Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 320(6): F1106-F1122, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33938239

RESUMEN

Ksp-cadherin (cadherin-16) is an atypical member of the cadherin superfamily of cell adhesion molecules that is ubiquitously expressed on the basolateral membrane of epithelial cells lining the nephron and the collecting system of the mammalian kidney. The principal aim of the present study was to determine if Ksp-cadherin played a critical role in the development and maintenance of the adult mammalian kidney by generating and evaluating a mouse line deficient in Ksp-cadherin. Ksp-null mutant animals were viable and fertile, and kidneys from both neonates and adults showed no evidence of structural abnormalities. Immunolocalization and Western blot analyses of Na+-K+-ATPase and E-cadherin indicated that Ksp-cadherin is not essential for either the genesis or maintenance of the polarized tubular epithelial phenotype. Moreover, E-cadherin expression was not altered to compensate for Ksp-cadherin loss. Plasma electrolytes, total CO2, blood urea nitrogen, and creatinine levels were also unaffected by Ksp-cadherin deficiency. However, a subtle but significant developmental delay in the ability to maximally concentrate urine was detected in Ksp-null mice. Expression analysis of the principal proteins involved in the generation of the corticomedullary osmotic gradient and the resultant movement of water identified misexpression of aquaporin-2 in the inner medullary collecting duct as the possible cause for the inability of young adult Ksp-cadherin-deficient animals to maximally concentrate their urine. In conclusion, Ksp-cadherin is not required for normal kidney development, but its absence leads to a developmental delay in maximal urinary concentrating ability.NEW & NOTEWORTHY Ksp-cadherin (cadherin-16) is an atypical member of the cadherin superfamily of cell adhesion molecules that is ubiquitously expressed on the basolateral membrane of epithelial cells lining the nephron and the collecting system. Using knockout mice, we found that Ksp-cadherin is in fact not required for kidney development despite its high and specific expression along the nephron. However, its absence leads to a developmental delay in maximal urinary concentrating ability.


Asunto(s)
Cadherinas/metabolismo , Capacidad de Concentración Renal/fisiología , Riñón/crecimiento & desarrollo , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Cadherinas/genética , Regulación del Desarrollo de la Expresión Génica , Riñón/fisiología , Capacidad de Concentración Renal/genética , Masculino , Ratones , Ratones Noqueados , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
2.
FASEB J ; 33(2): 2156-2170, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30252533

RESUMEN

cAMP is a universal second messenger regulating a plethora of processes in the kidney. Two downstream effectors of cAMP are PKA and exchange protein directly activated by cAMP (Epac), which, unlike PKA, is often linked to elevation of [Ca2+]i. While both Epac isoforms (Epac1 and Epac2) are expressed along the nephron, their relevance in the kidney remains obscure. We combined ratiometric calcium imaging with quantitative immunoblotting, immunofluorescent confocal microscopy, and balance studies in mice lacking Epac1 or Epac2 to determine the role of Epac in renal water-solute handling. Epac1-/- and Epac2-/- mice developed polyuria despite elevated arginine vasopressin levels. We did not detect major deficiencies in arginine vasopressin [Ca2+]i signaling in split-opened collecting ducts or decreases in aquaporin water channel type 2 levels. Instead, sodium-hydrogen exchanger type 3 levels in the proximal tubule were dramatically reduced in Epac1-/- and Epac2-/- mice. Water deprivation revealed persisting polyuria, impaired urinary concentration ability, and augmented urinary excretion of Na+ and urea in both mutant mice. In summary, we report a nonredundant contribution of Epac isoforms to renal function. Deletion of Epac1 and Epac2 decreases sodium-hydrogen exchanger type 3 expression in the proximal tubule, leading to polyuria and osmotic diuresis.-Cherezova, A., Tomilin, V., Buncha, V., Zaika, O., Ortiz, P. A., Mei, F., Cheng, X., Mamenko, M., Pochynyuk, O. Urinary concentrating defect in mice lacking Epac1 or Epac2.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Capacidad de Concentración Renal/genética , Animales , Acuaporina 2/metabolismo , Arginina Vasopresina/metabolismo , Señalización del Calcio , Diuresis , Eliminación de Gen , Riñón/metabolismo , Riñón/fisiología , Ratones , Ratones Noqueados , Ósmosis , Poliuria/genética , Intercambiador 3 de Sodio-Hidrógeno/metabolismo
3.
Cell Physiol Biochem ; 39(1): 172-82, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27336410

