Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Am J Physiol Renal Physiol ; 325(2): F224-F234, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37318989

RESUMO

The impact of chronic dietary K+ loading on proximal tubule (PT) function was measured using free-flow micropuncture along with measurements of overall kidney function, including urine volume, glomerular filtration rate, and absolute and fractional Na+ and K+ excretion in the rat. Feeding animals a diet with 5% KCl [high K+ (HK)] for 7 days reduced glomerular filtration rate by 29%, increased urine volume by 77%, and increased absolute K+ excretion by 202% compared with rats on a 1% KCl [control K+ (CK)] diet. HK did not change absolute Na+ excretion but significantly increased fraction excretion of Na+ (1.40% vs. 0.64%), indicating that fractional Na+ absorption is reduced by HK. PT reabsorption was assessed using free-flow micropuncture in anesthetized animals. At 80% of the accessible length of the PT, measurements of inulin concentration indicated volume reabsorption of 73% and 54% in CK and HK, respectively. At the same site, fractional PT Na+ reabsorption was 66% in CK animals and 37% in HK animals. Fractional PT K+ reabsorption was 66% in CK and 37% in HK. To assess the role of Na+/H+ exchanger isoform 3 (NHE3) in mediating these changes, we measured NHE3 protein expression in total kidney microsomes as well as surface membranes using Western blots. We found no significant changes in protein in either cell fraction. Expression of the Ser552 phosphorylated form of NHE3 was also similar in CK and HK animals. Reduction in PT transport may facilitate K+ excretion and help balance Na+ excretion by shifting Na+ reabsorption from K+-reabsorbing to K+-secreting nephron segments.NEW & NOTEWORTHY In rats fed a diet rich in K+, proximal tubules reabsorbed less fluid, Na+, and K+ compared with those in animals on a control diet. Glomerular filtration rates also decreased, probably due to glomerulotubular feedback. These reductions may help to maintain balance of the two ions simultaneously by shifting Na+ reabsorption to K+-secreting nephron segments.


Assuntos
Túbulos Renais Proximais , Néfrons , Ratos , Animais , Trocador 3 de Sódio-Hidrogênio/metabolismo , Túbulos Renais Proximais/metabolismo , Néfrons/metabolismo , Rim/metabolismo , Sódio/metabolismo , Taxa de Filtração Glomerular
2.
Am J Physiol Renal Physiol ; 322(2): F225-F244, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35001663

RESUMO

The renal response to acute hyperkalemia is mediated by increased K+ secretion within the connecting tubule (CNT), flux that is modulated by tubular effects (e.g., aldosterone) in conjunction with increased luminal flow. There is ample evidence that peritubular K+ blunts Na+ reabsorption in the proximal tubule, thick ascending Henle limb, and distal convoluted tubule (DCT). Although any such reduction may augment CNT delivery, the relative contribution of each is uncertain. The kidney model of this laboratory was recently advanced with representation of the cortical labyrinth and medullary ray. Model tubules capture the impact of hyperkalemia to blunt Na+ reabsorption within each upstream segment. However, this forces the question of the extent to which increased Na+ delivery is transmitted past the macula densa and its tubuloglomerular feedback (TGF) signal. Beyond increasing macula densa Na+ delivery, peritubular K+ is predicted to raise cytosolic Cl- and depolarize macula densa cells, which may also activate TGF. Thus, although the upstream reduction in Na+ transport may be larger, it appears that the DCT effect is critical to increasing CNT delivery. Beyond the flow effect, hyperkalemia reduces ammoniagenesis and reduced ammoniagenesis enhances K+ excretion. What this model provides is a possible mechanism. When cortical [Formula: see text] is taken up via peritubular Na+-K+([Formula: see text])-ATPase, it acidifies principal cells. Consequently, reduced ammoniagenesis increases principal cell pH, thereby increasing conductance of both the epithelial Na+ channel and renal outer medullary K+ channel, enhancing K+ excretion. In this model, the effect of aldosterone on principal cells, diminished DCT Na+ reabsorption, and reduced ammoniagenesis all provide relatively equal and additive contributions to renal K+ excretion.NEW & NOTEWORTHY Hyperkalemia blunts Na+ reabsorption along the nephron, and increased CNT Na+ delivery facilitates K+ secretion. The model suggests that tubuloglomerular feedback limits transmission of proximal effects past the macula densa, so that it is DCT transport that is critical. Hyperkalemia also reduces PCT ammoniagenesis, which enhances K+ excretion. The model suggests a mechanism, namely, that reduced cortical ammonia impacts CNT transport by raising cell pH and thus increasing both ENaC and ROMK conductance.


