Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 325(6): F733-F749, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37823196

RESUMEN

Chronic infusion of subpressor level of angiotensin II (ANG II) increases the abundance of Na+ transporters along the distal nephron, balanced by suppression of Na+ transporters along the proximal tubule and medullary thick ascending limb (defined as "proximal nephron"), which impacts K+ handling along the entire renal tubule. The objective of this study was to quantitatively assess the impact of chronic ANG II on the renal handling of Na+ and K+ in female rats, using a computational model of the female rat renal tubule. Our results indicate that the downregulation of proximal nephron Na+ reabsorption (TNa), which occurs in response to ANG II-triggered hypertension, involves changes in both transporter abundance and trafficking. Our model suggests that substantial (∼30%) downregulation of active NHE3 in proximal tubule (PT) microvilli is needed to reestablish the Na+ balance at 2 wk of ANG II infusion. The 35% decrease in SGLT2, a known NHE3 regulator, may contribute to this downregulation. Both depression of proximal nephron TNa and stimulation of distal ENaC raise urinary K+ excretion in ANG II-treated females, while K+ loss is slightly mitigated by cortical NKCC2 and NCC upregulation. Our model predicts that K+ excretion may be more significantly limited during ANG II infusion by ROMK inhibition in the distal nephron and/or KCC3 upregulation in the PT, which remain open questions for experimental validation. In summary, our analysis indicates that ANG II hypertension triggers a series of events from distal TNa stimulation followed by compensatory reduction in proximal nephron TNa and accompanying adjustments to limit excessive K+ secretion.NEW & NOTEWORTHY We used a computational model of the renal tubule to assess the impact of 2-wk angiotensin II (ANG II) infusion on the handling of Na+ and K+ in female rats. ANG II strongly stimulates distal Na+ reabsorption and K+ secretion. Simulations indicate that substantial downregulation of proximal tubule NHE3 is needed to reestablish Na+ balance at 2 wk. Proximal adaptations challenge K+ homeostasis, and regulation of distal NCC and specific K+ channels likely limit urinary K+ losses.


Asunto(s)
Angiotensina II , Hipertensión , Túbulos Renales , Potasio , Sodio , Femenino , Animales , Ratas , Túbulos Renales/fisiopatología , Hipertensión/fisiopatología , Angiotensina II/farmacología , Sodio/metabolismo , Potasio/metabolismo , Ratas Sprague-Dawley , Simulación por Computador , Masculino , Simportadores/metabolismo
2.
J Am Heart Assoc ; 11(19): e026581, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36172956

RESUMEN

Background A beneficial role for prostanoids in hypertension is suggested by clinical studies showing nonsteroidal anti-inflammatory drugs, which block the production of all prostanoids, cause sodium retention and exacerbate hypertension. Among prostanoids, prostaglandin E2 and its E-prostanoid receptor 4 receptor (EP4R) have been implicated in blood pressure control. Our previous study found that conditional deletion of EP4R from all tissues in adult mice exacerbates angiotensin II-dependent hypertension, suggesting a powerful effect of EP4R to resist blood pressure elevation. We also found that elimination of EP4R from vascular smooth muscle cells did not affect the severity of hypertension, suggesting nonvascular targets of prostaglandin E mediate this antihypertensive effect. Methods and Results Here we generated mice with cell-specific deletion of EP4R from macrophage-specific EP4 receptor knockouts or kidney epithelial cells (KEKO) to assess the contributions of EP4R in these cells to hypertension pathogenesis. Macrophage-specific EP4 receptor knockouts showed similar blood pressure responses to alterations in dietary sodium or chronic angiotensin II infusion as Controls. By contrast, angiotensin II-dependent hypertension was significantly augmented in KEKOs (mean arterial pressure: 146±3 mm Hg) compared with Controls (137±4 mm Hg; P=0.02), which was accompanied by impaired natriuresis in KEKOs. Because EP4R expression in the kidney is enriched in the collecting duct, we compared responses to amiloride in angiotensin II-infused KEKOs and Controls. Blockade of the epithelial sodium channel with amiloride caused exaggerated natriuresis in KEKOs compared with Controls (0.21±0.01 versus 0.15±0.02 mmol/24 hour per 20 g; P=0.015). Conclusions Our data suggest EP4R in kidney epithelia attenuates hypertension. This antihypertension effect of EP4R may be mediated by reducing the activity of the epithelial sodium channel, thereby promoting natriuresis.


