Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Pflugers Arch ; 470(7): 1127-1137, 2018 07.
Article in English | MEDLINE | ID: mdl-29455241

ABSTRACT

Pharmacological inhibition or genetic loss of function defects of the renin angiotensin aldosterone system (RAAS) causes compensatory renin cell hyperplasia and hyperreninemia. The triggers for the compensatory stimulation of renin synthesis and secretion in this situation may be multimodal. Since cyclooxygenase-2 (COX-2) expression in the macula densa is frequently increased in states of a defective RAAS, we have investigated a potential role of COX-2 and its derived prostaglandins for renin expression and secretion in aldosterone synthase-deficient mice (AS-/-) as a model for a genetic defect of the RAAS. In comparison with wild-type mice (WT), AS-/- mice had 9-fold and 30-fold increases of renin mRNA and of plasma renin concentrations (PRC), respectively. Renin immunoreactivity in the kidney cortex of AS-/- mice was 10-fold higher than in WT. Macula densa COX-2 expression was 5-fold increased in AS-/- kidneys relative to WT kidneys. Treatment of AS-/- mice with the COX-2 inhibitor SC-236 for 1 week lowered both renal renin mRNA and PRC by 70%. Hyperplastic renin cells in AS-/- kidneys were found to express the prostaglandin E2 receptors EP2 and EP4. Global deletion of EP2 receptors did not alter renin mRNA nor PRC values in AS-/- mice. Renin cell-specific inducible deletion of the EP4 receptor lowered renin mRNA and PRC by 25% in AS-/- mice. Renin cell-specific inducible deletion of the EP4 receptor in combination with global deletion of the EP2 receptor lowered renin mRNA and PRC by 70-75% in AS-/- mice. Lineage tracing of renin-expressing cells revealed that deletion of EP2 and EP4 leads to a preferential downregulation of perivascular renin expression. Our findings suggest that increased macula densa COX-2 activity in AS-/- mice triggers perivascular renin expression and secretion via prostaglandin E2.


Subject(s)
Cyclooxygenase 2/metabolism , Cytochrome P-450 CYP11B2/metabolism , Dinoprostone/metabolism , Hyperplasia/metabolism , Renin-Angiotensin System/physiology , Renin/metabolism , Animals , Cyclooxygenase Inhibitors/pharmacology , Down-Regulation/drug effects , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Renin-Angiotensin System/drug effects
2.
Hamostaseologie ; 36(4): 279-283, 2016 Nov 07.
Article in English | MEDLINE | ID: mdl-27487748

ABSTRACT

The platelet adenosine 5'-diphosphate (ADP) receptor P2Y12 (P2Y12R) plays a critical role in platelet aggregation. The present report illustrates an update of dysfunctional platelet P2Y12R mutations diagnosed with congenital lifelong bleeding problems. Described patients with heterozygous or homozygous substitution in the P2Y12R gene and qualitative abnormalities of the platelet P2Y12R are summarized. Recently, a further dysfunctional variant of P2Y12R has been identified in two brothers who presented with a lifelong severe bleeding disorder. During in vitro aggregation studies, the patient´s platelets show a markedly reduced and rapid reversible ADP-promoted aggregation. A homozygous c.561T>A substitution that changes the codon for His187 to Gln (p.His187Gln) in the P2Y12R gene has been identified. This mutation causes no change in receptor expression but decreases the affinity of the ligand for the receptor, even at high concentrations. Structure modelling studies indicated that the p.His187Gln mutation, located in the fifth transmembrane spanning domain (TM5), impairs conformational changes of the receptor. Structural integrity of the TM5 region is necessary for agonist and antagonist binding and for correct receptor function.


Subject(s)
Blood Platelet Disorders/immunology , Hemorrhage/genetics , Hemorrhage/immunology , Polymorphism, Single Nucleotide/genetics , Receptors, Purinergic P2Y12/genetics , Blood Platelet Disorders/genetics , Genetic Predisposition to Disease/genetics , Humans , Models, Genetic , Models, Immunological , Mutation/genetics
3.
Pflugers Arch ; 467(8): 1825-34, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25241776

ABSTRACT

Defects of the gap junction protein connexin 40 (Cx40) in renin-secreting cells (RSCs) of the kidney lead to a shift of the localization of RSCs from the media layer of afferent arterioles to the periglomerular interstitium. The dislocation of RSCs goes in parallel with elevated plasma renin levels, impaired pressure control of renin secretion, and hypertension. The reasons for the extravascular shift of RSCs and the blunted pressure regulation of renin secretion caused by the absence of Cx40 are still unclear. We have therefore addressed the question if Cx40 is essential for the metaplastic transformation of preglomerular vascular smooth muscle cells (SMCs) into RSCs and if Cx40 is essential for the pressure control of renin secretion from RSCs located in the media layer of afferent arterioles. For our study, we used mice lacking the angiotensin II type 1A (AT1A) receptors, which display a prominent and reversible salt-sensitive metaplastic transformation of SMCs into RSCs. This mouse line was crossed with Cx40-deficient mice to obtain AT1A and Cx40 double deleted mice. The kidneys of AT1A (-/-)Cx40(-/-) mice kept on normal salt (0.3 %) displayed RSCs both in the inner media layer of preglomerular vessels and in the periglomerular interstitium. In contrast to hypotensive AT1A (-/-) (mean bp syst 112 mmHg) and hypertensive Cx40(-/-) (mean bp syst 160 mmHg) mice AT1A (-/-)Cx40(-/-) mice were normotensive(mean bp syst 130 mmHg). Pressure regulation of renin secretion from isolated kidneys was normal in AT1A (-/-) mice, but was absent in AT1A (-/-)Cx40(-/-) mice alike in Cx40(-/-) mice. Low-salt diet (0.02 %) increased RSC numbers in the media layer, whilst high-salt diet (4 %) caused disappearance of RSCs in the media layer but not in the periglomerular interstitium. Blood pressure was clearly salt sensitive both in AT1A (-/-) and in AT1A (-/-)Cx40(-/-) mice but was shifted to higher pressure values in the latter genotype. Our data indicate that Cx40 is not a requirement for intramural vascular localization of RSCs nor for reversible metaplastic transformation of SMCs into RSCs. Therefore, the ectopic localization of RSCs in Cx40(-/-) kidneys is more likely due to a disturbed intercellular communication rather than being the result of chronic overactivation of the renin-angiotensin-aldosterone system or hypertension. Moreover, our findings suggest that Cx40 is a requirement for the pressure control of renin secretion irrespective of the localization of RSCs.


Subject(s)
Baroreflex , Cell Movement , Connexins/metabolism , Kidney/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pressoreceptors/metabolism , Renin-Angiotensin System , Renin/metabolism , Animals , Blood Pressure , Connexins/deficiency , Connexins/genetics , Diet, Sodium-Restricted , Female , Genotype , Hypertension/genetics , Hypertension/metabolism , Hypertension/physiopathology , Hypotension/genetics , Hypotension/metabolism , Hypotension/physiopathology , Kidney/blood supply , Mechanotransduction, Cellular , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Renin/genetics , Sodium Chloride, Dietary , Gap Junction alpha-5 Protein
4.
Am J Physiol Renal Physiol ; 307(11): F1215-26, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25339696

ABSTRACT

Both the processing and release of secretory granules involve water movement across granule membranes. It was hypothesized that the water channel aquaporin (AQP)1 directly contributes to the recruitment of renin-positive cells in the afferent arteriole. AQP1(-/-) and AQP1(+/+) mice were fed a low-salt (LS) diet [0.004% (wt/wt) NaCl] for 7 days and given enalapril [angiotensin-converting enzyme inhibitor (ACEI), 0.1 mg/ml] in drinking water for 3 days. There were no differences in plasma renin concentration at baseline. After LS-ACEI, plasma renin concentrations increased markedly in both genotypes but was significantly lower in AQP1(-/-) mice compared with AQP1(+/+) mice. Tissue renin concentrations were higher in AQP1(-/-) mice, and renin mRNA levels were not different between genotypes. Mean arterial blood pressure was not different at baseline and during LS diet but decreased significantly in both genotypes after the addition of ACEI; the response was faster in AQP1(-/-) mice but then stabilized at a similar level. Renin release after 200 µl blood withdrawal was not different. Isoprenaline-stimulated renin release from isolated perfused kidneys did not differ between genotypes. Cortical tissue norepinephrine concentrations were lower after LS-ACEI compared with baseline with no difference between genotypes. Plasma nitrite/nitrate concentrations were unaffected by genotype and LS-ACEI. In AQP1(-/-) mice, the number of afferent arterioles with recruitment was significantly lower compared with AQP1(+/+) mice after LS-ACEI. We conclude that AQP1 is not necessary for acutely stimulated renin secretion in vivo and from isolated perfused kidneys, whereas recruitment of renin-positive cells in response to chronic stimulation is attenuated or delayed in AQP1(-/-) mice.


Subject(s)
Aquaporin 1/metabolism , Kidney/metabolism , Renin/biosynthesis , Renin/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Aquaporin 1/genetics , Blood Pressure/genetics , Blood Pressure/physiology , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Diet, Sodium-Restricted , Female , In Vitro Techniques , Kidney/cytology , Kidney/drug effects , Male , Mice , Mice, Knockout , Nitrates/metabolism , Norepinephrine/metabolism , Pregnancy , Renal Circulation/drug effects
5.
Physiol Rep ; 1(5): e00132, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24303195

ABSTRACT

Apart from their endocrine functions renin-expressing cells play an important functional role as mural cells of the developing preglomerular arteriolar vessel tree in the kidney. The recruitment of renin-expressing cells from the mesenchyme to the vessel wall is not well understood. Assuming that it may follow more general lines of pericyte recruitment to endothelial tubes we have now investigated the relevance of the platelet-derived growth factor (PDGF)-B-PDGFR-ß signaling pathway in this context. We studied renin expression in kidneys lacking PDGFR-ß in these cells and in kidneys with reduced endothelial PDGF-B expression. We found that expression of renin in the kidneys under normal and stimulated conditions was not different from wild-type kidneys. As expected, PDGFR-ß immunoreactivity was found in mesangial, adventitial and tubulo-interstitial cells but not in renin-expressing cells. These findings suggest that the PDGF-B-PDGFR-ß signaling pathway is not essential for the recruitment of renin-expressing cells to preglomerular vessel walls in the kidney.

6.
Am J Physiol Renal Physiol ; 305(3): F355-61, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23761669

ABSTRACT

Renin-expressing cells in the kidney normally appear as mural cells of developing preglomerular vessels and finally impose as granulated juxtaglomerular cells in adult kidneys. The differentiation of renin-expressing cells from the metanephric mesenchyme in general and the potential role of special precursor stages in particular is not well understood. Therefore, it was the aim of this study to search for renin cell precursors in the kidney. As an experimental model, we used kidneys of aldosterone synthase-deficient mice, which display a prominent compensatory overproduction of renin cells that are arranged in multilayered perivascular cell clusters. We found that the perivascular cell clusters contained two apparently distinct cell types, one staining positive for renin and another one staining positive for type I procollagen (PC1). It appeared as if PC1 and renin expression were inversely related at the cellular level. The proportion of renin-positive to PC1-positive cells in the clusters was inversely linked to the rate of salt intake, as was overall renin expression. Our findings suggest that the cells in the perivascular cell clusters can reversibly switch between PC1 and renin expression and that PC1-expressing cells might be precursors of renin cells. A few of those PC1-positive cells were found also in adult wild-type kidneys in the juxtaglomerular lacis cell area, in which renin expression can be induced on demand.


Subject(s)
Collagen Type I/biosynthesis , Kidney/metabolism , Renin/physiology , Stem Cells/metabolism , Animals , Cell Lineage , Cytochrome P-450 CYP11B2/deficiency , Female , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Immunohistochemistry , Juxtaglomerular Apparatus/blood supply , Kidney/cytology , Kidney Cortex/cytology , Kidney Cortex/metabolism , Lac Operon/genetics , Mice , Mice, Knockout , Microscopy, Confocal , Real-Time Polymerase Chain Reaction
7.
Am J Physiol Renal Physiol ; 304(5): F578-84, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23303406

ABSTRACT

On the basis of evidence that within the adult kidney, the aldo-keto reductase AKR1B7 (aldo-keto reductase family 1, member 7, also known as mouse vas deferens protein, MVDP) is selectively expressed in renin-producing cells, we aimed to define a possible role of AKR1B7 for the regulation and function of renin cells in the kidney. We could confirm colocalization and corecruitment of renin and of AKR1B7 in wild-type kidneys. Renin cells in AKR1B7-deficient kidneys showed normal morphology, numbers, and intrarenal distribution. Plasma renin concentration (PRC) and renin mRNA levels of AKR1B7-deficient mice were normal at standard chow and were lowered by a high-salt diet directly comparable to wild-type mice. Treatment with a low-salt diet in combination with an angiotensin-converting enzyme inhibitor strongly increased PRC and renin mRNA in a similar fashion both in AKR1B7-deficient and wild-type mice. Under this condition, we also observed a strong retrograde recruitment of renin-expressing cell along the preglomerular vessels, however, without a difference between AKR1B7-deficient and wild-type mice. The isolated perfused mouse kidney model was used to study the acute regulation of renin secretion by ANG II and by perfusion pressure. Regarding these parameters, no differences were observed between AKR1B7-deficient and wild-type kidneys. In summary, our data suggest that AKR1B7 is not of major relevance for the regulation of renin production and secretion in spite of its striking coregulation with renin expression.


Subject(s)
Aldehyde Reductase/metabolism , Kidney/metabolism , Renin/metabolism , Aldehyde Reductase/genetics , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Enalapril/pharmacology , Kidney/drug effects , Mice , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Renin/genetics , Sodium Chloride, Dietary
8.
Hypertension ; 61(2): 400-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23297374

ABSTRACT

Chronic challenge of renin-angiotensin causes recruitment of renin-producing cells in the kidney along the media layer of afferent arterioles and hypertrophy of cells in the juxtaglomerular apparatus. This study aimed to define the role of nitric oxide (NO) with regard to the recruitment pattern of renin-producing cells and to the possible pathways along which NO could act. We considered the hypothesis that endothelium-derived NO acts via NO-sensitive guanylate cyclase. Mice were treated with low-salt diet in combination with the angiotensin I-converting enzyme inhibitor enalapril for 3 weeks, which led to a 13-fold increase in renin expression associated with marked recruitment of renin cells in afferent arterioles and hypertrophy of the juxtaglomerular apparatus in wild-type mice. In wild-type mice additionally treated with the nonselective NO synthase inhibitor L-NAME, the recruitment of renin-expressing cells along the afferent arterioles was absent and juxtaglomerular hypertrophy was diminished. An almost identical attenuation of renin cell recruitment as with L-NAME treatment in wild-type mice was found in mice lacking the endothelial isoform of NO synthase. Treatment of mice lacking NO-sensitive guanylate cyclase in renin-expressing cells and preglomerular smooth muscle cells with low-salt diet in combination with the angiotensin I-converting enzyme inhibitor enalapril for 3 weeks produced juxtaglomerular hypertrophy like in wild-type mice, but no recruitment in afferent arterioles. These findings suggest that endothelium-derived NO and concomitant formation of cGMP in preglomerular renin cell precursors supports recruitment of renin-expressing cells along preglomerular vessels, but not in the juxtaglomerular apparatus.


Subject(s)
Endothelium, Vascular/metabolism , Guanylate Cyclase/metabolism , Kidney/metabolism , Nitric Oxide/metabolism , Renin/metabolism , Signal Transduction/physiology , Animals , Blood Pressure/drug effects , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Juxtaglomerular Apparatus/metabolism , Kidney/drug effects , Mice , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Signal Transduction/drug effects
9.
Am J Physiol Renal Physiol ; 302(10): F1278-85, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22357914

ABSTRACT

The salt intake of an organism controls the number of renin-producing cells in the kidney by yet undefined mechanisms. This study aimed to assess a possible mediator role of preglomerular blood pressure in the control of renin expression by oral salt intake. We used wild-type (WT) mice and mice lacking angiotensin II type 1a receptors (AT(1a)-/-) displaying an enhanced salt sensitivity to renin expression. In WT kidneys, we found renin-expressing cells at the ends of all afferent arterioles. A low-salt diet (0.02%) led to a moderate twofold increase in renin-expressing cells along afferent arterioles. In AT(1a)-/- mice, lowering of salt content led to a 12-fold increase in renin expression. Here, the renin-expressing cells were distributed along the preglomerular vascular tree in a typical distal-to-proximal distribution gradient which was most prominent at high salt intake and was obliterated at low salt intake by the appearance of renin-expressing cells in proximal parts of the preglomerular vasculature. While lowering of salt intake produced only a small drop in blood pressure in WT mice, the marked reduction of systolic blood pressure in AT(1a)-/- mice was accompanied by the disappearance of the distribution gradient from afferent arterioles to arcuate arteries. Unilateral renal artery stenosis in AT(1a)-/- mice on a normal salt intake produced a similar distribution pattern of renin-expressing cells as did low salt intake. Conversely, increasing blood pressure by administration of the NOS inhibitor N-nitro-l-arginine methyl ester or of the adrenergic agonist phenylephrine in AT(1a)-/- mice kept on low salt intake produced a similar distribution pattern of renin-producing cells as did normal salt intake alone. These findings suggest that changes in preglomerular blood pressure may be an important mediator of the influence of salt intake on the number and distribution of renin-producing cells in the kidney.


Subject(s)
Blood Pressure/physiology , Kidney/physiology , Receptor, Angiotensin, Type 1/genetics , Renin/genetics , Sodium Chloride, Dietary/pharmacology , Adrenergic alpha-1 Receptor Agonists/pharmacology , Animals , Arterioles/physiology , Blood Pressure/drug effects , Enzyme Inhibitors/pharmacology , Homeostasis/physiology , Juxtaglomerular Apparatus/physiology , Kidney/blood supply , Mice , Mice, 129 Strain , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Phenylephrine/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Renin/metabolism
10.
J Am Soc Nephrol ; 22(6): 1031-40, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21597036

ABSTRACT

Deletion of the gap-junction-forming protein connexin40 leads to renin-dependent hypertension in mice, but whether observed human variants in connexin40, such as A96S, promote hypertension is unknown. Here, we generated mice with the A96S variant in the mouse connexin40 gene. Although mice homozygous for the A96S mutations had normal expression patterns of connexin40 in the kidney, they were hypertensive, had sixfold higher plasma renin concentrations, and had 40% higher levels of renin mRNA than controls. Renin-expressing cells were aberrantly located outside the media layer of afferent arterioles, and increased renal perfusion pressure did not inhibit renin secretion from kidneys isolated from homozygous A96S mice. Treatment with a low-salt diet in combination with an ACE inhibitor increased renin mRNA levels, plasma renin concentrations, and the number of aberrantly localized renin-producing cells. Taken together, these findings suggest that the A96S mutation in connexin40 leads to renin-dependent hypertension in mice. Modulation of renin secretion by BP critically depends on functional connexin40; with the A96S mutation, the aberrant extravascular localization of renin-secreting cells in the kidney likely impairs the pressure-mediated inhibition of renin secretion.


Subject(s)
Connexins/genetics , Hypertension/genetics , Hypertension/physiopathology , Mutation/genetics , Renin/physiology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Diet, Sodium-Restricted , Disease Models, Animal , Female , Gap Junctions/physiology , HeLa Cells , Humans , Hypertension/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Transfection , Treatment Outcome , Gap Junction alpha-5 Protein
11.
Am J Physiol Renal Physiol ; 301(1): F71-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21389089

ABSTRACT

During nephrogenesis, renin expression shifts from large renal arteries toward smaller vessels in a defined spatiotemporal pattern, finally becoming restricted to the juxtaglomerular position. Chronic stimulation in adult kidneys leads to a recruitment of renin expression in the upstream vasculature. The mechanisms that control this characteristic switch-on and switch-off in the immature and adult kidney are not well-understood. Previous studies in mice with juxtaglomerular cell-specific deletion of the adenylyl cyclase-stimulatory G protein Gsα suggested that signaling along the cAMP pathway plays an essential role for renin expression during nephrogenesis and in the adult kidney. To identify the Gsα-dependent receptor that might be involved in activating this pathway, the present studies were performed to compare renin expression in wild types with that in mice with targeted deletions of ß(1) and ß(2)-adrenoceptors. The sympathetic nervous system is an important regulator of the renin system in the adult kidney so that activation of ß-adrenenoceptors may also participate in the activation of renin expression along the developing arterial tree and in upstream vasculature in adulthood. Compared with wild-types, renin expression was found to be significantly lower at all developmental stages in the kidneys of ß(1)/ß(2) Adr(-/-) mice. Three-dimensional analysis showed reduced renin expression in all segments of the vascular tree in mutants and a virtual absence of renin expression in the large arcuate arteries. Adult mutant kidneys showed the typical upstream renin expression after chronic stimulation. Tyrosine hydroxylase staining in fetal and postnatal kidneys revealed that sympathetic innervation of renin-producing cells occurs early in fetal development. Our data indicate that genetic disruption of ß-adrenergic receptors reduces basal renin expression along the developing preglomerular tree and in adult kidneys. Furthermore, ß-adrenergic receptor input is critical for the expression of renin in large renal vessels during early fetal development.


Subject(s)
Blood Vessels/embryology , Blood Vessels/metabolism , Kidney/embryology , Kidney/metabolism , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/physiology , Renin/biosynthesis , Animals , Arterioles/metabolism , Female , Fetal Development , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Mice , Mice, Knockout , Pregnancy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics , Renin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sympathetic Nervous System/embryology , Sympathetic Nervous System/physiology , Tyrosine 3-Monooxygenase/metabolism
12.
Am J Physiol Renal Physiol ; 297(5): F1371-80, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19710239

ABSTRACT

During nephrogenesis, renin expression shifts from the vessel walls of interlobular arteries to the terminal portions of afferent arterioles in a wavelike pattern. Since the mechanisms responsible for the developmental deactivation of renin expression are as yet unknown, we hypothesized that the developing renin-angiotensin system (RAS) may downregulate itself via negative feedback to prevent overactivity of renin. To test for a possible role of angiotensin II in the developmental deactivation of renin expression, we studied the development of intrarenal renin expression in mice lacking ANG II AT1a, AT1b, or AT2 receptors and in animals with abolished circulating ANG II due to deletion of the gene for angiotensin I-converting enzyme (ACE). The development of intrarenal renin expression was normal in mice lacking ANG II AT1b or AT2 receptors. In animals lacking both ANG II AT1a and AT1b receptors, ACE, or ANG II AT1a receptors, renin expression was normal early and renin disappeared from mature vessels until development of cortical interlobular and afferent arterioles began. The development of cortical vessels in these genotypes was accompanied by a markedly increased number of renin-expressing cells, many of which were ectopically located and attached in a grapelike fashion to the outer vessel perimeter. Although the number of renin-expressing cells declined during final maturation of the kidneys, the atypical distribution pattern of renin cells was maintained. These findings suggest that ANG II does not play a central role in the typical developmental shift in renin expression from the arcuate vessels to the afferent arterioles. During postnatal maturation of mouse kidneys, interruption of the RAS causes severe hyperplasia of renin cells via a mechanism that centrally involves AT(1a) receptors. However, the distribution pattern of renin cells in adult kidneys with an interrupted RAS does not mimic any normal developmental stage since renin expression is frequently found in cells outside the arteriolar vessel walls in RAS mutants.


Subject(s)
Renin-Angiotensin System/genetics , Renin-Angiotensin System/physiology , Renin/biosynthesis , Actins/biosynthesis , Actins/genetics , Animals , Feedback, Physiological/physiology , Hyperplasia , Image Processing, Computer-Assisted , Immunohistochemistry , Kidney/growth & development , Kidney/metabolism , Mice , Mice, Knockout , Muscle, Smooth/metabolism , Peptidyl-Dipeptidase A/biosynthesis , Peptidyl-Dipeptidase A/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/physiology , Renin/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Nat Med ; 15(5): 545-52, 2009 May.
Article in English | MEDLINE | ID: mdl-19412173

ABSTRACT

In salt-sensitive hypertension, the accumulation of Na(+) in tissue has been presumed to be accompanied by a commensurate retention of water to maintain the isotonicity of body fluids. We show here that a high-salt diet (HSD) in rats leads to interstitial hypertonic Na(+) accumulation in skin, resulting in increased density and hyperplasia of the lymphcapillary network. The mechanisms underlying these effects on lymphatics involve activation of tonicity-responsive enhancer binding protein (TonEBP) in mononuclear phagocyte system (MPS) cells infiltrating the interstitium of the skin. TonEBP binds the promoter of the gene encoding vascular endothelial growth factor-C (VEGF-C, encoded by Vegfc) and causes VEGF-C secretion by macrophages. MPS cell depletion or VEGF-C trapping by soluble VEGF receptor-3 blocks VEGF-C signaling, augments interstitial hypertonic volume retention, decreases endothelial nitric oxide synthase expression and elevates blood pressure in response to HSD. Our data show that TonEBP-VEGF-C signaling in MPS cells is a major determinant of extracellular volume and blood pressure homeostasis and identify VEGFC as an osmosensitive, hypertonicity-driven gene intimately involved in salt-induced hypertension.


Subject(s)
Blood Pressure/physiology , Hypertension/physiopathology , Macrophages/physiology , Sodium Chloride/adverse effects , Sodium, Dietary/adverse effects , Transcription Factors/physiology , Vascular Endothelial Growth Factor C/physiology , Animals , Homeostasis , Hypertension/chemically induced , Mice , Phagocytes/drug effects , Phagocytes/physiology , Rats , Receptors, Vascular Endothelial Growth Factor/physiology , Signal Transduction , Skin/drug effects , Skin Physiological Phenomena , Vascular Endothelial Growth Factor C/antagonists & inhibitors
14.
Am J Physiol Renal Physiol ; 296(5): F1006-12, 2009 May.
Article in English | MEDLINE | ID: mdl-19261741

ABSTRACT

During metanephric kidney development, renin expression in the renal vasculature begins in larger vessels, shifting to smaller vessels and finally remaining restricted to the terminal portions of afferent arterioles at the entrance into the glomerular capillary network. The mechanisms determining the successive expression of renin along the vascular axis of the kidney are not well understood. Since the cAMP signaling cascade plays a central role in the regulation of both renin secretion and synthesis in the adult kidney, it seemed feasible that this pathway might also be critical for renin expression during kidney development. In the present study we determined the spatiotemporal development of renin expression and the development of the preglomerular arterial tree in mouse kidneys with renin cell-specific deletion of G(s)alpha, a core element for receptor activation of adenylyl cyclases. We found that in the absence of the G(s)alpha protein, renin expression was largely absent in the kidneys at any developmental stage, accompanied by alterations in the development of the preglomerular arterial tree. These data indicate that the maintenance of renin expression following a specific spatiotemporal pattern along the preglomerular vasculature critically depends on the availability of G(s)alpha. We infer from our data that the cAMP signaling pathway is not only critical for the regulation of renin synthesis and secretion in the mature kidney but that it also is critical for establishing the juxtaglomerular expression site of renin during development.


Subject(s)
Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Expression Regulation, Developmental/physiology , Kidney Glomerulus , Renal Circulation/physiology , Renin/genetics , Animals , Arterioles/embryology , Arterioles/physiology , Chromogranins , GTP-Binding Protein alpha Subunits, Gs/metabolism , Juxtaglomerular Apparatus/embryology , Juxtaglomerular Apparatus/physiology , Kidney Glomerulus/blood supply , Kidney Glomerulus/embryology , Kidney Glomerulus/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Renin/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...