RESUMEN

BACKGROUND/AIM: STIM1 is as an essential component in store operated Ca2+ entry. However give the paucity of information on the role of STIM1 in kidney, the aim was to study the function of STIM1 in the medulla of the kidney. METHODS: we crossed a Ksp-cre mouse with another mouse containing two loxP sites flanking Exon 6 of STIM1. The Ksp-cre mouse is based upon the Ksp-cadherin gene promoter which expresses cre recombinase in developing nephrons, collecting ducts (SD) and thick ascending limbs (TAL) of the loop of Henle. RESULTS: The offspring of these mice are viable without gross morphological changes, however, we noticed that the STIM1 Ksp-cre knockout mice produced more urine compared to control. To examine this more carefully, we fed mice low (LP) and high protein (HP) diets respectively. When mice were fed HP diet STIM1 ko mice had significantly increased urinary volume and lower specific gravity compared to wt mice. In STIM1 ko mice fed HP diet urine creatinine and urea were significantly lower compared to wt mice fed HP diet, however the fractional excretion was the same. CONCLUSION: These data support the idea that STIM1 ko mice have impaired urinary concentrating ability when challenged with HP diet is most likely caused by impaired Ca2+-dependent signal transduction through the vasopressin receptor cascade.


Asunto(s)
Riñón/metabolismo , Cinesinas/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Agua/metabolismo , Animales , Calcio/sangre , Calcio/orina , Creatinina/sangre , Creatinina/orina , Proteínas en la Dieta/administración & dosificación , Proteínas en la Dieta/farmacología , Capacidad de Concentración Renal/genética , Cinesinas/genética , Ratones Noqueados , Ratones Transgénicos , Insuficiencia Renal/genética , Insuficiencia Renal/metabolismo , Molécula de Interacción Estromal 1/genética , Urea/sangre , Urea/orina , Micción/efectos de los fármacos , Equilibrio Hidroelectrolítico/genética
4.
Proc Natl Acad Sci U S A ; 112(27): 8397-402, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26100911

RESUMEN

The antidiuretic hormone arginine vasopressin is a systemic effector in urinary concentration. However, increasing evidence suggests that other locally produced factors may also play an important role in the regulation of water reabsorption in renal collecting ducts. Recently, prostaglandin E2 (PGE2) receptor EP4 has emerged as a potential therapeutic target for the treatment of nephrogenic diabetes insipidus, but the underlying mechanism is unknown. To evaluate the role of EP4 in regulating water homeostasis, mice with renal tubule-specific knockout of EP4 (Ksp-EP4(-/-)) and collecting duct-specific knockout of EP4 (AQP2-EP4(-/-)) were generated using the Cre-loxP recombination system. Urine concentrating defect was observed in both Ksp-EP4(-/-) and AQP2-EP4(-/-) mice. Decreased aquaporin 2 (AQP2) abundance and apical membrane targeting in renal collecting ducts were evident in Ksp-EP4(-/-) mice. In vitro studies demonstrated that AQP2 mRNA and protein levels were significantly up-regulated in mouse primary inner medullary collecting duct (IMCD) cells after pharmacological activation or adenovirus-mediated overexpression of EP4 in a cAMP/cAMP-response element binding protein-dependent manner. In addition, EP4 activation or overexpression also increased AQP2 membrane accumulation in a mouse IMCD cell line (IMCD3) stably transfected with the AQP2 gene, mainly through the cAMP/protein kinase A and extracellular signal-regulated kinase pathways. In summary, the EP4 receptor in renal collecting ducts plays an important role in regulating urinary concentration under physiological conditions. The ability of EP4 to promote AQP2 membrane targeting and increase AQP2 abundance makes it a potential therapeutic target for the treatment of clinical disorders including acquired and congenital diabetes insipidus.


Asunto(s)
Acuaporina 2/genética , Capacidad de Concentración Renal/genética , Túbulos Renales Colectores/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/genética , Animales , Acuaporina 2/metabolismo , Western Blotting , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Dinoprostona/análogos & derivados , Dinoprostona/biosíntesis , Dinoprostona/farmacología , Túbulos Renales Colectores/citología , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Cultivo Primario de Células , Pirrolidinonas/farmacología , Interferencia de ARN , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Agua/metabolismo
5.
Subcell Biochem ; 73: 137-52, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25298343

RESUMEN

Urea transporter gene knockout mice have been created for the study of the urine-concentrating mechanism. The major findings in studies of the renal phenotype of these mice are as follows: (1) Urea accumulation in the inner medullary interstitium is dependent on intrarenal urea recycling mediated by urea transporters; (2) urea transporters are essential for preventing urea-induced osmotic diuresis and thus for water conservation; (3) NaCl concentration in the inner medullary interstitium is not significantly affected by the absence of IMCD, descending limb of Henle and descending vasa recta urea transporters. Studies in urea transporter knockout mouse models have highlighted the essential role of urea for producing maximally concentrated urine.


Asunto(s)
Médula Renal/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Urea/metabolismo , Animales , Transporte Biológico/genética , Capacidad de Concentración Renal/genética , Ratones Noqueados , Modelos Genéticos , Fenotipo , Transportadores de Urea
6.
Kidney Int ; 85(5): 1137-50, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24429399

RESUMEN

We have bred a Pkd1 floxed allele with a nestin-Cre expressing line to generate cystic mice with preserved glomerular filtration rate to address the pathogenesis of complex autosomal dominant polycystic kidney disease (ADPKD) phenotypes. Hypertension affects about 60% of these patients before loss of renal function, leading to significant morbimortality. Cystic mice were hypertensive at 5 and 13 weeks of age, a phenotype not seen in noncystic controls and Pkd1-haploinsufficient animals that do not develop renal cysts. Fractional sodium excretion was reduced in cystic mice at these ages. Angiotensinogen gene expression was higher in cystic than noncystic kidneys at 18 weeks, while ACE and the AT1 receptor were expressed in renal cyst epithelia. Cystic animals displayed increased renal cAMP, cell proliferation, and apoptosis. At 24 weeks, mean arterial pressure and fractional sodium excretion did not significantly differ between the cystic and noncystic groups, whereas cardiac mass increased in cystic mice. Renal concentrating deficit is also an early finding in ADPKD. Maximum urine osmolality and urine nitrite excretion were reduced in 10-13- and 24-week-old cystic mice, deficits not found in haploinsufficient and noncystic controls. A trend of higher plasma vasopressin was observed in cystic mice. Thus, cyst growth most probably plays a central role in early-stage ADPKD-associated hypertension, with activation of the intrarenal renin-angiotensin system as a key mechanism. Cyst expansion is also likely essential for the development of the concentrating deficit in this disease. Our findings are consistent with areas of reduced perfusion in the kidneys of patients with ADPKD.


Asunto(s)
Presión Arterial , Proliferación Celular , Hipertensión/etiología , Capacidad de Concentración Renal , Riñón/metabolismo , Riñón Poliquístico Autosómico Dominante/complicaciones , Canales Catiónicos TRPP/deficiencia , Animales , Apoptosis , Presión Arterial/genética , Biomarcadores/sangre , Biomarcadores/orina , Proliferación Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación de la Expresión Génica , Genotipo , Tasa de Filtración Glomerular , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Riñón/patología , Riñón/fisiopatología , Capacidad de Concentración Renal/genética , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Riñón Poliquístico Autosómico Dominante/fisiopatología , Sistema Renina-Angiotensina , Canales Catiónicos TRPP/genética , Factores de Tiempo
7.
Biochim Biophys Acta ; 1839(2): 97-106, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24389345

RESUMEN

Sfmbt2-hosted miR-466a-3p and its close relatives are often among the most significantly up-regulated or down-regulated miRNAs in responses to numerous deleterious environmental stimuli. The exact roles of these miRNAs in cellular stress responses, however, are not clear. Here we showed that many Sfmbt2-hosted miRNAs were highly hypertonic stress responsive in vitro and in vivo. In renal medulla, water deprivation induced alterations in the expression of miR-466(a/b/c/e/p)-3p in a pattern similar to that of miR-200b-3p, a known regulator of osmoresponsive transcription factor Nfat5. Remarkably, exposure of mIMCD3 cells to an arginine vasopressin analog time-dependently down-regulated the expression of miR-466(a/b/c/e/p)-3p and miR-200b-3p, which provides a novel regulatory mechanism for these osmoresponsive miRNAs. In cultured mIMCD3 cells we further demonstrated that miR-466a-3p and miR-466g were capable of targeting Nfat5 by interacting with its 3'UTR. In transgenic mice overexpressing miR-466a-3p, significant down-regulation of Nfat5 and many other osmoregulation-related genes was observed in both the renal cortex and medulla. Moreover, sustained transgenic over-expression of miR-466a-3p was found to be associated with polydipsia, polyuria and disturbed ion homeostasis and kidney morphology. Since the mature sequence of miR-466a-3p is completely equivalent to that of miR-466e-3p and that the seed sequence of miR-466a-3p is completely equivalent to that of miR-297(a/b/c)-3p, miR-466d-3p, miR-467g and miR-669d-3p, and that miR-466a-3p differs from miR-466(b/c/p)-3p only in a 5' nucleotide, we propose that miR-466a-3p and many of its close relatives are important epigenetic regulators of renal Nfat5 signaling, osmoregulation and urine concentration in mice.


Asunto(s)
Intrones/genética , Capacidad de Concentración Renal/genética , MicroARNs/genética , Factores de Transcripción NFATC/genética , Osmorregulación/genética , Factores de Transcripción/genética , Regiones no Traducidas 3'/genética , Animales , Arginina Vasopresina/análogos & derivados , Arginina Vasopresina/farmacología , Secuencia de Bases , Western Blotting , Línea Celular , Creatina/sangre , Creatina/orina , Epigénesis Genética , Expresión Génica/efectos de los fármacos , Corteza Renal/metabolismo , Médula Renal/metabolismo , Ratones , Ratones Transgénicos , Factores de Transcripción NFATC/metabolismo , Potasio/sangre , Potasio/orina , Proteínas Represoras , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , Transducción de Señal/genética , Sodio/sangre , Sodio/orina , Urea/sangre , Urea/orina
8.
Proc Natl Acad Sci U S A ; 111(6): 2277-82, 2014 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-24464484

RESUMEN

The farnesoid X receptor (FXR) is a ligand-activated transcription factor belonging to the nuclear receptor superfamily. FXR is mainly expressed in liver and small intestine, where it plays an important role in bile acid, lipid, and glucose metabolism. The kidney also has a high FXR expression level, with its physiological function unknown. Here we demonstrate that FXR is ubiquitously distributed in renal tubules. FXR agonist treatment significantly lowered urine volume and increased urine osmolality, whereas FXR knockout mice exhibited an impaired urine concentrating ability, which led to a polyuria phenotype. We further found that treatment of C57BL/6 mice with chenodeoxycholic acid, an FXR endogenous ligand, significantly up-regulated renal aquaporin 2 (AQP2) expression, whereas FXR gene deficiency markedly reduced AQP2 expression levels in the kidney. In vitro studies showed that the AQP2 gene promoter contained a putative FXR response element site, which can be bound and activated by FXR, resulting in a significant increase of AQP2 transcription in cultured primary inner medullary collecting duct cells. In conclusion, the present study demonstrates that FXR plays a critical role in the regulation of urine volume, and its activation increases urinary concentrating capacity mainly via up-regulating its target gene AQP2 expression in the collecting ducts.


Asunto(s)
Capacidad de Concentración Renal/genética , Receptores Citoplasmáticos y Nucleares/genética , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Secuencia de Bases , Cartilla de ADN , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Receptores Citoplasmáticos y Nucleares/metabolismo
9.
Pediatr Nephrol ; 29(8): 1297-303, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23736674

RESUMEN

Nephrogenic diabetes insipidus (NDI) provides an excellent model for the benefits and insights that can be gained from studying rare diseases. The discovery of underlying genes identified key molecules involved in urinary concentration, including the type 2 vasopressin receptor AVPR2 and the water channel AQP2, which constitute obvious pharmacologic targets. Subsequently developed drugs targeting AVPR2 not only provide potential benefit to some patients with NDI, but are now used for much more common clinical applications as diverse as nocturnal enuresis and heart failure. Yet, the story is still evolving: clinical observations and animal experiments continue to discover new ways to affect urinary concentration. These novel pathways can potentially be exploited for therapeutic gain. Here we review the (patho)physiology of water homoeostasis, the current status of clinical management, and potential new treatments.


Asunto(s)
Diabetes Insípida Nefrogénica/orina , Capacidad de Concentración Renal/genética , Agua Corporal/metabolismo , Niño , Diabetes Insípida Nefrogénica/genética , Humanos
10.
Am J Physiol Renal Physiol ; 303(4): F494-502, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22622456

RESUMEN

In aldosterone target tissues, 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2) is coexpressed with mineralocorticoid receptors (MR) and protects the receptor from activation by glucocorticoids. Null mutations in the encoding gene, HSD11B2, cause apparent mineralocorticoid excess, in which hypertension is thought to reflect volume expansion secondary to sodium retention. Hsd11b2(-/-) mice are indeed hypertensive, but impaired natriuretic capacity is associated with significant volume contraction, suggestive of a urine concentrating defect. Water turnover and the urine concentrating response to a 24-h water deprivation challenge were therefore assessed in Hsd11b2(-/-) mice and controls. Hsd11b2(-/-) mice have a severe and progressive polyuric/polydipsic phenotype. In younger mice (∼2 mo of age), polyuria was associated with decreased abundance of aqp2 and aqp3 mRNA. The expression of other genes involved in water transport (aqp4, slc14a2, and slc12a2) was not changed. The kidney was structurally normal, and the concentrating response to water deprivation was intact. In older Hsd11b2(-/-) mice (>6 mo), polyuria was associated with a severe atrophy of the renal medulla and downregulation of aqp2, aqp3, aqp4, slc14a2, and slc12a2. The concentrating response to water deprivation was impaired, and the natriuretic effect of the loop diuretic bumetanide was lost. In older Hsd11b2(-/-) mice, the V2 receptor agonist desmopressin did not restore full urine concentrating capacity. We find that Hsd11b2(-/-) mice develop nephrogenic diabetes insipidus. Gross changes to renal structure are observed, but these were probably secondary to sustained polyuria, rather than of developmental origin.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Diabetes Insípida/enzimología , Capacidad de Concentración Renal/fisiología , Riñón/fisiología , Envejecimiento , Animales , Diabetes Insípida/genética , Regulación de la Expresión Génica , Homeostasis , Riñón/anatomía & histología , Capacidad de Concentración Renal/genética , Ratones , Ratones Noqueados , Concentración Osmolar , Simportadores de Cloruro de Sodio-Potasio/genética , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Agua/metabolismo , Pérdida de Peso
12.
Am J Physiol Renal Physiol ; 303(1): F37-44, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22492943

RESUMEN

Regulation of water and urea transport in the inner medullary collecting duct is essential for urine concentration. Aquaporin (AQP)2 water channels and urea transporter (UT)-A1 are inserted into the apical membrane upon phosphorylation of the channels to allow the transcellular movement of water and urea. Since ANG II activates PKC in many cell types, we tested the hypothesis that ANG II-induced regulation of water and urea transport is mediated by PKC. Osmotic minipumps delivered ANG II to wild-type (WT) or PKC-α(-/-) mice for 7 days. Inner medullas were harvested, and protein abundance was determined by immunoblot. ANG II increased systolic blood pressure to a similar degree in WT and PKC-α(-/-) mice. ANG II had no effect on the urine output of WT mice but increased that of PKC-α(-/-) mice. In accordance with observed differences in urine output, AQP2 abundance was unchanged in ANG II-treated WT animals but was decreased in PKC-α(-/-) mice. No change in membrane accumulation was seen. Phosphorylation of the cAMP-induced transcription factor CREB was decreased in PKC-α(-/-) mice in response to ANG II with no change in overall CREB abundance. ANG II did not alter the abundance of UT-A1 protein in WT or PKC-α(-/-) mice. Phosphorylation and overall abundance of tonicity-responsive enhancer-binding protein, a transcription factor that regulates UT-A1, were also unaltered by ANG II in either group. We conclude that PKC-α protects against ANG II-induced decreases in urine concentrating ability by maintaining AQP2 levels through CREB phosphorylation.


Asunto(s)
Angiotensina II/toxicidad , Acuaporina 2/metabolismo , Hipertensión/metabolismo , Capacidad de Concentración Renal/genética , Riñón/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteína Quinasa C-alfa/genética , Animales , Acuaporina 2/genética , Presión Sanguínea/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hipertensión/inducido químicamente , Hipertensión/genética , Riñón/efectos de los fármacos , Capacidad de Concentración Renal/efectos de los fármacos , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Fosforilación , Proteína Quinasa C-alfa/metabolismo , Transportadores de Urea
13.
Endocrine ; 42(1): 63-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22374125

RESUMEN

In this mini-review, current evidence for how the vasopressin/V2-type receptor/aquaporin axis developed co-evolutionary as a crucial part of the urine-concentrating mechanism will be presented. The present-day human kidney, allowing the concentration of urine up to a maximal osmolality around 1200 mosmol kg(-1)-or urine to plasma osmolality ratio around 4-with essentially no sodium secreted is the result of up to 3 billion years evolution. Moving from aquatic to terrestrial habitats required profound changes in kidney morphology, most notable the loops of Henle modifying the kidneys from basically a water excretory system to a water conserving system. Vasopressin-like molecules has during the evolution played a significant role in body fluid homeostasis, more specifically, the osmolality of body liquids by controlling the elimination/reabsorption of fluid trough stimulating V2-type receptors to mobilize aquaporin water channels in the renal collector tubules. Recent evidence supports that all components of the vasopressin/V2-type receptor/aquaporin axis can be traced back to early precursors in evolutionary history. The potential clinical and pharmacological implications of a better phylogenetic understanding of these biological systems so essential for body fluid homeostasis relates to any pathological aspects of the urine-concentrating mechanism, in particular deficiencies of any part of the vasopressin-V2R-AQP2 axis causing central or nephrogenic diabetes insipidus-and for broader patient populations also in preventing and treating disturbances in human circadian regulation of urine volume and osmolality that may lead to enuresis and nocturia.


Asunto(s)
Acuaporinas/genética , Evolución Molecular , Capacidad de Concentración Renal/genética , Receptores de Vasopresinas/genética , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Acuaporina 2/fisiología , Acuaporinas/metabolismo , Acuaporinas/fisiología , Humanos , Modelos Biológicos , Receptores de Vasopresinas/metabolismo , Receptores de Vasopresinas/fisiología , Transducción de Señal/genética , Orina/química , Orina/fisiología
14.
Am J Physiol Renal Physiol ; 302(8): F1005-12, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22237797

RESUMEN

PGE(2) plays an important role in the regulation of fluid metabolism chiefly via antagonizing vasopressin-induced osmotic permeability in the distal nephron, but its enzymatic sources remain uncertain. The present study was undertaken to investigate the potential role of microsomal PGE synthase (mPGES)-1 in the regulation of urine concentrating ability after water deprivation (WD). Following 24-h WD, wild-type (WT) mice exhibited a significant reduction in urine volume, accompanied by a significant elevation in urine osmolality compared with control groups. In contrast, in response to WD, mPGES-1 knockout (KO) mice had much less urine volume and higher urine osmolality. Analysis of plasma volume by measurement of hematocrit and by using a nanoparticle-based method consistently demonstrated that dehydrated WT mice were volume depleted, which was significantly improved in the KO mice. WD induced a twofold increase in urinary PGE(2) output in WT mice, which was completely blocked by mPGES-1 deletion. At baseline, the KO mice had a 20% increase in V(2) receptor mRNA expression in the renal medulla but not the cortex compared with WT controls; the expression was unaffected by WD irrespective of the genotype. In response to WD, renal medullary aquaporin-2 (AQP2) mRNA exhibited a 60% increase in WT mice, and this increase was greater in the KO mice. Immunoblotting demonstrated increased renal medullary AQP2 protein abundance in both genotypes following WD, with a greater increase in the KO mice. Similar results were obtained by using immunohistochemistry. Paradoxically, plasma AVP response to WD seen in WT mice was absent in the KO mice. Taken together, these results suggest that mPGES-1-derived PGE(2) reduces urine concentrating ability through suppression of renal medullary expression of V(2) receptors and AQP2 but may enhance it by mediating the central AVP response.


Asunto(s)
Oxidorreductasas Intramoleculares/fisiología , Capacidad de Concentración Renal/fisiología , Privación de Agua/fisiología , Animales , Acuaporina 2/biosíntesis , Arginina Vasopresina/sangre , Dinoprostona/orina , Eliminación de Gen , Oxidorreductasas Intramoleculares/genética , Riñón/citología , Riñón/fisiología , Capacidad de Concentración Renal/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Concentración Osmolar , Prostaglandina-E Sintasas , Receptores de Vasopresinas/fisiología
16.
Am J Physiol Renal Physiol ; 301(1): F44-54, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21429969

RESUMEN

N-methyl-D-aspartate receptors (NMDARs) are Ca(2+)-permeable, ligand-gated, nonselective cation channels that function as neuronal synaptic receptors but which are also expressed in multiple peripheral tissues. Here, we show for the first time that NMDAR subunits NR3a and NR3b are highly expressed in the neonatal kidney and that there is continued expression of NR3a in the renal medulla and papilla of the adult mouse. NR3a was also expressed in mIMCD-3 cells, where it was found that hypoxia and hypertonicity upregulated NR3a expression. Using short-hairpin (sh) RNA-based knockdown, a stable inner medullary collecting duct (IMCD) cell line was established that had ∼80% decrease in NR3a. Knockdown cells exhibited an increased basal intracellular calcium concentration, reduced cell proliferation, and increased cell death. In addition, NR3a knockdown cells exhibited reduced water transport in response to the addition of vasopressin, suggesting an alteration in aquaporin-2 (AQP2) expression/function. Consistent with this notion, we demonstrate decreased surface expression of glycosylated AQP2 in IMCD cells transfected with NR3a shRNA. To determine whether this also occurred in vivo, we compared AQP2 levels in wild-type vs. in NR3a(-/-) mice. Total AQP2 protein levels in the outer and inner medulla were significantly reduced in knockout mice compared with control mice. Finally, NR3a(-/-) mice showed a significant delay in their ability to increase urine osmolality during water restriction. Thus NR3a may play a renoprotective role in collecting duct cells. Therefore, under conditions that are associated with high vasopressin levels, NR3a, by maintaining low intracellular calcium levels, protects the function of the principal cells to reabsorb water and thereby increase medullary osmolality.


Asunto(s)
Túbulos Renales Colectores/metabolismo , Receptores de N-Metil-D-Aspartato/biosíntesis , Receptores de N-Metil-D-Aspartato/fisiología , Animales , Acuaporina 2/metabolismo , Western Blotting , Calcio/metabolismo , Señalización del Calcio/fisiología , Células Cultivadas , Humanos , Soluciones Hipertónicas , Inmunohistoquímica , Capacidad de Concentración Renal/genética , Capacidad de Concentración Renal/fisiología , Médula Renal/citología , Médula Renal/metabolismo , Túbulos Renales Colectores/citología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/aislamiento & purificación , Ratones , Ratones Noqueados , Concentración Osmolar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina/farmacología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Regulación hacia Arriba/fisiología
17.
Physiol Genomics ; 43(1): 32-42, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-20923861

RESUMEN

Arginine vasopressin (AVP) and angiotensin II (ANG II) are distinct peptide hormones involved in multiple organs modulating renal, cardiovascular, and brain functions. They achieve these functions via specific G protein-coupled receptors, respectively. The AVR/NAVR locus encodes two overlapping V2-type vasopressin isoreceptors: angiotensin-vasopressin receptor (AVR) responding to ANG II and AVP equivalently, and nonangiotensin vasopressin receptor (NAVR), which binds vasopressin exclusively. AVR and NAVR are expressed from a single gene by alternative promoter usage that is synergistically upregulated by testosterone and estrogen. This study tested the hypothesis that AVR/NAVR modulates urinary concentrating ability, blood pressure, and cognitive performance in vivo in a sex-specific manner. We developed a C57BL/6 inbred AVR/NAVR(-/-) knockout mouse that showed lower blood pressure in both male and female subjects and a urinary-concentrating defect restricted to male mice. We also detected sex-specific effects on cognitive and anxiety-like behaviors. AVR/NAVR(-/-) male mice exhibited impaired visuospatial and associative learning, while female mice showed improved performance in both type of cognition. AVR/NAVR deficiency produced an anxiolytic-like effect in female mice, while males were unaffected. Analysis of AVR- and NAVR-mediated phosphorylation/dephosphorylation of signaling proteins revealed activation/deactivation of known modulators of cognitive function. Our studies identify AVR/NAVR as key receptors involved in blood pressure regulation and sex-specific modulation of renal water homeostasis, cognitive function, and anxiety-like behavior. As such, the AVR/NAVR receptor system provides a molecular mechanism for sexually diergic traits and a putative common pathway for the emerging association of hypertension and cognitive decline and dementia.


Asunto(s)
Ansiedad/fisiopatología , Presión Sanguínea/fisiología , Cognición/fisiología , Capacidad de Concentración Renal/fisiología , Receptores de Angiotensina/deficiencia , Receptores Acoplados a Proteínas G/deficiencia , Receptores de Vasopresinas/deficiencia , Animales , Ansiedad/genética , Presión Sanguínea/genética , Femenino , Capacidad de Concentración Renal/genética , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Fosforilación , Receptores de Angiotensina/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Vasopresinas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Clin Nephrol ; 74(6): 411-22, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21084044

RESUMEN

BACKGROUND: A family was identified with autosomal dominant inheritance of anemia, polyuria, hyperuricemia, and chronic kidney disease. Mutational analysis revealed a novel heterozygous mutation c.58T > C resulting in the amino acid substitution of cysteine for arginine in the preprorenin signal sequence (p.cys20Arg) occurring in all affected members. METHODS: Effects of the identified mutation were characterized using in vitro and in vivo studies. Affected individuals were clinically characterized before and after administration of fludrocortisone. RESULTS: The mutation affects endoplasmic reticulum co-translational translocation and posttranslational processing, resulting in massive accumulation of non-glycosylated preprorenin in the cytoplasm. This affects expression of intra-renal RAS components and leads to ultrastructural damage of the kidney. Affected individuals suffered from anemia, hyperuricemia, decreased urinary concentrating ability, and progressive chronic kidney disease. Treatment with fludrocortisone in an affected 10-year-old child resulted in an increase in blood pressure and estimated glomerular filtration rate. CONCLUSIONS: A novel REN gene mutation resulted in an alteration in the amino acid sequence of the renin signal sequence and caused childhood anemia, polyuria, and kidney disease. Treatment with fludrocortisone improved renal function in an affected child. Nephrologists should consider REN mutational analysis in families with autosomal dominant inheritance of chronic kidney disease, especially if they suffer from anemia, hyperuricemia, and polyuria in childhood.


Asunto(s)
Fludrocortisona/uso terapéutico , Genes Dominantes , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/genética , Mutación , Señales de Clasificación de Proteína/genética , Renina/genética , Adulto , Secuencia de Aminoácidos , Anemia/genética , Anemia/metabolismo , Secuencia de Bases , Biopsia , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Línea Celular , Niño , Enfermedad Crónica , Quimosina , Citoplasma/metabolismo , Análisis Mutacional de ADN , Retículo Endoplásmico/metabolismo , Precursores Enzimáticos , Femenino , Predisposición Genética a la Enfermedad , Tasa de Filtración Glomerular/efectos de los fármacos , Tasa de Filtración Glomerular/genética , Glicosilación , Heterocigoto , Humanos , Hiperuricemia/genética , Hiperuricemia/metabolismo , Hipoaldosteronismo/genética , Hipoaldosteronismo/metabolismo , Capacidad de Concentración Renal/genética , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Masculino , Datos de Secuencia Molecular , Linaje , Fenotipo , Poliuria/genética , Poliuria/metabolismo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Renina/metabolismo , Transfección , Resultado del Tratamiento
19.
Am J Physiol Renal Physiol ; 299(4): F712-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20668095

RESUMEN

Activation of V2 receptors (V2R) during antidiuresis increases the permeability of the inner medullary collecting duct to urea and water. Extracellular osmolality is elevated as the concentrating capacity of the kidney increases. Osmolality is known to contribute to the regulation of collecting duct water (aquaporin-2; AQP2) and urea transporter (UT-A1, UT-A3) regulation. AQP1KO mice are a concentrating mechanism knockout, a defect attributed to the loss of high interstitial osmolality. A V2R-specific agonist, deamino-8-D-arginine vasopressin (dDAVP), was infused into wild-type and AQP1KO mice for 7 days. UT-A1 mRNA and protein abundance were significantly increased in the medullas of wild-type and AQP1KO mice following dDAVP infusion. The mRNA and protein abundance of UT-A3, the basolateral urea transporter, was significantly increased by dDAVP in both wild-type and AQP1KO mice. Semiquantitative immunoblots revealed that dDAVP infusion induced a significant increase in the medullary expression of the endoplasmic reticulum (ER) chaperone GRP78. Immunofluorescence studies demonstrated that GRP78 expression colocalized with AQP2 in principal cells of the papillary tip of the renal medulla. Using immunohistochemistry and immunogold electron microscopy, we demonstrate that vasopressin induced a marked apical targeting of GRP78 in medullary principal cells. Urea-sensitive genes, GADD153 and ATF4 (components of the ER stress pathway), were significantly increased in AQP1KO mice by dDAVP infusion. These findings strongly support an important role of vasopressin in the activation of an ER stress response in renal collecting duct cells, in addition to its role in activating an increase in UT-A1 and UT-A3 abundance.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Capacidad de Concentración Renal/genética , Médula Renal/efectos de los fármacos , Médula Renal/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Vasopresinas/farmacología , Animales , Acuaporina 1/genética , Acuaporina 1/fisiología , Membrana Celular/metabolismo , Desamino Arginina Vasopresina/farmacología , Retículo Endoplásmico/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Capacidad de Concentración Renal/fisiología , Médula Renal/fisiopatología , Ratones , Ratones Noqueados , Modelos Animales , Concentración Osmolar , ARN Mensajero , Transportadores de Urea
20.
Horm Res Paediatr ; 73(5): 349-54, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20389105

RESUMEN

BACKGROUND: AVPR2 mutations cause most cases of nephrogenic diabetes insipidus (NDI); 211 AVPR2 mutations have been described, but only 7 are described causing partial NDI. METHODS: Two unrelated Thai boys had polyuria and polydipsia in infancy but had normal electrolytes and serum osmolality at 2 years of age. Patient 1 could not concentrate his urine in response to water deprivation or 1-desamino-8-D-arginine vasopressin (DDAVP); patient 2 could concentrate to approximately 600 mosm/l. The patients' AVPR2 genes were sequenced and the identified mutations were re-created in AVPR2 cDNA expression vectors. AVPR2 activities were measured by stimulating transfected HEK293T cells with arginine vasopressin (AVP) or DDAVP, and assessing the resulting cAMP production by the activation of a luciferase reporter. RESULTS: Patient 1 carried the previously described missense mutation R181C; patient 2 carried the novel missense mutation M311V. When transiently transfected into HEK293T cells, 6.8 x 10(-12) M AVP induced the half-maximal response (EC50) of the wild-type, whereas the EC50 value for R181C was 5.9 x 10(-9) M and for M311V was 2.6 x 10(-10)M. Responses to DDAVP were qualitatively similar but required 10-fold higher concentrations. CONCLUSION: The novel AVPR2 mutation M311V retains partial activity and results in a milder form of NDI.


Asunto(s)
Diabetes Insípida Nefrogénica/genética , Receptores de Vasopresinas/genética , Arginina Vasopresina , Pueblo Asiatico/genética , Secuencia de Bases , Línea Celular , Preescolar , Desamino Arginina Vasopresina , Humanos , Lactante , Capacidad de Concentración Renal/genética , Masculino , Mutación , Mutación Missense , Receptores de Vasopresinas/fisiología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...