Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Más filtros

Intervalo de año de publicación
1.
Can J Physiol Pharmacol ; 98(9): 579-586, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32437627

RESUMEN

Previously, we showed that global knockout (KO) of the circadian clock transcription factor PER1 in male, but not female, mice fed a high-salt diet plus mineralocorticoid treatment (HS/DOCP) resulted in nondipping hypertension and decreased night/day ratio of sodium (Na) excretion. Additionally, we have shown that the endothelin-1 (ET-1) gene is targeted by both PER1 and aldosterone. We hypothesized that ET-1 would exhibit a sex-specific response to HS/DOCP treatment in PER1 KO. Here we show that male, but not female, global PER1 KO mice exhibit a decreased night/day ratio of urinary ET-1. Gene expression analysis revealed significant genotype differences in ET-1 and endothelin A receptor (ETA) expression in male, but not female, mice in response to HS/DOCP. Additionally, both wild-type and global PER1 KO male mice significantly increase endothelin B receptor (ETB) expression in response to HS/DOCP, but female mice do not. Finally, siRNA-mediated knockdown of PER1 in mouse cortical collecting duct cells (mpkCCDc14) resulted in increased ET-1 mRNA expression and peptide secretion in response to aldosterone treatment. These data suggest that PER1 is a negative regulator of ET-1 expression in response to HS/DOCP, revealing a novel mechanism for the regulation of renal Na handling in response to HS/DOCP treatment.


Asunto(s)
Endotelina-1/metabolismo , Hipertensión/metabolismo , Túbulos Renales Colectores/fisiopatología , Proteínas Circadianas Period/metabolismo , Eliminación Renal/fisiología , Aldosterona/administración & dosificación , Aldosterona/efectos adversos , Animales , Relojes Circadianos/fisiología , Modelos Animales de Enfermedad , Endotelina-1/orina , Femenino , Humanos , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Túbulos Renales Colectores/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Proteínas Circadianas Period/genética , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Eliminación Renal/efectos de los fármacos , Factores Sexuales , Cloruro de Sodio Dietético/efectos adversos , Cloruro de Sodio Dietético/metabolismo
2.
J Physiol ; 596(23): 5873-5889, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29676801

RESUMEN

KEY POINTS: In the present study, we investigated whether hypoxia during late pregnancy impairs kidney development in mouse offspring, and also whether this has long-lasting consequences affecting kidney function in adulthood. Hypoxia disrupted growth of the kidney, particularly the collecting duct network, in juvenile male offspring. By mid-late adulthood, these mice developed early signs of kidney disease, notably a compromised response to water deprivation. Female offspring showed no obvious signs of impaired kidney development and did not develop kidney disease, suggesting an underlying protection mechanism from the hypoxia insult. These results help us better understand the long-lasting impact of gestational hypoxia on kidney development and the increased risk of chronic kidney disease. ABSTRACT: Prenatal hypoxia is a common perturbation to arise during pregnancy, and can lead to adverse health outcomes in later life. The long-lasting impact of prenatal hypoxia on postnatal kidney development and maturation of the renal tubules, particularly the collecting duct system, is relatively unknown. In the present study, we used a model of moderate chronic maternal hypoxia throughout late gestation (12% O2 exposure from embryonic day 14.5 until birth). Histological analyses revealed marked changes in the tubular architecture of male hypoxia-exposed neonates as early as postnatal day 7, with disrupted medullary development and altered expression of Ctnnb1 and Crabp2 (encoding a retinoic acid binding protein). Kidneys of the RARElacZ line offspring exposed to hypoxia showed reduced ß-galactosidase activity, indicating reduced retinoic acid-directed transcriptional activation. Wild-type male mice exposed to hypoxia had an early decline in urine concentrating capacity, evident at 4 months of age. At 12 months of age, hypoxia-exposed male mice displayed a compromised response to a water deprivation challenge, which was was correlated with an altered cellular composition of the collecting duct and diminished expression of aquaporin 2. There were no differences in the tubular structures or urine concentrating capacity between the control and hypoxia-exposed female offspring at any age. The findings of the present study suggest that prenatal hypoxia selectively disrupts collecting duct patterning through altered Wnt/ß-catenin and retinoic acid signalling and this results in impaired function in male mouse offspring in later life.


Asunto(s)
Hipoxia Fetal/fisiopatología , Túbulos Renales Colectores/fisiopatología , Animales , Animales Recién Nacidos , Femenino , Túbulos Renales Colectores/anatomía & histología , Túbulos Renales Colectores/crecimiento & desarrollo , Masculino , Ratones , Ratones Transgénicos , Embarazo , Factores Sexuales
3.
Am J Physiol Renal Physiol ; 314(5): F1020-F1025, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29357422

RESUMEN

Lithium is widely used in treatment of bipolar affective disorders but often causes nephrogenic diabetes insipidus (NDI), a disorder characterized by severe urinary-concentrating defects. Lithium-induced NDI is caused by lithium uptake by collecting duct principal cells and altered expression of aquaporin-2 (AQP2), which are essential for water reabsorption of tubular fluid in the collecting duct. Sex hormones have previously been shown to affect the regulation of AQP2, so we tested whether tamoxifen (TAM), a selective estrogen receptor modulator, would attenuate lithium-induced alterations on renal water homeostasis. Rats were treated for 14 days with lithium, and TAM treatment was initiated 1 wk after onset of lithium administration. Lithium treatment resulted in severe polyuria and reduced AQP2 expression, which were ameliorated by TAM. Consistent with this, TAM attenuated downregulation of AQP2 and increased phosphorylation of the cAMP-responsive element-binding protein, which induced AQP2 expression in freshly isolated inner-medullary collecting duct suspension prepared from lithium-treated rats. In conclusion, TAM attenuated polyuria dose dependently and impaired urine concentration and downregulation of AQP2 protein expression in rats with lithium-induced NDI. These findings suggest that TAM is likely to be a novel therapeutic option for lithium-induced NDI.


Asunto(s)
Diabetes Insípida Nefrogénica/prevención & control , Hipoglucemiantes/farmacología , Capacidad de Concentración Renal/efectos de los fármacos , Túbulos Renales Colectores/efectos de los fármacos , Cloruro de Litio , Tamoxifeno/farmacología , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Proteína de Unión a CREB/metabolismo , Diabetes Insípida Nefrogénica/inducido químicamente , Diabetes Insípida Nefrogénica/metabolismo , Diabetes Insípida Nefrogénica/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/fisiopatología , Masculino , Fosforilación , Poliuria/inducido químicamente , Poliuria/fisiopatología , Poliuria/prevención & control , Ratas Sprague-Dawley , Factores de Tiempo
4.
Am J Physiol Renal Physiol ; 314(2): F306-F316, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29046300

RESUMEN

Aquaporin-2 (AQP2) is a water channel protein expressed in principal cells (PCs) of the kidney collecting ducts (CDs) and plays a critical role in mediating water reabsorption and urine concentration. AQP2 undergoes both regulated trafficking mediated by vasopressin (VP) and constitutive recycling, which is independent of VP. For both pathways, actin cytoskeletal dynamics is a key determinant of AQP2 trafficking. We report here that manganese chloride (MnCl2) is a novel and potent regulator of AQP2 trafficking in cultured cells and in the kidney. MnCl2 treatment promoted internalization and intracellular accumulation of AQP2. The effect of MnCl2 on the intracellular accumulation of AQP2 was associated with activation of RhoA and actin polymerization without modification of AQP2 phosphorylation. Although the level of total and phosphorylated AQP2 did not change, MnCl2 treatment impeded VP-induced phosphorylation of AQP2 at its serine-256, -264, and -269 residues and dephosphorylation at serine 261. In addition, MnCl2 significantly promoted F-actin polymerization along with downregulation of RhoA activity and prevented VP-induced membrane accumulation of AQP2. Finally, MnCl2 treatment in mice resulted in significant polyuria and reduced urinary concentration, likely due to intracellular relocation of AQP2 in the PCs of kidney CDs. More importantly, the reduced urinary concentration caused by MnCl2 treatment in animals was not corrected by VP. In summary, our study identified a novel effect of MnCl2 on AQP2 trafficking through modifying RhoA activity and actin polymerization and uncovered its potent impact on water diuresis in vivo.


Asunto(s)
Citoesqueleto de Actina/efectos de los fármacos , Actinas/metabolismo , Acuaporina 2/metabolismo , Cloruros/toxicidad , Capacidad de Concentración Renal/efectos de los fármacos , Túbulos Renales Colectores/efectos de los fármacos , Poliuria/inducido químicamente , Citoesqueleto de Actina/metabolismo , Animales , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/fisiopatología , Células LLC-PK1 , Masculino , Compuestos de Manganeso , Ratones Endogámicos C57BL , Fosforilación , Polimerizacion , Poliuria/metabolismo , Poliuria/fisiopatología , Transporte de Proteínas , Transducción de Señal/efectos de los fármacos , Porcinos , Vasopresinas/farmacología , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
5.
Kidney Int ; 94(3): 514-523, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30146013

RESUMEN

Pseudohypoaldosteronism type II (PHAII) is a genetic disease characterized by association of hyperkalemia, hyperchloremic metabolic acidosis, hypertension, low renin, and high sensitivity to thiazide diuretics. It is caused by mutations in the WNK1, WNK4, KLHL3 or CUL3 gene. There is strong evidence that excessive sodium chloride reabsorption by the sodium chloride cotransporter NCC in the distal convoluted tubule is involved. WNK4 is expressed not only in distal convoluted tubule cells but also in ß-intercalated cells of the cortical collecting duct. These latter cells exchange intracellular bicarbonate for external chloride through pendrin, and therefore, account for renal base excretion. However, these cells can also mediate thiazide-sensitive sodium chloride absorption when the pendrin-dependent apical chloride influx is coupled to apical sodium influx by the sodium-driven chloride/bicarbonate exchanger. Here we determine whether this system is involved in the pathogenesis of PHAII. Renal pendrin activity was markedly increased in a mouse model carrying a WNK4 missense mutation (Q562E) previously identified in patients with PHAII. The upregulation of pendrin led to an increase in thiazide-sensitive sodium chloride absorption by the cortical collecting duct, and it caused metabolic acidosis. The function of apical potassium channels was altered in this model, and hyperkalemia was fully corrected by pendrin genetic ablation. Thus, we demonstrate an important contribution of pendrin in renal regulation of sodium chloride, potassium and acid-base homeostasis and in the pathophysiology of PHAII. Furthermore, we identify renal distal bicarbonate secretion as a novel mechanism of renal tubular acidosis.


Asunto(s)
Acidosis Tubular Renal/fisiopatología , Túbulos Renales Colectores/fisiopatología , Proteínas Serina-Treonina Quinasas/genética , Seudohipoaldosteronismo/complicaciones , Transportadores de Sulfato/metabolismo , Acidosis Tubular Renal/sangre , Acidosis Tubular Renal/etiología , Animales , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Humanos , Túbulos Renales Colectores/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Potasio/sangre , Potasio/metabolismo , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/fisiopatología , Eliminación Renal , Cloruro de Sodio/metabolismo , Simportadores de Sodio-Bicarbonato/metabolismo , Transportadores de Sulfato/genética , Regulación hacia Arriba
6.
Am J Physiol Renal Physiol ; 313(3): F666-F668, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28179257

RESUMEN

Alterations in extracellular fluid volume regulation and sodium balance may result in the development and maintenance of salt-dependent hypertension, a major risk factor for cardiovascular disease. Numerous pathways contribute to the regulation of sodium excretion and blood pressure, including endothelin and purinergic signaling. Increasing evidence suggests a link between purinergic receptor activation and endothelin production within the renal collecting duct as a means of promoting natriuresis. A better understanding of the relationship between these two systems, especially in regard to sodium homeostasis, will fill a significant knowledge gap and may provide novel antihypertensive treatment options. Therefore, this review focuses on the cross talk between endothelin and purinergic signaling as it relates to the renal regulation of sodium and blood pressure homeostasis.


Asunto(s)
Adenosina Trifosfato/metabolismo , Presión Sanguínea , Endotelina-1/metabolismo , Túbulos Renales Colectores/metabolismo , Natriuresis , Transducción de Señal , Sodio en la Dieta/metabolismo , Animales , Humanos , Hipertensión/metabolismo , Hipertensión/fisiopatología , Túbulos Renales Colectores/fisiopatología , Receptores de Endotelina/metabolismo , Receptores Purinérgicos P2/metabolismo , Sodio en la Dieta/sangre , Sodio en la Dieta/orina
7.
Am J Physiol Renal Physiol ; 313(6): F1243-F1253, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28814438

RESUMEN

Augmented intratubular angiotensin (ANG) II is a key determinant of enhanced distal Na+ reabsorption via activation of epithelial Na+ channels (ENaC) and other transporters, which leads to the development of high blood pressure (BP). In ANG II-induced hypertension, there is increased expression of the prorenin receptor (PRR) in the collecting duct (CD), which has been implicated in the stimulation of the sodium transporters and resultant hypertension. The impact of PRR deletion along the nephron on BP regulation and Na+ handling remains controversial. In the present study, we investigate the role of PRR in the regulation of renal function and BP by using a mouse model with specific deletion of PRR in the CD (CDPRR-KO). At basal conditions, CDPRR-KO mice had decreased renal function and lower systolic BP associated with higher fractional Na+ excretion and lower ANG II levels in urine. After 14 days of ANG II infusion (400 ng·kg-1·min-1), the increases in systolic BP and diastolic BP were mitigated in CDPRR-KO mice. CDPRR-KO mice had lower abundance of cleaved αENaC and γENaC, as well as lower ANG II and renin content in urine compared with wild-type mice. In isolated CD from CDPRR-KO mice, patch-clamp studies demonstrated that ANG II-dependent stimulation of ENaC activity was reduced because of fewer active channels and lower open probability. These data indicate that CD PRR contributes to renal function and BP responses during chronic ANG II infusion by enhancing renin activity, increasing ANG II, and activating ENaC in the distal nephron segments.


Asunto(s)
Angiotensina II , Presión Sanguínea , Hipertensión/metabolismo , Túbulos Renales Colectores/metabolismo , Natriuresis , ATPasas de Translocación de Protón/deficiencia , Receptores de Superficie Celular/deficiencia , Eliminación Renal , Sodio/metabolismo , Animales , Modelos Animales de Enfermedad , Canales Epiteliales de Sodio/metabolismo , Predisposición Genética a la Enfermedad , Hipertensión/genética , Hipertensión/fisiopatología , Hipertensión/prevención & control , Túbulos Renales Colectores/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Proteinuria/metabolismo , Proteinuria/fisiopatología , ATPasas de Translocación de Protón/genética , Receptores de Superficie Celular/genética , Renina/metabolismo , Cloruro de Sodio Dietético/administración & dosificación , Cloruro de Sodio Dietético/metabolismo , Factores de Tiempo
8.
Kidney Int ; 91(5): 1070-1087, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28139295

RESUMEN

Hypercalcemia can cause renal dysfunction such as nephrogenic diabetes insipidus (NDI), but the mechanisms underlying hypercalcemia-induced NDI are not well understood. To elucidate the early molecular changes responsible for this disorder, we employed mass spectrometry-based proteomic analysis of inner medullary collecting ducts (IMCD) isolated from parathyroid hormone-treated rats at onset of hypercalcemia-induced NDI. Forty-one proteins, including the water channel aquaporin-2, exhibited significant changes in abundance, most of which were decreased. Bioinformatic analysis revealed that many of the downregulated proteins were associated with cytoskeletal protein binding, regulation of actin filament polymerization, and cell-cell junctions. Targeted LC-MS/MS and immunoblot studies confirmed the downregulation of 16 proteins identified in the initial proteomic analysis and in additional experiments using a vitamin D treatment model of hypercalcemia-induced NDI. Evaluation of transcript levels and estimated half-life of the downregulated proteins suggested enhanced protein degradation as the possible regulatory mechanism. Electron microscopy showed defective intercellular junctions and autophagy in the IMCD cells from both vitamin D- and parathyroid hormone-treated rats. A significant increase in the number of autophagosomes was confirmed by immunofluorescence labeling of LC3. Colocalization of LC3 and Lamp1 with aquaporin-2 and other downregulated proteins was found in both models. Immunogold electron microscopy revealed aquaporin-2 in autophagosomes in IMCD cells from both hypercalcemia models. Finally, parathyroid hormone withdrawal reversed the NDI phenotype, accompanied by termination of aquaporin-2 autophagic degradation and normalization of both nonphoshorylated and S256-phosphorylated aquaporin-2 levels. Thus, enhanced autophagic degradation of proteins plays an important role in the initial mechanism of hypercalcemic-induced NDI.


Asunto(s)
Acuaporina 2/metabolismo , Autofagia , Diabetes Insípida Nefrogénica/fisiopatología , Hipercalcemia/complicaciones , Túbulos Renales Colectores/fisiopatología , Animales , Cromatografía Liquida , Diabetes Insípida Nefrogénica/etiología , Diabetes Insípida Nefrogénica/metabolismo , Dihidrotaquisterol/toxicidad , Modelos Animales de Enfermedad , Regulación hacia Abajo , Técnica del Anticuerpo Fluorescente , Semivida , Humanos , Hipercalcemia/inducido químicamente , Uniones Intercelulares/metabolismo , Uniones Intercelulares/ultraestructura , Túbulos Renales Colectores/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Masculino , Microscopía Inmunoelectrónica , Proteínas Asociadas a Microtúbulos/metabolismo , Hormona Paratiroidea/farmacología , Fosforilación , Proteolisis , Proteómica/métodos , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem
9.
Curr Hypertens Rep ; 19(8): 62, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28695400

RESUMEN

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.


Asunto(s)
Hipertensión/metabolismo , Hipertensión/fisiopatología , Túbulos Renales Colectores/metabolismo , Renina/metabolismo , Angiotensina II/metabolismo , Animales , Humanos , Hipertensión/etiología , Túbulos Renales Colectores/fisiopatología , Túbulos Renales Distales/metabolismo , Túbulos Renales Distales/fisiopatología , Túbulos Renales Proximales/fisiopatología , Renina/biosíntesis , Sistema Renina-Angiotensina/fisiología
10.
Curr Hypertens Rep ; 18(4): 29, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26951246

RESUMEN

Recent evidence suggests that the renal tubular renin angiotensin system regulates urinary Na(+) and water excretion and blood pressure. Three key components of the tubular renin angiotensin system, namely renin, prorenin receptor, and angiotensin-II type 1 receptor, are localized to the collecting duct. This system may modulate collecting duct Na(+) and water reabsorption via angiotensin-II-dependent and angiotensin-II-independent pathways. Further, the system may be of greatest relevance in hypertensive states and particularly those characterized by high circulating angiotensin-II. In this review, we summarize the current knowledge on the synthesis, regulation, and function of collecting duct-derived renin angiotensin system components and examine recent developments with regard to regulation of blood pressure and renal fluid and Na(+) excretion.


Asunto(s)
Presión Sanguínea , Túbulos Renales Colectores/fisiopatología , Sistema Renina-Angiotensina , Animales , Humanos , Hipertensión/fisiopatología , Receptores de Superficie Celular/metabolismo , Renina/sangre , ATPasas de Translocación de Protón Vacuolares/metabolismo
11.
Clin Exp Hypertens ; 38(1): 1-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26151827

RESUMEN

We elucidated the role of collecting duct kinin B2 receptor (B2R) in the development of salt-sensitivity and angiotensin II (ANG II)-induced hypertension. To this end, we used a Cre-Lox recombination strategy to generate mice lacking Bdkrb2 gene for B2R in the collecting duct (Hoxb7-Cre(tg/+):Bdkrb2(flox/flox)). In 3 groups of control (Bdkrb2(flox/flox)) and 3 groups of UB(Bdkrb2-/-) mice, systolic blood pressure (SBP) responses to high salt intake (4 or 8% NaCl; HS) were monitored by radiotelemetry in comparison with standard salt diet (0.4% NaCl) prior to and during subcutaneous ANG II infusion (1000 ng/min/kg) via osmotic minipumps. High salt intakes alone for 2 weeks did not alter SBP in either strain. ANG II significantly increased SBP equally in control (121 ± 2 to 156 ± 3 mmHg) and UB(Bdkrb2-/-) mice (120 ± 2 to 153 ± 2 mmHg). The development of ANG II-induced hypertension was exacerbated by 4%HS in both control (125 ± 3 to 164 ± 5 mmHg) and UB(Bdkrb2-/-) mice (124 ± 2 to 162 ± 3 mmHg) during 2 weeks. Interestingly, 8%HS caused a more profound and earlier ANG II-induced hypertension in UB(Bdkrb2-/-) (129 ± 2 to 166 ± 3 mmHg) as compared to control (128 ± 2 to 158 ± 2 mmHg) and it was accompanied by body weight loss and increased mortality. In conclusion, targeted inactivation of B2R in the renal collecting duct does not cause salt-sensitivity; however, collecting duct B2R attenuates the hypertensive actions of ANG II under conditions of very high salt intake.


Asunto(s)
Angiotensina II/metabolismo , Presión Sanguínea , Hipertensión , Túbulos Renales Colectores , Cloruro de Sodio Dietético/efectos adversos , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Hipertensión/metabolismo , Hipertensión/fisiopatología , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/fisiopatología , Masculino , Ratones , Receptor de Bradiquinina B2/genética
12.
J Am Soc Nephrol ; 26(12): 2978-87, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25855780

RESUMEN

P2Y12 receptor (P2Y12-R) signaling is mediated through Gi, ultimately reducing cellular cAMP levels. Because cAMP is a central modulator of arginine vasopressin (AVP)-induced water transport in the renal collecting duct (CD), we hypothesized that if expressed in the CD, P2Y12-R may play a role in renal handling of water in health and in nephrogenic diabetes insipidus. We found P2Y12-R mRNA expression in rat kidney, and immunolocalized its protein and aquaporin-2 (AQP2) in CD principal cells. Administration of clopidogrel bisulfate, an irreversible inhibitor of P2Y12-R, significantly increased urine concentration and AQP2 protein in the kidneys of Sprague-Dawley rats. Notably, clopidogrel did not alter urine concentration in Brattleboro rats that lack AVP. Clopidogrel administration also significantly ameliorated lithium-induced polyuria, improved urine concentrating ability and AQP2 protein abundance, and reversed the lithium-induced increase in free-water excretion, without decreasing blood or kidney tissue lithium levels. Clopidogrel administration also augmented the lithium-induced increase in urinary AVP excretion and suppressed the lithium-induced increase in urinary nitrates/nitrites (nitric oxide production) and 8-isoprostane (oxidative stress). Furthermore, selective blockade of P2Y12-R by the reversible antagonist PSB-0739 in primary cultures of rat inner medullary CD cells potentiated the expression of AQP2 and AQP3 mRNA, and cAMP production induced by dDAVP (desmopressin). In conclusion, pharmacologic blockade of renal P2Y12-R increases urinary concentrating ability by augmenting the effect of AVP on the kidney and ameliorates lithium-induced NDI by potentiating the action of AVP on the CD. This strategy may offer a novel and effective therapy for lithium-induced NDI.


Asunto(s)
Arginina Vasopresina/metabolismo , Diabetes Insípida Nefrogénica/metabolismo , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/fisiopatología , Receptores Purinérgicos P2Y12/metabolismo , Animales , Acuaporina 2/análisis , Acuaporina 2/efectos de los fármacos , Acuaporina 2/orina , Arginina Vasopresina/efectos de los fármacos , Arginina Vasopresina/orina , Clopidogrel , Desamino Arginina Vasopresina/metabolismo , Diabetes Insípida Nefrogénica/inducido químicamente , Diabetes Insípida Nefrogénica/fisiopatología , Capacidad de Concentración Renal/efectos de los fármacos , Médula Renal/química , Túbulos Renales Colectores/química , Litio , Masculino , Antagonistas del Receptor Purinérgico P2Y/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Brattleboro , Ratas Sprague-Dawley , Receptores Purinérgicos P2Y12/análisis , Receptores Purinérgicos P2Y12/genética , Ticlopidina/análogos & derivados , Ticlopidina/farmacología , Agua/metabolismo
13.
Clin Exp Pharmacol Physiol ; 42(1): 14-21, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25371190

RESUMEN

The intrarenal renin-angiotensin system (RAS) plays a critical role in the pathogenesis and progression of hypertension and kidney disease. In angiotensin (Ang) II-dependent hypertension, collecting duct renin synthesis and secretion are stimulated despite suppression of juxtaglomerular (JG) renin. This effect is mediated by the AngII type I receptor (AT1 R), independent of blood pressure. Although the regulation of JG renin has been extensively studied, the mechanisms by which renin is regulated in the collecting duct remain unclear. The augmentation of renin synthesis and activity in the collecting duct may provide a pathway for additional generation of intrarenal and intratubular AngII formation due to the presence of angiotensinogen substrate and angiotensin-converting enzyme in the nephron. The recently described (pro)renin receptor ((P)RR) binds renin or prorenin, enhancing renin activity and fully activating the biologically inactive prorenin peptide. Stimulation of (P)RR also activates intracellular pathways related to fibrosis. Renin and the (P)RR are augmented in renal tissues of AngII-dependent hypertensive rats. However, the functional contribution of the (P)RR to enhanced renin activity in the collecting duct and its contribution to the development of hypertension and kidney disease have not been well elucidated. This review focuses on recent evidence demonstrating the mechanism of renin regulation in the collecting ducts and its interaction with the (P)RR. The data suggest that renin-(P)RR interactions may induce stimulation of intracellular pathways associated with the development of hypertension and kidney disease.


Asunto(s)
Hipertensión/fisiopatología , Túbulos Renales Colectores/fisiopatología , Receptores de Superficie Celular/fisiología , ATPasas de Translocación de Protón Vacuolares/fisiología , Animales , Humanos , Hipertensión/diagnóstico , Renina/fisiología , Sistema Renina-Angiotensina/fisiología
14.
Biochemistry (Mosc) ; 80(5): 582-5, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26071776

RESUMEN

Chronic progressive nephropathy is a common noninfectious disease in aging (mice, rats) and non-aging (naked mole rat) rodents, sometimes resulting in death. The etiology and pathogenesis of the disease remain mysterious. For instance, it remains unclear what is the immediate cause of the disease and where exactly in the kidneys, glomerular or tubulointerstitial compartment, do primary and secondary changes occur. Here, I propose a potential scenario for development of progressive nephropathy that is based on an assumption that the disease is caused by occurrence and spread of mutant cellular clones from tubular epithelium secreting proinflammatory and prosclerotic cytokines. The hypothesis considers some features of the disease that have never been discussed earlier. According to the proposed concept, a clone of mutant cells secretes cytokines inducing chronic inflammation, proliferation of fibroblasts, and active collagen production that eventually results in sclerosis and thickening of tubular basement membranes. Sclerosis of interstitium and thickening of tubular basement membranes cause narrowing of some parts of the nephron, especially collecting ducts, which hinders passage of the urine, elevates tubular hydrostatic pressure, and impairs filtration and reabsorption in the kidneys. High hydrostatic pressure and reabsorption-induced elevated concentration of macromolecular substances in the primary urine result in development of large cysts and glomerular hyalinosis followed by renal failure. Based on this, it might be concluded that chronic progressive nephropathy in rodents represents a special type of tubulointerstitial dysplasia (or "non-tumorous neoplasia") in kidneys with secondary glomerular disorder at late stage of the disease. The concept for development of the disease proposed here may be of special importance from the viewpoint of toxicological pathology and gerontology, particularly for analysis of pathological features resulting in death of non-aging animals (naked mole rats).


Asunto(s)
Envejecimiento , Túbulos Renales Colectores , Mutación , Insuficiencia Renal Crónica , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Presión Hidrostática , Inflamación , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/patología , Túbulos Renales Colectores/fisiopatología , Ratones , Ratas , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/fisiopatología
15.
J Am Soc Nephrol ; 25(2): 232-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24158982

RESUMEN

cAMP is an important mediator of cystogenesis in polycystic kidney disease (PKD). Several adenylyl cyclase (AC) isoforms could mediate cAMP accumulation in PKD, and identification of a specific pathogenic AC isoform is of therapeutic interest. We investigated the role of AC6 in a mouse model of PKD that is homozygous for the loxP-flanked PKD1 gene and heterozygous for an aquaporin-2-Cre recombinase transgene to achieve collecting duct-specific gene targeting. Collecting duct-specific knockout of polycystin-1 caused massive renal cyst formation, kidney enlargement, and severe kidney failure, with a mean survival time of 2 months. In contrast, coincident collecting duct-specific knockout of polycystin-1 and AC6 (also homozygous for the floxed ADCY6 gene) markedly decreased kidney size and cystogenesis, improved renal function, reduced activation of the B-Raf/ERK/MEK pathway, and greatly increased survival. Absence of collecting duct AC6 did not alter urinary cAMP excretion or kidney cAMP concentration. In conclusion, AC6 is a key mediator of cyst formation and renal injury in a model of PKD.


Asunto(s)
Adenilil Ciclasas/deficiencia , Riñón Poliquístico Autosómico Dominante/enzimología , Adenilil Ciclasas/genética , Adenilil Ciclasas/fisiología , Animales , Acuaporina 2/genética , AMP Cíclico/fisiología , Modelos Animales de Enfermedad , Femenino , Genotipo , Integrasas/genética , Riñón/patología , Fallo Renal Crónico/etiología , Fallo Renal Crónico/prevención & control , Túbulos Renales Colectores/fisiopatología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Canales Catiónicos TRPP/deficiencia , Canales Catiónicos TRPP/genética , Transgenes
16.
Am J Physiol Renal Physiol ; 307(5): F551-9, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24899059

RESUMEN

Polycystic kidney disease (PKD) is a common genetic disorder leading to cyst formation in the kidneys and other organs that ultimately results in kidney failure and death. Currently, there is no therapy for slowing down or stopping the progression of PKD. In this study, we identified the disintegrin metalloenzyme 17 (ADAM17) as a key regulator of cell proliferation in kidney tissues of conditional knockout Ift88(-/-) mice and collecting duct epithelial cells from Ift88°(rpk) mice, animal models of autosomal recessive polycystic kidney disease (ARPKD). Using Western blotting, an enzyme activity assay, and a growth factor-shedding assay in the presence or absence of the specific ADAM17 inhibitor TMI-005, we show that increased expression and activation of ADAM17 in the cystic kidney and in collecting duct epithelial cells originating from the Ift88°(rpk) mice (designated as PKD cells) lead to constitutive shedding of several growth factors, including heparin-binding EGF-like growth factor (HB-EGF), amphiregulin, and transforming growth factor-α (TGF-α). Increased growth factor shedding induces activation of the EGFR/MAPK/ERK pathway and maintains higher cell proliferation rate in PKD cells compared with control cells. PKD cells also displayed increased lactate formation and extracellular acidification indicative of aerobic glycolysis (Warburg effect), which was blocked by ADAM17 inhibition. We propose that ADAM17 is a key promoter of cellular proliferation in PKD cells by activating the EGFR/ERK axis and a proproliferative glycolytic phenotype.


Asunto(s)
Proteínas ADAM/fisiología , Proliferación Celular/fisiología , Células Epiteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Glucólisis/fisiología , Túbulos Renales Colectores/patología , Enfermedades Renales Poliquísticas/fisiopatología , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/efectos de los fármacos , Proteína ADAM17 , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Receptores ErbB/fisiología , Femenino , Factor de Crecimiento Similar a EGF de Unión a Heparina/fisiología , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/fisiopatología , Masculino , Ratones , Ratones Noqueados , Morfolinas/farmacología , Fenotipo , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Factor de Crecimiento Transformador alfa/fisiología , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
17.
Crit Care Med ; 42(10): e663-72, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25072760

RESUMEN

OBJECTIVE: Acute renal tubular injury is a serious complication in the postoperative period, which is associated with high mortality and increased ICU stay. We aimed to demonstrate the protective effect of rhEPO against acute tubular injury induced by ischemia-reperfusion and to explore the mechanism of canonical transient receptor potential channel-6. DESIGN: Randomized laboratory animal study. SETTINGS: Animal research laboratory. INTERVENTIONS: Male Sprague-Dawley rats were randomly divided into three groups: the sham group, the control group, and the rhEPO group. Experimental acute tubular injury was established in rats by bilateral renal arterial occlusion for 30 minutes followed by reperfusion. MEASUREMENTS AND MAIN RESULTS: Blood samples were obtained for cystatin-C and neutrophil gelatinase-associated lipocalin measurements by enzyme-linked immunosorbance assays. Seventy-two hours after reperfusion, urine samples were collected for osmolality and fractional excretion of sodium (%) assays on a chemistry analyzer. Kidneys were harvested at 24, 48, and 72 hours after reperfusion. Transient receptor potential channel-6, aquaporin-2, and Na,K-ATPase expression in collecting ducts were studied by immunofluorescence and Western blot. Coimmunoprecipitations were also performed to identify the possible signalplex relation between transient receptor potential channel-6 and aquaporin-2 or Na,K-ATPase channels. RhEPO pretreatment significantly inhibited serum cystatin-C (2 hr: 453 ± 64 µg/L vs 337 ± 28 µg/L, p < 0.01), serum neutrophil gelatinase-associated lipocalin (72 hr: 1,175 ± 107 ng/L vs 1,737 ± 402 ng/L, p < 0.05), and urinary fractional excretion of sodium (%) increase (0.9 ± 0.1 vs 2.2 ± 0.8, p < 0.05) and alleviated the decrease of urinary osmolality (1,293 ± 101 mosmol/kg H2O vs 767 ± 91 mosmol/kg H2O, p < 0.05) induced by ischemia-reperfusion injury. Meanwhile, recombinant human erythropoietin greatly improved the ischemia-reperfusion-induced attenuation of transient receptor potential channel-6 expression (48 hr: 42% ± 2% vs 67% ± 2% and 72 hr: 55% ± 2% vs 66% ± 2%), as well as aquaporin-2 and Na,K-ATPase expression in collecting ducts. Transient receptor potential channel-6 functionally interacted with Na,K-ATPase but not aquaporin-2. CONCLUSIONS: Recombinant human erythropoietin pretreatment at the dose of 5,000 IU/kg potently prevented ischemia-reperfusion-induced acute tubular injury, which might be partly attributed to the restoring the effect of transient receptor potential channel-6 expression and collecting duct function.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Eritropoyetina/uso terapéutico , Túbulos Renales Colectores/fisiopatología , Daño por Reperfusión/tratamiento farmacológico , Canales Catiónicos TRPC/fisiología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/fisiopatología , Proteínas de Fase Aguda/fisiología , Animales , Acuaporina 2/análisis , Acuaporina 2/metabolismo , Acuaporina 2/fisiología , Cistatina C/sangre , Cistatina C/fisiología , Humanos , Túbulos Renales Colectores/química , Túbulos Renales Colectores/metabolismo , Lipocalina 2 , Lipocalinas/sangre , Lipocalinas/fisiología , Masculino , Proteínas Proto-Oncogénicas/sangre , Proteínas Proto-Oncogénicas/fisiología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/uso terapéutico , Daño por Reperfusión/metabolismo , Daño por Reperfusión/fisiopatología , ATPasa Intercambiadora de Sodio-Potasio/análisis , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Canales Catiónicos TRPC/análisis
18.
Minerva Endocrinol ; 39(1): 33-41, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24513602

RESUMEN

Hyponatremia is the most frequent electrolyte disorder in hospitalized patients. Acute and severe hyponatremia can be a life-threatening condition, but recent evidence indicates that also mild and chronic hyponatremia is associated with neurological and extra-neurological signs, such as gait disturbances, attention deficits, falls and fracture occurrence, and bone loss. The syndrome of inappropriate ADH secretion (SIADH) is the most frequent cause of hyponatremia. Hyponatremia secondary to SIADH may result for instance from ectopic release of ADH in lung cancer, from diseases affecting the central nervous system, from pneumonia or other pneumopathies or as a side-effect of various drugs In SIADH, hyponatremia results from a pure disorder of water handling by the kidney, whereas external sodium balance is usually well regulated. Despite increased total body water, only minor changes of urine output and modest oedema are usually seen. Neurological impairment may range from subclinical to life-threatening, depending on the degree and mostly on the rate of serum sodium reduction. The management of hyponatremia secondary to SIADH is largely dependent on the symptomatology of the patient. This review briefly summarizes the main aspects related to hyponatremia and then discusses the available treatment options for the management of SIADH, including vaptans, which are vasopressin receptor antagonists targeted for the correction of euvolemic hyponatremia, such as that observed in SIADH.


Asunto(s)
Hiponatremia/etiología , Síndrome de Secreción Inadecuada de ADH/complicaciones , Antagonistas de los Receptores de Hormonas Antidiuréticas , Benzazepinas/efectos adversos , Benzazepinas/uso terapéutico , Volumen Sanguíneo , Agua Corporal , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Ensayos Clínicos como Asunto , Demeclociclina/efectos adversos , Demeclociclina/uso terapéutico , Diabetes Insípida Nefrogénica/inducido químicamente , Manejo de la Enfermedad , Interacciones Farmacológicas , Hospitalización , Humanos , Hidrocortisona/deficiencia , Hiponatremia/inducido químicamente , Hiponatremia/tratamiento farmacológico , Hipotiroidismo/complicaciones , Síndrome de Secreción Inadecuada de ADH/tratamiento farmacológico , Túbulos Renales Colectores/fisiopatología , Litio/efectos adversos , Litio/uso terapéutico , Estudios Multicéntricos como Asunto , Concentración Osmolar , Síndromes Paraneoplásicos/complicaciones , Solución Salina Hipertónica/uso terapéutico , Tolvaptán
19.
J Am Soc Nephrol ; 24(4): 604-16, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23411787

RESUMEN

The molecular mechanism of cyst formation and expansion in autosomal recessive polycystic kidney disease (ARPKD) is poorly understood, but impaired mechanosensitivity to tubular flow and dysfunctional calcium signaling are important contributors. The activity of the mechanosensitive Ca(2+)-permeable TRPV4 channel underlies flow-dependent Ca(2+) signaling in murine collecting duct (CD) cells, suggesting that this channel may contribute to cystogenesis in ARPKD. Here, we developed a method to isolate CD-derived cysts and studied TRPV4 function in these cysts laid open as monolayers and in nondilated split-open CDs in a rat model of ARPKD. In freshly isolated CD-derived cyst monolayers, we observed markedly impaired TRPV4 activity, abnormal subcellular localization of the channel, disrupted TRPV4 glycosylation, decreased basal [Ca(2+)]i, and loss of flow-mediated [Ca(2+)]i signaling. In contrast, nondilated CDs of these rats exhibited functional TRPV4 with largely preserved mechanosensitive properties. Long-term systemic augmentation of TRPV4 activity with a selective TRPV4 activator significantly attenuated the renal manifestations of ARPKD in a time-dependent manner. At the cellular level, selective activation of TRPV4 restored mechanosensitive Ca(2+) signaling as well as the function and subcellular distribution of TRPV4. In conclusion, the functional status of TRPV4, which underlies mechanosensitive Ca(2+) signaling in CD cells, inversely correlates with renal cystogenesis in ARPKD. Augmenting TRPV4 activity may have therapeutic potential in ARPKD.


Asunto(s)
Calcio/metabolismo , Quistes/metabolismo , Túbulos Renales Colectores/patología , Riñón Poliquístico Autosómico Recesivo/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Quistes/fisiopatología , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/fisiopatología , Riñón Poliquístico Autosómico Recesivo/fisiopatología , Ratas , Ratas Sprague-Dawley , Transducción de Señal
20.
J Am Soc Nephrol ; 24(9): 1435-50, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23766535

RESUMEN

Ciliogenesis and cystogenesis require the exocyst, a conserved eight-protein trafficking complex that traffics ciliary proteins. In culture, the small GTPase Cdc42 co-localizes with the exocyst at primary cilia and interacts with the exocyst component Sec10. The role of Cdc42 in vivo, however, is not well understood. Here, knockdown of cdc42 in zebrafish produced a phenotype similar to sec10 knockdown, including tail curvature, glomerular expansion, and mitogen-activated protein kinase (MAPK) activation, suggesting that cdc42 and sec10 cooperate in ciliogenesis. In addition, cdc42 knockdown led to hydrocephalus and loss of photoreceptor cilia. Furthermore, there was a synergistic genetic interaction between zebrafish cdc42 and sec10, suggesting that cdc42 and sec10 function in the same pathway. Mice lacking Cdc42 specifically in kidney tubular epithelial cells died of renal failure within weeks of birth. Histology revealed cystogenesis in distal tubules and collecting ducts, decreased ciliogenesis in cyst cells, increased tubular cell proliferation, increased apoptosis, increased fibrosis, and led to MAPK activation, all of which are features of polycystic kidney disease, especially nephronophthisis. Taken together, these results suggest that Cdc42 localizes the exocyst to primary cilia, whereupon the exocyst targets and docks vesicles carrying ciliary proteins. Abnormalities in this pathway result in deranged ciliogenesis and polycystic kidney disease.


Asunto(s)
Cilios/metabolismo , Cilios/patología , Enfermedades Renales Quísticas/patología , Enfermedades Renales Quísticas/fisiopatología , Fenotipo , Proteína de Unión al GTP cdc42/deficiencia , Animales , Apoptosis , Proliferación Celular , Modelos Animales de Enfermedad , Fibrosis , Técnicas In Vitro , Enfermedades Renales Quísticas/metabolismo , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/patología , Túbulos Renales Colectores/fisiopatología , Túbulos Renales Distales/metabolismo , Túbulos Renales Distales/patología , Túbulos Renales Distales/fisiopatología , Ratones , Ratones Noqueados , Ratones Transgénicos , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Transducción de Señal/fisiología , Proteínas de Transporte Vesicular/deficiencia , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Pez Cebra , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA