a magnetized microsphere/sieving technique; cytosolic Ca2+ ([Ca2+]i) was measured with fura-2 ratiometric fluorescence. RESULTS: Ryanodine (3 micromol/L) increased [Ca2+]i from 79 to 138 nmol/L (P = 0.01). Nifedipine (Nif), given before or after ryanodine, was without effect. The addition of calcium (1 mmol/L) to VSMCs in calcium-free buffer did not alter resting [Ca2+]i. In Ca-free buffer containing Nif, [Ca2+]i rose from 61 to 88 nmol/L after the addition of the Ca2+-ATPase inhibitor cyclopiazonic acid and to 159 nmol/L after the addition of Ca2+ (1 mmol/L). Mn2+ quenched the Ca/fura signal, confirming divalent cation entry. In Ca-free buffer with Nif, [Ca2+]i increased from 80 to 94 nmol/L with the addition of ryanodine and further to 166 nmol/L after the addition of Ca2+ (1 mmol/L). Mn2+ quenching was again shown. Thus, emptying of the sarcoplasmic reticulum (SR) with ryanodine stimulated capacitative Ca2+ entry. CONCLUSION: Preglomerular VSMCs have functional RyR, and a capacitative (store-operated) entry mechanism is activated by the depletion of SR Ca2+ with ryanodine, as is the case with inhibitors of SR Ca2+-ATPase.


Subject(s)
Calcium/metabolism , Kidney Glomerulus/blood supply , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Caffeine/pharmacology , Calcium-Transporting ATPases/metabolism , Chelating Agents/pharmacology , Cytosol/metabolism , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Indoles/pharmacology , Kidney Glomerulus/metabolism , Phosphodiesterase Inhibitors/pharmacology , Rats , Rats, Inbred WKY , Renal Circulation/physiology , Ryanodine/pharmacology , Sarcoplasmic Reticulum/metabolism
7.
Am J Physiol Renal Physiol ; 278(6): F940-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10836981

ABSTRACT

Previous studies have demonstrated that arginine vasopressin (AVP) produces exaggerated renal vasoconstriction in young spontaneously hypertensive rats (SHR) relative to normotensive rats. The exaggerated renal vascular reactivity does not appear to be due to a primary defect in postreceptor calcium signal transduction. Although the magnitudes of vascular responses differ, the relative proportions of calcium entry and mobilization pathways evoked by AVP in renal resistance vessels are similar in these rat strains. The purpose of the present study was to evaluate possible differences in V(1) mRNA and receptor density and affinity in preglomerular resistance vessels (<50 microm) obtained from young Wistar-Kyoto (WKY) and SHR. Quantitative RT-PCR analysis revealed twofold greater expression of the V(1a) receptor gene in preglomerular arterioles of 7-wk-old SHR compared with WKY. In vitro radiolabeled ligand binding studies were performed under equilibrium conditions on preglomerular resistance arterioles freshly isolated from kidneys of 7-wk-old rats. The results indicate that AVP receptor density (B(max)) is two to three times greater in SHR than in WKY (248 +/- 24 vs. 91 +/- 11 fmol/mg protein, P < 0.001). The affinity does not differ between strains (K(d) = 0.5 nM). Displacement studies yielded similar results for SHR and WKY. Unlabeled AVP completely displaced [(3)H]AVP binding, with an IC(50) of 2.5 x 10(-10) M. Expression of AVP receptor types in afferent arterioles was evaluated using the V(1) receptor agonist, [Phe(2), Ile(3),Org(8)]vasopressin, the V(1) receptor antagonist, [d(CH(2))(5), Tyr(Me)(2), Tyr(NH(2))(9)]Arg(8)-vasopressin, and the V(2) receptor agonist, desamino-[D-Arg(8)]vasopressin. Both the V(1) agonist and antagonist displaced up to 90% of the AVP binding with IC(50) values of 4 x 10(-8) and 8 x 10(-7) M, respectively. The V(2) receptor agonist was a weak inhibitor, displacing less than 15% of AVP binding at a high concentration of 10(-4) M. These results demonstrate that virtually all AVP receptors in the preglomerular arterioles are of the V(1) type. Collectively, our results provide evidence that the enhanced renal reactivity to AVP is mediated by a higher density of V(1) receptors associated with increased gene expression in renal resistance vessels of SHR developing genetic hypertension.


Subject(s)
Hypertension/genetics , Hypertension/metabolism , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Renal Circulation/physiology , Animals , Arginine Vasopressin/metabolism , Arterioles/physiopathology , Base Sequence , Binding, Competitive , DNA Primers/genetics , Hypertension/physiopathology , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Renal Circulation/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , Vascular Resistance/genetics , Vascular Resistance/physiology
8.
Am J Physiol Renal Physiol ; 278(1): F138-47, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10644665

ABSTRACT

This study provides new information about the relative importance of different alpha(1)-adrenoceptors during norepinephrine (NE) activation in rat renal resistance vessels. In Sprague-Dawley rats, we measured renal blood flow (RBF) using electromagnetic flowmetry in vivo and the intracellular free calcium concentration ([Ca(2+)](i)) utilizing ratiometric photometry of fura 2 fluorescence in isolated afferent arterioles. Renal arterial bolus injection of NE produced a transient 46% decrease in RBF. In microdissected afferent arterioles, NE (1 microM) elicited an immediate square-shaped increase in [Ca(2+)](i), from 90 to 175 nM (P < 0.001). Chloroethylclonidine (CEC) (50 microM) had no chronic irreversible alkylating effect in vitro but exerted acute reversible blockade on norepinephrine (NE) responses both on [Ca(2+)](i) in vitro and on RBF in vivo. The RBF response was attenuated by approximately 50% by the putative alpha(1A)-adrenoceptor and alpha(1D)-adrenoceptor antagonists 5-methylurapidil (5-MU), and 8-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-8-azaspiro[4. 5]decane-7,9-dione dihydrochloride (BMY-7378) (12.5 and 62.5 microg/h), respectively. The in vitro [Ca(2+)](i) response to NE was blocked approximately 25% and 50% by 5-MU (100 nM and 1 microM). BMY-7378 (100 nM and 1 microM) attenuated the NE-induced response by approximately 40% and 100%. The degree of inhibition in vitro was similar to the in vivo experiments. In conclusion, 5-MU and BMY-7378 attenuated the NE-induced responses, although relatively high concentrations were required, suggesting involvement of both the alpha(1A)-adrenoceptor and alpha(1D)-adrenoceptor. Participation of the alpha(1B)-adrenoceptor is less likely, as we found no evidence for CEC-induced alkylation.


Subject(s)
Kidney/blood supply , Muscle, Smooth, Vascular/metabolism , Receptors, Adrenergic, alpha-1/drug effects , Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Animals , Arterioles/metabolism , Calcium/metabolism , Clonidine/analogs & derivatives , Clonidine/pharmacology , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Norepinephrine/pharmacology , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Vascular Resistance/drug effects
9.
J Am Soc Nephrol ; 10(12): 2620-30, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10589703

ABSTRACT

The present study describes methodology and its application to evaluate renal reactivity in acute studies on anesthetized mice. Renal blood flow (RBF) was measured using an ultrasonic transit-time flowmeter and a non-cannulating V-shaped probe. An intrarenal artery injection technique established feasibility and reproducibility of studies of renal vascular reactivity to angiotensin II (AngII) in adult wild-type mice. The study also examined whether AngII would affect RBF in mice lacking AT1A receptors due to gene targeting. Mean arterial pressure averaged 83 and 62 mmHg, respectively, in mice with and without AT1A receptors. The RBF was similar in both groups, averaging 7 ml/min per g kidney wt. AngII injection (10-microl bolus) into the renal artery produced transient, dose-dependent, selective reductions in RBF in AT1A knockout mice as well as wild-type mice. The response was considerably greater in mice with AT1A receptors: 10% for 0.1 ng, 30% for 1 ng, and 45% for 5 ng AngII in control animals versus respective decreases of 6, 15, and 17% in knockout mice. In other studies, angiotensin-converting enzyme (captopril) or renin (CP-71362-14) was inhibited. During inhibition of AngII formation, renal vascular reactivity to AngII increased twofold in both groups. Coadministration of the AT1 receptor antagonist losartan (1 to 1000 ng) elicited dose-dependent inhibition of AngII effects, with near maximum blockage of 80 to 90% in both groups of mice. The putative AT2 receptor antagonist PD 123319 inhibited 30 to 40% of AngII-induced vasoconstriction, whereas CGP 42112 had no effect in either group. In conclusion, AngII can elicit renal vasoconstriction, albeit attenuated, in AT1A knockout mice. The weaker RBF effects are most likely due to the absence of the AT1A receptor. Inhibition of the response by AT1 receptor antagonist suggests mediation by the AT1B receptor in these animals. The residual constrictor effect observed during AT1 receptor blockade and sensitive to PD 123319 appears to be mediated by a non-AT1 receptor.


Subject(s)
Angiotensin II/pharmacology , Receptors, Angiotensin/genetics , Receptors, Angiotensin/physiology , Renal Circulation/drug effects , Renal Circulation/physiology , Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Captopril/pharmacology , Female , Imidazoles/pharmacology , Losartan/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligopeptides/pharmacology , Pyridines/pharmacology , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Renin/antagonists & inhibitors , Vasoconstriction/drug effects , Vasoconstriction/physiology
10.
Am J Physiol ; 277(6): F850-8, 1999 12.
Article in English | MEDLINE | ID: mdl-10600931

ABSTRACT

In order to exert an appropriate biological effect, the action of the vasoconstrictive hormone angiotensin II (ANG II) is modulated by vasoactive factors such as prostaglandins PGE2 and PGI2. The present study investigates whether prostaglandins alter ANG II-mediated increases in cytosolic calcium concentration ([Ca2+]i) in vascular smooth muscle cells (VSMC) isolated from rat renal preglomerular arterioles. [Ca2+]i was assessed using the calcium-sensitive dye fura 2 and a microscope-based photometer system. ANG II (10(-7) M) caused a biphasic, time-dependent [Ca2+]i response: an initial peak increase from 52 +/- 7 to 264 +/- 25 nM, followed by a sustained plateau of 95 +/- 9 nM in cultured VSMC. Coadministration of PGE2 or PGI2 or synthetic mimetics caused dose-dependent decreases in the peak [Ca2+]i response to ANG II, with attenuation of 40-50%. This degree of inhibition was even more pronounced in individual freshly isolated preglomerular VSMC. Increasing cAMP levels in cultured VSMC, by using either a cell-permeable analog or inhibiting phosphodiesterase activity, mirrored the antagonistic effects of prostaglandins on ANG II-stimulated increases in [Ca2+]i. Radioimmunoassays demonstrate that ANG II (10(-7) M) stimulates production of PGI2 and PGE2; the stable prostacyclin metabolite 6-keto-PGF(1alpha) was released in 10-fold greater concentrations than PGE(2.) Indomethacin blockade of prostaglandin production potentiated both the peak (264 to 337 +/- 26 nM) and sustained [Ca2+]i responses (95 to 181 +/- 22 nM) to ANG II. When prostaglandin analogs were added during indomethacin treatment, the ANG II response was restored to the typical pattern. In conclusion, we demonstrate that modulation of intracellular calcium levels is one mechanism by which prostaglandins can buffer ANG II-mediated constriction in renal preglomerular VSMC. PGI2 is more potent than PGE2 in this regard.


Subject(s)
Angiotensin II/pharmacology , Arterioles/physiology , Calcium/metabolism , Dinoprostone/pharmacology , Epoprostenol/pharmacology , Kidney/blood supply , Muscle, Smooth, Vascular/physiology , Renal Circulation/physiology , Animals , Buffers , Cells, Cultured , Cytosol/metabolism , Iloprost/pharmacology , Kinetics , Male , Rats , Rats, Sprague-Dawley , Vascular Resistance
11.
Hypertension ; 34(5): 1134-40, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10567195

ABSTRACT

The protective effect of vasodilator agents linked to the cAMP pathway is less effective for buffering the vasoconstrictor effect of angiotensin II in young animals with genetic hypertension. To determine the underlying cellular mechanism, experiments were performed on freshly isolated preglomerular resistance arterioles obtained from kidneys of 7-week-old spontaneously hypertensive rats (SHR) and normotensive Wistar-Kyoto rats (WKY). Specific high-affinity saturable binding of (3)H-prostaglandin (PG) E(2) revealed 1 receptor class in renal microvessels; PGE(2) receptor density was similar in SHR and WKY (106 versus 115 fmol/mg; P>0.8), as was receptor affinity (3.6 versus 3.5 nmol/L; P>0.7). Basal cAMP activity was similar in renal arterioles from SHR and WKY. A major finding was that PGE(2), PGI(2), and isoproterenol produced weaker stimulation of cAMP formation in arteriolar cells of SHR (P<0.02). In contrast, GTPgammas and forskolin stimulated cAMP generation to a similar degree in both rat strains, which suggests normal adenylate cyclase activity in hypertension-prone SHR. Immunoblots revealed the presence of 3 classes of G proteins (G(s), G(i), and G(q)) in preglomerular arterioles. The relative amounts of discernible G-protein alpha-subunits in renal resistance vessels did not differ between SHR and WKY. These results extend previous in vivo studies of abnormal renal vascular reactivity in SHR and more directly localize defective coupling of the prostaglandin and beta-adrenergic receptors to a stimulatory G protein and cAMP production in freshly isolated preglomerular arteriolar cells of young SHR. This dysfunction may be due to an abnormal interaction between prostaglandin receptors and G(s) protein that leads to inefficient coupling of initiating steps in the cAMP-protein kinase A cascade during the development of hypertension.


Subject(s)
Cyclic AMP/physiology , GTP-Binding Protein alpha Subunits, Gs/physiology , Hypertension/physiopathology , Kidney/blood supply , Receptors, Prostaglandin E/physiology , Receptors, Prostaglandin/physiology , Animals , Arterioles/physiopathology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , In Vitro Techniques , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Epoprostenol
12.
Am J Physiol ; 277(4): F533-42, 1999 10.
Article in English | MEDLINE | ID: mdl-10516277

ABSTRACT

Calcium entry via voltage-gated L-type channels is responsible for at least half of the increase in cytosolic calcium ([Ca(2+)](i)) in afferent arterioles following agonist stimulation. We sought the presence of capacitative calcium entry in fresh vascular smooth muscle cells (VSMC) derived from rat preglomerular vessels. [Ca(2+)](i) was measured using fura-2 ratiometric fluorescence. Vasopressin V1 receptor agonist (V1R) (10(-7) M) increased [Ca(2+)](i) by approximately 100 nM. A calcium channel blocker (CCB), nifedipine or verapamil (10(-7) M), inhibited the response by approximately 50%. V1R in the presence of CCB increased [Ca(2+)](i) from 106 to 176 nM, confirming that calcium mobilization and/or entry may occur independent of voltage-gated channels. In nominally Ca(2+)-free buffer, V1R increased [Ca(2+)](i) from 94 to 129 nM, denoting mobilization; addition of CaCl(2) (1 mM) further elevated [Ca(2+)](i) to 176 nM, indicating a secondary phase of Ca(2+) entry. Similar responses were obtained when CCB was present in calcium-free buffer or when EGTA was present. In nominally Ca(2+)-free medium, the sarcoplasmic reticulum Ca(2+)-ATPase inhibitors (SRCAI), thapsigargin and cyclopiazonic acid (CPA), increased [Ca(2+)](i) from 97 to 128 and 143 nM, respectively, and to 214 and 220 nM, respectively, when 1 mM extracellular Ca(2+) was added. In the presence of verapamil, the results with CPA acid were nearly identical. In Ca(2+)-free buffer, the stimulatory effect of V1R or SRCAI on the Ca(2+)/fura signal was quenched by the addition of Mn(2+) (1 mM), demonstrating divalent cation entry. These studies provide evidence for capacitative (store- operated) calcium entry in VSMC freshly isolated from rat preglomerular arterioles.


Subject(s)
Calcium/metabolism , Kidney Glomerulus/blood supply , Muscle, Smooth, Vascular/metabolism , Animals , Arterioles/cytology , Arterioles/drug effects , Arterioles/metabolism , Arterioles/physiology , Calcium/physiology , Calcium Channel Blockers/pharmacology , Calcium-Transporting ATPases/antagonists & inhibitors , Electric Conductivity , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Intracellular Membranes/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Osmolar Concentration , Rats , Rats, Inbred WKY , Receptors, Vasopressin/agonists , Sarcoplasmic Reticulum/enzymology , Thapsigargin/pharmacology
13.
Am J Physiol ; 276(5): F700-10, 1999 05.
Article in English | MEDLINE | ID: mdl-10330052

ABSTRACT

This study provides new information about the relative importance of Ca2+ mobilization and entry in the renal vascular response to adrenoceptor activation. We measured renal blood flow (RBF) in Sprague-Dawley rats in vivo using electromagnetic flowmetry. We measured intracellular free Ca2+ concentration ([Ca2+]i) in isolated afferent arterioles utilizing ratiometric photometry of fura-2 fluorescence. Renal arterial injection of NE produced a transient decrease in RBF. The response was attenuated, in a dose-dependent manner, up to approximately 50% by nifedipine, an antagonist of L-type Ca2+ entry channels. Inhibition of Ca2+ mobilization by 3,4, 5-trimethoxybenzoic acid-8-(diethylamino)octyl ester (TMB-8) inhibited the renal vascular effects of NE in a dose-dependent manner, with maximal blockade of approximately 80%. No additional attenuation was observed when nifedipine and TMB-8 were administered together. In microdissected afferent arterioles, norepinephrine (NE; 10(-6) M) elicited an immediate square-shaped increase in [Ca2+]i, from 110 to 240 nM. This in vitro response was blocked by nifedipine (10(-6) M) and TMB-8 (10(-5) M) to a degree similar to that of the in vivo experiments. A nominally calcium-free solution blocked 80-90% of the [Ca2+]i response to NE. The increased [Ca2+]i elicited by depolarization with medium containing 50 mM KCl was totally blocked by nifedipine. In contrast, TMB-8 had no effect. Our results indicate that both Ca2+ entry and mobilization play important roles in the renal vascular Ca2+ and contractile response to adrenoceptor activation. The entry and mobilization mechanisms activated by NE may interact. That a calcium-free solution caused a larger inhibition of the NE effects on afferent arterioles than nifedipine suggests more than one Ca2+ entry pathway.


Subject(s)
Calcium/metabolism , Norepinephrine/pharmacology , Renal Circulation/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Arterioles/chemistry , Arterioles/drug effects , Arterioles/metabolism , Calcium/analysis , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Calcium Channels, L-Type , Chelating Agents/pharmacology , Cytosol/chemistry , Egtazic Acid/pharmacology , Electromagnetic Fields , Fluorescent Dyes , Fura-2 , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Male , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Nifedipine/pharmacology , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum/enzymology , Type C Phospholipases/metabolism
14.
Am J Physiol ; 276(5): F758-66, 1999 05.
Article in English | MEDLINE | ID: mdl-10330058

ABSTRACT

We performed micropuncture studies to determine the role of thromboxane A2 in the exaggerated tubuloglomerular feedback (TGF) activity in young spontaneously hypertensive rats (SHR). Glomerular function was assessed by changes in proximal tubular stop-flow pressure (SFP) produced by different rates of orthograde perfusion through Henle's loop. Seven-week-old SHR exhibited an exaggerated TGF activity compared with Wistar-Kyoto rats (WKY) during euvolemia, confirming earlier studies. During control periods, the feedback-induced maximal SFP response (DeltaSFP) was greater in SHR (18-19 vs. 12-13 mmHg in WKY), whereas basal SFP and proximal tubular free-flow pressure were similar in both strains. In one series, the thromboxane A2 agonist U-46619 was added to the tubular perfusate for a final concentration of 10(-6) M. In WKY, DeltaSFP was increased by 100% to 26 mmHg. In contrast, DeltaSFP in young SHR was unaffected by the thromboxane A2 agonist. In other animals, the thromboxane synthase inhibitor pirmagrel (50 mg/kg) was injected intravenously to inhibit thromboxane production. In SHR, pirmagrel decreased DeltaSFP by 8.5 mmHg and reduced reactivity. Less attenuation was observed in WKY; DeltaSFP was reduced by 3 mmHg, whereas reactivity was unchanged. In other studies, tubular perfusion with the thromboxane receptor inhibitor SQ-29548 (10(-6) M) reduced DeltaSFP more in SHR (7 vs. 3 mmHg in WKY) and also decreased reactivity more in SHR (2.3 vs. 0.5 mmHg. nl-1. min-1). Coperfusion of SQ-29548 and U-46619 resulted in an 85% block of the effect of U-46619 on DeltaSFP. Tubular perfusion with the agonist U-46619 during thromboxane synthase inhibition markedly enhanced DeltaSFP in both strains, with a greater effect in WKY. These results suggest that elevated levels of thromboxane A2 in young SHR contribute to the exaggerated TGF control of glomerular function in SHR during the developmental phase of hypertension.


Subject(s)
Feedback/physiology , Juxtaglomerular Apparatus/physiology , Rats, Inbred SHR/physiology , Thromboxane A2/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Age Factors , Animals , Blood Pressure , Body Weight , Bridged Bicyclo Compounds, Heterocyclic , Capillaries/drug effects , Capillaries/physiology , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated , Feedback/drug effects , Hematocrit , Hydrazines/pharmacology , Imidazoles/pharmacology , Juxtaglomerular Apparatus/drug effects , Kidney Tubules, Proximal/physiology , Loop of Henle/physiology , Pyridines/pharmacology , Rats , Rats, Inbred WKY , Renal Circulation/drug effects , Renal Circulation/physiology , Thromboxane A2/antagonists & inhibitors , Vasoconstrictor Agents/pharmacology
15.
Am J Physiol ; 276(3): F409-16, 1999 03.
Article in English | MEDLINE | ID: mdl-10070164

ABSTRACT

One purpose of the present study was to evaluate the ability of 7-wk-old spontaneously hypertensive rats (SHR) to reset tubuloglomerular feedback (TGF) activity in response to acute volume expansion (VE). Second, we evaluated the contribution of ANG II, via its action on AT1 receptors, to TGF control of glomerular function during VE. TGF was assessed by micropuncture methods and proximal tubular stop-flow pressure (SFP) determinations in SHR, Wistar-Kyoto rats (WKY), and Sprague-Dawley rats (SD). During euvolemia SHR exhibited enhanced TGF activity. In the same animals acute VE was achieved by infusion of saline (5 ml. h-1. 100 g body wt-1). VE led to resetting of TGF in all three strains. Maximal SFP responses, elicited by a 30-40 nl/min loop of Henle perfusion rate, decreased from 19 to 12 mmHg in SHR and, on average, from 11 to 5 mmHg in WKY and SD (P < 0.001). Tubular flow rate producing a half-maximal response (turning point) shifted to higher flow rates during VE, from 12 to 14 nl/min in SHR and from 15 to 19 nl/min in WKY. Administration of the AT1 receptor blocker candesartan (0.05 mg/kg iv) during sustained VE decreased TGF-mediated reductions in SFP in SHR and slightly increased the turning point in WKY. Nevertheless, other parameters of TGF activity were unaffected by AT1 receptor blockade. In conclusion, young SHR possess the ability to reset TGF activity in response to VE to a degree similar to compensatory adjustments in WKY. However, TGF remains enhanced in SHR during VE. ANG II and its action on AT1 receptors are in part responsible for the exaggerated SFP responses in young SHR during VE.


Subject(s)
Kidney Glomerulus/physiology , Kidney Tubules/physiology , Plasma Substitutes/pharmacology , Rats, Inbred SHR/physiology , Angiotensin II/physiology , Angiotensin Receptor Antagonists , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Feedback , Loop of Henle/physiology , Perfusion , Pressure , Punctures , Rats , Rats, Inbred WKY , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/physiology , Sodium Chloride/pharmacology , Tetrazoles/pharmacology
16.
Circ Res ; 84(2): 186-92, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-9933250

ABSTRACT

This study aimed to characterize the interaction between nitric oxide (NO)- and cAMP-related pathways in the control of renal blood flow. Using the isolated perfused rat kidney model, we determined the effects of inhibition of NO formation by Nomega-nitro-L-arginine methyl ester (L-NAME; 1 mmol/L) and of NO administration by sodium nitroprusside (SNP, 10 micromol/L) on renal vascular resistance under conditions of elevated vascular cAMP levels. cAMP levels were increased either by adenylate cyclase activation via isoproterenol or by inhibition of cAMP phosphodiesterases (PDEs) 1, 3, and 4. We found that L-NAME markedly increased vascular resistance and that this effect was completely reversed by SNP. Both isoproterenol and inhibitors of the cAMP PDEs lowered basal vascular resistance. In the presence of isoproterenol (3 nmol/L) and inhibitors of PDE-1 [8-methoxymethyl-l-methyl-3-(2-methylpropyl)-xanthine; 8-MM-IBMX, 20 micromol/L] and PDE-4 (rolipram, 20 micromol/L), L-NAME again substantially increased vascular resistance, and this effect of L-NAME was completely reversed by SNP. In the presence of the PDE-3 inhibitors milrinone (20 micromol/L) and trequinsin (200 nmol/L), however, both L-NAME and SNP failed to exert any additional effects. Because PDE-3 is a cGMP-inhibited cAMP PDE and because the vasodilatory effect of SNP was abrogated by the guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ) (20 micromol/L), our findings are compatible with the idea that an action of NO on PDE-3 could account for the vasodilatory properties of NO on the renal vasculature. Moreover, our findings suggest that PDE-3 activity is an important determinant of renal vascular resistance.


Subject(s)
Cyclic AMP/metabolism , Kidney/blood supply , Nitric Oxide/metabolism , Adenylyl Cyclases/drug effects , Animals , Arterioles/drug effects , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitroprusside/pharmacology , Perfusion , Rats , Rats, Sprague-Dawley , Stimulation, Chemical
17.
Am J Physiol ; 276(2): F260-70, 1999 02.
Article in English | MEDLINE | ID: mdl-9950957

ABSTRACT

Experiments were conducted to gain insight into mechanisms responsible for exaggerated renal vascular reactivity to ANG II and vasopressin (AVP) in spontaneously hypertensive rats (SHR) during the development of hypertension. Cytosolic calcium concentration ([Ca2+]i) was measured by ratiometric fura 2 fluorescence and a microscope-based photometer. Vascular smooth muscle cells (SMC) from preglomerular arterioles were isolated and dispersed using an iron oxide-sieving method plus collagenase treatment. ANG II and AVP produced rapid and sustained increases in [Ca2+]i. ANG II elicited similar dose-dependent increases in [Ca2+]i in SMC from SHR and Wistar-Kyoto rats (WKY). In contrast, AVP caused almost twofold larger responses in afferent arteriolar SMC from SHR. ANG II effects were inhibited by the AT1 receptor antagonist losartan. AVP action was blocked by the V1 receptor antagonist [d(CH2)5,Tyr(NH2)9]AVP. In SMC pretreated with nifedipine, neither ANG II nor AVP elicited [Ca2+]i responses. Poststimulation nifedipine reversed elevated [Ca2+]i to basal levels. Short-term reductions in external [Ca2+]i (EGTA) mimicked the nifedipine effects. Our study shows that AT1 and V1 receptors stimulate [Ca2+]i by a common mechanism characterized by preferential action on voltage-gated L-type channels sensitive to dihydropyridines. Calcium signaling elicited by AT1 receptors does not differ between SHR and WKY; thus the in vivo exaggerated reactivity may be dependent on interactions with other cell types, e. g., endothelium. In contrast, AVP produced larger changes in [Ca2+]i in arteriolar SMC from SHR, and such direct effects can account for the exaggerated renal blood flow responses.


Subject(s)
Calcium Signaling/physiology , Hypertension/physiopathology , Kidney Glomerulus/blood supply , Muscle, Smooth, Vascular/physiology , Rats, Inbred SHR/physiology , Angiotensin II/pharmacology , Animals , Arginine Vasopressin/pharmacology , Arterioles/drug effects , Arterioles/physiology , Calcium/metabolism , Calcium Channels/physiology , Calcium Channels, L-Type , Cytosol/metabolism , In Vitro Techniques , Male , Muscle, Smooth, Vascular/drug effects , Osmolar Concentration , Rats , Rats, Inbred WKY
18.
J Am Soc Nephrol ; 10 Suppl 11: S84-9, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9892145

ABSTRACT

Experiments were conducted to gain insight into calcium signaling mechanisms triggered by angiotensin II (AngII) stimulation in vascular smooth muscle cells (SMC) freshly isolated from preglomerular vessels of normotensive Wistar Kyoto rats (WKY) and spontaneously hypertensive rats (SHR). Cytosolic calcium concentration ([Ca2+]i) was measured using ratiometric Fura-2 fluorescence and a microscope-based photometer. Vascular SMC from preglomerular vessels were isolated and dispersed using an iron oxide-sieving method combined with collagenase treatment. AngII produced rapid increases in [Ca2+]i that remained elevated for the duration of continued stimulation. The same pattern of time response was observed in WKY and in SHR. AngII elicited dose-dependent increases in [Ca2+]i in groups of individual preglomerular arteriolar SMC from both strains. AngII (10(-10) M) induced an increase from baseline levels in WKY and SHR (37+/-9 and 32+/-13 nM; P < 0.05). In response to 10(-6) M AngII, steady-state responses were 165+/-30 and 170+/-35 nM (P < 0.01). The responses did not differ between strains (P > 0.4). The effects of AngII were inhibited by 88% by the AT1 receptor blocker candesartan in renal SMC. In SMC pretreated with calcium-free medium, baseline [Ca2+]i fell by about 60 nM. Thereafter, AngII did not elicit any [Ca2+]i response either in WKY or in SHR when calcium entry was prevented. Also, after prestimulation by AngII, a calcium-free solution completely reversed the effects of AngII. This study shows that AngII acts through AT1 receptors to stimulate [Ca2+]i by a predominant action on calcium entry with no evidence for calcium mobilization. Other studies have demonstrated that calcium entry in these SMC is mediated by voltage-gated, L-type entry channels sensitive to dihydropyridine agents. No strain differences were noted between the actions of AngII on individual renal SMC from SHR and normotensive control animals.


