Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 598
Filtrar
1.
FASEB J ; 35(10): e21912, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34533842

RESUMO

High salt diet (HSD) is a hallmark of blood pressure elevations, weight gain and diabetes onset in the metabolic syndrome. In kidney, compensatory mechanisms are activated to balance salt turnover and maintain homeostasis. Data on the long-term effects of HSD with respect to tubular cell functions and kidney architecture that exclude confounding indirect blood pressure effects are scarce. Additionally we focus on cold shock Y-box binding protein-1 as a tubular cell protective factor. A HSD model (4% NaCl in chow; 1% NaCl in water) was compared to normal salt diet (NSD, standard chow) over 16 months using wild type mice and an inducible conditional whole body knockout for cold shock Y-box binding protein-1 (BL6J/N, Ybx1). HSD induced no difference in blood pressure over 16 months, comparing NSD/HSD and Ybx1 wild type/knockout. Nevertheless, marked phenotypic changes were detected. Glucosuria and subnephrotic albuminuria ensued in wild type animals under HSD, which subsided in Ybx1-deficient animals. At the same time megalin receptors were upregulated. The sodium-glucose cotransporter-2 (SGLT2) was completely downregulated in wild type HSD animals that developed glucosuria. In Ybx1 knockouts, expression of AQP1 and SGLT2 was maintained under HSD; proximal tubular widening and glomerular tubularization developed. Concurrently, amino aciduria of neutral and hydrophobic amino acids was seen. In vitro translation confirmed that YB-1 translationally represses Sglt2 transcripts. Our data reveal profound effects of HSD primarily within glomeruli and proximal tubular segments. YB-1 is regulated by HSD and orchestrates HSD-dependent changes; notably, sets reabsorption thresholds for amino acids, proteins and glucose.


Assuntos
Resposta ao Choque Frio/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Túbulos Renais Proximais/efeitos dos fármacos , Sódio na Dieta/farmacologia , Transportador 2 de Glucose-Sódio/genética , Fatores de Transcrição/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Feminino , Túbulos Renais Proximais/citologia , Leucócitos/citologia , Macrófagos/citologia , Masculino , Fenótipo , Podócitos/efeitos dos fármacos , Renina/biossíntese , Renina/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Regulação para Cima/efeitos dos fármacos
2.
EBioMedicine ; 64: 103209, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33508746

RESUMO

BACKGROUND: Erythrocyte mass contributes to maintaining systemic oxygen delivery and blood viscosity, with the latter being one of the determinants of blood pressure. However, the physiological response to blood pressure changes under anaemic conditions remain unknown. METHODS AND FINDINGS: We show that anaemia decreases blood pressure in human patients and mouse models. Analyses of pathways related to blood pressure regulation demonstrate that anaemia enhances the expression of the gene encoding the vasopressor substance renin in kidneys. Although kidney juxtaglomerular cells are known to continuously produce renin, renal interstitial fibroblasts are identified in the present study as a novel site of renin induction under anaemic hypotensive conditions in mice and rats. Notably, some renal interstitial fibroblasts are found to simultaneously express renin and the erythroid growth factor erythropoietin in the anaemic mouse kidney. Antihypertensive agents but not hypoxic stimuli induced interstitial renin expression, suggesting that blood pressure reduction triggers interstitial renin induction in anaemic mice. The interstitial renin expression was also detected in injured fibrotic kidneys of the mouse and human, and the renin-expressing interstitial cells in murine fibrotic kidneys were identified as myofibroblasts originating from renal interstitial fibroblasts. Since the elevated expression levels of renin in fibrotic kidneys along with progression of renal fibrosis were well correlated to the systemic blood pressure increase, the renal interstitial renin production seemed to affect systemic blood pressure. INTERPRETATION: Renal interstitial fibroblasts function as central controllers of systemic oxygen delivery by producing both renin and erythropoietin. FUNDING: Grants-in-Aid from Japan Society for the Promotion of Science (JSPS) KAKENHI (17K19680, 15H04691, and 26111002) and the Takeda Science Foundation.


Assuntos
Anemia/metabolismo , Eritropoetina/biossíntese , Fibroblastos/metabolismo , Rim/metabolismo , Renina/biossíntese , Idoso , Anemia/complicações , Animais , Biomarcadores , Pressão Sanguínea , Doença Crônica , Modelos Animais de Doenças , Eritropoetina/genética , Feminino , Fibrose , Expressão Gênica , Humanos , Hipotensão/complicações , Hipóxia/etiologia , Hipóxia/metabolismo , Nefropatias/etiologia , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , Renina/genética , Transdução de Sinais
3.
Mol Cell Endocrinol ; 522: 111118, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33340569

RESUMO

Pregnancy demands major cardiovascular, renal and endocrine changes to provide an adequate blood supply for the growing fetus. The renin-angiotensin-aldosterone system plays a key role in this adaptation process. One of its components, prorenin, is released in significant amounts from the ovary and uteroplacental unit. This review describes the sources of prorenin in the periconception period and in pregnancy, including its modulation by in-vitro fertilization protocols, and discusses its potential effects, among others focusing on preeclampsia. It ends with discussing the long-term consequences, even in later life, of inappropriate renin-angiotensin-aldosterone system activity in pregnancy and offers directions for future research. Ultimately, a full understanding of the role of prorenin periconceptionally and during pregnancy will help to develop tools to diagnose and/or prevent reproductive complications.


Assuntos
Renina/metabolismo , Animais , Feminino , Hemodinâmica , Humanos , Modelos Biológicos , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/metabolismo , Gravidez , Biossíntese de Proteínas , Renina/biossíntese , Sistema Renina-Angiotensina
4.
J Renin Angiotensin Aldosterone Syst ; 21(2): 1470320320923977, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32419571

RESUMO

INTRODUCTION: The angiotensin converting enzyme inhibitor ramipril is a standard antihypertensive therapy for many patients. Because angiotensin II may promote inflammation, we were interested in whether basal pretreatment with ramipril may modify renal function and inflammation as well as systemic outcome in experimentally induced sepsis in mice. MATERIAL AND METHODS: Ramipril (10 mg/kg/day) pretreatment or placebo (NaCl) was given intraperitoneally for 5 days to C57BL6/J mice, followed by either sham operation or cecal ligation and puncture sepsis induction. Real-time polymerase chain reaction and immunological stains were used to evaluate renal gene and protein expression, respectively. Plasma creatinine, neutrophil-gelatinase associated lipocalin, and blood urea nitrogen were used as markers for renal function. A clinical severity score was determined. RESULTS: Administration of ramipril before cecal ligation and puncture surgery was associated with reduced renal inflammation but did not improved renal function and structure and even worsened the clinical status of septic mice. CONCLUSIONS: The data suggest that the effects of ramipril pretreatment are complex. Additional studies including monitoring of hemodynamic parameters are necessary to elucidate the exact mechanism(s) of this observation. In addition, the timing of the ramipril administration could be of importance.


Assuntos
Injúria Renal Aguda/patologia , Inibidores da Enzima Conversora de Angiotensina/efeitos adversos , Inflamação/patologia , Rim/patologia , Ramipril/efeitos adversos , Sepse/complicações , Sepse/patologia , Injúria Renal Aguda/genética , Injúria Renal Aguda/mortalidade , Animais , Apoptose/efeitos dos fármacos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Testes de Função Renal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/biossíntese , Renina/biossíntese
5.
Exp Eye Res ; 189: 107828, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31589840

RESUMO

Several lines of evidence support the existence of a renin-angiotensin system (RAS) in the retina that is separated from the blood stream by the retinal pigment epithelium (RPE). Under physiological conditions, increased activity of intraretinal RAS regulates neuronal activity of the retina but patho-physiologically participates in retinal degeneration such as hypertensive or diabetic retinopathy. Interestingly, the RPE appears to be a modulator of intraretinal RAS in response to changes in systemic RAS. As increased systemic RAS activity is associated with increased sympathetic tonus, we investigated whether systemic ß-adrenergic stimulation of the RPE also modulates renin expression in the RPE. In vivo, the mouse RPE expresses the ß-adrenergic receptor subtypes 1 and 2. Staining of retina sagittal sections showed tyrosine hydroxylase positive nerve endings in the choroid indicating adrenaline/noradrenaline production sites in close proximity to the RPE. Systemic infusion of isoproterenol increased renin expression in the RPE but not in the retina. This increase was sensitive to concomitant systemic application of the angiotensin-2 receptor-type-1 blocker losartan. In vitro analysis of renin gene expression using polarized porcine RPE showed that the activity of the renin promoter can be increased by cAMP stimulation (IBMX/forskolin) but was not influenced by angiotensin-2. Thus, with the identification of the ß-adrenergic system we added a new regulator of the retinal RAS with relevance for retinal function and pathology. Furthermore, it appears that the RPE is not only a close interaction partner of the photoreceptors but also a regulator or retinal activity in general.


Assuntos
Receptores Adrenérgicos beta/biossíntese , Sistema Renina-Angiotensina/fisiologia , Epitélio Pigmentado da Retina/metabolismo , Sistema Nervoso Simpático/fisiologia , Animais , Células Cultivadas , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Renina/biossíntese , Epitélio Pigmentado da Retina/citologia , Estimulação Química
6.
Exp Eye Res ; 189: 107838, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31622617

RESUMO

As many other organs, the retina has a local renin-angiotensin-system (RAS). All main elements of the RAS are active in the retina: renin, angiotensinogen, angiotensin-converting enzymes. The functional role of the intraretinal RAS is not fully understood. So far, histological and functional analysis point to a regulation of ganglion cell activity and maybe also of bipolar cell activity, but it is not clear how RAS contributes to retinal signal processing. In contrast to local RAS in other organs, the retinal RAS is clearly separated from the systemic RAS. The angiotensin-2 (AngII)/AngI ratio in the retina is different to that in the plasma. However, it appears that the retinal pigment epithelium (RPE), that forms the outer blood/retina barrier, is a major regulator of the retinal RAS by producing renin. Interestingly, comparable to the kidney, the renin production in the RPE is under control of the angiotensin-2 receptor type-1 (AT1). AT1 localizes to the basolateral membrane of the RPE and faces the blood side of the blood/retina barrier. Increases in systemic AngII reduce renin production in the RPE and therefore decrease the intraretinal RAS activity. The relevance of the local RAS for retinal function remains unclear. Nevertheless, it is of fundamental significance to understand the pathology of systemically induced retinal diseases such as hypertension or diabetes.


Assuntos
Barreira Hematorretiniana/metabolismo , Sistema Renina-Angiotensina/fisiologia , Renina/biossíntese , Epitélio Pigmentado da Retina/metabolismo , Animais , Humanos
7.
Naunyn Schmiedebergs Arch Pharmacol ; 392(11): 1401-1410, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31243492

RESUMO

It has been reported that the serine protease kallikrein stimulates and that aprotinin, a protease inhibitor, inhibits renal renin secretion. Since direct stimulation of the protease-activated receptor (PAR) 2 increases renin secretion in isolated perfused mouse kidneys, we hypothesized that activation of PAR2 receptors by serine proteases could be involved in the synthesis and secretion of renin in vivo. We therefore determined the response of plasma renin concentration (PRC) to acute intraperitoneal administration of the PAR2 agonist SLIGRL, isoproterenol, hydralazine, furosemide, losartan, or lipopolysaccharide in conscious wild-type (WT) and Par2-deficient mice. Basal PRC was not different in Par2-deficient mice compared with WT mice. All six acute treatments caused significant increases of PRC in both WT and Par2-deficient mice. The response was significantly lower only in endotoxin-treated Par2-deficient mice. Chronic treatment with losartan, low salt intake, the combination of both, or furosemide caused an increase of PRC and renin mRNA in WT mice, whereas a high salt intake caused a decrease. Alterations in PRC and renal renin mRNA expression were not different between WT and Par2 -/- mice in response to chronic treatments. Par2-deficiency did not alter furosemide-induced diuresis and natriuresis. Systolic blood pressure responses to chronic treatments were not different between WT and Par2 -/- mice. In conclusion, deficiency of Par2 receptors does not alter renin secretion and renin gene expression modulated by a variety of typical maneuvers. However, activation of Par2 receptors by serine proteases seems to be of importance for renin secretion in the context of inflammation.


Assuntos
Rim/metabolismo , Receptor PAR-2/fisiologia , Renina/biossíntese , Animais , Relação Dose-Resposta a Droga , Furosemida/farmacologia , Expressão Gênica/efeitos dos fármacos , Hidralazina/farmacologia , Isoproterenol/farmacologia , Rim/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Losartan/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligopeptídeos/farmacologia , Receptor PAR-2/agonistas , Receptor PAR-2/genética , Renina/sangue , Renina/genética , Cloreto de Sódio/farmacologia
8.
Am J Physiol Renal Physiol ; 317(2): F512-F517, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31188032

RESUMO

Diabetic nephropathy (DN) is the most common cause of end-stage renal disease associated with high mortality worldwide. Increases in iron levels have been reported in diabetic rat kidneys as well as in human urine of patients with diabetes. In addition, a low-iron diet or iron chelators delay the progression of DN in patients with diabetes and in animal models of diabetes. Possible maladaptive mechanisms of organ damage by tissue iron accumulation have not been well studied. We recently reported that iron induced the retinal renin-angiotensin system (RAS) and accelerated the progression of diabetic retinopathy. However, whether iron regulates the systemic RAS is unknown. To explore if iron alters the expression of intrarenal RAS and its role in the progression of DN, we used the high Fe iron (HFE) knockout mouse, a genetic model of systemic iron overload. We found that diabetes upregulated the expression of iron regulatory proteins and augmented tissue iron accumulation in the kidneys of both type 1 and type 2 diabetic mouse models. Iron accumulation in the kidneys of HFE knockout mice was associated with increase in serum and intrarenal renin expression. Induction of diabetes in HFE knockout mice using streptozotocin caused a much higher accumulation of renal iron and accelerated the progression of nephropathy compared with diabetic wild-type mice. Treatment of diabetic mice with the iron chelator deferiprone reversed the renin upregulation and reduced kidney injury. Thus, our results establish a new link between renal iron and RAS activity. Exploring the mechanisms of iron-induced RAS activation further may have a significant therapeutic impact on hypertension and DN.


Assuntos
Nefropatias Diabéticas/genética , Nefropatias Diabéticas/metabolismo , Proteína da Hemocromatose/genética , Proteína da Hemocromatose/metabolismo , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Ferro/metabolismo , Rim/metabolismo , Animais , Deferiprona/farmacologia , Deferiprona/uso terapêutico , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/tratamento farmacológico , Progressão da Doença , Quelantes de Ferro/farmacologia , Quelantes de Ferro/uso terapêutico , Masculino , Camundongos , Camundongos Knockout , Renina/biossíntese , Sistema Renina-Angiotensina/efeitos dos fármacos
9.
Exp Physiol ; 104(5): 740-754, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30790372

RESUMO

NEW FINDINGS: What is the central question of this study? Can a single bone marrow mononuclear cell (BMMC) transplant into the subcapsular region of kidney improve cellular communication and adhesion, while restoring renal tissue cytoarchitecture and function during renovascular hypertension? What is the main finding and its importance? The BMMC transplantation restored connexin 40 expression and led to recovery of N- and E-cadherin levels within 15 days. It was observed, for the first time, that BMMC transplantation restores expression of nephrin, a component of the glomerular filtration barrier related to podocytes and the glomerular basal membrane. ABSTRACT: Stem cell therapy has emerged as a potential treatment for renal diseases owing to the regenerative potential of stem cells. However, a better understanding of the morphological and functional changes of damaged renal cells in the presence of transplanted stem cells is needed. The aim of this study was to investigate cell-cell communication and adhesion in renal parenchyma, with analysis of fibrosis, to evaluate renal morphology and function after bone marrow mononuclear cell (BMMC) transplantation in two-kidney-one-clip rats. The BMMC therapy significantly decreased blood pressure and renin expression, improved renal morphology and restored the glomerular filtration barrier, with remodelling of podocytes. In addition, there was a reduction in fibrosis, and connexin 40 and nephrin expression were significantly increased after 7 and 15 days of transplantation. Plasma creatinine, urea and total protein levels were restored, and proteinuria was reduced. Furthermore, N- and E-cadherin expression was increased soon after BMMC therapy. Green fluorescent protein-positive BMMCs were found in the renal cortex 24 and 48 h after transplantation into the renal subcapsule, and at 7 and 15 days after transplantation, these cells were observed throughout the renal medulla, indicating cellular migration. Therefore, these data suggest that transplanted BMMCs improve cell-cell communication and adhesion between damaged cells, which is accompanied by a recovery of renal morphology and function.


Assuntos
Transplante de Medula Óssea/métodos , Barreira de Filtração Glomerular/patologia , Hipertensão Renovascular/patologia , Hipertensão Renovascular/terapia , Junções Intercelulares/patologia , Animais , Pressão Sanguínea , Caderinas/metabolismo , Comunicação Celular , Fibrose , Rim/patologia , Córtex Renal/patologia , Masculino , Monócitos/transplante , Podócitos/patologia , Ratos , Ratos Wistar , Renina/biossíntese
10.
J Clin Invest ; 128(11): 4787-4803, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30130256

RESUMO

Renin cells are crucial for survival - they control fluid-electrolyte and blood pressure homeostasis, vascular development, regeneration, and oxygen delivery to tissues. During embryonic development, renin cells are progenitors for multiple cell types that retain the memory of the renin phenotype. When there is a threat to survival, those descendants are transformed and reenact the renin phenotype to restore homeostasis. We tested the hypothesis that the molecular memory of the renin phenotype resides in unique regions and states of these cells' chromatin. Using renin cells at various stages of stimulation, we identified regions in the genome where the chromatin is open for transcription, mapped histone modifications characteristic of active enhancers such as H3K27ac, and tracked deposition of transcriptional activators such as Med1, whose deletion results in ablation of renin expression and low blood pressure. Using the rank ordering of super-enhancers, epigenetic rewriting, and enhancer deletion analysis, we found that renin cells harbor a unique set of super-enhancers that determine their identity. The most prominent renin super-enhancer may act as a chromatin sensor of signals that convey the physiologic status of the organism, and is responsible for the transformation of renin cell descendants to the renin phenotype, a fundamental process to ensure homeostasis.


Assuntos
Epigênese Genética , Código das Histonas , Histonas/metabolismo , Homeostase , Subunidade 1 do Complexo Mediador/metabolismo , Renina/biossíntese , Células-Tronco/metabolismo , Animais , Histonas/genética , Subunidade 1 do Complexo Mediador/genética , Camundongos , Camundongos Transgênicos , Renina/genética , Células-Tronco/citologia
11.
J Biol Chem ; 293(4): 1151-1162, 2018 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-29123029

RESUMO

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.


Assuntos
Teste de Complementação Genética , Sistema Justaglomerular/enzimologia , Renina/biossíntese , Transgenes , Animais , Humanos , Sistema Justaglomerular/patologia , Camundongos , Camundongos Knockout , Renina/genética
12.
PLoS One ; 12(12): e0189084, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29232382

RESUMO

Renin producing cells of the juxtaglomerulus, herein called cells of renin lineage (CoRL), have garnered recent interest for their propensity to act as a progenitor source for various kidney cell types including podocytes. Despite recent advances, the process of transdifferentiation of CoRL to podocytes is poorly understood. In this study, we employed a transgenic reporter mouse line which permanently labels CoRL with ZsGreen fluorescent protein, allowing for isolation by fluorescence-activated cell sorting. At 5 days following induction of abrupt podocyte ablation via anti-podocyte sheep IgG, mice were sacrificed and CoRL were isolated by FACS. RNA was subsequently analyzed by microarray. Gene set enrichment analysis (GSEA) was performed and revealed that CoRL display a distinct phenotype following podocyte ablation, primarily consisting of downregulation of metabolic processes and upregulation of immuno-modulatory processes. Additionally, RNA-biology and cell cycle-related processes were also upregulated. Changes in gene expression or activity of a core set of transcription factors including HNF1 and E2F were identified through changes in enrichment of their respective target genes. However, integration of results from transcription factor and canonical pathway analysis indicated that ERR1 and PU-box family members may be the major contributors to the post-podocyte ablation phenotype of CoRL. Finally, top ranking genes were selected from the microarray-based analysis and confirmed by qPCR. Collectively, our results provide valuable insights into the transcriptional regulation of CoRL following abrupt podocyte ablation.


Assuntos
Linhagem da Célula , Podócitos/metabolismo , Renina/biossíntese , Transcrição Gênica , Animais , Separação Celular , Citometria de Fluxo , Regulação da Expressão Gênica , Córtex Renal/citologia , Córtex Renal/metabolismo , Camundongos , Camundongos Transgênicos , Podócitos/citologia , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição/metabolismo
13.
Curr Hypertens Rep ; 19(8): 62, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28695400

RESUMO

The presence of renin production by the principal cells of the collecting duct has opened new perspectives for the regulation of intrarenal angiotensin II (Ang II). Angiotensinogen (AGT) and angiotensin-converting enzyme (ACE) are present in the tubular fluid coming from the proximal tubule and collecting duct. All the components needed for Ang II formation are present along the nephron, and much is known about the mechanisms regulating renin in juxtaglomerular cells (JG); however, those in the collecting duct remain unclear. Ang II suppresses renin via protein kinase C (PKC) and calcium (Ca2+) in JG cells, but in the principal cells, Ang II increases renin synthesis and release through a pathophysiological mechanism that increases further intratubular Ang II de novo formation to enhance distal Na + reabsorption. Transgenic mice overexpressing renin in the collecting duct demonstrate the role of collecting duct renin in the development of hypertension. The story became even more interesting after the discovery of a specific receptor for renin and prorenin: the prorenin receptor ((P)RR), which enhances renin activity and fully activates prorenin. The interactions between (P)RR and prorenin/renin may further increase intratubular Ang II levels. In addition to Ang II, other mechanisms have been described in the regulation of renin in the collecting duct, including vasopressin (AVP), bradykinin (BK), and prostaglandins. Current active investigations are aimed at elucidating the mechanisms regulating renin in the distal nephron segments and understand its role in the pathogenesis of hypertension.


Assuntos
Hipertensão/metabolismo , Hipertensão/fisiopatologia , Túbulos Renais Coletores/metabolismo , Renina/metabolismo , Angiotensina II/metabolismo , Animais , Humanos , Hipertensão/etiologia , Túbulos Renais Coletores/fisiopatologia , Túbulos Renais Distais/metabolismo , Túbulos Renais Distais/fisiopatologia , Túbulos Renais Proximais/fisiopatologia , Renina/biossíntese , Sistema Renina-Angiotensina/fisiologia
14.
Hypertension ; 69(6): 1145-1155, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28396539

RESUMO

Renin, a key component in the regulation of blood pressure in mammals, is produced by the rare and highly specialized juxtaglomerular cells of the kidney. Chronic stimulation of renin release results in a recruitment of new juxtaglomerular cells by the apparent conversion of adjacent smooth muscle cells along the afferent arterioles. Because juxtaglomerular cells rapidly dedifferentiate when removed from the kidney, their developmental origin and the mechanism that explains their phenotypic plasticity remain unclear. To overcome this limitation, we have performed RNA expression analysis on 4 human renin-producing tumors. The most highly expressed genes that were common between the reninomas were subsequently used for in situ hybridization in kidneys of 5-day-old mice, adult mice, and adult mice treated with captopril. From the top 100 genes, 10 encoding for ligands were selected for further analysis. Medium of human embryonic kidney 293 cells transfected with the mouse cDNA encoding these ligands was applied to (pro)renin-synthesizing As4.1 cells. Among the ligands, only platelet-derived growth factor B reduced the medium and cellular (pro)renin levels, as well as As4.1 renin gene expression. In addition, platelet-derived growth factor B-exposed As4.1 cells displayed a more elongated and aligned shape with no alteration in viability. This was accompanied by a downregulated expression of α-smooth muscle actin and an upregulated expression of interleukin-6, suggesting a phenotypic shift from myoendocrine to inflammatory. Our results add 36 new genes to the list that characterize renin-producing cells and reveal a novel role for platelet-derived growth factor B as a regulator of renin-synthesizing cells.


Assuntos
Perfilação da Expressão Gênica , Sistema Justaglomerular/citologia , Nefropatias/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Renina/biossíntese , Análise de Variância , Animais , Células Cultivadas , Modelos Animais de Doenças , Expressão Gênica , Humanos , Hibridização In Situ , Sistema Justaglomerular/metabolismo , Nefropatias/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Renina/genética , Transdução de Sinais
15.
J Hypertens ; 35(2): 330-337, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28005703

RESUMO

BACKGROUND: The function of prorenin, the inactive precursor of renin, remains unclear after many decades of research. The discovery of a (pro)renin receptor suggested that prorenin, by binding to this receptor, would become active, that is, obtain an 'open' conformation. However, the receptor only interacted with prorenin at levels that were many orders of magnitude above its normal levels, making such interaction in-vivo unlikely. Prorenin occurs in two conformations, an open, active form, and a closed, inactive form. Under physiological conditions (pH 7.4, 37 °C), virtually all prorenin occurs in the closed conformation. This study investigated to what degree prorenin-synthesizing cells release prorenin in an open conformation. METHODS AND RESULTS: Renin plus prorenin-synthesizing human mast cells, and prorenin-synthesizing HEK293 cells (transfected with the mammalian expression vector pRhR1100, containing human prorenin) and human decidua cells were incubated with the renin inhibitor VTP-27999. This inhibitor will trap open prorenin, as after VTP-27999 binding, prorenin can no longer return to its closed conformation, thus allowing its detection in a renin immunoradiometric assay. No evidence for the release of open prorenin was found. Moreover, incubating decidua cells with angiotensinogen yielded low angiotensin levels, corresponding with the activity of ≈1% of prorenin in the medium, that is, the amount of open prorenin expected based upon the equilibrium between open and closed prorenin under physiological conditions. CONCLUSION: Our study does not reveal evidence for the release of open, active prorenin by prorenin-synthesizing cells, at least under cell culture conditions. This argues against prorenin activity at the site of its release.


Assuntos
Renina/biossíntese , Renina/química , Adulto , Angiotensinogênio/farmacologia , Angiotensinas/metabolismo , Anti-Hipertensivos , Carbamatos/farmacologia , Decídua/citologia , Decídua/efeitos dos fármacos , Decídua/metabolismo , Feminino , Células HEK293/efeitos dos fármacos , Células HEK293/metabolismo , Humanos , Masculino , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Pessoa de Meia-Idade , Piperidinas/farmacologia , Renina/antagonistas & inibidores
16.
Swiss Med Wkly ; 146: w14365, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27922171

RESUMO

Efficient cell communication is a prerequisite for the coordinated function of tissues and organs. In vertebrates, this communication is mediated by a variety of mechanisms, including the exchange of molecules between cells, and between cells and the extracellular medium, via membrane channels made of connexin and pannexin proteins. These channels are a necessary component of all human tissues. Here, we review the biological essentials of the connexin and pannexin families, and the roles of these proteins in the function of cells which are central to major human diseases. We then discuss how connexins and pannexins participate in human pathology, and the clinical perspectives that this knowledge opens.


Assuntos
Conexinas/genética , Conexinas/metabolismo , Células Endoteliais/metabolismo , Células Secretoras de Insulina/metabolismo , Canais Iônicos/metabolismo , Neovascularização Patológica/metabolismo , Transporte Biológico/fisiologia , Comunicação Celular , Junções Comunicantes/metabolismo , Humanos , Neoplasias/fisiopatologia , Renina/biossíntese
17.
Physiol Rep ; 4(8)2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27122048

RESUMO

Short-term maternal corticosterone (Cort) administration at mid-gestation in the mouse reduces nephron number in both sexes while programming renal and cardiovascular dysfunction in 12-month male but not female offspring. The renal renin-angiotensin-aldosterone system (RAAS), functions in a sexually dimorphic manner to regulate both renal and cardiovascular physiology. This study aimed to identify if there are sex-specific differences in basal levels of the intrarenal RAAS and to determine the impact of maternal Cort exposure on the RAAS in male and female offspring at 6 months of age. While intrarenal renin concentrations were higher in untreated females compared to untreated males, renal angiotensin II concentrations were higher in males than females. Furthermore, basal plasma aldosterone concentrations were greater in females than males. Cort exposed male but not female offspring had reduced water intake and urine excretion. Cort exposure increased renal renin concentrations and elevated mRNA expression of Ren1, Ace2, and Mas1 in male but not female offspring. In addition, male Cort exposed offspring had increased expression of the aldosterone receptor, Nr3c2 and renal sodium transporters. In contrast, Cort exposure increased Agtr1a mRNA levels in female offspring only. This study demonstrates that maternal Cort exposure alters key regulators of renal function in a sex-specific manner at 6 months of life. These finding likely contribute to the disease outcomes in male but not female offspring in later life and highlights the importance of renal factors other than nephron number in the programming of renal and cardiovascular disease.


Assuntos
Corticosterona/toxicidade , Rim/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Caracteres Sexuais , Angiotensina II/biossíntese , Animais , Western Blotting , Modelos Animais de Doenças , Feminino , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Proto-Oncogene Mas , RNA Mensageiro/análise , Renina/biossíntese , Sistema Renina-Angiotensina/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
PLoS One ; 11(2): e0148542, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26866372

RESUMO

We studied Mesenchymal Stromal Cells (MSC) effects in experimental Unilateral Ureteral Obstruction (UUO), a fibrogenic renal disease. Rats were divided in 5 groups: sham, UUO, MSC treated-UUO, ACEi treated-UUO, MSC+ACEi treated- UUO. Data were collected at 1, 7, 21 days. UUO induced monocyte renal infiltration, tubular cell apoptosis, tubular atrophy, interstitial fibrosis and overexpression of TGFß, Renin mRNA (RENmRNA), increase of Renin, Angiotensin II (AII) and aldosterone serum levels. Both lisinopril (ACEi) and MSC treatment prevented monocyte infiltration, reduced tubular cell apoptosis, renal fibrosis and TGFß expression. Combined therapy provided a further suppression of monocyte infiltration and tubular injury. Lisinopril alone caused a rebound activation of Renin-Angiotensin System (RAS), while MSC suppressed RENmRNA and Renin synthesis and induced a decrease of AII and aldosterone serum levels. Furthermore, in in-vitro and in-vivo experiments, MSC inhibit Human antigen R (HuR) trascription, an enhancer of RENmRNA stability by IL10 release. In conclusion, we demonstrate that in UUO MSC prevent fibrosis, by decreasing HuR-dependent RENmRNA stability. Our findings give a clue to understand the molecular mechanism through which MSC may prevent fibrosis in a wide and heterogeneous number of diseases that share RAS activation as common upstream pathogenic mechanism.


Assuntos
Proteína Semelhante a ELAV 1/fisiologia , Fibrose/fisiopatologia , Rim/fisiopatologia , Células-Tronco Mesenquimais/citologia , Sistema Renina-Angiotensina , Obstrução Ureteral/fisiopatologia , Aldosterona/metabolismo , Angiotensina II/metabolismo , Animais , Animais Geneticamente Modificados , Apoptose , Diferenciação Celular , Linhagem Celular , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunofenotipagem , Interleucina-10/metabolismo , Túbulos Renais/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Renina/biossíntese , Fator de Crescimento Transformador beta/metabolismo , Obstrução Ureteral/terapia
19.
Am J Physiol Renal Physiol ; 310(4): F284-93, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26608789

RESUMO

Renin is synthesized in the principal cells of the collecting duct (CD), and its production is increased via cAMP in angiotensin (ANG) II-dependent hypertension, despite suppression of juxtaglomerular (JG) renin. Vasopressin, one of the effector hormones of the renin-angiotensin system (RAS) via the type 2-receptor (V2R), activates the cAMP/PKA/cAMP response element-binding protein (CREB) pathway and aquaporin-2 expression in principal cells of the CD. Accordingly, we hypothesized that activation of V2R increases renin synthesis via PKA/CREB, independently of ANG II type 1 (AT1) receptor activation in CD cells. Desmopressin (DDAVP; 10(-6) M), a selective V2R agonist, increased renin mRNA (∼3-fold), prorenin (∼1.5-fold), and renin (∼2-fold) in cell lysates and cell culture media in the M-1 CD cell line. Cotreatment with DDAVP+H89 (PKA inhibitor) or CREB short hairpin (sh) RNA prevented this response. H89 also blunted DDAVP-induced CREB phosphorylation and nuclear localization. In 48-h water-deprived (WD) mice, prorenin-renin protein levels were increased in the renal inner medulla (∼1.4- and 1.8-fold). In WD mice treated with an ACE inhibitor plus AT1 receptor blockade, renin mRNA and prorenin protein levels were still higher than controls, while renin protein content was not changed. In M-1 cells, ANG II or DDAVP increased prorenin-renin protein levels; however, there were no further increases by combined treatment. These results indicate that in the CD the activation of the V2R stimulates renin synthesis via the PKA/CREB pathway independently of RAS, suggesting a critical role for vasopressin in the regulation of renin in the CD.


Assuntos
Túbulos Renais Coletores/efeitos dos fármacos , Túbulos Renais Coletores/metabolismo , Receptores de Vasopressinas/agonistas , Renina/biossíntese , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Desamino Arginina Vasopressina/farmacologia , Técnicas de Silenciamento de Genes , Isoquinolinas/farmacologia , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Camundongos , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/biossíntese , RNA Interferente Pequeno/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Sulfonamidas/farmacologia
20.
J Mol Med (Berl) ; 94(1): 61-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26256830

RESUMO

UNLABELLED: In the heart, secretory renin promotes hypertrophy, apoptosis, necrosis, fibrosis, and cardiac failure through angiotensin generation from angiotensinogen. Thus, inhibitors of the renin-angiotensin system are among the most potent drugs in the treatment of cardiac failure. Renin transcripts have been identified encoding a renin isoform with unknown targets and unknown functions that are localized to the cytosol and mitochondria. We hypothesize that this isoform, in contrast to secretory renin, exerts cardioprotective effects in an angiotensin-independent manner. Cells overexpressing cytosolic renin were generated by transfection or obtained from CX(exon2-9)renin transgenic rats. Overexpression of cytosolic renin reduced the rate of necrosis in H9c2 cardiomyoblasts and in primary cardiomyocytes after glucose depletion. These effects were not mediated by angiotensin generation since an inhibitor of renin activity did not influence the in vitro effects. siRNA-mediated knockdown of endogenous cytosolic renin increased the rate of necrosis and aggravated the pro-necrotic effects of glucose depletion. Isolated perfused hearts obtained from transgenic rats overexpressing cytosolic renin exhibited a 50% reduction of infarct size after ischemia-reperfusion injury. Cytosolic renin is essential for survival, both under basal conditions and during glucose starvation. The protective effects are angiotensin-independent and contrary to the known actions of secretory renin. KEY MESSAGES: A cytosolic isoform of renin with unknown functions is expressed in the heart. Cytosolic renin diminishes ischemia induced damage to the heart. The protective effects of cytosolic renin contradict the known function of secretory renin. The effects of cytosolic renin are not mediated via angiotensin generation. Renin-binding protein is a potential target for cytosolic renin.


Assuntos
Cardiotônicos/metabolismo , Isquemia Miocárdica/prevenção & controle , Necrose/prevenção & controle , Renina/metabolismo , Angiotensinogênio/metabolismo , Animais , Células Cultivadas , Citosol/metabolismo , Glucose/metabolismo , Coração/fisiopatologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Isoformas de Proteínas/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Ratos Wistar , Renina/antagonistas & inibidores , Renina/biossíntese , Renina/genética , Sistema Renina-Angiotensina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...