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










Publication year range
1.
Am J Physiol Regul Integr Comp Physiol ; 280(4): R1177-84, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11247842

ABSTRACT

The objective was to determine the central nervous system (CNS) responses to dehydration (c-Fos and vasopressin mRNA) in mice lacking the ANG AT(1a) receptor [ANG AT(1a) knockout (KO)]. Control and AT(1a) KO mice were dehydrated for 24 or 48 h. Baseline plasma vasopressin (VP) was not different between the groups; however, the response to dehydration was attenuated in AT(1a) KO (24 +/- 11 vs. 10.6 +/- 2.7 pg/ml). Dehydration produced similar increases in plasma osmolality and depletion of posterior pituitary VP content. Neuronal activation was observed as increases in c-Fos protein and VP mRNA. The supraoptic responses were not different between groups. In the paraventricular nucleus (PVN), c-Fos-positive neurons (57.4 +/- 10.7 vs. 98.4 +/- 7.4 c-Fos cells/PVN, control vs. AT(1a) KO) and VP mRNA levels (1.0 +/- 0.1 vs. 1.4 +/- 0.1 microCi, control vs. AT(1a) KO) were increased with greater responses in AT(1a) KO. A comparison of 1- to 2-day water deprivation showed that plasma VP, brain c-Fos, and VP mRNA returned toward control on day 2, although plasma osmolality remained high. Data demonstrate that AT(1a) KO mice show a dichotomous response to dehydration, reduced for plasma VP and enhanced for PVN c-Fos protein and VP mRNA. The results illustrate the importance of ANG AT(1a) receptors in the regulation of osmotic and endocrine balance.


Subject(s)
Dehydration/physiopathology , Gene Expression Regulation/physiology , Neurons/physiology , Receptors, Angiotensin/physiology , Animals , Crosses, Genetic , Dehydration/genetics , Female , Genes, fos , Genotype , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Knockout , Pituitary Gland/physiology , Pituitary Gland/physiopathology , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/deficiency , Receptors, Angiotensin/genetics , Transcription, Genetic , Vasopressins/analysis , Vasopressins/genetics
2.
Physiol Behav ; 74(4-5): 457-64, 2001.
Article in English | MEDLINE | ID: mdl-11790405

ABSTRACT

Angiotensin II (ANG II) type 1 receptors are found in the mouse suprachiasmatic nucleus (SCN), the site of the circadian pacemaker, but their significance for circadian timekeeping is unknown. We examined circadian rhythms of wheel running and drinking in angiotensin AT(1a) receptor knockout (KO) mice. Mean daily running and drinking activity were elevated in KO mice under a light-dark (LD) cycle and in constant dark (DD). These increases were confined to the usual active (dark) period, thus, the 'amplitude' of running and drinking rhythms was higher in KO mice. The phase of entrainment to LD (measured by the onset of the daily active period) did not differ between groups, either in LD or on the first day of DD ('unmasked' phase). KO mice showed a modestly shorter free-running period (tau) in DD. The direction and magnitude of phase shifts to light pulses at two circadian times (CTs) in DD did not differ between groups. Core functions of the circadian system appear intact following AT(1a) receptor KO. The modestly shorter tau and increased rhythm amplitude in KO mice may be secondary to an effect of the mutation on the level of running and drinking activity.


Subject(s)
Circadian Rhythm/physiology , Drinking/physiology , Receptors, Angiotensin/physiology , Animals , Female , Male , Mice , Mice, Knockout , Motor Activity/physiology , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/genetics , Suprachiasmatic Nucleus/physiology
3.
Trends Endocrinol Metab ; 11(7): 263-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10920382

ABSTRACT

The renin-angiotensin system (RAS) has a wide range of actions in biological processes ranging from development and reproduction to cardiovascular and renal functions. Most of these actions are mediated by the octapeptide hormone angiotensin II. The identified family of angiotensin II receptors is divided into two pharmacological classes: type 1 (AT1) and type 2 (AT2). The classically recognized actions of the RAS are primarily mediated by the AT1 subtype of angiotensin receptors, and these receptors are the targets of a new class of anti-hypertensive agents. In recent years, our understanding of the physiological functions of AT1 receptors has been advanced through the use of gene-targeting technology. In this review, we will summarize the emerging picture of AT1 receptor functions that has been provided by gene-targeting experiments.


Subject(s)
Gene Targeting , Receptors, Angiotensin/genetics , Receptors, Angiotensin/physiology , Angiotensin II/metabolism , Angiotensin II/physiology , Animals , Blood Pressure/physiology , Humans , Immunity/physiology , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Water-Electrolyte Balance/genetics , Water-Electrolyte Balance/physiology
4.
J Clin Invest ; 106(1): 103-6, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10880053

ABSTRACT

The renin-angiotensin system (RAS) plays a critical role in cardiovascular and fluid homeostasis. The major biologically active peptide of the RAS is angiotensin II, which acts through G protein-coupled receptors of two pharmacological classes, AT(1) and AT(2). AT(1) receptors, expressed in brain and peripheral tissues, mediate most classically recognized actions of the RAS, including blood pressure homeostasis and regulation of drinking and water balance. In rodents, two highly homologous AT(1) receptor isoforms, termed AT(1A) and AT(1B) receptors, are expressed at different levels in major forebrain cardiovascular and fluid regulatory centers, with AT(1A) expression generally exceeding AT(1B) expression, but the relative contributions of these receptor subtypes to central angiotensin II responses are not known. We used gene targeting in combination with a unique system for maintaining catheters in the cerebral ventricles of conscious mice to test whether there are differential roles for AT(1A) and AT(1B) receptors in responses elicited by angiotensin II in the brain. Here we show that the blood pressure increase elicited by centrally administered angiotensin II can be selectively ascribed to the AT(1A) receptor. However, the drinking response requires the presence of AT(1B) receptors. To our knowledge, this is the first demonstration of a primary and nonredundant physiological function for AT(1B) receptors.


Subject(s)
Angiotensin II/pharmacology , Brain/drug effects , Receptors, Angiotensin/physiology , Animals , Blood Pressure/drug effects , Brain/physiology , Drinking/drug effects , Mice , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2
5.
Am J Physiol Renal Physiol ; 278(1): F75-82, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10644657

ABSTRACT

Mice lacking AT(1A) receptors for ANG II have a defect in urinary concentration manifested by an inability to increase urinary osmolality to levels seen in controls after thirsting. This defect results in extreme serum hypertonicity during water deprivation. In the basal state, plasma vasopressin levels are similar in wild-type controls and Agtr1a -/- mice. Plasma vasopressin levels increase normally in the AT(1A) receptor-deficient mice after 24 h of water deprivation, suggesting that the defect in urine concentration is intrinsic to the kidney. Using magnetic resonance microscopy, we find that the absence of AT(1A) receptors is associated with a modest reduction in the distance from the kidney surface to the tip of the papilla. However, this structural abnormality seems to play little role in the urinary concentrating defect in Agtr1a -/- mice since the impairment is largely reproduced in wild-type mice by treatment with an AT(1)-receptor antagonist. These studies demonstrate a critical role for the AT(1A) receptor in maintaining inner medullary structures in the kidney and in regulating renal water excretion.


Subject(s)
Kidney Concentrating Ability/physiology , Kidney/metabolism , Receptors, Angiotensin/deficiency , Water/metabolism , Angiotensin Receptor Antagonists , Animals , Body Weight , Deamino Arginine Vasopressin/pharmacology , Female , Genotype , Kidney/anatomy & histology , Kidney Concentrating Ability/drug effects , Losartan/pharmacology , Male , Mice , Osmolar Concentration , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/genetics , Urine/chemistry , Urodynamics , Vasopressins/blood , Water/pharmacology , Water Deprivation
6.
Hypertension ; 35(2): 550-4, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10679496

ABSTRACT

To examine the role of the angiotensin II (AT)(1A) receptor in the regulation of blood pressure and sodium balance, we measured systolic blood pressure responses in AT(1A) receptor-deficient (Agtr1a-/-) and wild-type (Agtr1a+/+) mice while dietary sodium content was systematically altered. On a 0.4% sodium diet, systolic blood pressures were significantly lower in Agtr1a-/- than in +/+ mice. In Agtr1a+/+ mice, changing dietary sodium content did not affect blood pressure. In contrast, when Agtr1a-/- mice were fed a high-salt diet (6% NaCl), their systolic blood pressures increased significantly from 79+/-4 to 94+/-4 mm Hg (P<0.006). The low blood pressures of Agtr1a-/- mice decreased further while on a low-salt diet from 82+/-3 to 69+/-3 mm Hg (P<0.03). On the high-salt diet, urinary sodium excretion increased to similar levels in Agtr1a+/+ and -/- mice. Although urinary sodium excretion was substantially reduced in both groups during the low-salt diet, cumulative sodium balances became negative in Agtr1a-/- mice despite a 6-fold increase in urinary aldosterone. We infer, therefore, that the reduced blood pressures in Agtr1a-/- mice on a normal diet are caused by depletion of sodium and extracellular volume. Their "sodium sensitivity" suggests a critical role for renal AT(1A) receptors to modulate sodium handling.


Subject(s)
Blood Pressure/physiology , Receptors, Angiotensin/physiology , Sodium, Dietary/administration & dosage , Aldosterone/urine , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/genetics , Sodium/urine , Systole
7.
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
8.
J Clin Invest ; 104(12): 1693-701, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10606623

ABSTRACT

The renin-angiotensin system (RAS) is a key regulator of vascular tone and blood pressure. In addition, angiotensin II also has a number of cellular effects that may contribute to disease pathogenesis. Using Agtr1a(-/-) mice, which lack AT(1A) receptors for angiotensin II, we have identified a novel function of the RAS to modulate the immune system. We find that angiotensin II, acting through type 1 (AT(1)) receptors on immune cells, triggers the proliferation of splenic lymphocytes. These actions contribute to the vigor of cellular alloimmune responses. Within lymphoid organs, sufficient components of the RAS are present to activate AT(1) receptors during an immune response, promoting cell growth. These actions require activation of calcineurin phosphatase. In an in vivo model of cardiac transplantation, the absence of AT(1) signaling accentuates the immunosuppressive effects of the calcineurin inhibitor cyclosporine. We conclude that inhibition of AT(1) receptor signaling should be useful as an anti-inflammatory and immunosuppressive therapy. Furthermore, the actions of the RAS to promote lymphocyte activation may contribute to inflammation that characterizes a number of diseases of the heart and the vascular system.


Subject(s)
Angiotensin II/physiology , Calcineurin/physiology , Lymphocyte Activation , Animals , Mice , Mice, Inbred C57BL , Peptidyl-Dipeptidase A/metabolism , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/physiology , Renin-Angiotensin System/physiology
9.
Kidney Int ; 56(5): 1855-62, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10571794

ABSTRACT

BACKGROUND: Genetically altered mice lacking the AT1A angiotensin II (Ang II) receptor were used to examine the role of AT1A receptors in regulating renal hemodynamics, sodium excretion, glomerulotubular balance, and Ang II levels in plasma and kidney during normal and volume-expanded conditions. METHODS: AT1A receptor-deficient mice and their wild-type controls were anesthetized with inactin and ketamine, and were prepared to allow intravenous infusions of solutions and measurements of aortic pressure and urine collections. Inulin and para-aminohippurate (PAH) solutions were infused intravenously for clearance determinations under conditions of euvolemia (2.5 microliter/min infusion of isotonic saline) or volume-expansion conditions (12.5 microliter/min). After three 30-minute urine collections, blood samples were collected, and kidneys were harvested. Plasma and kidney Ang II measurements were made by radioimmunoassay. RESULTS: In the euvolemic state, mean arterial pressures (MAPs) were significantly lower in the AT1A receptor-deficient mice (68 +/- 4 mm Hg) compared with wild-type controls (89 +/- 3 mm Hg). Despite the lower MAP, the glomerular filtration rate (GFR), renal plasma flow (RPF), absolute sodium excretion, and fractional sodium excretion were not significantly different between wild-type and AT1A-/- mice. Volume expansion did not alter MAP in wild-type mice, but significantly increased MAP in the AT1A-/- mice (68 +/- 4 to 83 +/- 5 mm Hg). Similar increases in GFR, RPF, absolute sodium excretion, and fractional sodium excretion in AT1A+/+ and AT1A-/- mice were observed. Glomerulotubular balance was not disrupted by the absence of AT1A receptors. During euvolemia, plasma Ang II concentrations were significantly higher in the AT1A-/- mice compared with wild-type mice (536 +/- 172 vs. 198 +/- 36 fmol/ml). Although volume expansion had no effect on plasma Ang II levels in the AT1A+/+ group, plasma Ang II concentrations were markedly suppressed in the AT1A-/- mice to levels that were not different from those in wild-type mice. In contrast, kidney tissue Ang II contents were reduced in the AT1A-/- mice and were not significantly altered during volume expansion in either the AT1A-/- or the AT1A+/+ mice. CONCLUSIONS: The absence of AT1A receptors does not impair chronic regulation of renal blood flow, GFR, or glomerulotubular balance. The prompt restoration of MAP following volume expansion suggests that low blood pressure in the AT1A receptor-deficient mice is primarily due to reduced effective plasma and extracellular fluid volume. Normalization of plasma Ang II levels with volume expansion demonstrates a dominant effect of extracellular fluid volume and blood pressure over AT1A receptor-mediated short-loop feedback in the regulation of plasma Ang II levels.


Subject(s)
Extracellular Space/physiology , Kidney/physiology , Receptors, Angiotensin/physiology , Angiotensin II/blood , Animals , Blood Pressure , Glomerular Filtration Rate , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/genetics , Renal Circulation , Sodium/metabolism
10.
Hypertension ; 33(1 Pt 2): 482-6, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9931152

ABSTRACT

Angiotensin (Ang) type 1a (AT1a) receptors are critical in the control of blood pressure and water balance. Experiments were performed to determine the influence of dehydration on brain Ang receptors and plasma vasopressin (VP) in mice lacking this receptor. Control or AT1a knockout (AT1aKO) male mice were give water ad libitum or deprived of water for 48 hours. Animals were anesthetized with halothane, blood samples were collected by heart puncture, and brains were processed for Ang-receptor autoradiography with 125I-sarthran (0.4 nmol/L). Dehydration produced an increase in AT1 receptors in the paraventricular nucleus (PVN) and anterior pituitary (AP) in control mice (PVN: 70+/-16 versus 146+/-10 fmol/mg protein; AP: 41+/-7 versus 86+/-15 fmol/mg protein). No changes were noted in the median preoptic nucleus. The majority of the brain receptors were of the AT1 subtype. There was little or no specific Ang binding in AT1aKO mice and no effect of dehydration. Plasma VP levels were elevated in the halothane-anesthetized animals (>200 pg/mL) with no significant effect of dehydration. A separate experiment was performed with decapitated mice anesthetized with pentobarbital. Dehydration increased plasma VP in control mice, from 3.3+/-0.6 to 13.3+/-4.7 pg/mL, whereas no change was noted in the AT1aKO mice, 5.1+/-0.3 versus 6.1+/-0.7 pg/mL (water versus dehydration). These results demonstrate a differential response to dehydration in mice lacking AT1a receptors. There was no evidence for AT1 receptors of any subtype in the brain regions examined and no effect of dehydration on VP secretion or brain Ang receptors.


Subject(s)
Brain/metabolism , Dehydration/physiopathology , Receptors, Angiotensin/physiology , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacokinetics , Animals , Autoradiography , Dehydration/genetics , Iodine Radioisotopes , Losartan/pharmacology , Male , Mice , Mice, Knockout , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary Gland, Anterior/metabolism , Preoptic Area/metabolism , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/deficiency , Receptors, Angiotensin/genetics , Reference Values , Vasopressins/blood , Water Deprivation
11.
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
12.
Proc Natl Acad Sci U S A ; 95(26): 15496-501, 1998 Dec 22.
Article in English | MEDLINE | ID: mdl-9860997

ABSTRACT

The classically recognized functions of the renin-angiotensin system are mediated by type 1 (AT1) angiotensin receptors. Whereas man possesses a single AT1 receptor, there are two AT1 receptor isoforms in rodents (AT1A and AT1B) that are products of separate genes (Agtr1a and Agtr1b). We have generated mice lacking AT1B (Agtr1b -/-) and both AT1A and AT1B receptors (Agtr1a -/-Agtr1b -/-). Agtr1b -/- mice are healthy, without an abnormal phenotype. In contrast, Agtr1a -/-Agtr1b -/- mice have diminished growth, vascular thickening within the kidney, and atrophy of the inner renal medulla. This phenotype is virtually identical to that seen in angiotensinogen-deficient (Agt-/-) and angiotensin-converting enzyme-deficient (Ace -/-) mice that are unable to synthesize angiotensin II. Agtr1a -/-Agtr1b -/- mice have no systemic pressor response to infusions of angiotensin II, but they respond normally to another vasoconstrictor, epinephrine. Blood pressure is reduced substantially in the Agtr1a -/- Agtr1b -/- mice and following administration of an angiotensin converting enzyme inhibitor, their blood pressure increases paradoxically. We suggest that this is a result of interruption of AT2-receptor signaling. In summary, our studies suggest that both AT1 receptors promote somatic growth and maintenance of normal kidney structure. The absence of either of the AT1 receptor isoforms alone can be compensated in varying degrees by the other isoform. These studies reaffirm and extend the importance of AT1 receptors to mediate physiological functions of the renin-angiotensin system.


Subject(s)
Angiotensin II/physiology , Blood Pressure/genetics , Growth/genetics , Kidney/abnormalities , Receptors, Angiotensin/physiology , Adrenal Glands/metabolism , Angiotensin II/pharmacology , Angiotensinogen/deficiency , Angiotensinogen/genetics , Angiotensinogen/physiology , Animals , Atrophy , Blood Pressure/drug effects , Crosses, Genetic , Epinephrine/pharmacology , Female , Homozygote , Humans , Kidney/pathology , Kidney/physiology , Kidney Medulla/pathology , Male , Mice , Mice, Knockout , Phenotype , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA, Messenger/genetics , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/deficiency , Receptors, Angiotensin/genetics , Renal Circulation/genetics , Restriction Mapping , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
13.
Am J Physiol ; 275(6): F922-7, 1998 12.
Article in English | MEDLINE | ID: mdl-9843909

ABSTRACT

The effect of the adenosine type 1 receptor agonist N6-cyclohexyladenosine (CHA) on glomerular vascular reactivity was studied in male angiotensin II type 1A (AT1A) receptor knockout mice (9). Vascular reactivity was assessed as the response of stop-flow pressure (PSF) to infusion of CHA into loops of Henle using micropuncture techniques. In AT1A +/+ mice at ambient arterial blood pressure (96.7 +/- 2.8 mmHg), the presence of CHA (10 (-5) M) in the perfusate increased PSF responses from 6.8 +/- 0.6 to 14.3 +/- 0.9 mmHg when the loop of Henle of the index nephron was perfused and from 0.7 +/- 0.3 to 12.3 +/- 1.0 mmHg when the loop of an adjacent nephron was perfused. At reduced arterial blood pressure (82.8 +/- 1. 3 mmHg), index nephron perfusion with CHA increased PSF responses from 4.5 +/- 0.3 to 9.4 +/- 0.4 mmHg. In AT1A -/- mice with a mean arterial blood pressure of 80 +/- 1.9 mmHg, CHA increased PSF responses only from 0.1 +/- 0.3 to 3.6 +/- 0.54 mmHg during index nephron perfusion and from 0.25 +/- 0.2 to 2.7 +/- 0.55 mmHg during adjacent nephron perfusion, significantly less than in wild-type animals (P < 0.001). Responses to CHA were intermediate in AT1A +/- mice. Thus AT1A receptor knockout mice show a markedly reduced constrictor response to CHA both in the presence and absence of simultaneous activation of the tubuloglomerular feedback system. These data support the notion of a functional interaction between adenosine and angiotensin II in the regulation of afferent arteriolar tone.


Subject(s)
Kidney Glomerulus/blood supply , Receptors, Angiotensin/deficiency , Receptors, Purinergic P1/physiology , Renal Circulation/physiology , Vasoconstriction/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Blood Pressure/physiology , Male , Mice , Mice, Knockout/genetics , Perfusion , Pressure , RNA, Messenger/metabolism , Receptors, Angiotensin/genetics , Receptors, Purinergic P1/genetics , Vasomotor System/drug effects
14.
Am J Physiol ; 275(6): F955-61, 1998 12.
Article in English | MEDLINE | ID: mdl-9843913

ABSTRACT

The actions of prostaglandin (PG) E2 are mediated by four distinct classes of PGE2 E-prostanoid (EP) receptors (EP1 through EP4). However, the in vivo functions of the individual EP receptor subtypes have not been delineated. To study the functions of one of these subtypes, the EP3 receptor, we generated EP3-deficient (-/-) mice by gene targeting. EP3 -/- animals survived in expected numbers, reproduced, and had no obvious abnormalities in their major organ systems. Because the EP3 receptor is expressed at high levels in the renal medulla and cortical collecting duct, and because previous studies have suggested that the EP3 receptor might antagonize the effects of vasopressin in the distal nephron, we examined urinary concentrating functions in EP3 -/- mice. Basal urine osmolality (UOsm) was similar in groups of EP3 -/- and wild-type (EP3 +/+) mice. However, after inhibition of endogenous PGE2 production by indomethacin, UOsm increased significantly in EP3 +/+ but not in EP3 -/- mice. Despite this insensitivity to acute inhibition of prostanoid production, EP3 -/- mice concentrated and diluted their urine normally in response to a series of physiological stimuli. This suggests that PGE2 acts through the EP3 receptor to modulate urinary concentrating mechanisms in the kidney, but these effects are not essential for normal regulation of urinary osmolality.


Subject(s)
Kidney Concentrating Ability/physiology , Receptors, Prostaglandin E/physiology , Animals , Cyclooxygenase Inhibitors/pharmacology , Deamino Arginine Vasopressin/pharmacology , Indomethacin/pharmacology , Kidney/physiology , Mice , Mice, Knockout/genetics , Osmolar Concentration , RNA, Messenger/metabolism , Receptors, Prostaglandin E/genetics , Renal Agents/pharmacology , Urine/chemistry
15.
Am J Physiol ; 274(3): H868-73, 1998 03.
Article in English | MEDLINE | ID: mdl-9530198

ABSTRACT

Mechanisms controlling cardiac growth are under intense investigation. Among these, the renin-angiotensin system has received great interest. In the current study, we tested the hypothesis that the renin-angiotensin system was not an obligate factor in cardiac hypertrophy. We examined the left ventricular hypertrophic response to a pressure overload in mice devoid of the AT1A receptor, the putative major effector of the growth response of the renin-angiotensin system. Aortic banding produced similar transband gradients in wild-type and AT1A knockout mice. The left ventricular mass-to-body weight ratio increased from 3.44 +/- 0.08 to 5.62 +/- 0.25 in wild-type ascending aortic-banded mice. The response in the knockout mice was not different (from 2.97 +/- 0.13 to 5.24 +/- 0.37). We conclude that the magnitude of cardiac hypertrophy is not affected by the absence of the AT1A receptor and its signaling pathway and that this component of the renin-angiotensin system is not necessary in cardiac hypertrophy.


Subject(s)
Angiotensin II/physiology , Cardiomegaly/etiology , Receptors, Angiotensin/deficiency , Animals , Blood Pressure , Body Weight , Heterozygote , Mice , Mice, Knockout , Receptor, Angiotensin, Type 1 , Renin/physiology
16.
Am J Physiol ; 274(1): F43-50, 1998 01.
Article in English | MEDLINE | ID: mdl-9458822

ABSTRACT

To examine the role of the type 1A (AT1A) angiotensin receptor in renal growth and development, we analyzed F2 progeny from a series of crosses between F1 mice that were heterozygous for a targeted disruption of the AT1A receptor gene [Agtr1A-(+/-)]. Among 21-day-old weanling F2 mice, we found that 194 (32%) were homozygous for the wild-type allele Agtr1A-(+/+), 299 (49%) were Agtr1A-(+/-), and 119 (19%) were Agtr1A-(-/-). This differed significantly from the proportions predicted by Mendelian genetics (P = 0.01), suggesting that the complete absence of AT1A receptors is associated with a mild survival disadvantage. Agtr1A-(-/-) mice grew normally, and we found no significant differences in body weight or heart and kidney weights in Agtr1A-(+/+) and Agtr1A-(-/-) mice examined at 21, 60, and 100 days. Protein and DNA content of kidneys and hearts were also similar in weanling or adult Agtr1A-(+/+) and Agtr1A-(-/-) mice. By light microscopy with immunohistochemistry, kidneys from Agtr1A-(-/-) were essentially normal, with two exceptions: 1) there was marked hypertrophy of the juxtaglomerular apparatus (JGA) and proximal expansion of renin-producing cells along the afferent arterioles, and 2) some glomeruli showed evidence of mesangial expansion. We did not find the severe renal vascular lesions or papillary atrophy that have been observed in angiotensinogen- or angiotensin converting enzyme-deficient animals. We conclude that the AT1A receptor is not essential for the normal organogenesis of the kidney; however, its absence is associated with mild mesangial expansion and JGA hypertrophy.


Subject(s)
Angiotensin II/metabolism , Kidney/growth & development , Receptors, Angiotensin/deficiency , Receptors, Angiotensin/genetics , Animals , Arterioles/pathology , Atrophy , Crosses, Genetic , Glomerular Mesangium/pathology , Glomerular Mesangium/ultrastructure , Heterozygote , Hypertrophy , Juxtaglomerular Apparatus/pathology , Kidney/cytology , Kidney/pathology , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Kidney Tubules, Proximal/pathology , Mice , Mice, Knockout , Receptor, Angiotensin, Type 1 , Renal Circulation , Renin/biosynthesis
17.
Am J Physiol ; 273(2 Pt 2): F315-20, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9277593

ABSTRACT

Experiments were performed in a recently generated strain of mice with an angiotensin II AT1A-receptor null mutation (M. Ito, M. I. Oliverio, P. J. Mannon, C. F. Best, N. Maeda, O. Smithies, and T. M. coffman. Proc. Natl. Acad. Sci. USA 92: 3521-3525, 1995) to examine the effects of chronic AT1A receptor deficiency on tubuloglomerular feeback (TGF) responses. All animals were genotyped by polymerase chain reaction using primers designed to amplify sequences from the deleted AT1A gene and from the neomycin resistance gene. Normal mice (AT1A +/+) and mice heterozygous (AT1A +/-) and homozygous (AT1A -/-) for the gene disruption were anesthetized, and stop-flow pressures (PSF) were determined during changes in loop perfusion rate with previously established micropuncture methods. In five AT1A +/+ mice (26 tubules) mean PSF at zero loop flow was 37.2 +/- 1.5 mmHg, falling to 28.2 +/- 1.9 mmHg at a flow of 45 nl/min (P < 0.0001). Flow rate causing the half-maximum response (V1/2) was 8.7 +/- 0.4 nl/min. In four AT1A +/- animals (19 tubules) mean PSF at zero flow was 39.9 +/- 2.4 mmHg, falling to 34.8 +/- 2.7 mmHg at 45 nl/min (mean V1/2 8.6 +/- 1.04 nl/min). In five AT1A -/- mice (24 tubules) PSF was not significantly affected by loop flow with PSF averaging 33.9 +/- 1.7 mmHg at zero flow and 33.2 +/- 1.6 mmHg at 45 nl/min (not significant). Mean arterial blood pressures in the anesthetized and laparotomized mice were 91.8 +/- 2.2, 97.1 +/- 3, and 80.7 +/- 3.2 mmHg in the AT1A +/+, AT1A +/-, and AT1A -/- animals, respectively. Blood pressure responses to exogenous angiotensin II were greatly blunted in the AT1A -/- mice. We conclude that AT1A receptor-mediated effects of angiotensin II are in essential component of TGF responsiveness under chronic conditions. Our studies show the feasibility of using complex micropuncture methods in mice, an approach that widens the potential of genetically altered mouse strains as experimental models.


Subject(s)
Kidney Glomerulus/physiology , Kidney Tubules/physiology , Receptors, Angiotensin/deficiency , Animals , Feedback , Glomerular Filtration Rate , Loop of Henle/physiology , Mice , Mice, Knockout , Nephrons/physiology , Perfusion , Punctures
18.
Am J Physiol ; 272(4 Pt 2): F515-20, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9140053

ABSTRACT

Most of the classic functions of the renin-angiotensin system are mediated by type 1 (AT1) angiotensin receptors, of which two subtypes, AT1A and AT1B, have been identified. However, distinct functions for these two AT1 receptors have been difficult to separate. We examined the pressor effects of angiotensin II in Agtr1A -/- mice, which lack AT1A receptors. In enalapril-pretreated Agtr1A -/- mice, angiotensin II caused significant and dose-proportional increases in mean arterial pressure. This pressor response was not blocked by pretreatment with sympatholytic agents but was completely inhibited by the AT1-receptor antagonists, losartan and candesartan, suggesting that it is directly mediated by AT1B receptors. Chronic treatment of Agtr1A -/- mice with losartan reduced systolic blood pressure from 80 +/- 5 to 72 +/- 4 mmHg (P < 0.04), suggesting a role for AT1B receptors in chronic blood pressure regulation. These studies provide the first demonstration of in vivo pressor effects mediated by AT1B receptors and demonstrate that, when AT1A receptors are absent, the AT1B receptor contributes to the regulation of resting blood pressure.


Subject(s)
Angiotensin II/pharmacology , Blood Pressure/drug effects , Kidney/physiology , Receptors, Angiotensin/deficiency , Receptors, Angiotensin/physiology , Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds/pharmacology , Enalapril/pharmacology , Hexamethonium/pharmacology , Imidazoles/pharmacology , Kidney/drug effects , Losartan , Mice , Mice, Knockout , Phentolamine/pharmacology , RNA, Messenger/biosynthesis , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/drug effects , Renin/biosynthesis , Sympatholytics/pharmacology , Tetrazoles/pharmacology , Time Factors , Transcription, Genetic
19.
Clin Cardiol ; 20(1): 3-6, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8994730

ABSTRACT

The renin-angiotensin system regulates blood pressure and sodium homeostasis through a series of coordinated substrate-enzyme interactions. These interactions result in the production of angiotensin II (AII), which exerts a number of diverse biologic effects mediated through AII cell-surface receptors. Dysregulation of this system is implicated in the pathogenesis of various forms of hypertension. Traditional therapy for hypertension has included angiotensin-converting enzyme inhibitors, which block the production of AII. However, a new class of drugs, AT1-receptor blockers, now offers a number of benefits by specifically blocking the effects of AII at its physiologically relevant receptor.


Subject(s)
Angiotensin II/physiology , Angiotensin Receptor Antagonists , Hypertension/drug therapy , Antihypertensive Agents/therapeutic use , Blood Pressure/physiology , Humans , Hypertension/physiopathology , Receptors, Angiotensin/physiology , Renin-Angiotensin System/physiology
20.
Proc Natl Acad Sci U S A ; 92(8): 3521-5, 1995 Apr 11.
Article in English | MEDLINE | ID: mdl-7724593

ABSTRACT

The renin-angiotensin system plays a critical role in sodium and fluid homeostasis. Genetic or acquired alterations in the expression of components of this system are strongly implicated in the pathogenesis of hypertension. To specifically examine the physiological and genetic functions of the type 1A receptor for angiotensin II, we have disrupted the mouse gene encoding this receptor in embryonic stem cells by gene targeting. Agtr1A(-/-) mice were born in expected numbers, and the histomorphology of their kidneys, heart, and vasculature was normal. AT1 receptor-specific angiotensin II binding was not detected in the kidneys of homozygous Agtr1A(-/-) mutant animals, and Agtr1A(+/-) heterozygotes exhibited a reduction in renal AT1 receptor-specific binding to approximately 50% of wild-type [Agtr1A(+/+)] levels. Pressor responses to infused angiotensin II were virtually absent in Agtr1A(-/-) mice and were qualitatively altered in Agtr1A(+/-) heterozygotes. Compared with wild-type controls, systolic blood pressure measured by tail cuff sphygmomanometer was reduced by 12 mmHg (1 mmHg = 133 Pa) in Agtr1A(+/-) mice and by 24 mmHg in Agtr1A(-/-) mice. Similar differences in blood pressure between the groups were seen when intraarterial pressures were measured by carotid cannulation. These studies demonstrate that type 1A angiotensin II receptor function is required for vascular and hemodynamic responses to angiotensin II and that altered expression of the Agtr1A gene has marked effects on blood pressures.


Subject(s)
Angiotensin II/metabolism , Blood Pressure/genetics , Hypertension/etiology , Receptors, Angiotensin/genetics , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Animals , Blood Pressure/drug effects , Electroporation , Female , Gene Targeting , Heterozygote , Homozygote , Kidney/anatomy & histology , Kidney/chemistry , Male , Mice , Mice, Knockout , Perfusion , Receptors, Angiotensin/deficiency , Systole
SELECTION OF CITATIONS
SEARCH DETAIL