Assuntos
Amônia/metabolismo , Hiperpotassemia/metabolismo , Rim/metabolismo , Modelos Biológicos , Potássio/sangue , Eliminação Renal , Reabsorção Renal , Sódio/metabolismo , Animais , Canais Epiteliais de Sódio/metabolismo , Retroalimentação Fisiológica , Concentração de Íons de Hidrogênio , Hiperpotassemia/sangue , Hiperpotassemia/fisiopatologia , Rim/fisiopatologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ratos
3.
Am J Physiol Renal Physiol ; 323(6): F642-F653, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36108052

RESUMO

The water channel aquaporin-1 (AQP1) is the principal water pathway for isotonic water reabsorption in the kidney proximal tubule (PT). We investigated flow-mediated fluid (Jv) and [Formula: see text] ([Formula: see text]) reabsorption in PTs of the mouse kidney by microperfusion in wild-type (WT) and AQP1 knockout (KO) mice. Experiments were simulated in an adaptation of a mathematical model of the rat PT. An increase in perfusion rate from 5 to 20 nL/min increased Jv and [Formula: see text] in PTs of WT mice. AQP1 KO mice significantly decreased Jv at low and high flow rates compared with control. In contrast, [Formula: see text] was not reduced at either low or high flow rates. Cell volume showed no significant difference between WT and AQP1 KO mice. Renal clearance experiments showed significantly higher urine flow in AQP1 KO mice, but there was no significant difference in either Na+ and K+ or [Formula: see text] excretion. Acid-base parameters of blood pH, Pco2, [Formula: see text], and urine pH were the same in both WT and KO mice. In model calculations, tubules whose tight junction (TJ) water permeability (Pf) was that assigned to the rat TJ, showed no difference in Jv between WT and KO, whereas TJ Pf set to 25% of the rat predicted Jv concordant with our observations from AQP1 KO. These results affirm the dominance of AQP1 in mediating isotonic water reabsorption by the mouse PT and demonstrate that flow-stimulated [Formula: see text] reabsorption is intact and independent of AQP1. With reference to the model, the findings also suggest that TJ water flux in the PT is less prominent in the mouse than in the rat kidney.NEW & NOTEWORTHY We found an absence of flow-dependent modulation of fluid absorption but no effect on either proximal tubule (PT) [Formula: see text] absorption or acid-base parameters in the aquaporin 1 (AQP1) knockout mouse. We affirmed the dominance of the water channel AQP1 in mediating isotonic water reabsorption by the mouse PT and demonstrated that flow-stimulated [Formula: see text] reabsorption is independent of AQP1. With reference to the model, the findings also suggest that tight junctional water flux in the PT is less prominent in the mouse than rat kidney.


Assuntos
Aquaporina 1 , Túbulos Renais Proximais , Camundongos , Ratos , Animais , Aquaporina 1/genética , Aquaporina 1/metabolismo , Túbulos Renais Proximais/metabolismo , Camundongos Knockout , Tamanho Celular , Água/metabolismo
4.
J Cell Sci ; 133(2)2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31843759

RESUMO

The fast turnover of membrane components through endocytosis and recycling allows precise control of the composition of the plasma membrane. Endocytic recycling can be rapid, with some molecules returning to the plasma membrane with a half time <5 min. Existing methods to study these trafficking pathways utilize chemical, radioactive or fluorescent labeling of cell surface receptors in pulse-chase experiments, which require tedious washing steps and manual collection of samples. Here, we introduce a live-cell endocytic recycling assay based on a newly designed cell-impermeable fluorogenic ligand for HaloTag, Janelia Fluor 635i (JF635i, where i indicates impermeant), which allows real-time detection of membrane receptor recycling at steady state. We used this method to study the effect of iron depletion on transferrin receptor (TfR) recycling using the chelator desferrioxamine. We found that this perturbation significantly increases the TfR recycling rate. The high temporal resolution and simplicity of this assay provides a clear advantage over extant methods and makes it ideal for large scale cellular imaging studies. This assay can be adapted to examine other cellular kinetic parameters such as protein turnover and biosynthetic trafficking.


Assuntos
Endocitose/genética , Humanos , Cinética , Transporte Proteico
5.
Pflugers Arch ; 473(11): 1749-1760, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34455480

RESUMO

We compared the regulation of the NaCl cotransporter (NCC) in adaptation to a low-K (LK) diet in male and female mice. We measured hydrochlorothiazide (HCTZ)-induced changes in urine volume (UV), glomerular filtration rate (GFR), absolute (ENa, EK), and fractional (FENa, FEK) excretion in male and female mice on control-K (CK, 1% KCl) and LK (0.1% KCl) diets for 7 days. With CK, NCC-dependent ENa and FENa were larger in females than males as observed previously. However, with LK, HCTZ-induced ENa and FENa increased in males but not in females, abolishing the sex differences in NCC function as observed in CK group. Despite large diuretic and natriuretic responses to HCTZ, EK was only slightly increased in response to the drug when animals were on LK. This suggests that the K-secretory apparatus in the distal nephron is strongly suppressed under these conditions. We also examined LK-induced changes in Na transport protein expression by Western blotting. Under CK conditions females expressed more NCC protein, as previously reported. LK doubled both total (tNCC) and phosphorylated NCC (pNCC) abundance in males but had more modest effects in females. The larger effect in males abolished the sex-dependence of NCC expression, consistent with the measurements of function by renal clearance. LK intake did not change NHE3, NHE2, or NKCC2 expression, but reduced the amount of the cleaved (presumably active) form of γENaC. LK reduced plasma K to lower levels in females than males. These results indicated that males had a stronger NCC-mediated adaptation to LK intake than females.


Assuntos
Cátions/metabolismo , Transporte de Íons/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Tiazidas/farmacologia , Animais , Diuréticos/farmacologia , Feminino , Taxa de Filtração Glomerular/efeitos dos fármacos , Transporte de Íons/efeitos dos fármacos , Túbulos Renais Distais/efeitos dos fármacos , Túbulos Renais Distais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Néfrons/efeitos dos fármacos , Néfrons/metabolismo , Caracteres Sexuais , Sódio/metabolismo , Membro 3 da Família 12 de Carreador de Soluto/metabolismo
6.
Am J Physiol Renal Physiol ; 320(6): F1059-F1079, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33779315

RESUMO

Ammonia generated within the kidney is partitioned into a urinary fraction (the key buffer for net acid excretion) and an aliquot delivered to the systemic circulation. The physiology of this partitioning has yet to be examined in a kidney model, and that was undertaken in this work. This involves explicit representation of the cortical labyrinth, so that cortical interstitial solute concentrations are computed rather than assigned. A detailed representation of cortical vasculature has been avoided by making the assumption that solute concentrations within the interstitium and peritubular capillaries are likely to be identical and that there is little to no modification of venous composition as blood flows to the renal vein. The model medullary ray has also been revised to include a segment of proximal straight tubule, which supplies ammonia to this region. The principal finding of this work is that cortical labyrinth interstitial ammonia concentration is likely to be several fold higher than systemic arterial ammonia. This elevation of interstitial ammonia enhances ammonia secretion in both the proximal convoluted tubule and distal convoluted tubule, with uptake by Na+-K+-ATPases of both segments. Model prediction of urinary ammonia excretion was concordant with measured values, but at the expense of greater ammoniagenesis, with high rates of renal venous ammonia flux. This derives from a limited capability of the model medulla to replicate the high interstitial ammonia concentrations that are required to drive collecting duct ammonia secretion. Thus, renal medullary ammonia trapping appears key to diverting ammonia from the renal vein to urine, but capturing the underlying physiology remains a challenge.NEW & NOTEWORTHY This is the first mathematical model to estimate solute concentrations within the kidney cortex. The model predicts cortical ammonia to be several fold greater than in the systemic circulation. This higher concentration drives ammonia secretion in proximal and distal tubules. The model reveals a gap in our understanding of how ammonia generated within the cortex is channeled efficiently into the final urine.


Assuntos
Amônia/metabolismo , Rim/fisiologia , Modelos Biológicos , Amônia/urina , Animais , Transporte Biológico , Rim/irrigação sanguínea , Ratos
7.
Am J Physiol Renal Physiol ; 318(4): F936-F955, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32088967

RESUMO

Kidney water conservation requires a hypertonic medullary interstitium, NaCl in the outer medulla and NaCl and urea in the inner medulla, plus a vascular configuration that protects against washout. In this work, a multisolute model of the rat kidney is revisited to examine its capacity to simulate antidiuresis. The first step was to streamline model computation by parallelizing its Jacobian calculation, thus allowing finer medullary spatial resolution and more extensive examination of model parameters. It is found that outer medullary NaCl is modestly increased when transporter density in ascending Henle limbs from juxtamedullary nephrons is scaled to match the greater juxtamedullary solute flow. However, higher NaCl transport produces greater CO2 generation and, by virtue of countercurrent vascular flows, establishment of high medullary Pco2. This CO2 gradient can be mitigated by assuming that a fraction of medullary transport is powered anaerobically. Reducing vascular flows or increasing vessel permeabilities does little to further increase outer medullary solute gradients. In contrast to medullary models of others, vessels in this model have solute reflection coefficients close to zero; increasing these coefficients provides little enhancement of solute profiles but does generate high interstitial pressures, which distort tubule architecture. Increasing medullary urea delivery via entering vasa recta increases inner medullary urea, although not nearly to levels found in rats. In summary, 1) medullary Na+ and urea gradients are not captured by the model and 2) the countercurrent architecture that provides antidiuresis also produces exaggerated Pco2 profiles and is an unappreciated constraint on models of medullary function.


Assuntos
Simulação por Computador , Rim/irrigação sanguínea , Rim/metabolismo , Modelos Biológicos , Natriurese , Circulação Renal , Reabsorção Renal , Sódio/urina , Animais , Dióxido de Carbono/metabolismo , Pressão Hidrostática , Oxigênio/metabolismo , Permeabilidade , Potássio/urina , Ratos , Ureia/metabolismo
8.
Am J Physiol Renal Physiol ; 317(4): F967-F977, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31390232

RESUMO

We studied sex differences in response to high K+ (HK) intake on thiazide-sensitive cation (Na+ and K+) excretion in wild-type (WT) and ANG II receptor subtype 1a (AT1aR) knockout (KO) mice. Renal clearance experiments were performed to examine Na+-Cl- cotransporter (NCC) activity on mice fed with control and HK (5% KCl, 7 days) diets. Hydrochlorothiazide (HCTZ)-induced changes in urine volume, glomerular filtration rate, absolute Na+ and K+ excretion, and fractional excretion were compared. HK-induced changes in NCC, Na+/H+ exchanger isoform 3 (NHE3), and ENaC expression were examined by Western blot analysis. In WT animals under the control diet, HCTZ-induced cation excretion was greater in female animals, reflecting larger increases in Na+ excretion, since there was little sex difference in HCTZ-induced K+ excretion. Under the HK diet, the sex difference in HCTZ-induced cation excretion was reduced because of larger increments in K+ excretion in male animals. The fraction of K+ excretion was 57 ± 5% in male WT animals and 36 ± 4% in female WT animals (P < 0.05), but this difference was absent in AT1aR KO mice. NCC abundance was higher in female animals than in male animals but decreased by similar fractions on HK diet. NHE3 abundance decreased, whereas cleaved forms of γ-ENaC increased, with HK in all groups; these changes were similar in male and female animals and were not significantly affected by AT1aR ablation. These results indicate that, with the HK diet, male animals display greater distal Na+ delivery and greater activation of K+ secretion mechanisms, all suggesting a more powerful male adaptation to HK intake.


Assuntos
Cátions/urina , Diuréticos/farmacologia , Complexo II de Transporte de Elétrons/metabolismo , Hidroclorotiazida/farmacologia , Rim/metabolismo , Potássio/farmacologia , Animais , Feminino , Taxa de Filtração Glomerular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Potássio/urina , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Caracteres Sexuais , Trocador 3 de Sódio-Hidrogênio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Urodinâmica
9.
Am J Physiol Renal Physiol ; 315(4): F1032-F1041, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29923764

RESUMO

Changes in the expression of Na transport proteins were measured in the kidneys of mice with increased dietary K intake for 1 wk. The epithelial Na channel (ENaC) was upregulated, with enhanced expression of full-length and cleaved forms of α-ENaC and cleaved γ-ENaC. At the same time, the amount of the NaCl cotransporter NCC and its phosphorylated form decreased by ~50% and ~80%, respectively. The expression of the phosphorylated form of the Na-K-2Cl cotransporter NKCC2 also decreased, despite an increase in overall protein content. The effect was stronger in males (80%) than in females (40%). This implies that less Na+ is reabsorbed in the thick ascending limb of Henle's loop and distal convoluted tubule along with Cl-, whereas more is reabsorbed in the aldosterone-sensitive distal nephron in exchange for secreted K+. The abundance of the proximal tubule Na/H exchanger NHE3 decreased by ~40%, with similar effects in males and females. Time-course studies indicated that NCC and NHE3 proteins decreased progressively over 7 days on a high-K diet. Expression of mRNA encoding these proteins increased, implying that the decreased protein levels resulted from decreased rates of synthesis or increased rates of degradation. The potential importance of changes in NHE3, NKCC2, and NCC in promoting K+ excretion was assessed with a mathematical model. Simulations indicated that decreased NHE3 produced the largest effect. Regulation of proximal tubule Na+ transport may play a significant role in achieving K homeostasis.


Assuntos
Túbulos Renais Proximais/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Néfrons/metabolismo , Sódio/metabolismo , Animais , Transporte Biológico/fisiologia , Proteínas de Transporte/metabolismo , Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Distais/metabolismo , Camundongos , Trocadores de Sódio-Hidrogênio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Membro 3 da Família 12 de Carreador de Soluto/metabolismo
11.
Pflugers Arch ; 469(5-6): 643-654, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28271233

RESUMO

The purpose of this review is to summarize our knowledge and understanding of the physiological importance and the mechanisms underlying flow-activated proximal tubule transport. Since the earliest micropuncture studies of mammalian proximal tubule, it has been recognized that tubular flow is an important regulator of sodium, potassium, and acid-base transport in the kidney. Increased fluid flow stimulates Na+ and HCO3- absorption in the proximal tubule via stimulation of Na/H-exchanger isoform 3 (NHE3) and H+-ATPase. In the proximal tubule, brush border microvilli are the major flow sensors, which experience changes in hydrodynamic drag and bending moment as luminal flow velocity changes and which transmit the force of altered flow to cytoskeletal structures within the cell. The signal to NHE3 depends upon the integrity of the actin cytoskeleton; the signal to the H+-ATPase depends upon microtubules. We have demonstrated that alterations in fluid drag impact tubule function by modulating ion transporter availability within the brush border membrane of the proximal tubule. Beyond that, there is evidence that transporter activity within the peritubular membrane is also modulated by luminal flow. Secondary messengers that regulate the flow-mediated tubule function have also been delineated. Dopamine blunts the responsiveness of proximal tubule transporters to changes in luminal flow velocity, while a DA1 antagonist increases flow sensitivity of solute reabsorption. IP3 receptor-mediated intracellular Ca2+ signaling is critical to transduction of microvillus drag. In this review, we summarize our findings of the regulatory mechanism of flow-mediated Na+ and HCO3- transport in the proximal tubule and review available information about flow sensing and regulatory mechanism of glomerulotubular balance.


Assuntos
Taxa de Filtração Glomerular , Túbulos Renais Proximais/metabolismo , Reabsorção Renal , Animais , Humanos , Túbulos Renais Proximais/fisiologia , Trocador 3 de Sódio-Hidrogênio/genética , Trocador 3 de Sódio-Hidrogênio/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo
12.
Am J Physiol Renal Physiol ; 312(6): F925-F950, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28179254

RESUMO

A model of the rat nephron (Weinstein. Am J Physiol Renal Physiol 308: F1098-F1118, 2015) has been extended with addition of medullary vasculature. Blood vessels contain solutes from the nephron model, plus additional species from the model of Atherton et al. (Am J Physiol Renal Fluid Electrolyte Physiol 247: F61-F72, 1984), representing hemoglobin buffering. In contrast to prior models of the urine-concentrating mechanism, reflection coefficients for DVR are near zero. Model unknowns are initial proximal tubule pressures and flows, connecting tubule pressure, and medullary interstitial pressures and concentrations. The model predicts outer medullary (OM) interstitial gradients for Na+, K+, CO2, and [Formula: see text], such that at OM-IM junction, the respective concentrations relative to plasma are 1.2, 3.0, 2.7, and 8.0; within IM, there is high urea and low [Formula: see text], with concentration ratios of 11 and 0.5 near the papillary tip. Quantitative similarities are noted between K+ and urea handling (medullary delivery and permeabilities). The model K+ gradient is physiologic, and the urea gradient is steeper due to restriction of urea permeability to distal collecting duct. Nevertheless, the predicted urea gradient is less than expected, suggesting reconsideration of proposals of an unrecognized reabsorptive urea flux. When plasma K+ is increased from 5.0 to 5.5 mM, Na+ and K+ excretion increase 2.3- and 1.3-fold, respectively. The natriuresis derives from a 3.3% decrease in proximal Na+ reabsorption and occurs despite delivery-driven increases in Na+ reabsorption in distal segments; kaliuresis derives from a 30% increase in connecting tubule Na+ delivery. Thus this model favors the importance of proximal over distal events in K+-induced diuresis.


Assuntos
Diurese , Capacidade de Concentração Renal , Modelos Biológicos , Néfrons/irrigação sanguínea , Néfrons/metabolismo , Potássio/metabolismo , Sódio/metabolismo , Animais , Bicarbonatos/metabolismo , Dióxido de Carbono/metabolismo , Concentração de Íons de Hidrogênio , Microcirculação , Potássio/sangue , Potássio/urina , Ratos , Circulação Renal , Eliminação Renal , Reabsorção Renal , Sódio/sangue , Sódio/urina , Ureia/metabolismo
13.
Am J Physiol Renal Physiol ; 313(1): F62-F73, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28356292

RESUMO

We assessed effects of acute volume reductions induced by administration of diuretics in rats. Direct block of Na+ transport produced changes in urinary electrolyte excretion. Adaptations to these effects appeared as alterations in the expression of protein for the distal nephron Na+ transporters NCC and ENaC. Two hours after a single injection of furosemide (6 mg/kg) or hydrochlorothiazide (HCTZ; 30 mg/kg) Na+ and K+ excretion increased but no changes in the content of activated forms of NCC (phosphorylated on residue T53) or ENaC (cleaved γ-subunit) were detected. In contrast, amiloride (0.6 mg/kg) evoked a similar natriuresis that coincided with decreased pT53NCC and increased cleaved γENaC. Alterations in posttranslational membrane protein processing correlated with an increase in plasma K+ of 0.6-0.8 mM. Decreased pT53NCC occurred within 1 h after amiloride injection, whereas changes in γENaC were slower and were blocked by the mineralocorticoid receptor antagonist spironolactone. Increased γENaC cleavage correlated with elevation of the surface expression of the subunit as assessed by in situ biotinylation. Na depletion induced by 2 h of furosemide or HCTZ treatment increases total NCC expression without affecting ENaC protein. However, restriction of Na intake for 10 h (during the day) or 18 h (overnight) increased the abundance of both total NCC and of cleaved α- and γENaC. We conclude that the kidneys respond acutely to hyperkalemic challenges by decreasing the activity of NCC while increasing that of ENaC. They respond to hypovolemia more slowly, increasing Na+ reabsorptive capacities of both of these transporters.


Assuntos
Diuréticos/farmacologia , Canais Epiteliais de Sódio/efeitos dos fármacos , Hiperpotassemia/metabolismo , Hipovolemia/metabolismo , Néfrons/efeitos dos fármacos , Potássio/metabolismo , Sódio/metabolismo , Amilorida/farmacologia , Animais , Diuréticos/toxicidade , Canais Epiteliais de Sódio/metabolismo , Feminino , Furosemida/farmacologia , Hidroclorotiazida/farmacologia , Hiperpotassemia/sangue , Hiperpotassemia/induzido quimicamente , Hiperpotassemia/urina , Hipovolemia/sangue , Hipovolemia/induzido quimicamente , Hipovolemia/urina , Masculino , Modelos Biológicos , Néfrons/metabolismo , Fosforilação , Potássio/sangue , Potássio/urina , Ratos Sprague-Dawley , Eliminação Renal/efeitos dos fármacos , Sódio/sangue , Sódio/urina , Membro 3 da Família 12 de Carreador de Soluto/efeitos dos fármacos , Membro 3 da Família 12 de Carreador de Soluto/metabolismo , Espironolactona/farmacologia
14.
Am J Physiol Renal Physiol ; 313(2): F505-F513, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28566500

RESUMO

We studied gender differences in Na+-Cl- cotransporter (NCC) activity and expression in wild-type (WT) and AT1a receptor knockout (KO) mice. In renal clearance experiments, urine volume (UV), glomerular filtration rate, absolute Na+ (ENa) and K+ (EK), and fractional Na+ (FENa) and K+ excretion were measured and compared at peak changes after bolus intravenous injection of hydrochlorothiazide (HCTZ; 30 mg/kg). In WT, females responded more strongly than males to HCTZ, with larger fractional increases of UV (7.8- vs. 3.4-fold), ENa (11.7- vs. 5.7-fold), FENa (7.9- vs. 4.9-fold), and EK (2.8- vs. 1.4-fold). In contrast, there were no gender differences in the responses to the diuretic in KO mice; HCTZ produced greater effects on male KO than on WT but similar effects on females. In WT, total (tNCC) and phosphorylated (pNCC) NCC protein expressions were 1.8- and 4.6-fold higher in females compared with males (P < 0.05), consistent with the larger response to HCTZ. In KO mice, tNCC and pNCC increased significantly in males to levels not different from those in females. There were no gender differences in the expression of the Na+/H+ exchanger (NHE3) in WT; NHE3 protein decreased to similar extents in male and female KO animals, suggesting AT1a-mediated NHE3 expression in proximal tubules. The resulting increase in delivery of NaCl to the distal nephron may underlie increased NCC expression and activity in mice lacking the AT1a receptor.


Assuntos
Angiotensina II/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Caracteres Sexuais , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Diurese , Feminino , Hidroclorotiazida , Rim/metabolismo , Masculino , Camundongos Knockout , Natriurese , Fenótipo , Proteínas Serina-Treonina Quinases/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Receptores de Droga/metabolismo , Simportadores de Cloreto de Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , Membro 3 da Família 12 de Carreador de Soluto/metabolismo
15.
Am J Physiol Renal Physiol ; 308(10): F1076-97, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25694479

RESUMO

Proximal tubule and loop of Henle function are coupled, with proximal transport determining loop fluid composition, and loop transport modulating glomerular filtration via tubuloglomerular feedback (TGF). To examine this interaction, we begin with published models of the superficial rat proximal convoluted tubule (PCT; including flow-dependent transport in a compliant tubule), and the rat thick ascending Henle limb (AHL). Transport parameters for this PCT are scaled down to represent the proximal straight tubule (PST), which is connected to the thick AHL via a short descending limb. Transport parameters for superficial PCT and PST are scaled up for a juxtamedullary nephron, and connected to AHL via outer and inner medullary descending limbs, and inner medullary thin AHL. Medullary interstitial solute concentrations are specified. End-AHL hydrostatic pressure is determined by distal nephron flow resistance, and the TGF signal is represented as a linear function of end-AHL cytosolic Cl concentration. These two distal conditions required iterative solution of the model. Model calculations capture inner medullary countercurrent flux of urea, and also suggest the presence of an outer medullary countercurrent flux of ammonia, with reabsorption in AHL and secretion in PST. For a realistically strong TGF signal, there is the expected homeostatic impact on distal flows, and in addition, a homeostatic effect on proximal tubule pressure. The model glycosuria threshold is compatible with rat data, and predicted glucose excretion with selective 1Na(+):1glucose cotransporter (SGLT2) inhibition comports with observations in the mouse. Model calculations suggest that enhanced proximal tubule Na(+) reabsorption during hyperglycemia is sufficient to activate TGF and contribute to diabetic hyperfiltration.


Assuntos
Túbulos Renais Proximais/fisiologia , Alça do Néfron/fisiologia , Modelos Animais , Modelos Teóricos , Animais , Transporte Biológico/fisiologia , Taxa de Filtração Glomerular/fisiologia , Glucose/metabolismo , Homeostase/fisiologia , Ratos , Sódio/metabolismo
16.
Am J Physiol Renal Physiol ; 308(10): F1098-118, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25694480

RESUMO

Mathematical models of the proximal tubule (PT), loop of Henle (LOH), and distal nephron have been combined to simulate transport by rat renal tubules. The ensemble is composed of 24,000 superficial (SF) nephrons and 12,000 juxtamedullary (JM) nephrons in 5 classes (according to LOH length); all coalesce into 7,200 connecting tubules (CNT). Medullary interstitial solute concentrations are specified. The model equations require that each nephron glomerular filtration rate (GFR) satisfies a tubuloglomerular feedback (TGF) relationship, and each initial hydrostatic pressure yields a common CNT pressure; that common CNT pressure is determined from an overall distal hydraulic resistance to flow. By virtue of the greater GFR for JM nephrons, fluid delivery to SF and JM tubules is comparable. Glucose reabsorption is restricted to the PT, cotransported with one Na in the convoluted tubule (SGLT2), and two Na in the straight tubule (SGLT1). Increasing ambient glucose from 5 to 10 mM increases proximal Na reabsorption and decreases distal delivery. This is mitigated by a TGF-mediated increase in GFR, and may thus be an etiology for TGF-mediated glomerular hyperfiltration. With SGLT2 inhibition by 95%, the model predicts that under normoglycemic conditions about 60% of filtered glucose will still be reabsorbed, so that profound glycosuria is not to be expected. Compared with glucose-driven osmotic diuresis, SGLT2 inhibition provokes greater natriuresis. When hyperglycemia is superimposed on SGLT2 inhibition, the model suggests that natriuresis may be severe, reflecting synergy of a proximal diuretic and osmotic diuresis. In sum, the model captures TGF-mediated diabetic hyperfiltration and predicts glomerular protection with SGLT2 inhibition.


Assuntos
Glucose/metabolismo , Modelos Animais , Modelos Teóricos , Néfrons/fisiologia , Animais , Transporte Biológico/fisiologia , Taxa de Filtração Glomerular/fisiologia , Ratos , Sódio/metabolismo , Transportador 2 de Glucose-Sódio , Inibidores do Transportador 2 de Sódio-Glicose
17.
Am J Physiol Renal Physiol ; 308(8): F839-47, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25651568

RESUMO

In the proximal tubule, axial flow (drag on brush-border microvilli) stimulates Na(+) and HCO3 (-) reabsorption by modulating both Na/H exchanger 3 (NHE3) and H-ATPase activity, a process critical to glomerulotubular balance. We have also demonstrated that blocking the angiotensin II receptor decreases baseline transport, but preserves the flow effect; dopamine leaves baseline fluxes intact, but abrogates the flow effect. In the current work, we provide evidence implicating cytosolic calcium in flow-dependent transport. Mouse proximal tubules were microperfused in vitro at perfusion rates of 5 and 20 nl/min, and reabsorption of fluid (Jv) and HCO3 (-) (JHCO3) were measured. We examined the effect of high luminal Ca(2+) (5 mM), 0 mM Ca(2+), the Ca(2+) chelator BAPTA-AM, the inositol 1,4,5-trisphosphate (IP3) receptor antagonist 2-aminoethoxydiphenyl borate (2-APB), and the Ca-ATPase inhibitor thapsigargin. In control tubules, increasing perfusion rate from 5 to 20 nl/min increased Jv by 62% and JHCO3 by 104%. With respect to Na(+) reabsorption, high luminal Ca(2+) decreased transport at low flow, but preserved the flow-induced increase; low luminal Ca(2+) had little impact; both BAPTA and 2-APB had no effect on baseline flux, but abrogated the flow effect; thapsigargin decreased baseline flow, leaving the flow effect intact. With respect to HCO3 (-) reabsorption, high luminal Ca(2+) decreased transport at low flow and mildly diminished the flow-induced increase; low luminal Ca(2+) had little impact; both BAPTA and 2-APB had no effect on baseline flux, but abrogated the flow effect. These data implicate IP3 receptor-mediated intracellular Ca(2+) signaling as a critical step in transduction of microvillous drag to modulate Na(+) and HCO3 (-) transport.


Assuntos
Bicarbonatos/metabolismo , Cálcio/metabolismo , Citosol/metabolismo , Túbulos Renais Proximais/metabolismo , Reabsorção Renal , Trocadores de Sódio-Hidrogênio/metabolismo , Sódio/metabolismo , Animais , Transporte Biológico , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Quelantes/farmacologia , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Cinética , Camundongos Endogâmicos C57BL , Perfusão , Reabsorção Renal/efeitos dos fármacos , Trocador 3 de Sódio-Hidrogênio
18.
Am J Physiol Renal Physiol ; 309(3): F259-68, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26017972

RESUMO

Epithelial Na(+) channel (ENaC) blockade stimulates stilbene-sensitive conductive Cl(-) secretion in the mouse cortical collecting duct (CCD). This study's purpose was to determine the co-ion that accompanies benzamil- and DIDS-sensitive Cl(-) flux. Thus transepithelial voltage, VT, as well as total CO2 (tCO2) and Cl(-) flux were measured in CCDs from aldosterone-treated mice consuming a NaCl-replete diet. We reasoned that if stilbene inhibitors (DIDS) reduce conductive anion secretion they should reduce the lumen-negative VT. However, during ENaC blockade (benzamil, 3 µM), DIDS (100 µM) application to the perfusate reduced net H(+) secretion, which increased the lumen-negative VT. Conversely, ENaC blockade alone stimulated H(+) secretion, which reduced the lumen-negative VT. Application of an ENaC inhibitor to the perfusate reduced the lumen-negative VT, increased intercalated cell intracellular pH, and reduced net tCO2 secretion. However, benzamil did not change tCO2 flux during apical H(+)-ATPase blockade (bafilomycin, 5 nM). The increment in H(+) secretion observed with benzamil application contributes to the fall in VT observed with application of this diuretic. As such, ENaC blockade reduces the lumen-negative VT by inhibiting conductive Na(+) absorption and by stimulating H(+) secretion by type A intercalated cells. In conclusion, 1) in CCDs from aldosterone-treated mice, benzamil application stimulates HCl secretion mediated by the apical H(+)-ATPase and a yet to be identified conductive Cl(-) transport pathway; 2) benzamil-induced HCl secretion is reversed with the application of stilbene inhibitors or H(+)-ATPase inhibitors to the perfusate; and 3) benzamil reduces VT not only by inhibiting conductive Na(+) absorption, but also by stimulating H(+) secretion.


Assuntos
Ácido Clorídrico/metabolismo , Túbulos Renais Coletores/metabolismo , Macrolídeos/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Proteínas de Transporte de Ânions/genética , Proteínas de Transporte de Ânions/metabolismo , Cloretos/metabolismo , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Feminino , Concentração de Íons de Hidrogênio , Túbulos Renais Coletores/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , ATPases Translocadoras de Prótons/antagonistas & inibidores , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Estilbenos/antagonistas & inibidores , Estilbenos/farmacologia , Transportadores de Sulfato
19.
Am J Physiol Renal Physiol ; 309(3): F251-8, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25925258

RESUMO

Inhibition of the epithelial Na(+) channel (ENaC) reduces Cl(-) absorption in cortical collecting ducts (CCDs) from aldosterone-treated rats and mice. Since ENaC does not transport Cl(-), the purpose of the present study was to explore how ENaC modulates Cl(-) absorption in mouse CCDs perfused in vitro. Therefore, we measured transepithelial Cl(-) flux and transepithelial voltage in CCDs perfused in vitro taken from mice that consumed a NaCl-replete diet alone or the diet with aldosterone administered by minipump. We observed that application of an ENaC inhibitor [benzamil (3 µM)] to the luminal fluid unmasks conductive Cl(-) secretion. During ENaC blockade, this Cl(-) secretion fell with the application of a nonselective Cl(-) channel blocker [DIDS (100 µM)] to the perfusate. While single channel recordings of intercalated cell apical membranes in split-open CCDs demonstrated a Cl(-) channel with properties that resemble the ClC family of Cl(-) channels, ClC-5 is not the primary pathway for benzamil-sensitive Cl(-) flux. In conclusion, first, in CCDs from aldosterone-treated mice, most Cl(-) absorption is benzamil sensitive, and, second, benzamil application stimulates stilbene-sensitive conductive Cl(-) secretion, which occurs through a ClC-5-independent pathway.


Assuntos
Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Cloretos/metabolismo , Ácido Clorídrico/metabolismo , Túbulos Renais Coletores/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/antagonistas & inibidores , Algoritmos , Amilorida/farmacologia , Animais , Membrana Celular/efeitos dos fármacos , Diuréticos/farmacologia , Canais Epiteliais de Sódio/genética , Feminino , Masculino , Camundongos , Camundongos Knockout
20.
Kidney Int ; 86(1): 139-45, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24573316

RESUMO

The large-conductance, calcium-activated BK-α/ß4 potassium channel, localized to the intercalated cells of the distal nephron, mediates potassium secretion during high-potassium, alkaline diets. Here we determine whether BK-α/ß4-mediated potassium transport is dependent on epithelial sodium channel (ENaC)-mediated sodium reabsorption. We maximized sodium-potassium exchange in the distal nephron by feeding mice a low-sodium, high-potassium diet. Wild-type and BK-ß4 knockout mice were maintained on a low-sodium, high-potassium, alkaline diet or a low-sodium, high-potassium, acidic diet for 7-10 days. Wild-type mice maintained potassium homeostasis on the alkaline, but not acid, diet. BK-ß4 knockout mice could not maintain potassium homeostasis on either diet. During the last 12 h of diet, wild-type mice on either a regular, alkaline, or an acid diet, or knockout mice on an alkaline diet, were administered amiloride (an ENaC inhibitor). Amiloride enhanced sodium excretion in all wild-type and knockout groups to similar values; however, amiloride diminished potassium excretion by 59% in wild-type but only by 33% in knockout mice on an alkaline diet. Similarly, amiloride decreased the trans-tubular potassium gradient by 68% in wild-type but only by 42% in knockout mice on an alkaline diet. Amiloride treatment equally enhanced sodium excretion and diminished potassium secretion in knockout mice on an alkaline diet and wild-type mice on an acid diet. Thus, the enhanced effect of amiloride on potassium secretion in wild-type compared to knockout mice on the alkaline diet clarify a BK- α/ß4-mediated potassium secretory pathway in intercalated cells driven by ENaC-mediated sodium reabsorption linked to bicarbonate secretion.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Néfrons/metabolismo , Sódio/metabolismo , Amilorida/farmacologia , Animais , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Hidroclorotiazida/farmacologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/deficiência , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/deficiência , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Néfrons/efeitos dos fármacos , Potássio/metabolismo , Potássio na Dieta/administração & dosagem , Inibidores de Simportadores de Cloreto de Sódio/farmacologia , Sódio na Dieta/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA