Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 399
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 128(7): 887-907, 2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33793334

RESUMEN

Renin cells are essential for survival perfected throughout evolution to ensure normal development and defend the organism against a variety of homeostatic threats. During embryonic and early postnatal life, they are progenitors that participate in the morphogenesis of the renal arterial tree. In adult life, they are capable of regenerating injured glomeruli, control blood pressure, fluid-electrolyte balance, tissue perfusion, and in turn, the delivery of oxygen and nutrients to cells. Throughout life, renin cell descendants retain the plasticity or memory to regain the renin phenotype when homeostasis is threatened. To perform all of these functions and maintain well-being, renin cells must regulate their identity and fate. Here, we review the major mechanisms that control the differentiation and fate of renin cells, the chromatin events that control the memory of the renin phenotype, and the major pathways that determine their plasticity. We also examine how chronic stimulation of renin cells alters their fate leading to the development of a severe and concentric hypertrophy of the intrarenal arteries and arterioles. Lastly, we provide examples of additional changes in renin cell fate that contribute to equally severe kidney disorders.


Asunto(s)
Hipertensión/etiología , Riñón/citología , Renina/fisiología , Animales , Arteriolas/embriología , Presión Sanguínea/fisiología , Comunicación Celular , Diferenciación Celular , Plasticidad de la Célula , Cromatina/fisiología , Ensamble y Desensamble de Cromatina/fisiología , Conexinas/fisiología , Homeostasis , Humanos , Integrinas/fisiología , Aparato Yuxtaglomerular/citología , Riñón/irrigación sanguínea , Riñón/embriología , Glomérulos Renales/fisiología , Ratones , MicroARNs/fisiología , Fenotipo , Regeneración/fisiología , Arteria Renal , Renina/metabolismo , Sistema Renina-Angiotensina/fisiología , Células Madre/fisiología , Equilibrio Hidroelectrolítico
2.
Am J Physiol Renal Physiol ; 321(6): F689-F704, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34693742

RESUMEN

Macula densa (MD) cells, a chief sensory cell type in the nephron, are endowed with unique microanatomic features including a high density of protein synthetic organelles and secretory vesicles in basal cell processes ("maculapodia") that suggest a so far unknown high rate of MD protein synthesis. This study aimed to explore the rate and regulation of MD protein synthesis and their effects on glomerular function using novel transgenic mouse models, newly established fluorescence cell biology techniques, and intravital microscopy. Sox2-tdTomato kidney tissue sections and an O-propargyl puromycin incorporation-based fluorescence imaging assay showed that MD cells have the highest level of protein synthesis within the kidney cortex followed by intercalated cells and podocytes. Genetic gain of function of mammalian target of rapamycin (mTOR) signaling specifically in MD cells (in MD-mTORgof mice) or their physiological activation by low-salt diet resulted in further significant increases in the synthesis of MD proteins. Specifically, these included both classic and recently identified MD-specific proteins such as cyclooxygenase 2, microsomal prostaglandin E2 synthase 1, and pappalysin 2. Intravital imaging of the kidney using multiphoton microscopy showed significant increases in afferent and efferent arteriole and glomerular capillary diameters and blood flow in MD-mTORgof mice coupled with an elevated glomerular filtration rate. The presently identified high rate of MD protein synthesis that is regulated by mTOR signaling is a novel component of the physiological activation and glomerular hemodynamic regulatory functions of MD cells that remains to be fully characterized.NEW & NOTEWORTHY This study discovered the high rate of protein synthesis in macula densa (MD) cells by applying direct imaging techniques with single cell resolution. Physiological activation and mammalian target of rapamycin signaling played important regulatory roles in this process. This new feature is a novel component of the tubuloglomerular cross talk and glomerular hemodynamic regulatory functions of MD cells. Future work is needed to elucidate the nature and (patho)physiological role of the specific proteins synthesized by MD cells.


Asunto(s)
Aparato Yuxtaglomerular/metabolismo , Biosíntesis de Proteínas , Animales , Comunicación Autocrina , Dieta Hiposódica , Tasa de Filtración Glomerular , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Microscopía Intravital , Aparato Yuxtaglomerular/citología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Comunicación Paracrina , Renina/metabolismo , Transducción de Señal , Sodio en la Dieta/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteína Fluorescente Roja
3.
Gen Comp Endocrinol ; 296: 113533, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32561435

RESUMEN

Renin or a renin-like enzyme evolved in ancestral vertebrates and is conserved along the vertebrate phylogeny. The ontogenic development of renin, however, is not well understood in nonmammalian vertebrates. We aimed to determine the expression patterns and relative abundance of renin mRNA in pre- and postnatal chickens (Gallus gallus, White Leghorn breed). Embryonic day 13 (E13) embryos show renal tubules, undifferentiated mesenchymal structures, and a small number of developing glomeruli. Maturing glomeruli are seen in post-hatch day 4 (D4) and day 30 (D30) kidneys, indicating that nephrogenic activity still exists in kidneys of 4-week-old chickens. In E13 embryos, renin mRNA measured by quantitative polymerase chain reaction in the adrenal glands is equivalent to the expression in the kidneys, whereas in post-hatch D4 and D30 maturing chicks, renal renin expressions increased 2-fold and 11-fold, respectively. In contrast, relative renin expression in the adrenals became lower than in the kidneys. Furthermore, renin expression is clearly visible by in situ hybridization in the juxtaglomerular (JG) area in D4 and D30 chicks, but not in E13 embryos. The results suggest that in chickens, renin evolved in both renal and extrarenal organs at an early stage of ontogeny and, with maturation, became localized to the JG area. Clear JG structures are not morphologically detectable in E13 embryos, but are visible in 30-day-old chicks, supporting this concept.


Asunto(s)
Pollos/genética , Regulación de la Expresión Génica , Renina/genética , Animales , Embrión de Pollo , Aparato Yuxtaglomerular/citología , Aparato Yuxtaglomerular/metabolismo , Organogénesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Renina/metabolismo , Sistema Renina-Angiotensina
4.
Int J Mol Sci ; 21(12)2020 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-32585970

RESUMEN

Connexin hemichannels play an important role in the control of cellular signaling and behaviors. Given that lowering extracellular Ca2+, a condition that activates hemichannels, is a well-characterized stimulator of renin in juxtaglomerular cells, we, therefore, tested a potential implication of hemichannels in the regulation of renin in As4.1 renin-secreting cells. Lowering extracellular Ca2+ induced hemichannel opening, which was associated with cAMP signaling pathway activation and increased renin production. Blockade of hemichannels with inhibitors or downregulation of Cxs with siRNAs abrogated the activation of cAMP pathway and the elevation of renin. Further analysis revealed that cAMP pathway activation was blocked by adenylyl cyclase inhibitor SQ 22536, suggesting an implication of adenyl cyclase. Furthermore, the participation of hemichannels in the activation of the cAMP signaling pathway was also observed in a renal tubular epithelial cell line NRK. Collectively, our results characterized the hemichannel opening as a presently unrecognized molecular event involved in low Ca2+-elicited activation of cAMP pathway and renin production. Our findings thus provide novel mechanistic insights into the low Ca2+-initiated cell responses. Given the importance of cAMP signaling pathway in the control of multiple cellular functions, our findings also highlight the importance of Cx-forming channels in various pathophysiological situations.


Asunto(s)
Calcio/metabolismo , Conexinas/metabolismo , AMP Cíclico/metabolismo , Uniones Comunicantes/fisiología , Aparato Yuxtaglomerular/metabolismo , Renina/metabolismo , Adenosina Trifosfato , Animales , Células Cultivadas , Aparato Yuxtaglomerular/citología , Ratones , Transducción de Señal
5.
Pflugers Arch ; 470(6): 969-978, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29427253

RESUMEN

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.


Asunto(s)
Calcio/metabolismo , Conexinas/metabolismo , Aparato Yuxtaglomerular/metabolismo , Renina/metabolismo , Animales , Conexinas/genética , Femenino , Aparato Yuxtaglomerular/citología , Masculino , Ratones , Proteína alfa-5 de Unión Comunicante
6.
Curr Hypertens Rep ; 19(2): 14, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28233238

RESUMEN

During development, renin cells are precursors for arteriolar smooth muscle, mesangial cells, and interstitial pericytes. Those seemingly differentiated descendants retain the memory to re-express renin when there is a threat to homeostasis. Understanding how such molecular memory is constructed and regulated would be crucial to comprehend cell identity which is, in turn, intimately linked to homeostasis.


Asunto(s)
Plasticidad de la Célula , Homeostasis/fisiología , Aparato Yuxtaglomerular/citología , Riñón/irrigación sanguínea , Renina/fisiología , Animales , Humanos , Aparato Yuxtaglomerular/fisiología
7.
Development ; 140(24): 4850-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24227652

RESUMEN

COUP-TFI is an orphan nuclear receptor acting as a strong transcriptional regulator in different aspects of forebrain embryonic development. In this study, we investigated COUP-TFI expression and function in the mouse olfactory bulb (OB), a highly plastic telencephalic region in which continuous integration of newly generated inhibitory interneurons occurs throughout life. OB interneurons belong to different populations that originate from distinct progenitor lineages. Here, we show that COUP-TFI is highly expressed in tyrosine hydroxylase (TH)-positive dopaminergic interneurons in the adult OB glomerular layer (GL). We found that odour deprivation, which is known to downregulate TH expression in the OB, also downregulates COUP-TFI in dopaminergic cells, indicating a possible correlation between TH- and COUP-TFI-activity-dependent action. Moreover, we demonstrate that conditional inactivation of COUP-TFI in the EMX1 lineage results in a significant reduction of both TH and ZIF268 expression in the GL. Finally, lentiviral vector-mediated COUP-TFI deletion in adult-generated interneurons confirmed that COUP-TFI acts cell-autonomously in the control of TH and ZIF268 expression. These data indicate that COUP-TFI regulates TH expression in OB cells through an activity-dependent mechanism involving ZIF268 induction and strongly argue for a maintenance rather than establishment function of COUP-TFI in dopaminergic commitment. Our study reveals a previously unknown role for COUP-TFI in the adult brain as a key regulator in the control of sensory-dependent plasticity in olfactory dopaminergic neurons.


Asunto(s)
Factor de Transcripción COUP I/metabolismo , Neuronas Dopaminérgicas/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Bulbo Olfatorio/metabolismo , Tirosina 3-Monooxigenasa/biosíntesis , Animales , Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Proteínas de Homeodominio/metabolismo , Aparato Yuxtaglomerular/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Privación Sensorial , Olfato/fisiología , Factores de Transcripción/metabolismo
8.
BMC Med Genet ; 17: 21, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26969407

RESUMEN

BACKGROUND: Plasma coagulation Factor XIIa (Hageman factor; encoded by F12) and kallikrein (KAL or Fletcher factor; encoded by KLKB1) are proteases of the kallikerin-kinin system involved in converting the inactive circulating prorenin to renin. Renin is a key enzyme in the formation of angiotensin II, which regulates blood pressure, fluid and electrolyte balance and is a biomarker for cardiovascular, metabolic and renal function. The renin-angiotensin system is implicated in extinction learning in posttraumatic stress disorder. METHODS & RESULTS: Active plasma renin was measured from two independent cohorts- civilian twins and siblings, as well as U.S. Marines, for a total of 1,180 subjects. Genotyping these subjects revealed that the carriers of the minor alleles at the two loci- F12 and KLKB1 had a significant association with reduced levels of active plasma renin. Meta-analyses confirmed the association across cohorts. In vitro studies verified digestion of human recombinant pro-renin by kallikrein (KAL) to generate active renin. Subsequently, the active renin was able to digest the synthetic substrate angiotensinogen to angiotensin-I. Examination of mouse juxtaglomerular cell line and mouse kidney sections showed co-localization of KAL with renin. Expression of either REN or KLKB1 was regulated in cell line and rodent models of hypertension in response to oxidative stress, interleukin or arterial blood pressure changes. CONCLUSIONS: The functional variants of KLKB1 (rs3733402) and F12 (rs1801020) disrupted the cascade of enzymatic events, resulting in diminished formation of active renin. Using genetic, cellular and molecular approaches we found that conversion of zymogen prorenin to renin was influenced by these polymorphisms. The study suggests that the variant version of protease factor XIIa due to the amino acid substitution had reduced ability to activate prekallikrein to KAL. As a result KAL has reduced efficacy in converting prorenin to renin and this step of the pathway leading to activation of renin affords a potential therapeutic target.


Asunto(s)
Factor XIIa/genética , Calicreínas/genética , Polimorfismo de Nucleótido Simple , Sistema Renina-Angiotensina/genética , Renina/sangre , Adolescente , Adulto , Anciano , Alelos , Angiotensina I/sangre , Angiotensinógeno/sangre , Animales , Presión Sanguínea , Proteínas de Ciclo Celular , Línea Celular , Regulación de la Expresión Génica , Sitios Genéticos , Estudio de Asociación del Genoma Completo , Técnicas de Genotipaje , Humanos , Hipertensión/genética , Aparato Yuxtaglomerular/citología , Calicreínas/sangre , Masculino , Ratones , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Precalicreína/metabolismo , Renina/genética , Serina Endopeptidasas/metabolismo , Transferasas , Adulto Joven
9.
J Neurosci ; 33(7): 2916-26, 2013 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-23407950

RESUMEN

Evidence for coexpression of two or more classic neurotransmitters in neurons has increased, but less is known about cotransmission. Ventral tegmental area (VTA) neurons corelease dopamine (DA), the excitatory transmitter glutamate, and the inhibitory transmitter GABA onto target cells in the striatum. Olfactory bulb (OB) short axon cells (SACs) form interglomerular connections and coexpress markers for DA and GABA. Using an optogenetic approach, we provide evidence that mouse OB SACs release both GABA and DA onto external tufted cells (ETCs) in other glomeruli. Optical activation of channelrhodopsin specifically expressed in DAergic SACs produced a GABA(A) receptor-mediated monosynaptic inhibitory response, followed by DA-D(1)-like receptor-mediated excitatory response in ETCs. The GABA(A) receptor-mediated hyperpolarization activates I(h) current in ETCs; synaptically released DA increases I(h), which enhances postinhibitory rebound spiking. Thus, the opposing actions of synaptically released GABA and DA are functionally integrated by I(h) to generate an inhibition-to-excitation "switch" in ETCs. Consistent with the established role of I(h) in ETC burst firing, we show that endogenous DA release increases ETC spontaneous bursting frequency. ETCs transmit sensory signals to mitral/tufted output neurons and drive intraglomerular inhibition to shape glomerulus output to downstream olfactory networks. GABA and DA cotransmission from SACs to ETCs may play a key role in regulating output coding across the glomerular array.


Asunto(s)
Axones/metabolismo , Dopamina/metabolismo , Neuronas/metabolismo , Bulbo Olfatorio/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Axones/fisiología , Channelrhodopsins , Estimulación Eléctrica , Glutamato Descarboxilasa/metabolismo , Humanos , Inmunohistoquímica , Aparato Yuxtaglomerular/citología , Rayos Láser , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/fisiología , Bulbo Olfatorio/citología , Técnicas de Placa-Clamp , Estimulación Luminosa , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Transmisión Sináptica/fisiología , Tirosina 3-Monooxigenasa/genética
10.
Am J Physiol Regul Integr Comp Physiol ; 307(5): R505-13, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24965790

RESUMEN

Renin, an aspartyl protease that catalyzes the rate-limiting step of the renin-angiotensin system, is first synthesized as an inactive precursor, prorenin. Prorenin is activated by the proteolytic removal of an amino terminal prosegment in the dense granules of the juxtaglomerular (JG) cells of the kidney by one or more proteases whose identity is uncertain but commonly referred to as the prorenin-processing enzyme (PPE). Because several extrarenal tissues secrete only prorenin, we tested the hypothesis that the unique ability of JG cells to produce active renin might be explained by the existence of a PPE whose expression is restricted to JG cells. We found that inducing renin production by the mouse kidney by up to 20-fold was not associated with the concomitant induction of candidate PPEs. Because the renin-containing granules of JG cells also contain several lysosomal hydrolases, we engineered mouse Ren1 prorenin to be targeted to the classical vesicular lysosomes of cultured HEK-293 cells, where it was accurately processed and stored. Furthermore, we found that HEK cell lysosomes hydrolyzed any artificial extensions placed on the protein and that active renin was extraordinarily resistant to proteolytic degradation. Altogether, our results demonstrate that accurate processing of prorenin is not restricted to JG cells but can occur in classical vesicular lysosomes of heterologous cells. The implication is that renin production may not require a specific PPE but rather can be achieved by general hydrolysis in the lysosome-like granules of JG cells.


Asunto(s)
Aparato Yuxtaglomerular/metabolismo , Lisosomas/metabolismo , Renina/metabolismo , Animales , Células Cultivadas , Cisteína Endopeptidasas/metabolismo , Células HEK293 , Humanos , Hidrólisis , Aparato Yuxtaglomerular/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Sistema Renina-Angiotensina/fisiología
11.
J Am Soc Nephrol ; 24(8): 1263-73, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23744888

RESUMEN

The renin-angiotensin-aldosterone system (RAAS) regulates BP and salt-volume homeostasis. Juxtaglomerular (JG) cells synthesize and release renin, which is the first and rate-limiting step in the RAAS. Intense pathologic stresses cause a dramatic increase in the number of renin-producing cells in the kidney, termed JG cell recruitment, but how this occurs is not fully understood. Here, we isolated renal CD44(+) mesenchymal stem cell (MSC)-like cells and found that they differentiated into JG-like renin-expressing cells both in vitro and in vivo. Sodium depletion and captopril led to activation and differentiation of these cells into renin-expressing cells in the adult kidney. In summary, CD44(+) MSC-like cells exist in the adult kidney and can differentiate into JG-like renin-producing cells under conditions that promote JG cell recruitment.


Asunto(s)
Células Madre Adultas/metabolismo , Captopril/farmacología , Diferenciación Celular/fisiología , Aparato Yuxtaglomerular/citología , Riñón/citología , Células Madre Mesenquimatosas/metabolismo , Sistema Renina-Angiotensina/fisiología , Renina/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Aparato Yuxtaglomerular/metabolismo , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Renina-Angiotensina/efectos de los fármacos
12.
Pflugers Arch ; 465(1): 71-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22744230

RESUMEN

Renin synthesis and renin secretion at the level of renal juxtaglomerular cells are regulated by neurotransmitters, hormones, paracrine, and mechanical signals. Although morphological evidence has indicated an intense intercellular communication of renin cells via connexins between the cells composing the juxtaglomerlar area, the functional behavior of renin-secreting cells has been considered of that of individual isolated cells for a long time. Findings obtained during recent years shed first light on the functional relevance of connexins for the control of renin secretion and also for the positioning of renin-secreting cells in the kidney. This short review aims to summarize these findings and tries to set them into a functional context.


Asunto(s)
Conexinas/metabolismo , Aparato Yuxtaglomerular/metabolismo , Renina/metabolismo , Animales , Uniones Comunicantes/metabolismo , Humanos , Aparato Yuxtaglomerular/citología , Sistema Renina-Angiotensina
13.
Pflugers Arch ; 465(1): 25-37, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22733355

RESUMEN

A major rate-limiting step in the renin-angiotensin-aldosterone system is the release of active renin from endocrine cells (juxtaglomerular (JG) cells) in the media layer of the afferent glomerular arterioles. The number and distribution of JG cells vary with age and the physiological level of stimulation; fetal life and chronic stimulation by extracellular volume contraction is associated with recruitment of renin-producing cells. Upon stimulation of renin release, labeled renin granules "disappear;" the number of granules decrease; cell membrane surface area increases in single cells, and release is quantal. Together, this indicates exocytosis as the predominant mode of release. JG cells release few percent of total renin content by physiological stimulation, and recruitment of renin cells is preferred to recruitment of granules during prolonged stimulation. Several endocrine and paracrine agonists, neurotransmitters, and cell swelling converge on the stimulatory cyclic AMP (cAMP) pathway. Renin secretion is attenuated in mice deficient in beta-adrenoceptors, prostaglandin E(2)-EP4 receptors, Gsα protein, and adenylyl cyclases 5 and 6. Phosphodiesterases (PDE) 3 and 4 degrade cAMP in JG cells, and PDE3 is inhibited by cyclic GMP (cGMP) and couples the cGMP pathway to the cAMP pathway. Cyclic AMP enhances K(+)-current in JG cells and is permissive for secretion by stabilizing membrane potential far from threshold that activates L-type voltage-gated calcium channels. Intracellular calcium paradoxically inhibits renin secretion likely through attenuated formation and enhanced degradation of cAMP; by activation of chloride currents and interaction with calcineurin. Connexin 40 is necessary for localization of JG cells in the vascular wall and for pressure- and macula densa-dependent suppression of renin release.


Asunto(s)
Aparato Yuxtaglomerular/metabolismo , Sistema Renina-Angiotensina , Renina/metabolismo , Animales , Diferenciación Celular , Humanos , Aparato Yuxtaglomerular/citología , Aparato Yuxtaglomerular/fisiología , Potenciales de la Membrana , Vías Secretoras , Transducción de Señal
14.
Pflugers Arch ; 465(6): 895-905, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23229015

RESUMEN

The protease renin is the key enzyme in the renin-angiotensin system (RAS) that regulates extracellular volume and blood pressure. Renin is synthesized in renal juxtaglomerular cells (JG cells) as the inactive precursor prorenin. Activation of prorenin by cleavage of the prosegment occurs in renin storage vesicles that have lysosomal properties. To characterize the renin storage vesicles more precisely, the expression and functional relevance of the major lysosomal membrane proteins lysosomal-associated membrane protein 1 (LAMP-1), LAMP-2, and lysosomal integral membrane protein 2 (LIMP-2) were determined in JG cells. Immunostaining experiments revealed strong coexpression of renin with the LIMP-2 (SCARB2), while faint staining of LAMP-1 and LAMP-2 was detected in some JG cells only. Stimulation of the renin system (ACE inhibitor, renal hypoperfusion) resulted in the recruitment of renin-producing cells in the afferent arterioles and parallel upregulation of LIMP-2, but not LAMP-1 or LAMP-2. Despite the coregulation of renin and LIMP-2, LIMP-2-deficient mice had normal renal renin mRNA levels, renal renin and prorenin contents, and plasma renin and prorenin concentrations under control conditions and in response to stimulation with a low salt diet (with or without angiotensin-converting enzyme (ACE) inhibition). No differences in the size or number of renin vesicles were detected using electron microscopy. Acute stimulation of renin release by isoproterenol exerted similar responses in both genotypes in vivo and in isolated perfused kidneys. Renin and the major lysosomal protein LIMP-2 are colocalized and coregulated in renal JG cells, further corroborating the lysosomal nature of renin storage vesicles. LIMP-2 does not appear to play an obvious role in the regulation of renin synthesis or release.


Asunto(s)
Antígenos CD36/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Renina/metabolismo , Vesículas Secretoras/metabolismo , Regulación hacia Arriba , Animales , Antígenos CD36/genética , Dieta Hiposódica , Aparato Yuxtaglomerular/citología , Aparato Yuxtaglomerular/metabolismo , Aparato Yuxtaglomerular/fisiología , Proteínas de Membrana de los Lisosomas/genética , Ratones , Ratones Noqueados , Renina/sangre , Transcripción Genética
15.
Am J Physiol Renal Physiol ; 304(5): F498-504, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23269646

RESUMEN

Renin, the rate-limiting enzyme in the formation of angiotensin II, is synthesized and stored in granules in juxtaglomerular (JG) cells. Therefore, the controlled mechanism involved in renin release is essential for the regulation of blood pressure. Exocytosis of renin-containing granules is likely involved in renin release; a process stimulated by cAMP. We found that the "soluble NSF (N-ethylmaleimide-sensitive factor) attachment protein receptor" (SNARE) protein VAMP2 mediates cAMP-stimulated renin release and exocytosis in JG cells. To mediate exocytosis, VAMP2 must interact with a synaptosome-associated protein (SNAP). In the renal cortex, the isoform SNAP23 is abundantly expressed. We hypothesized that SNAP23 mediates cAMP-stimulated renin release from primary cultures of mouse JG cells. We found that SNAP23 protein is expressed and colocalized with renin-containing granules in primary cultures of mouse JG cell lysates. Thus, we then tested the involvement of SNAP23 in cAMP-stimulated renin release by transducing JG cells with a dominant-negative SNAP23 construct. In control JG cells transduced with a scrambled sequence, increasing cAMP stimulated renin release from 1.3 ± 0.3 to 5.3 ± 1.2% of renin content. In cells transduced with dominant-negative SNAP23, cAMP increased renin from 1.0 ± 0.1 to 3.0 ± 0.6% of renin content, a 50% blockade. Botulinum toxin E, which cleaves and inactivates SNAP23, reduced cAMP-stimulated renin release by 42 ± 17%. Finally, adenovirus-mediated silencing of SNAP23 significantly blocked cAMP-stimulated renin release by 50 ± 13%. We concluded that the SNARE protein SNAP23 mediates cAMP-stimulated renin release. These data show that renin release is a SNARE-dependent process.


Asunto(s)
AMP Cíclico/metabolismo , Aparato Yuxtaglomerular/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Renina/metabolismo , Animales , Células Cultivadas , Colforsina/farmacología , Aparato Yuxtaglomerular/citología , Aparato Yuxtaglomerular/efectos de los fármacos , Ratones , Proteínas Qb-SNARE/genética , Proteínas Qc-SNARE/genética
16.
Am J Physiol Renal Physiol ; 304(3): F248-56, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23220722

RESUMEN

The calcium-sensing receptor (CaSR) is a G-coupled protein expressed in renal juxtaglomerular (JG) cells. Its activation stimulates calcium-mediated decreases in cAMP content and inhibits renin release. The postreceptor pathway for the CaSR in JG cells is unknown. In parathyroids, CaSR acts through G(q) and/or G(i). Activation of G(q) stimulates phospholipase C (PLC), and inositol 1,4,5-trisphosphate (IP(3)), releasing calcium from intracellular stores. G(i) stimulation inhibits cAMP formation. In afferent arterioles, the ryanodine receptor (RyR) enhances release of stored calcium. We hypothesized JG cell CaSR activation inhibits renin via the PLC/IP(3) and also RyR activation, increasing intracellular calcium, suppressing cAMP formation, and inhibiting renin release. Renin release from primary cultures of isolated mouse JG cells (n = 10) was measured. The CaSR agonist cinacalcet decreased renin release 56 ± 7% of control (P < 0.001), while the PLC inhibitor U73122 reversed cinacalcet inhibition of renin (104 ± 11% of control). The IP(3) inhibitor 2-APB also reversed inhibition of renin from 56 ± 6 to 104 ± 11% of control (P < 0.001). JG cells were positively labeled for RyR, and blocking RyR reversed CaSR-mediated inhibition of renin from 61 ± 8 to 118 ± 22% of control (P < 0.01). Combining inhibition of IP(3) and RyR was not additive. G(i) inhibition with pertussis toxin plus cinacalcet did not reverse renin inhibition (65 ± 12 to 41 ± 8% of control, P < 0.001). We conclude stimulating JG cell CaSR activates G(q), initiating the PLC/IP(3) pathway, activating RyR, increasing intracellular calcium, and resulting in calcium-mediated renin inhibition.


Asunto(s)
Inositol 1,4,5-Trifosfato/metabolismo , Aparato Yuxtaglomerular/metabolismo , Receptores Sensibles al Calcio/metabolismo , Renina/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transducción de Señal/fisiología , Fosfolipasas de Tipo C/metabolismo , Animales , Calcimiméticos/farmacología , Calcio/metabolismo , Células Cultivadas , Cinacalcet , AMP Cíclico/metabolismo , Inhibidores Enzimáticos/farmacología , Estrenos/farmacología , Aparato Yuxtaglomerular/citología , Aparato Yuxtaglomerular/efectos de los fármacos , Ratones , Modelos Animales , Naftalenos/farmacología , Toxina del Pertussis/farmacología , Pirrolidinonas/farmacología , Receptores Sensibles al Calcio/agonistas , Receptores Sensibles al Calcio/efectos de los fármacos , Fosfolipasas de Tipo C/antagonistas & inhibidores
17.
J Biol Chem ; 286(32): 28608-18, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21708949

RESUMEN

Renin is essential for blood pressure control. Renin is stored in granules in juxtaglomerular (JG) cells, located in the pole of the renal afferent arterioles. The second messenger cAMP stimulates renin release. However, it is unclear whether fusion and exocytosis of renin-containing granules is involved. In addition, the role of the fusion proteins, SNAREs (soluble N-ethylmaleimide-sensitive factor attachment proteins), in renin release from JG cells has not been studied. The vesicle SNARE proteins VAMP2 (vesicle associated membrane protein 2) and VAMP3 mediate cAMP-stimulated exocytosis in other endocrine cells. Thus, we hypothesized that VAMP2 and/or -3 mediate cAMP-stimulated renin release from JG cells. By fluorescence-activated cell sorting, we isolated JG cells expressing green fluorescent protein and compared the relative abundance of VAMP2/3 in JG cells versus total mouse kidney mRNA by quantitative PCR. We found that VAMP2 and VAMP3 mRNA are expressed and enriched in JG cells. Confocal imaging of primary cultures of JG cells showed that VAMP2 (but not VAMP3) co-localized with renin-containing granules. Cleavage of VAMP2 and VAMP3 with tetanus toxin blocked cAMP-stimulated renin release from JG cells by ~50% and impaired cAMP-stimulated exocytosis by ~50%, as monitored with FM1-43. Then we specifically knocked down VAMP2 or VAMP3 by adenoviral-mediated delivery of short hairpin silencing RNA. We found that silencing VAMP2 blocked cAMP-induced renin release by ~50%. In contrast, silencing VAMP3 had no effect on basal or cAMP-stimulated renin release. We conclude that VAMP2 and VAMP3 are expressed in JG cells, but only VAMP2 is targeted to renin-containing granules and mediates the stimulatory effect of cAMP on renin exocytosis.


Asunto(s)
AMP Cíclico/metabolismo , Exocitosis/fisiología , Aparato Yuxtaglomerular/metabolismo , Renina/metabolismo , Vesículas Secretoras/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/metabolismo , Animales , Células Cultivadas , Citometría de Flujo , Aparato Yuxtaglomerular/citología , Ratones , Microscopía Confocal , ARN Mensajero/metabolismo , Proteína 3 de Membrana Asociada a Vesículas/metabolismo
18.
Am J Physiol Renal Physiol ; 303(8): F1157-65, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22896038

RESUMEN

Parathyroid hormone (PTH) is positively coupled to the generation of cAMP via its actions on the PTH1R and PTH2R receptors. Renin secretion from juxtaglomerular (JG) cells is stimulated by elevated intracellular cAMP, and every stimulus that increases renin secretion is thought to do so via increasing cAMP. Thus we hypothesized that PTH increases renin release from primary cultures of mouse JG cells by elevating intracellular cAMP via the PTH1R receptor. We found PTH1R, but not PTH2R, mRNA expressed in JG cells. While PTH increased JG cell cAMP content from (log(10) means ± SE) 3.27 ± 0.06 to 3.92 ± 0.12 fmol/mg protein (P < 0.001), it did not affect renin release. The PTH1R-specific agonist, parathyroid hormone-related protein (PTHrP), also increased JG cell cAMP from 3.13 ± 0.09 to 3.93 ± 0.09 fmol/mg protein (P < 0.001), again without effect on renin release. PTH2R receptor agonists had no effect on cAMP or renin release. PTHrP increased cAMP in the presence of both low and high extracellular calcium from 3.31 ± 0.17 to 3.83 ± 0.20 fmol/mg protein (P < 0.01) and from 3.29 ± 0.18 to 3.63 ± 0.22 fmol/mg protein (P < 0.05), respectively, with no effect on renin release. PTHrP increased JG cell cAMP in the presence of adenylyl cyclase-V inhibition from 2.85 ± 0.17 to 3.44 ± 0.14 fmol/mg protein (P < 0.001) without affecting renin release. As a positive control, forskolin increased JG cell cAMP from 3.39 ± 0.13 to 4.48 ± 0.07 fmol/mg protein (P < 0.01) and renin release from 2.96 ± 0.10 to 3.29 ± 0.08 ng ANG I·mg prot(-1)·h(-1) (P < 0.01). Thus PTH increases JG cell cAMP via non-calcium-sensitive adenylate cyclases without affecting renin release. These data suggest compartmentalization of cAMP signaling in JG cells.


Asunto(s)
AMP Cíclico/metabolismo , Aparato Yuxtaglomerular/efectos de los fármacos , Hormona Paratiroidea/farmacología , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Receptor de Hormona Paratiroídea Tipo 2/metabolismo , Renina/metabolismo , Animales , Células Cultivadas , Aparato Yuxtaglomerular/citología , Aparato Yuxtaglomerular/metabolismo , Ratones , Hormona Paratiroidea/metabolismo
19.
Clin Exp Nephrol ; 16(1): 17-24, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22124804

RESUMEN

Control of the renin system by physiological mechanisms such as the baroreceptor or the macula densa (MD) is characterized by asymmetry in that the capacity for renin secretion and expression to increase is much larger than the magnitude of the inhibitory response. The large stimulatory reserve of the renin-angiotensin system may be one of the causes for the remarkable salt-conserving power of the mammalian kidney. Physiological stimulation of renin secretion and expression relies on the activation of regulatory pathways that converge on the cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) pathway. Mice with selective Gs-alpha (Gsα) deficiency in juxtaglomerular granular cells show a marked reduction of basal renin secretion, and an almost complete unresponsiveness of renin release to furosemide, hydralazine, or isoproterenol. Cyclooxygenase-2 generating prostaglandin E(2) (PGE(2)) and prostacyclin (PGI(2)) in MD and thick ascending limb cells is one of the main effector systems utilizing Gsα-coupled receptors to stimulate the renin-angiotensin system. In addition, ß-adrenergic receptors are critical for the expression of high basal levels of renin and for its release response to lowering blood pressure or MD sodium chloride concentration. Nitric oxide generated by nitric oxide synthases in the MD and in endothelial cells enhances cAMP-dependent signaling by stabilizing cAMP through cyclic guanosine monophosphate-dependent inhibition of phosphodiesterase 3. The stimulation of renin secretion by drugs that inhibit angiotensin II formation or action results from the convergent activation of cAMP probably through indirect augmentation of the activity of PGE(2) and PGI(2) receptors, ß-adrenergic receptors, and nitric oxide.


Asunto(s)
AMP Cíclico/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gs/fisiología , Renina/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Ciclooxigenasa 2/fisiología , Diuréticos/farmacología , Furosemida/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gs/deficiencia , Aparato Yuxtaglomerular/citología , Aparato Yuxtaglomerular/metabolismo , Túbulos Renales Distales/metabolismo , Ratones , Óxido Nítrico Sintasa/metabolismo , Prostaglandinas/fisiología , Receptores Adrenérgicos beta/metabolismo , Sistema Renina-Angiotensina/fisiología , Transducción de Señal/efectos de los fármacos
20.
Physiol Genomics ; 43(17): 1021-8, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21750232

RESUMEN

Renin-expressing cells are crucial in the control of blood pressure and fluid-electrolyte homeostasis. Notch receptors convey cell-cell signals that may regulate the renin cell phenotype. Because the common downstream effector for all Notch receptors is the transcription factor RBP-J, we used a conditional knockout approach to delete RBP-J in cells of the renin lineage. The resultant RBP-J conditional knockout (cKO) mice displayed a severe reduction in the number of renin-positive juxtaglomerular apparatuses (JGA) and a reduction in the total number of renin positive cells per JGA and along the afferent arterioles. This reduction in renin protein was accompanied by a decrease in renin mRNA expression, decreased circulating renin, and low blood pressure. To investigate whether deletion of RBP-J altered the ability of mice to increase the number of renin cells normally elicited by a physiological threat, we treated RBP-J cKO mice with captopril and sodium depletion for 10 days. The resultant treated RBP-J cKO mice had a 65% reduction in renin mRNA levels (compared with treated controls) and were unable to increase circulating renin. Although these mice attempted to increase the number of renin cells, the cells were unusually thin and had few granules and barely detectable amounts of immunoreactive renin. As a consequence, the cells were incapable of fully adopting the endocrine phenotype of a renin cell. We conclude that RBP-J is required to maintain basal renin expression and the ability of smooth muscle cells along the kidney vasculature to regain the renin phenotype, a fundamental mechanism to preserve homeostasis.


Asunto(s)
Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Renina/metabolismo , Animales , Células Cultivadas , Genotipo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Inmunohistoquímica , Aparato Yuxtaglomerular/citología , Aparato Yuxtaglomerular/metabolismo , Ratones , Ratones Noqueados , ARN Mensajero/genética , Renina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA