Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Hypertension ; 71(6): 1075-1082, 2018 06.
Article in English | MEDLINE | ID: mdl-29661841

ABSTRACT

The activity of the renin-angiotensin-aldosterone system is triggered by the release of the protease renin from the kidneys, which in turn is controlled in the sense of negative feedback loops. It is widely assumed that Ang II (angiotensin II) directly inhibits renin expression and secretion via a short-loop feedback by an effect on renin-producing cells (RPCs) mediated by AT1 (Ang II type 1) receptors. Because the concept of such a direct short-loop negative feedback control, which originates mostly from in vitro experiments, has not yet been systematically proven in vivo, we aimed to test the validity of this concept by studying the regulation of renin synthesis and secretion in mice lacking Ang II-AT1 receptors on RPCs. We found that RPCs of the kidney express Ang II-AT1 receptors. Mice with conditional deletion of Ang II-AT1 receptors in RPCs were normal with regard to the number of renin cells, renal renin mRNA, and plasma renin concentrations. Renin expression and secretion of these mice responded to Ang I (angiotensin I)-converting enzyme inhibition and to Ang II infusion like in wild-type (WT) controls. In summary, we did not obtain evidence that Ang II-AT1 receptors on RPCs are of major relevance for the normal regulation of renin expression and secretion in mice. Therefore, we doubt the existence of a direct negative feedback function of Ang II on RPCs.


Subject(s)
Angiotensin II/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Blood Pressure/physiology , Hypertension/metabolism , Receptors, Angiotensin/metabolism , Renin-Angiotensin System/physiology , Renin/blood , Animals , Disease Models, Animal , Female , Hypertension/drug therapy , Hypertension/physiopathology , Immunohistochemistry , Male , Mice , Renin-Angiotensin System/drug effects
2.
J Am Soc Nephrol ; 29(5): 1383-1396, 2018 05.
Article in English | MEDLINE | ID: mdl-29444905

ABSTRACT

BACKGROUND: The kidney is considered to be a structurally stable organ with limited baseline cellular turnover. Nevertheless, single cells must be constantly replaced to conserve the functional integrity of the organ. PDGF chain B (PDGF-BB) signaling through fibroblast PDGF receptor-ß (PDGFRß) contributes to interstitial-epithelial cell communication and facilitates regenerative functions in several organs. However, the potential role of interstitial cells in renal tubular regeneration has not been examined. METHODS: In mice with fluorescent protein expression in renal tubular cells and PDGFRß-positive interstitial cells, we ablated single tubular cells by high laser exposure. We then used serial intravital multiphoton microscopy with subsequent three-dimensional reconstruction and ex vivo histology to evaluate the cellular and molecular processes involved in tubular regeneration. RESULTS: Single-tubular cell ablation caused the migration and division of dedifferentiated tubular epithelial cells that preceded tubular regeneration. Moreover, tubular cell ablation caused immediate calcium responses in adjacent PDGFRß-positive interstitial cells and the rapid migration thereof toward the injury. These PDGFRß-positive cells enclosed the injured epithelium before the onset of tubular cell dedifferentiation, and the later withdrawal of these PDGFRß-positive cells correlated with signs of tubular cell redifferentiation. Intraperitoneal administration of trapidil to block PDGFRß impeded PDGFRß-positive cell migration to the tubular injury site and compromised the recovery of tubular function. CONCLUSIONS: Ablated tubular cells are exclusively replaced by resident tubular cell proliferation in a process dependent on PDGFRß-mediated communication between the renal interstitium and the tubular system.


Subject(s)
Cell Dedifferentiation , Epithelial Cells/physiology , Kidney Tubules, Proximal/physiology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Regeneration , Urothelium/physiology , Animals , Calcium/metabolism , Cell Communication , Cell Movement/drug effects , Female , Intravital Microscopy , Kidney/cytology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/injuries , Lymphokines/metabolism , Male , Mice , Phosphodiesterase Inhibitors/pharmacology , Platelet-Derived Growth Factor/metabolism , Recovery of Function , Trapidil/pharmacology , Urothelium/injuries
3.
Pflugers Arch ; 470(6): 969-978, 2018 06.
Article in English | MEDLINE | ID: mdl-29427253

ABSTRACT

The so-called calcium paradoxon of renin describes the phenomenon that exocytosis of renin from juxtaglomerular cells of the kidney is stimulated by lowering of the extracellular calcium concentration. The yet poorly understood effect of extracellular calcium on renin secretion appears to depend on the function of the gap junction protein connexin 40 (Cx40) in renin-producing cells. This study aimed to elucidate the role of Cx40 for the calcium dependency of renin secretion in more detail by investigating if Cx40 function is really essential for the influence of extracellular calcium on renin secretion, if and how Cx40 affects intracellular calcium dynamics in renin-secreting cells and if Cx40-mediated gap junctional coupling of renin-secreting cells with the mesangial cell area is relevant for the influence of extracellular calcium on renin secretion. Renin secretion was studied in isolated perfused mouse kidneys. Calcium measurements were performed in renin-producing cells of microdissected glomeruli. The ultrastructure of renin-secreting cells was examined by electron microscopy. We found that Cx40 was not essential for stimulation of renin secretion by lowering of the extracellular calcium concentration. Instead, Cx40 increased the sensitivity of renin secretion response towards lowering of the extracellular calcium concentration. In line, the sensitivity and dynamics of intracellular calcium in response to lowering of extracellular calcium were dampened when renin-secreting cells lacked Cx40. Disruption of gap junctional coupling of renin-secreting cells by selective deletion of Cx40 from mesangial cells, however, did not change the stimulation of renin secretion by lowering of the extracellular calcium concentration. Deletion of Cx40 from renin cells but not from mesangial cells was associated with a shift of renin expression from perivascular cells of afferent arterioles to extraglomerular mesangial cells. Our findings suggest that Cx40 is not directly involved in the regulation of renin secretion by extracellular calcium. Instead, it appears that in renin-secreting cells of the kidney lacking Cx40, intracellular calcium dynamics and therefore also renin secretion are desensitized towards changes of extracellular calcium. Whether the dampened calcium response of renin-secreting cells lacking Cx40 function results from a direct involvement of Cx40 in intracellular calcium regulation or from the cell type shift of renin expression from perivascular to mesangial cells remains to be clarified. In any case, Cx40-mediated gap junctional coupling between renin and mesangial cells is not relevant for the calcium paradoxon of renin secretion.


Subject(s)
Calcium/metabolism , Connexins/metabolism , Juxtaglomerular Apparatus/metabolism , Renin/metabolism , Animals , Connexins/genetics , Female , Juxtaglomerular Apparatus/cytology , Male , Mice , Gap Junction alpha-5 Protein
4.
J Biol Chem ; 293(4): 1151-1162, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29123029

ABSTRACT

Normal renin synthesis and secretion is important for the maintenance of juxtaglomerular apparatus architecture. Mice lacking a functional Ren1d gene are devoid of renal juxtaglomerular cell granules and exhibit an altered macula densa morphology. Due to the species-specificity of renin activity, transgenic mice are ideal models for experimentally investigating and manipulating expression patterns of the human renin gene in a native cellular environment without confounding renin-angiotensin system interactions. A 55-kb transgene encompassing the human renin locus was crossed onto the mouse Ren1d-null background, restoring granulation in juxtaglomerular cells. Correct processing of human renin in dense core granules was confirmed by immunogold labeling. After stimulation of the renin-angiotensin system, juxtaglomerular cells contained rhomboid protogranules with paracrystalline contents, dilated rough endoplasmic reticulum, and electron-lucent granular structures. However, complementation of Ren1d-/- mice with human renin was unable to rescue the abnormality seen in macula densa structure. The juxtaglomerular apparatus was still able to respond to tubuloglomerular feedback in isolated perfused juxtaglomerular apparatus preparations, although minor differences in glomerular tuft contractility and macula densa cell calcium handling were observed. This study reveals that the human renin protein is able to complement the mouse Ren1d-/- non-granulated defect and suggests that granulopoiesis requires a structural motif that is conserved between the mouse Ren1d and human renin proteins. It also suggests that the altered macula densa phenotype is related to the activity of the renin-1d enzyme in a local juxtaglomerular renin-angiotensin system.


Subject(s)
Genetic Complementation Test , Juxtaglomerular Apparatus/enzymology , Renin/biosynthesis , Transgenes , Animals , Humans , Juxtaglomerular Apparatus/pathology , Mice , Mice, Knockout , Renin/genetics
5.
Am J Pathol ; 187(7): 1496-1511, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28527294

ABSTRACT

Intrauterine hypoxia is a reason for impaired kidney development. The cellular and molecular pathways along which hypoxia exerts effects on nephrogenesis are not well understood. They are likely triggered by hypoxia-inducible transcription factors (HIFs), and their effects appear to be dependent on the cell compartment contributing to kidney formation. In this study, we investigated the effects of HIF activation in the developing renal stroma, which also essentially modulates nephron development from the metanephric mesenchyme. HIF activation was achieved by conditional deletion of the von Hippel-Lindau tumor suppressor (VHL) protein in the forkhead box FOXD1 cell lineage, from which stromal progenitors arise. The resulting kidneys showed maturation defects associated with early postnatal death. In particular, nephron formation, tubular maturation, and the differentiation of smooth muscle, renin, and mesangial cells were impaired. Erythropoietin expression was strongly enhanced. Codeletion of VHL together with HIF2A but not with HIF1A led to apparently normal kidneys, and the animals reached normal age but were anemic because of low erythropoietin levels. Stromal deletion of HIF2A or HIF1A alone did not affect kidney development. These findings emphasize the relevance of sufficient intrauterine oxygenation for normal renal stroma differentiation, suggesting that chronic activity of HIF2 in stromal progenitors impairs kidney development. Finally, these data confirm the concept that normal stroma function is essential for normal tubular differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Forkhead Transcription Factors/genetics , Kidney/embryology , Oxygen/metabolism , Signal Transduction , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Hypoxia , Cell Lineage , Erythropoietin/genetics , Erythropoietin/metabolism , Female , Forkhead Transcription Factors/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Rats , Stem Cells/metabolism , Stromal Cells/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
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.
Kidney Int ; 83(2): 233-41, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23235568

ABSTRACT

The mode of renin release from renal juxtaglomerular cells into circulation is still unsolved in several aspects. Here we studied the intracellular organization of renin-storage vesicles and their changes during controlled stimulation of renin release. This was accomplished using isolated perfused mouse kidneys with 3-dimensional electron microscopic analyses of renin-producing cells. Renin was found to be stored in a network of single granules and cavern-like structures, and dependent on the synthesis of glycosylated prorenin. Acute stimulation of renin release led to increased exocytosis in combination with intracellular fusion of vesicles to larger caverns and their subsequent emptying. Renin release from the kidneys of SCID-beige mice, which contain few but gigantic renin-storage vesicles, was no different from that of kidneys from wild-type mice. Thus, our findings suggest that renin is released by mechanisms similar to compound exocytosis.


Subject(s)
Exocytosis , Renin/metabolism , Animals , Isoproterenol/pharmacology , Juxtaglomerular Apparatus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, SCID
8.
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
9.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...