Asunto(s)
Hipertensión , Subtipo EP4 de Receptores de Prostaglandina E , Amilorida/uso terapéutico , Angiotensina II/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Antihipertensivos/uso terapéutico , Dinoprostona/metabolismo , Células Epiteliales , Canales Epiteliales de Sodio/genética , Hipertensión/tratamiento farmacológico , Riñón , Macrófagos/metabolismo , Ratones , Prostaglandinas , Subtipo EP4 de Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Sodio/metabolismo , Cloruro de Sodio Dietético/metabolismo
3.
Am J Physiol Cell Physiol ; 321(5): C897-C909, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34613843

RESUMEN

Kidneys continuously filter an enormous amount of sodium and adapt kidney Na+ reabsorption to match Na+ intake to maintain circulatory volume and electrolyte homeostasis. Males (M) respond to high-salt (HS) diet by translocating proximal tubule Na+/H+ exchanger isoform 3 (NHE3) to the base of the microvilli, reducing activated forms of the distal NaCl cotransporter (NCC) and epithelial Na+ channel (ENaC). Males (M) and females (F) on normal-salt (NS) diet present sex-specific profiles of "transporters" (cotransporters, channels, pumps, and claudins) along the nephron, e.g., F exhibit 40% lower NHE3 and 200% higher NCC abundance than M. We tested the hypothesis that adaptations to HS diet along the nephron will, likewise, exhibit sexual dimorphisms. C57BL/6J mice were fed for 15 days with 4% NaCl diet (HS) versus 0.26% NaCl diet (NS). On HS, M and F exhibited normal plasma [Na+] and [K+], similar urine volume, Na+, K+, and osmolal excretion rates normalized to body weight. In F, like M, HS lowered abundance of distal NCC, phosphorylated NCC, and cleaved (activated) forms of ENaC. The adaptations associated with achieving electrolyte homeostasis exhibit sex-dependent and independent mechanisms. Sex differences in baseline "transporters" abundance persist during HS diet, yet the fold changes during HS diet (normalized to NS) are similar along the distal nephron and collecting duct. Sex-dependent differences observed along the proximal tubule during HS show that female kidneys adapt differently from patterns reported in males, yet achieve and maintain fluid and electrolyte homeostasis.


Asunto(s)
Adaptación Fisiológica , Proteínas de Transporte de Membrana/metabolismo , Nefronas/metabolismo , Cloruro de Sodio Dietético/metabolismo , Equilibrio Hidroelectrolítico , Animales , Biomarcadores/sangre , Biomarcadores/orina , Femenino , Túbulos Renales Colectores/metabolismo , Túbulos Renales Proximales/metabolismo , Masculino , Ratones Endogámicos C57BL , Fosforilación , Caracteres Sexuales , Factores Sexuales , Cloruro de Sodio Dietético/efectos adversos , Intercambiador 3 de Sodio-Hidrógeno/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
5.
Am J Physiol Renal Physiol ; 321(1): F69-F81, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34056928

RESUMEN

The renal nephron consists of a series of distinct cell types that function in concert to maintain fluid and electrolyte balance and blood pressure. The renin-angiotensin system (RAS) is central to Na+ and volume balance. We aimed to determine how loss of angiotensin II signaling in the proximal tubule (PT), which reabsorbs the bulk of filtered Na+ and volume, impacts solute transport throughout the nephron. We hypothesized that PT renin-angiotensin system disruption would not only depress PT Na+ transporters but also impact downstream Na+ transporters. Using a mouse model in which the angiotensin type 1a receptor (AT1aR) is deleted specifically within the PT (AT1aR PTKO), we profiled the abundance of Na+ transporters, channels, and claudins along the nephron. Absence of PT AT1aR signaling was associated with lower abundance of PT transporters (Na+/H+ exchanger isoform 3, electrogenic Na+-bicarbonate cotransporter 1, and claudin 2) as well as lower abundance of downstream transporters (total and phosphorylated Na+-K+-2Cl- cotransporter, medullary Na+-K+-ATPase, phosphorylated NaCl cotransporter, and claudin 7) versus controls. However, transport activities of Na+-K+-2Cl- cotransporter and NaCl cotransporter (assessed with diuretics) were similar between groups in order to maintain electrolyte balance. Together, these results demonstrate the primary impact of angiotensin II regulation on Na+ reabsorption in the PT at baseline and the associated influence on downstream Na+ transporters, highlighting the ability of the nephron to integrate Na+ transport along the nephron to maintain homeostasis.NEW & NOTEWORTHY Our study defines a novel role for proximal tubule angiotensin receptors in regulating the abundance of Na+ transporters throughout the nephron, thereby contributing to the integrated control of fluid balance in vivo.


Asunto(s)
Angiotensina II/farmacología , Proteínas de Transporte de Membrana/metabolismo , Nefronas/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Animales , Riñón/metabolismo , Natriuresis/efectos de los fármacos , Intercambiadores de Sodio-Hidrógeno/metabolismo
6.
Am J Physiol Renal Physiol ; 320(6): F1080-F1092, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33969697

RESUMEN

A major pathway in hypertension pathogenesis involves direct activation of ANG II type 1 (AT1) receptors in the kidney, stimulating Na+ reabsorption. AT1 receptors in tubular epithelia control expression and stimulation of Na+ transporters and channels. Recently, we found reduced blood pressure and enhanced natriuresis in mice with cell-specific deletion of AT1 receptors in smooth muscle (SMKO mice). Although impaired vasoconstriction and preserved renal blood flow might contribute to exaggerated urinary Na+ excretion in SMKO mice, we considered whether alterations in Na+ transporter expression might also play a role; therefore, we carried out proteomic analysis of key Na+ transporters and associated proteins. Here, we show that levels of Na+-K+-2Cl- cotransporter isoform 2 (NKCC2) and Na+/H+ exchanger isoform 3 (NHE3) are reduced at baseline in SMKO mice, accompanied by attenuated natriuretic and diuretic responses to furosemide. During ANG II hypertension, we found widespread remodeling of transporter expression in wild-type mice with significant increases in the levels of total NaCl cotransporter, phosphorylated NaCl cotransporter (Ser71), and phosphorylated NKCC2, along with the cleaved, activated forms of the α- and γ-epithelial Na+ channel. However, the increases in α- and γ-epithelial Na+ channel with ANG II were substantially attenuated in SMKO mice. This was accompanied by a reduced natriuretic response to amiloride. Thus, enhanced urinary Na+ excretion observed after cell-specific deletion of AT1 receptors from smooth muscle cells is associated with altered Na+ transporter abundance across epithelia in multiple nephron segments. These findings suggest a system of vascular-epithelial in the kidney, modulating the expression of Na+ transporters and contributing to the regulation of pressure natriuresis.NEW & NOTEWORTHY The use of drugs to block the renin-angiotensin system to reduce blood pressure is common. However, the precise mechanism for how these medications control blood pressure is incompletely understood. Here, we show that mice lacking angiotensin receptors specifically in smooth muscle cells lead to alternation in tubular transporter amount and function. Thus, demonstrating the importance of vascular-tubular cross talk in the control of blood pressure.


Asunto(s)
Angiotensina II/farmacología , Células Epiteliales/metabolismo , Riñón/irrigación sanguínea , Miocitos del Músculo Liso/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Amilorida/farmacología , Animales , Bloqueadores del Canal de Sodio Epitelial/farmacología , Femenino , Furosemida/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes , Hipertensión/inducido químicamente , Proteínas Luminiscentes , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Receptor de Angiotensina Tipo 1/genética , Sodio/metabolismo , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Proteína Fluorescente Roja
7.
Am J Physiol Cell Physiol ; 319(4): C757-C770, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32845718

RESUMEN

Extracellular fluid (ECF) potassium concentration ([K+]) is maintained by adaptations of kidney and skeletal muscle, responses heretofore studied separately. We aimed to determine how these organ systems work in concert to preserve ECF [K+] in male C57BL/6J mice fed a K+-deficient diet (0K) versus 1% K+ diet (1K) for 10 days (n = 5-6/group). During 0K feeding, plasma [K+] fell from 4.5 to 2 mM; hindlimb muscle (gastrocnemius and soleus) lost 28 mM K+ (from 115 ± 2 to 87 ± 2 mM) and gained 27 mM Na+ (from 27 ± 0.4 to 54 ± 2 mM). Doubling of muscle tissue [Na+] was not associated with inflammation, cytokine production or hypertension as reported by others. Muscle transporter adaptations in 0K- versus 1K-fed mice, assessed by immunoblot, included decreased sodium pump α2-ß2 subunits, decreased K+-Cl- cotransporter isoform 3, and increased phosphorylated (p) Na+,K+,2Cl- cotransporter isoform 1 (NKCC1p), Ste20/SPS-1-related proline-alanine rich kinase (SPAKp), and oxidative stress-responsive kinase 1 (OSR1p) consistent with intracellular fluid (ICF) K+ loss and Na+ gain. Renal transporters' adaptations, effecting a 98% reduction in K+ excretion, included two- to threefold increased phosphorylated Na+-Cl- cotransporter (NCCp), SPAKp, and OSR1p abundance, limiting Na+ delivery to epithelial Na+ channels where Na+ reabsorption drives K+ secretion; and renal K sensor Kir 4.1 abundance fell 25%. Mass balance estimations indicate that over 10 days of 0K feeding, mice lose ~48 µmol K+ into the urine and muscle shifts ~47 µmol K+ from ICF to ECF, illustrating the importance of the concerted responses during K+ deficiency.


Asunto(s)
Adaptación Fisiológica/genética , Hipertensión/genética , Riñón/metabolismo , Potasio/metabolismo , Animales , Presión Sanguínea/genética , Canales Epiteliales de Sodio/genética , Líquido Extracelular/metabolismo , Humanos , Hipertensión/patología , Riñón/patología , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Fosforilación/genética , Canales de Potasio de Rectificación Interna/genética , Proteínas Serina-Treonina Quinasas/genética , Simportadores de Cloruro de Sodio-Potasio/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Simportadores/genética , Factores de Transcripción/genética , Cotransportadores de K Cl
8.
J Am Soc Nephrol ; 31(4): 783-798, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32086277

RESUMEN

BACKGROUND: Increased nerve activity causes hypertension and kidney disease. Recent studies suggest that renal denervation reduces BP in patients with hypertension. Renal NE release is regulated by prejunctional α2A-adrenoceptors on sympathetic nerves, and α2A-adrenoceptors act as autoreceptors by binding endogenous NE to inhibit its own release. However, the role of α2A-adrenoceptors in the pathogenesis of hypertensive kidney disease is unknown. METHODS: We investigated effects of α2A-adrenoceptor-regulated renal NE release on the development of angiotensin II-dependent hypertension and kidney disease. In uninephrectomized wild-type and α2A-adrenoceptor-knockout mice, we induced hypertensive kidney disease by infusing AngII for 28 days. RESULTS: Urinary NE excretion and BP did not differ between normotensive α2A-adrenoceptor-knockout mice and wild-type mice at baseline. However, NE excretion increased during AngII treatment, with the knockout mice displaying NE levels that were significantly higher than those of wild-type mice. Accordingly, the α2A-adrenoceptor-knockout mice exhibited a systolic BP increase, which was about 40 mm Hg higher than that found in wild-type mice, and more extensive kidney damage. In isolated kidneys, AngII-enhanced renal nerve stimulation induced NE release and pressor responses to a greater extent in kidneys from α2A-adrenoceptor-knockout mice. Activation of specific sodium transporters accompanied the exaggerated hypertensive BP response in α2A-adrenoceptor-deficient kidneys. These effects depend on renal nerves, as demonstrated by reduced severity of AngII-mediated hypertension and improved kidney function observed in α2A-adrenoceptor-knockout mice after renal denervation. CONCLUSIONS: Our findings reveal a protective role of prejunctional inhibitory α2A-adrenoceptors in pathophysiologic conditions with an activated renin-angiotensin system, such as hypertensive kidney disease, and support the concept of sympatholytic therapy as a treatment.


Asunto(s)
Hipertensión Renal/etiología , Hipertensión Renal/prevención & control , Nefritis/etiología , Nefritis/prevención & control , Receptores Adrenérgicos alfa 2/fisiología , Sistema Nervioso Simpático/fisiopatología , Transmisión Sináptica/fisiología , Angiotensina II , Animales , Modelos Animales de Enfermedad , Hipertensión Renal/fisiopatología , Ratones , Ratones Noqueados , Nefritis/fisiopatología , Simpatectomía
9.
Acta Physiol (Oxf) ; 229(1): e13448, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31994810

RESUMEN

AIM: Sexual dimorphisms are evident along the nephron: Females (F) exhibit higher ratios of renal distal to proximal Na+ transporters' abundance, greater lithium clearance (CLi ) more rapid natriuresis in response to saline infusion and lower plasma [K+ ] vs. males (M). During angiotensin II infusion hypertension (AngII-HTN) M exhibit distal Na+ transporter activation, lower proximal and medullary loop transporters, blunted natriuresis in response to saline load, and reduced plasma [K+ ]. This study aimed to determine whether responses of F to AngII-HTN mimicked those in M or were impacted by sexual dimorphisms evident at baseline. METHODS: Sprague Dawley rats and C57BL/6 mice were AngII infused via osmotic minipumps 2 and 3 weeks, respectively, and assessed by metabolic cage collections, tail-cuff sphygmomanometer, semi-quantitative immunoblotting of kidney and patch-clamp electrophysiology. RESULTS: In F rats, AngII-infusion increased BP to 190 mm Hg, increased phosphorylation of cortical NKCC2, NCC and cleavage of ENaC two to threefold, increased ENaC channel activity threefold and aldosterone 10-fold. K+ excretion increased and plasma [K+ ] decreased. Evidence of natriuresis in F included increased urine Na+ excretion and CLi , and decreased medullary NHE3, NKCC2 and Na,K-ATPase abundance. In C57BL/6 mice, AngII-HTN increased abundance of distal Na+ transporters, suppressed proximal-medullary transporters and reduced plasma [K+ ] in both F and M. CONCLUSION: Despite baseline sexual dimorphisms, AngII-HTN provokes similar increases in BP, aldosterone, distal transporters, ENaC channel activation and K+ loss accompanied by similar suppression of proximal and loop Na+ transporters, natriuresis and diuresis in females and males.


Asunto(s)
Angiotensina II/farmacología , Electrólitos/metabolismo , Hipertensión/metabolismo , Canales Iónicos/metabolismo , Animales , Canales Epiteliales de Sodio/metabolismo , Femenino , Hipertensión/inducido químicamente , Transporte Iónico , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Potasio/metabolismo , Ratas , Ratas Sprague-Dawley , Sodio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
10.
J Am Soc Nephrol ; 28(12): 3504-3517, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28774999

RESUMEN

Compared with males, females have lower BP before age 60, blunted hypertensive response to angiotensin II, and a leftward shift in pressure natriuresis. This study tested the concept that this female advantage associates with a distinct sexual dimorphic pattern of transporters along the nephron. We applied quantitative immunoblotting to generate profiles of transporters, channels, claudins, and selected regulators in both sexes and assessed the physiologic consequences of the differences. In rats, females excreted a saline load more rapidly than males did. Compared with the proximal tubule of males, the proximal tubule of females had greater phosphorylation of Na+/H+ exchanger isoform 3 (NHE3), distribution of NHE3 at the base of the microvilli, and less abundant expression of Na+/Pi cotransporter 2, claudin-2, and aquaporin 1. These changes associated with less bicarbonate reabsorption and higher lithium clearance in females. The distal nephrons of females had a higher abundance of total and phosphorylated Na+/Cl- cotransporter (NCC), claudin-7, and cleaved forms of epithelial Na+ channel (ENaC) α and γ subunits, which associated with a lower baseline plasma K+ concentration. A K+-rich meal increased the urinary K+ concentration and decreased the level of renal phosphorylated NCC in females. Notably, we observed similar abundance profiles in female versus male C57BL/6 mice. These results define sexual dimorphic phenotypes along the nephron and suggest that lower proximal reabsorption in female rats expedites excretion of a saline load and enhances NCC and ENaC abundance and activation, which may facilitate K+ secretion and set plasma K+ at a lower level.


Asunto(s)
Electrólitos/metabolismo , Túbulos Renales/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Caracteres Sexuales , Animales , Transporte Biológico , Presión Sanguínea , Femenino , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Microvellosidades/metabolismo , Nefronas/metabolismo , Fosforilación , Potasio/metabolismo , Ratas , Ratas Sprague-Dawley , Sodio/metabolismo
11.
Am J Physiol Endocrinol Metab ; 312(4): E348-E356, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28174181

RESUMEN

The World Health Organization ranks hypertension the leading global risk factor for disease, specifically, cardiovascular disease. Blood pressure (BP) is higher in Westernized populations consuming Na+-rich processed foods than in isolated societies consuming K+-rich natural foods. Evidence suggests that lowering dietary Na+ is particularly beneficial in hypertensive individuals who consume a high-Na+ diet. Nonetheless, numerous population studies demonstrate a relationship between higher dietary K+, estimated from urinary excretion or dietary recall, and lower BP, regardless of Na+ intake. Interventional studies with K+ supplementation suggest that it provides a direct benefit; K+ may also be a marker for other beneficial components of a "natural" diet. Recent studies in rodent models indicate mechanisms for the K+ benefit: the distal tubule Na+-Cl- cotransporter (NCC) controls Na+ delivery downstream to the collecting duct, where Na+ reabsorbed by epithelial Na+ channels drives K+ secretion and excretion through K+ channels in the same region. High dietary K+ provokes a decrease in NCC activity to drive more K+ secretion (and Na+ excretion, analogous to the actions of a thiazide diuretic) whether Na+ intake is high or low; low dietary K+ provokes an increase in NCC activity and Na+ retention, also independent of dietary Na+ Together, the findings suggest that public health efforts directed toward increasing consumption of K+-rich natural foods would reduce BP and, thus, cardiovascular and kidney disease.


Asunto(s)
Presión Sanguínea/fisiología , Enfermedades Cardiovasculares/prevención & control , Riñón/fisiopatología , Potasio en la Dieta , Sodio en la Dieta , Animales , Enfermedades Cardiovasculares/fisiopatología , Humanos
12.
Hypertension ; 68(4): 904-12, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27600183

RESUMEN

Angiotensin II (AngII) hypertension increases distal tubule Na-Cl cotransporter (NCC) abundance and phosphorylation (NCCp), as well as epithelial Na(+) channel abundance and activating cleavage. Acutely raising plasma [K(+)] by infusion or ingestion provokes a rapid decrease in NCCp that drives a compensatory kaliuresis. The first aim tested whether acutely raising plasma [K(+)] with a single 3-hour 2% potassium meal would lower NCCp in Sprague-Dawley rats after 14 days of AngII (400 ng/kg per minute). The potassium-rich meal neither decreased NCCp nor increased K(+) excretion. AngII-infused rats exhibited lower plasma [K(+)] versus controls (3.6±0.2 versus 4.5±0.1 mmol/L; P<0.05), suggesting that AngII-mediated epithelial Na(+) channel activation provokes K(+) depletion. The second aim tested whether doubling dietary potassium intake from 1% (A1K) to 2% (A2K) would prevent K(+) depletion during AngII infusion and, thus, prevent NCC accumulation. A2K-fed rats exhibited normal plasma [K(+)] and 2-fold higher K(+) excretion and plasma [aldosterone] versus A1K. In A1K rats, NCC, NCCpS71, and NCCpT53 abundance increased 1.5- to 3-fold versus controls (P<0.05). The rise in NCC and NCCp abundance was prevented in the A2K rats, yet blood pressure did not significantly decrease. Epithelial Na(+) channel subunit abundance and cleavage increased 1.5- to 3-fold in both A1K and A2K; ROMK (renal outer medulla K(+) channel abundance) abundance was unaffected by AngII or dietary K(+) In summary, the accumulation and phosphorylation of NCC seen during chronic AngII infusion hypertension is likely secondary to potassium deficiency driven by epithelial Na(+) channel stimulation.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Hipertensión/fisiopatología , Potasio en la Dieta/farmacología , Simportadores del Cloruro de Sodio/metabolismo , Equilibrio Hidroelectrolítico/fisiología , Angiotensina II/farmacología , Animales , Modelos Animales de Enfermedad , Hipertensión/inducido químicamente , Infusiones Intravenosas , Pruebas de Función Renal , Masculino , Análisis Multivariante , Fosforilación , Deficiencia de Potasio/prevención & control , Potasio en la Dieta/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Simportadores del Cloruro de Sodio/efectos de los fármacos , Intercambiadores de Sodio-Hidrógeno/metabolismo
13.
Physiol Rep ; 3(9)2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26347505

RESUMEN

In Sprague Dawley rats, 2-week angiotensin II (AngII) infusion increases Na(+) transporter abundance and activation from cortical thick ascending loop of Henle (TALH) to medullary collecting duct (CD) and raises blood pressure associated with a pressure natriuresis, accompanied by depressed Na(+) transporter abundance and activation from proximal tubule (PT) through medullary TALH. This study tests the hypothesis that early during AngII infusion, before blood pressure raises, Na(+) transporters' abundance and activation increase all along the nephron. Male Sprague Dawley rats were infused via osmotic minipumps with a subpressor dose of AngII (200 ng/kg/min) or vehicle for 3 days. Overnight urine was collected in metabolic cages and sodium transporters' abundance and phosphorylation were determined by immunoblotting homogenates of renal cortex and medulla. There were no significant differences in body weight gain, overnight urine volume, urinary Na(+) and K(+) excretion, or rate of excretion of a saline challenge between AngII and vehicle infused rats. The 3-day nonpressor AngII infusion significantly increased the abundance of PT Na(+)/H(+) exchanger 3 (NHE3), cortical TALH Na-K-2Cl cotransporter 2 (NKCC2), distal convoluted tubule (DCT) Na-Cl cotransporter (NCC), and cortical CD ENaC subunits. Additionally, phosphorylation of cortical NKCC2, NCC, and STE20/SPS1-related proline-alanine-rich kinase (SPAK) were increased; medullary NKCC2 and SPAK were not altered. In conclusion, 3-day AngII infusion provokes PT NHE3 accumulation as well as NKCC2, NCC, and SPAK accumulation and activation in a prehypertensive phase before evidence for intrarenal angiotensinogen accumulation.

14.
Am J Physiol Cell Physiol ; 309(8): C541-50, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26246427

RESUMEN

Physiological concentrations of angiotensin II (ANG II) upregulate the activity of Na(+)/H(+) exchanger isoform 3 (NHE3) in the renal proximal tubule through activation of the ANG II type I (AT1) receptor/G protein-coupled signaling. This effect is key for maintenance of extracellular fluid volume homeostasis and blood pressure. Recent findings have shown that selective activation of the beta-arrestin-biased AT1 receptor signaling pathway induces diuresis and natriuresis independent of G protein-mediated signaling. This study tested the hypothesis that activation of this AT1 receptor/beta-arrestin signaling inhibits NHE3 activity in proximal tubule. To this end, we determined the effects of the compound TRV120023, which binds to the AT1R, blocks G-protein coupling, and stimulates beta-arrestin signaling on NHE3 function in vivo and in vitro. NHE3 activity was measured in both native proximal tubules, by stationary microperfusion, and in opossum proximal tubule (OKP) cells, by Na(+)-dependent intracellular pH recovery. We found that 10(-7) M TRV120023 remarkably inhibited proximal tubule NHE3 activity both in vivo and in vitro. Additionally, stimulation of NHE3 by ANG II was completely suppressed by TRV120023 both in vivo as well as in vitro. Inhibition of NHE3 activity by TRV120023 was associated with a decrease in NHE3 surface expression in OKP cells and with a redistribution from the body to the base of the microvilli in the rat proximal tubule. These findings indicate that biased signaling of the beta-arrestin pathway through the AT1 receptor inhibits NHE3 activity in the proximal tubule at least in part due to changes in NHE3 subcellular localization.


Asunto(s)
Arrestinas/metabolismo , Túbulos Renales Proximales/citología , Receptor de Angiotensina Tipo 1/fisiología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Humanos , Concentración de Iones de Hidrógeno , Túbulos Renales Proximales/fisiología , Masculino , Oligopéptidos/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Sodio/metabolismo , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/genética , beta-Arrestinas
15.
Physiol Rep ; 2(12)2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25472608

RESUMEN

Na,K-ATPase generates the driving force for sodium reabsorption in the kidney. Na,K-ATPase functional properties are regulated by small proteins belonging to the FXYD family. In kidney FXYD2 is the most abundant: it is an inhibitory subunit expressed in almost every nephron segment. Its absence should increase sodium pump activity and promote Na(+) retention, however, no obvious renal phenotype was detected in mice with global deletion of FXYD2 (Arystarkhova et al. 2013). Here, increased total cortical Na,K-ATPase activity was documented in the Fxyd2(-/-) mouse, without increased α1ß1 subunit expression. We tested the hypothesis that adaptations occur in distal convoluted tubule (DCT), a major site of sodium adjustments. Na,K-ATPase immunoreactivity in DCT was unchanged, and there was no DCT hypoplasia. There was a marked activation of thiazide-sensitive sodium chloride cotransporter (NCC; Slc12a3) in DCT, predicted to increase Na(+) reabsorption in this segment. Specifically, NCC total increased 30% and NCC phosphorylated at T53 and S71, associated with activation, increased 4-6 fold. The phosphorylation of the closely related thick ascending limb (TAL) apical NKCC2 (Slc12a1) increased at least twofold. Abundance of the total and cleaved (activated) forms of ENaC α-subunit was not different between genotypes. Nonetheless, no elevation of blood pressure was evident despite the fact that NCC and NKCC2 are in states permissive for Na(+) retention. Activation of NCC and NKCC2 may reflect an intracellular linkage to elevated Na,K-ATPase activity or a compensatory response to Na(+) loss proximal to the TAL and DCT.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...