Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Curr Med Chem ; 23(42): 4735-4752, 2016.
Article in English | MEDLINE | ID: mdl-27839506

ABSTRACT

BACKGROUND: Renin-angiotensin system (RAS) and its main product Angiotensin II (AngII) are in the focus of the pharmacological industry mainly because of hypertension treatment. Up-regulated RAS is generally associated with cardiovascular diseases and consequent organs injuries. The classic inhibition of RAS is based on the blocking of the type 1 AngII receptors and inhibition of ACE. The concept of the circulating and tissue RAS opens new challenges for the drug targeting. In spite of a big effort invested, in some cases a traditional RAS manipulation is struggling with unwanted side effects and/or resistance to treatment. OBJECTIVE: To improve the efficiency of the classic RAS inhibitors specific complications issuing from feed-back circuits inside the RAS have to be elucidated. Moreover, new peptidases identified in the AngII biosynthesis and Angiotensin 1-7/MAS pathways with opposing effects to AngII are tested for the clinical use. The aim of this review is also to bring attention to new tools in RAS manipulation based on the RNA interference (RNAi). RNAi employs small non-coding nucleic acids that interfere with the mRNA translation. The usefulness of this approach has been demonstrated in the treatment of oncological diseases and progress was also made in the field of the cardiovascular medicine. CONCLUSION: We suppose that in the near future, in addition to traditional pharmacological tools, RNAi will contribute to the control of RAS and AngII production. RNAi may also be of importance in the manipulation of tissue RAS that is not easily accessible by the traditional chemical substances.


Subject(s)
Angiotensin II/metabolism , Drug Discovery/methods , Angiotensin II/deficiency , Angiotensin II/genetics , Animals , Blood Pressure/drug effects , Blood Pressure/genetics , Humans , RNA Interference , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/genetics , Renin-Angiotensin System/physiology
2.
J Mol Cell Cardiol ; 97: 180-90, 2016 08.
Article in English | MEDLINE | ID: mdl-27210827

ABSTRACT

Angiotensin II (AngII) type 1 receptor blockers (ARBs) have been effectively used in hypertension and cardiac remodeling. However, the differences among them are still unclear. We designed this study to examine and compare the effects of several ARBs widely used in clinics, including Olmesartan, Candesartan, Telmisartan, Losartan, Valsartan and Irbesartan, on the ACE-AngII-AT1 axis and the ACE2-Ang(1-7)-Mas axis during the development of cardiac remodeling after pressure overload. Although all of the six ARBs, attenuated the development of cardiac hypertrophy and heart failure induced by transverse aortic constriction (TAC) for 2 or 4weeks in the wild-type mice evaluated by echocardiography and hemodynamic measurements, the degree of attenuation by Olmesartan, Candesartan and Losartan tended to be larger than that of the other three drugs tested. Additionally, the degree of downregulation of the ACE-AngII-AT1 axis and upregulation of the ACE2-Ang(1-7)-Mas axis was higher in response to Olmesartan, Candesartan and Losartan administration in vivo and in vitro. Moreover, in angiotensinogen-knockdown mice, TAC-induced cardiac hypertrophy and heart failure were inhibited by Olmesartan, Candesartan and Losartan but not by Telmisartan, Valsartan and Irbesartan administration. Furthermore, only Olmesartan and Candesartan could downregulate the ACE-AngII-AT1 axis and upregulate the ACE2-Ang(1-7)-Mas axis in vitro. Our data suggest that Olmesartan, Candesartan and Losartan could effectively inhibit pressure overload-induced cardiac remodeling even when with knockdown of Ang II, possibly through upregulation of the expression of the ACE2-Ang(1-7)-Mas axis and downregulation of the expression of the ACE-AngII-AT1 axis. In contrast, Telmisartan, Valsartan and Irbesartan only played a role in the presence of AngII, and Losartan had no effect in the presence of AngII in vitro.


Subject(s)
Angiotensin II/metabolism , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Ventricular Remodeling/drug effects , Angiotensin II/deficiency , Angiotensin II/genetics , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Blood Pressure/drug effects , Cardiomegaly/etiology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Disease Models, Animal , Echocardiography , Hemodynamics/drug effects , Hypertension/complications , Hypertension/physiopathology , Male , Mice , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Proto-Oncogene Mas , RNA, Small Interfering/chemistry
3.
J Am Soc Nephrol ; 23(7): 1181-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22518004

ABSTRACT

Angiotensin II content in the kidney is much higher than in the plasma, and it increases more in kidney diseases through an uncertain mechanism. Because the kidney abundantly expresses angiotensinogen mRNA, transcriptional dysregulation of angiotensinogen within the kidney is one potential cause of increased renal angiotensin II in the setting of disease. Here, we observed that kidney-specific angiotensinogen knockout mice had levels of renal angiotensinogen protein and angiotensin II that were similar to those levels of control mice. In contrast, liver-specific knockout of angiotensinogen nearly abolished plasma and renal angiotensinogen protein and renal tissue angiotensin II. Immunohistochemical analysis in mosaic proximal tubules of megalin knockout mice revealed that angiotensinogen protein was incorporated selectively in megalin-intact cells of the proximal tubule, indicating that the proximal tubule reabsorbs filtered angiotensinogen through megalin. Disruption of the filtration barrier in a transgenic mouse model of podocyte-selective injury increased renal angiotensin II content and markedly increased both tubular and urinary angiotensinogen protein without an increase in renal renin activity, supporting the dependency of renal angiotensin II generation on filtered angiotensinogen. Taken together, these data suggest that liver-derived angiotensinogen is the primary source of renal angiotensinogen protein and angiotensin II. Furthermore, an abnormal increase in the permeability of the glomerular capillary wall to angiotensinogen, which characterizes proteinuric kidney diseases, enhances the synthesis of renal angiotensin II.


Subject(s)
Angiotensin II/metabolism , Angiotensinogen/metabolism , Kidney/metabolism , Liver/metabolism , Angiotensin II/deficiency , Angiotensin II/genetics , Angiotensinogen/deficiency , Angiotensinogen/genetics , Animals , Kidney/pathology , Liver/pathology , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Animal , Phenotype , RNA, Messenger/metabolism
4.
Circ Res ; 104(1): 113-23, 2009 Jan 02.
Article in English | MEDLINE | ID: mdl-19023134

ABSTRACT

By mimicking sympathetic stimulation in vivo, we previously reported that mice globally lacking serotonin 5-HT(2B) receptors did not develop isoproterenol-induced left ventricular hypertrophy. However, the exact cardiac cell type(s) expressing 5-HT(2B) receptors (cardiomyocytes versus noncardiomyocytes) involved in pathological heart hypertrophy was never addressed in vivo. We report here that mice expressing the 5-HT(2B) receptor solely in cardiomyocytes, like global 5-HT(2B) receptor-null mice, are resistant to isoproterenol-induced cardiac hypertrophy and dysfunction, as well as to isoproterenol-induced increases in cytokine plasma-levels. These data reveal a key role of noncardiomyocytes in isoproterenol-induced hypertrophy in vivo. Interestingly, we show that primary cultures of angiotensinogen null adult cardiac fibroblasts are releasing cytokines on stimulation with either angiotensin II or serotonin, but not in response to isoproterenol stimulation, demonstrating a critical role of angiotensinogen in adrenergic-dependent cytokine production. We then show a functional interdependence between AT(1)Rs and 5-HT(2B) receptors in fibroblasts by revealing a transinhibition mechanism that may involve heterodimeric receptor complexes. Both serotonin- and angiotensin II-dependent cytokine production occur via a Src/heparin-binding epidermal growth factor-dependent transactivation of epidermal growth factor receptors in cardiac fibroblasts, supporting a common signaling pathway. Finally, we demonstrate that 5-HT(2B) receptors are overexpressed in hearts from patients with congestive heart failure, this overexpression being positively correlated with cytokine and norepinephrine plasma levels. Collectively, these results reveal for the first time that interactions between AT(1) and 5-HT(2B) receptors coexpressed by noncardiomyocytes are limiting key events in adrenergic agonist-induced, angiotensin-dependent cardiac hypertrophy. Accordingly, antagonists of 5-HT(2B) receptors might represent novel therapeutics for sympathetic overstimulation-dependent heart failure.