Subject(s)
Calcium Signaling , Kidney Glomerulus/blood supply , Muscle, Smooth, Vascular/metabolism , Receptors, Angiotensin/metabolism , Angiotensin II/pharmacology , Angiotensin Receptor Antagonists , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Calcium/metabolism , Cells, Cultured , Egtazic Acid , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Tetrazoles/pharmacology
19.
J Am Soc Nephrol ; 10 Suppl 11: S149-61, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9892156

ABSTRACT

Angiotensin II (AngII) exerts powerful effects on the renal microcirculation to influence a variety of functions. This review summarizes some of the major findings over the past 10 years as they elucidate the multiple roles that AngII plays in the regulation of whole kidney blood flow, perfusion of cortical and medullary regions, and renal autoregulation. Topics of discussion include localization of AngII receptor types and subtypes in the renal vasculature, action of AngII on vascular smooth muscle cells of the afferent and efferent arterioles, and intracellular signaling pathways. Within the microvasculature, AngII causes potent constriction in both the afferent and efferent arterioles, with responses modulated by paracrine and autocrine factors of endothelial and macula densa origins. With regard to renal autoregulatory mechanisms consisting of the myogenic response and the tubuloglomerular feedback mechanism, the myogenic response appears to operate independent of the renin-angiotensin system. On the other hand, tubuloglomerular feedback activity is often directly proportional to concentrations of AngII, especially in high renin states. Of the two types defined to date, the AT1 is the predominant receptor in the adult rat kidney mediating the vascular effects of AngII. AT2 receptor is highly expressed in the fetal kidney and is important for renal development, but is very weakly expressed in adult animals. Nevertheless, AT2 receptors may mediate vasodilation under certain conditions. The signaling transduction pathways for AT1 receptors include Gq/11-protein and protein kinase C activation. AngII causes constriction of the afferent arteriole primarily by stimulation of calcium entry via voltage-sensitive, L-type channels, whereas AngII effects on the efferent arteriole are due to calcium release from intracellular stores and calcium entry through voltage-independent calcium entry channels. Future experiments should contribute to a more in-depth understanding of the modulation of AngII effects by other vasoactive agents and interactions between different second-messenger signaling pathways in health and disease.


Subject(s)
Angiotensin II/pharmacology , Kidney/drug effects , Receptors, Angiotensin/drug effects , Angiotensin II/antagonists & inhibitors , Angiotensin Receptor Antagonists , Animals , Cells, Cultured , Hemodynamics/drug effects , Homeostasis/drug effects , Humans , Kidney/blood supply , Kidney/growth & development , Microcirculation/drug effects , Muscle, Smooth, Vascular/drug effects , Regional Blood Flow/drug effects , Signal Transduction/drug effects
20.
J Am Soc Nephrol ; 10 Suppl 11: S202-7, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9892164

ABSTRACT

This study determined the inhibitory effect of the angiotensin II (AngII) type I (AT1) receptor blocker candesartan on renal vascular reactivity in vivo. Reactivity to AngII before and during candesartan administration was assessed by measuring (by electromagnetic or ultrasonic flowmetry) renal blood flow responses to AngII in rats and mice. AngII produced greater renal vasoconstriction in 7-wk-old, spontaneously hypertensive rats than in Wistar-Kyoto rats. After indomethacin treatment, AngII (2 ng) produced 40% reductions in renal blood flow in both rat strains, without affecting systemic arterial pressure. Coadministration of candesartan blocked AngII effects in a dose-dependent manner, with similar levels of inhibition in spontaneously hypertensive rats and Wistar-Kyoto rats; maximal inhibition was 80%. In rats that had been pretreated (for 30 min) with intravenous candesartan, AngII-induced renal vasoconstriction was inhibited dose dependently up to 98%. To evaluate receptor subtype mediation, responses were compared in mice with or without the AT1A receptor (deleted by gene targeting). Intrarenal AngII (1 ng) caused a 32% reduction of renal blood flow in wild-type mice and an 8% reduction of renal blood flow in AT1A receptor-knockout mice. Ten nanograms of AngII were required to elicit 20% renal vasoconstriction in these mutant mice. Concurrent injection of candesartan caused dose-dependent inhibition of AngII up to 80%. The candesartan IC50 values for percentage changes in renal blood flow did not differ in the two groups of mice. These studies establish that candesartan is an effective, highly selective, AT1 receptor blocker, inhibiting renal vasoconstriction in rodents in a concentration- and time-dependent manner. Candesartan effectively blocks AT1A and AT1B receptors in renal resistance vessels of rodents, with similar efficacies in rats and mice.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin Receptor Antagonists , Benzimidazoles/pharmacology , Kidney/drug effects , Tetrazoles/pharmacology , Angiotensin II/administration & dosage , Animals , Benzimidazoles/therapeutic use , Biphenyl Compounds , Dose-Response Relationship, Drug , Hypertension/drug therapy , Hypertension/genetics , Kidney/blood supply , Mice , Mice, Knockout , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/genetics , Regional Blood Flow/drug effects , Renal Artery , Tetrazoles/therapeutic use , Time Factors , Vasoconstriction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...