Subject(s)
Fibroblasts/physiology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Myocardium/pathology , Receptor, Angiotensin, Type 1/physiology , Receptor, Serotonin, 5-HT2B/physiology , Adult , Angiotensin II/deficiency , Angiotensin II/physiology , Angiotensin II/toxicity , Animals , Cells, Cultured/metabolism , Cytokines/blood , Cytokines/metabolism , ErbB Receptors/physiology , Female , Fibroblasts/drug effects , Heart Failure/chemically induced , Heart Failure/drug therapy , Heart Failure/pathology , Heparin-binding EGF-like Growth Factor , Humans , Hypertrophy, Left Ventricular/chemically induced , Hypertrophy, Left Ventricular/prevention & control , Intercellular Signaling Peptides and Proteins/physiology , Isoproterenol/toxicity , Male , Mice , Mice, Knockout , Mice, Transgenic , Middle Aged , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Norepinephrine/physiology , Protein Interaction Mapping , Serotonin 5-HT2 Receptor Antagonists , Serotonin Antagonists/therapeutic use , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/physiology
5.
Cell Metab ; 6(6): 506-12, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18054319

ABSTRACT

An overactive renin-angiotensin system is associated with obesity and the metabolic syndrome. However, the mechanisms behind it are unclear. Cleaving angiotensinogen to angiotensin I by renin is a rate-limiting step of angiotensin II production, but renin is suggested to have angiotensin-independent effects. We generated mice lacking renin (Ren1c) using embryonic stem cells from C57BL/6 mice, a strain prone to diet-induced obesity. Ren1c(-/-) mice are lean, insulin sensitive, and resistant to diet-induced obesity without changes in food intake and physical activity. The lean phenotype is likely due to a higher metabolic rate and gastrointestinal loss of dietary fat. Most of the metabolic changes in Ren1c(-/-) mice were reversed by angiotensin II administration. These results support a role for angiotensin II in the pathogenesis of diet-induced obesity and insulin resistance.


Subject(s)
Dietary Fats/metabolism , Energy Metabolism , Obesity/prevention & control , Renin/deficiency , Adipose Tissue/metabolism , Angiotensin II/deficiency , Angiotensin II/pharmacology , Animals , Basal Metabolism , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Dietary Fats/administration & dosage , Insulin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Phenotype , Renin/genetics , Thinness/genetics , Thinness/metabolism
6.
Cell Tissue Res ; 323(2): 313-20, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16189718

ABSTRACT

We have attempted to elucidate the mechanism by which endothelial-type nitric oxide synthase (eNOS) is regulated in the kidney, with special reference to the role of renal hemodynamics and angiotensin II (Ang II). We compared angiotensinogen gene knockout (Atg-/-) mice, which lacked Ang II (resulting in sodium/water depletion and severe hypotension), with wild-type (Atg+/+) mice. Using Western blot analysis and the NADPH diaphorase histochemical reaction, we found that the expression and activity of eNOS were markedly lower in the renal vessels of Atg-/- mice compared with wild-type (Atg+/+) mice. Dietary salt loading significantly enhanced renal eNOS levels and increased blood pressure in Atg-/- mice, but severe hypotension almost abolished the effects of salt loading. In contrast, in Atg+/+ mice, altered salt intake or hydralazine had no effect on renal eNOS levels. These results suggest that perfusion pressure plays an essential role in maintaining renal vascular eNOS activity, whereas Ang II plays a supportive role, especially when renal circulation is impaired.


Subject(s)
Angiotensin II/genetics , Blood Vessels/enzymology , Kidney/blood supply , Kidney/enzymology , Nitric Oxide Synthase Type II/metabolism , Angiotensin II/deficiency , Animals , Blood Pressure/drug effects , Blood Vessels/drug effects , Blotting, Western , Hydralazine/pharmacology , Hypotension/chemically induced , Hypotension/genetics , Hypotension/physiopathology , Immunoenzyme Techniques , Kidney/physiopathology , Mice , Mice, Inbred ICR , Mice, Knockout , NADPH Dehydrogenase/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III , Sodium/administration & dosage , Sodium Chloride, Dietary , Water-Electrolyte Balance/genetics
7.
Am J Physiol Regul Integr Comp Physiol ; 289(6): R1763-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16109805

ABSTRACT

Angiotensinogen (AGT) is mainly expressed in glial cells in close proximity to renin-expressing neurons in the brain. We previously reported that glial-specific overexpression of ANG II results in mild hypertension. Here, we tested the hypothesis that glial-derived AGT plays an important role in blood pressure regulation in hypertensive mice carrying human renin (hREN) and human AGT transgenes under the control of their own endogenous promoters. To perform a glial-specific deletion of AGT, we used an AGT transgene containing loxP sites (hAGT(flox)), so the gene can be permanently ablated in the presence of cre-recombinase expression, driven by the glial fibrillary acidic protein (GFAP) promoter. Triple transgenic mice (RAC) containing a: 1) systemically expressed hREN transgene, 2) systemically expressed hAGT(flox) transgene, and 3) GFAP-cre-recombinase were generated and compared with double transgenic mice (RA) lacking cre-recombinase. Liver and kidney hAGT mRNA levels were unaltered in RAC and RA mice, as was the level of hAGT in the systemic circulation, consistent with the absence of cre-recombinase expression in those tissues. Whereas hAGT mRNA was present in the brain of RA mice (lacking cre-recombinase), it was absent from the brain of RAC mice expressing cre-recombinase, confirming brain-specific elimination of AGT. Immunohistochemistry revealed a loss of AGT immunostaining glial cells throughout the brain in RAC mice. Arterial pressure measured by radiotelemetry was significantly lower in RAC than RA mice and unchanged from nontransgenic control mice. These data suggest that there is a major contribution of glial-AGT to the hypertensive state in mice carrying systemically expressed hREN and hAGT genes and confirm the importance of a glial source of ANG II substrate in the brain.


Subject(s)
Angiotensin II/deficiency , Angiotensinogen/deficiency , Blood Pressure/physiology , Brain/physiology , Neuroglia/physiology , Renin/metabolism , Angiotensinogen/genetics , Animals , Baroreflex/physiology , Mice , Mice, Transgenic , Renin/genetics , Renin-Angiotensin System/physiology
8.
Eur J Pharmacol ; 493(1-3): 161-5, 2004 Jun 16.
Article in English | MEDLINE | ID: mdl-15189777

ABSTRACT

To explore the role of angiotensin II, we assessed hemodynamics and cardiac function in angiotensinogen-deficient mice in comparison to wild-type animals. Left ventricular end-diastolic diameter and wall thickness were evaluated by echocardiography and systolic and diastolic left ventricular function by pressure-volume relations using a micro-conductance catheter. Compared to wild-type animals, the angiotensinogen-deficient mice were hypotensive and showed impaired systolic function. The hearts were dilated, demonstrated by echocardiography and by a right-ward shift of the pressure-volume loops, but end-diastolic pressure, isovolumic relaxation (tau) and diastolic stiffness were unchanged. Afterload, however, was reduced leading to maintained cardiac output. Although a blockade of the renin-angiotensin system via angiotensin converting enzyme inhibitors or angiotensin AT1 receptor antagonist is beneficial after cardiac failure, the absence of angiotensin peptides during the ontogenesis leads to dilated cardiomyopathy.


Subject(s)
Angiotensin II/deficiency , Cardiomyopathy, Dilated/etiology , Disease Models, Animal , Angiotensin II/genetics , Angiotensinogen/deficiency , Angiotensinogen/genetics , Animals , Cardiomyopathy, Dilated/diagnosis , Echocardiography/instrumentation , Echocardiography/methods , Hemodynamics/physiology , Hypotension/etiology , Male , Mice , Mice, Knockout , Netherlands , Receptors, Angiotensin/physiology , Stroke Volume/physiology , Ventricular Function, Left/physiology
9.
Eur J Appl Physiol ; 91(2-3): 342-8, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14595564

ABSTRACT

The deletion (D) rather than insertion (I) allele of the angiotensin-converting enzyme (ACE) gene is associated with greater ACE activity. We examined: (1) the influence of posture change (recumbent to seated) and acute exercise on serum ACE and angiotensin II (Ang II) activity; (2) the relationship between ACE and Ang II levels; and (3) the influence of ACE genotype on changes in ACE and Ang II levels with posture and exercise. Recreationally active young male Caucasians (10 each of II, ID and DD genotypes) rested for 35 min supine then 15 min upright, took 20 min bicycle ergometric exercise at 70% maximum oxygen uptake, then rested for 40 min. Samples were taken throughout for ACE activity and Ang II levels. Supine ACE levels were dependent upon ACE genotype [24.8 (5.7), 26.9 (4.5), 45.5 (6.4) nmol His-Leu ml(-1) min(-1); II, ID, DD, respectively; P<0.00005] and thereafter. ACE activity rose with assumption of a seated posture [from 32.4 (10.9) nmol His-Leu ml(-1) min(-1) to 35.0 (11.5) nmol His-Leu ml(-1) min(-1), P<0.00001], the absolute rise being independent of genotype [3.22 (1.92), 1.6 (1.6), 2.4 (2.3) nmol His-Leu ml(-1) min(-1); II, ID, DD; P=0.22], unlike percentage change [12.8 (6.8), 5.6 (5.5), 5.3 (5.0)%; II, ID, DD; P<0.01, and P=0.004 for II vs presence of the D allele]. A further genotype-independent rise occurred with exercise [+2.9 (3.7) units, P<0.0003]. An associated rise in Ang II levels [30.3 (15.9), or 2587.9 (489.76)%, P<0.00001] was independent of ACE genotype or activity. Upright posture increases ACE activity, and this may be influenced by ACE genotype. ACE activity and Ang II levels rise independently with exercise in a non-genotype-dependent fashion.


Subject(s)
Angiotensin II/genetics , Angiotensin II/metabolism , Exercise/physiology , Peptidyl-Dipeptidase A/blood , Posture/physiology , Adaptation, Physiological/physiology , Adolescent , Adult , Angiotensin II/deficiency , Exercise Test/methods , Genotype , Humans , Male , Physical Exertion/physiology
10.
J Mol Cell Cardiol ; 34(11): 1455-62, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12431444

ABSTRACT

The local generation of all components of the renin-angiotensin system (RAS) in the heart has been the basis for the postulation of a tissue RAS in this organ. Since angiotensin II is involved in the induction of cardiac hypertrophy and fibrosis the local generation of this peptide may be of highest clinical importance. Several transgenic animal models have been generated to evaluate the functional importance of the cardiac RAS. We have established a new hypertensive mouse model lacking local angiotensinogen expression in the heart. In these animals, cardiac weight and collagen synthesis are increased compared to normotensive control mice but to a lesser extent than in mice with equally enhanced blood pressure but intact cardiac angiotensinogen generation. Thus, we have shown that local synthesis of this protein is involved but not essential in the development of cardiac hypertrophy and fibrosis.


Subject(s)
Cardiomegaly/etiology , Hypertension/complications , Renin-Angiotensin System/physiology , Angiotensin II/deficiency , Angiotensin II/pharmacology , Animals , Animals, Genetically Modified , Cardiomegaly/physiopathology , Fibrosis , Heart/drug effects , Hypertension/genetics , Hypertension/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Myocardial Infarction/etiology , Myocardium/metabolism , Myocardium/pathology , Organ Specificity , Peptidyl-Dipeptidase A/physiology , Rats , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/deficiency , Receptors, Angiotensin/physiology , Stress, Mechanical , Ventricular Remodeling
11.
Regul Pept ; 99(2-3): 209-16, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11384784

ABSTRACT

Angiotensin II (Ang II) is a potent vasoactive peptide and displays growth factor-like properties. Different high-affinity Ang II receptor subtypes (AT1A, AT1B and AT2) have been cloned. They are expressed in various brain structures. Additionally, it has been assumed that Mas could interact directly or indirectly with the renin-angiotensin system. The AT1 receptor mediates pressor and mitogenic effects of Ang II, whereas physiological function and signaling mechanisms of the AT2 receptor remain poorly understood. Recent reports have shown that Ang II could mediate apoptosis through AT2 receptors. Since the AT1A, AT2 and Mas knockout mice provide new tools for uncovering potential actions of Ang II, the cell number in different brain structures of male adult wild-type mice and mice deficient for AT1A, AT2 or Mas was evaluated to get more insight into the role of Ang II in central nervous system development. In nearly all investigated brain structures (cortex, hippocampus, amygdala, thalamus), the cell number was significantly higher in AT2-deficient mice in comparison to wild-type mice. To the contrary, in AT1A-deficient mice the cell number was significantly less than in controls in the lateral geniculate and the medial amygdaloid nucleus. However, cell numbers were not changed in Mas-knockout mice compared to their wild-types. These results show the contrary effects of both angiotensin receptors on cell growth and represent the first demonstration of their action on neuronal cell development evidenced in the adult mouse brain.


Subject(s)
Gene Deletion , Receptors, Angiotensin/deficiency , Receptors, Angiotensin/genetics , Angiotensin II/deficiency , Angiotensin II/physiology , Animals , Brain/cytology , Brain/enzymology , Brain/metabolism , Brain Chemistry/genetics , Cell Count , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/enzymology , Neurons/metabolism , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/physiology , Receptors, G-Protein-Coupled
12.
Behav Brain Res ; 100(1-2): 1-4, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10212048

ABSTRACT

Angiotensin II in the brain was shown to be involved in mechanisms influencing cardiovascular and electrolyte homeostasis, anxiety and learning. Here, we report behavioural studies in mice lacking angiotensinogen. We analysed learning and anxiety related behaviour using the Morris water maze task and the elevated plus maze task, respectively. In both tests no differences were found between control mice and angiotensin-deficient mice. This implicates that angiotensin does not influence learning and anxiety-related behaviour in mice under normal conditions.


Subject(s)
Angiotensin II/physiology , Arousal/physiology , Brain/physiology , Maze Learning/physiology , Mental Recall/physiology , Angiotensin II/deficiency , Animals , Escape Reaction/physiology , Male , Mice , Mice, Knockout
13.
Kidney Int ; 53(3): 548-55, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9507198

ABSTRACT

Angiotensinogen gene-knockout (Atg-/-) mice lacking angiotensin II exhibit chronic hypotension. The present study was designed to investigate pathophysiology of Atg-/- mice from the renal functional view. Wild-type (Atg+/+) and Atg-/- mice at 10 weeks of age were housed in metabolic cages for 24-hour urine collection. When provided free access to water, Atg-/- mice showed an increased urine output and a decreased urine osmolality compared with Atg+/+ mice. Urinary excretion and plasma levels of vasopressin were significantly higher in mutant mice than in wild-type mice. On the other hand, urinary excretion of aldosterone in mutant mice was suppressed to the levels under the detection limit of the assay system. The mean plasma aldosterone level of Atg-/- mice was suppressed to 30% of that of Atg+/+ mice. Plasma levels of creatinine, endogenous creatinine clearance, and urinary electrolyte excretion were not different between these mice. In Atg+/+ mice, urine osmolality was markedly increased from 1929 +/- 21 to 3314 +/- 402 mOsm/kg during water deprivation, whereas this parameter in Atg-/- mice did not change significantly (from 1413 +/- 121 to 1590 +/- 92 mOsm/kg). Urinary vasopressin excretion increased during water deprivation from 0.24 +/- 0.04 and 0.70 +/- 0.08 to 0.42 +/- 0.06 and 2.31 +/- 0.35 ng/mg creatinine in wild-type and mutant mice, respectively. Histologic study revealed interstitial inflammation, and atrophic changes in the tubules and papilla in Atg-/- mice. In conclusion, a genetic deficiency of angiotensinogen produced an impaired urine concentrating ability and tubulointerstitial lesions, indicating the critical role of angiotensinogen in developing normal tubular function and construction.


Subject(s)
Angiotensinogen/deficiency , Angiotensinogen/genetics , Kidney Concentrating Ability/genetics , Kidney Concentrating Ability/physiology , Aldosterone/blood , Aldosterone/urine , Angiotensin II/deficiency , Angiotensinogen/physiology , Animals , Creatinine/blood , Creatinine/urine , Kidney/pathology , Kidney/physiopathology , Mice , Mice, Knockout , Potassium/urine , Sodium/urine , Vasopressins/blood , Vasopressins/urine , Water Deprivation/physiology
14.
Kidney Int ; 53(3): 617-25, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9507206

ABSTRACT

Chronic volume depletion by dietary salt restriction causes marked decrease in glomerular filtration rate (GFR) with little increase in urine osmolality in angiotensinogen gene null mutant (Agt-/-) mice. Moreover, urine osmolality is insensitive to both water and vasopressin challenge. In contrast, in normal wild-type (Agt+/+) mice, GFR remains remarkably constant and urine osmolality is adjusted promptly. Changes in volume status also cause striking divergence in renal structure between Agt-/- and Agt+/+ mice. Thus, in contrast to the remarkably stable glomerular size of Agt+/+ mice, glomeruli of Agt-/- mice are atrophied during a low salt and hypertrophied during a high salt diet. Moreover, the renal papilla, a structure unique to mammals and essential for urine diluting and concentrating mechanisms, is hypoplastic in Agt-/- mice. Thus, angiotensin is essential for the two fundamental homeostatic functions of the mammalian kidney, namely stable GFR and high urine diluting and concentrating capacity during alteration in extracellular fluid (ECF) volume. This is not only accompanied by angiotensin's tonic effects on renal vasomotor tone and tubule transporters, but also accomplished through its capacity to affect the structure of both the glomerulus and the papilla directly or indirectly.


Subject(s)
Angiotensinogen/genetics , Angiotensinogen/physiology , Kidney Glomerulus/physiopathology , Kidney Tubules/physiopathology , Actins/metabolism , Angiotensin II/deficiency , Angiotensin II/physiology , Animals , Atrial Natriuretic Factor/blood , Blood Pressure/genetics , Blood Pressure/physiology , Female , Glomerular Filtration Rate/genetics , Glomerular Filtration Rate/physiology , Homeostasis , In Situ Hybridization , Kidney Concentrating Ability/genetics , Kidney Concentrating Ability/physiology , Kidney Glomerulus/pathology , Male , Mice , Mice, Knockout , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transforming Growth Factor beta/genetics
15.
Kidney Int Suppl ; 63: S201-4, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9407459

ABSTRACT

To localize angiotensin II type 1a (AT-1a) receptor and to reveal the physiological roles of angiotensin II in the renal microcirculation, we investigated the AT-1a gene deficient mice, generated by a targeted replacement of the AT-1a receptor loci by the lacZ gene (Sugaya et al, J Biol Chem 270: 18719, 1995). Immunohistochemical localization of beta-galactosidase was performed in the heterozygous mutant mice to reveal the expression sites of AT-1a. The AT-1a receptor (that is, beta-galactosidase) was expressed both in the afferent and efferent arteriolar smooth muscles and also in the mesangial cells. The effect of angiotensin II on glomerular arterioles was directly observed using the hydronephrotic mice. Angiotensin II similarly constricted both the afferent and efferent arterioles in the wild-type and heterozygous mutant mice in a dose-dependent manner. This constriction was completely abolished by an AT-1 antagonist, CV-11974. In the homozygous null mutant mice, however, angiotensin II did not affect the arterioles at all. Electron microscopic studies revealed that the mesangial cells made contact with the glomerular basement membrane (GBM) at the capillary neck and also with each other in the wild-type mice. However, in the homozygous null mutant mice, the mesangial cells lost the contact either with GBM or with each other and thus the capillary neck became remarkably wider. The mesangial matrix area appeared loose and enlarged, suggesting impaired mesangial matrix formation. In conclusion, via the AT-1a receptor, angiotensin II equally constricts both the afferent and efferent arterioles and plays an essential role in maintaining the normal glomerular function and structure.


Subject(s)
Angiotensin II/metabolism , Receptors, Angiotensin/metabolism , Renal Circulation/drug effects , Angiotensin II/deficiency , Angiotensin II/pharmacology , Animals , Arterioles/drug effects , Glomerular Mesangium/cytology , Glomerular Mesangium/drug effects , Glomerular Mesangium/metabolism , Hydronephrosis/genetics , Hydronephrosis/pathology , Immunohistochemistry , Mice , Microcirculation/drug effects , Receptors, Angiotensin/drug effects , Receptors, Angiotensin/genetics , beta-Galactosidase/metabolism
16.
Cell Tissue Res ; 278(1): 21-8, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7954700

ABSTRACT

Transgenic rats for the murine Ren-2 gene display high blood pressure, low circulating levels of angiotensin II, and high renin content in the adrenal glands. Moreover, transgenic rats possess an increased aldosterone secretion (maximal from 6 to 18 weeks of age), paralleling the development of hypertension. To investigate further the cytophysiology of the adrenal glands of this strain of rats, we performed a combined morphometric and functional study of the zona glomerulosa of 10-week-old female transgenic rats. Morphometry did not reveal notable differences between zona glomerulosa cells of transgenic and age- and sex-matched Sprague-Dawley rats, with the exception of a marked accumulation of lipid droplets, in which cholesterol and cholesterol esters are stored. The volume of the lipid-droplet compartment underwent a significant decrease when transgenic rats were previously injected with angiotensin II or ACTH. Dispersed zona glomerulosa cells of transgenic rats showed a significantly higher basal aldosterone secretion, but their response to angiotensin II and ACTH was similar to that of Sprague-Dawley animals. Angiotensin II-receptor number and affinity were not dissimilar in zona glomerulosa cells of transgenic and Sprague-Dawley rats. These data suggest that the sustained stimulation of the adrenal renin-angiotensin system in transgenic animals causes an increase in the accumulation in zona glomerulosa cells of cholesterol available for steroidogenesis, as indicated by the expanded volume of the lipid-droplet compartment and the elevated basal steroidogenesis. However, the basal hyperfunction of the zona glomerulosa in transgenic animals does not appear to be coupled with an enhanced responsivity to its main secretagogues, at least in terms of aldosterone secretion.


Subject(s)
Hyperaldosteronism/genetics , Hypertension/etiology , Mice/genetics , Rats/anatomy & histology , Renin-Angiotensin System/physiology , Renin/metabolism , Zona Glomerulosa/pathology , Adrenocorticotropic Hormone/pharmacology , Aldosterone/metabolism , Angiotensin II/deficiency , Angiotensin II/pharmacology , Animals , Animals, Genetically Modified , Cell Size , Cholesterol/analysis , Cholesterol Esters/analysis , Female , Hyperaldosteronism/complications , Hyperaldosteronism/pathology , Male , Rats/physiology , Rats, Sprague-Dawley , Receptors, Angiotensin/analysis , Secretory Rate , Zona Glomerulosa/chemistry , Zona Glomerulosa/metabolism
17.
Brain Res ; 632(1-2): 287-95, 1993 Dec 31.
Article in English | MEDLINE | ID: mdl-8149234

ABSTRACT

Stimulation of the peripheral renin-angiotensin system has been shown previously to decrease the voluntary intake of ethanol in the rat. The existence of a separate brain renin-angiotensin system, independent from that of the periphery, has been widely demonstrated. The brain renin-angiotensin system plays an important role in the regulation of water and electrolyte balance and neuroendocrine function. However, the role played by this system in the regulation of voluntary alcohol consumption has not yet been studied. The goal of the present work was to assess the feasibility of decreasing the voluntary alcohol intake in a strain of rats (Rapp SS/Jr rats) that have a genetic deficiency responsible for a low activity of the renin-angiotensin system and elevated alcohol intake. Adult Rapp SS/Jr rats received intraventricular transplants of fetal hypothalamic grafts (from normal donors), known to contain angiotensin-immunoreactive cell bodies. Our studies revealed that angiotensin-immunoreactivity in the cell bodies and fibres in the paraventricular, supraoptic and suprachiasmatic nuclei of the hypothalamus in Rapp SS/Jr rats was markedly reduced. Animals that had surviving grafts containing angiotensin-immunoreactive cell bodies in the dorsal third ventricle--but not in the ventral third ventricle, in the lateral ventricles, or sham operated animals--had a 40% decrease of their voluntary alcohol intake, when compared to their intake before surgery, or to the control group. However, water consumption was not reduced in both the sham and transplanted animals.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Alcohol Drinking , Angiotensin II/metabolism , Brain Tissue Transplantation , Drinking Behavior , Hypothalamus/transplantation , Renin-Angiotensin System/genetics , Alcohol Drinking/genetics , Alcoholism/prevention & control , Angiotensin II/deficiency , Animals , Cerebral Ventricles/metabolism , Fetal Tissue Transplantation , Male , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Suprachiasmatic Nucleus/metabolism , Supraoptic Nucleus/metabolism , Water
19.
South Med J ; 70(5): 634-6, 1977 May.
Article in English | MEDLINE | ID: mdl-193197

ABSTRACT

Hyperkalemia secondary to hyporeninemic hypoaldosteronism with a normal glucocorticoid function was diagnosed in a 47-year-old man with moderate renal insufficiency. Mineralocorticoid administration corrected the hyperkalemia. A probable explanation for hyporeninemia and hypoaldosteronism in this syndrome is that the primary defect is an inability to release renin and the resultant angiotensin deficiency leads to an aldosterone deficiency.


Subject(s)
Aldosterone/deficiency , Hyperkalemia/etiology , Acidosis/complications , Adrenocorticotropic Hormone/therapeutic use , Aldosterone/metabolism , Angiotensin II/deficiency , Chlorides/blood , Humans , Hyperkalemia/complications , Kidney Failure, Chronic/complications , Male , Middle Aged , Renin/blood , Renin/metabolism , Sodium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...