Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 226
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Physiol Renal Physiol ; 320(5): F719-F733, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33719576

ABSTRACT

Phosphorylation of the thiazide-sensitive NaCl cotransporter (NCC) in the distal convoluted tubule (DCT) is altered rapidly in response to changes in extracellular K+ concentration ([K+]). High extracellular [K+] is believed to activate specific phosphatases to dephosphorylate NCC, thereby reducing its activity. This process is defective in the human disease familial hyperkalemic hypertension, in which extracellular [K+] fails to dephosphorylate NCC, suggesting an interplay between NCC-activating and NCC-inactivating switches. Here, we explored the role of STE20/SPS1-related proline-alanine-rich protein kinase (SPAK) and intracellular Cl- concentration in the rapid effects of extracellular K+ on NCC phosphorylation. SPAK was found to be rapidly dephosphorylated in vitro in human embryonic kidney cells and ex vivo in kidney slices by high [K+]. Acute high-K+ challenge resulted in DCT1-specific SPAK dephosphorylation in vivo and dissolution of SPAK puncta. In line with the postulate of interplay between activating and inactivating switches, we found that the "on" switch, represented by with no lysine kinase 4 (WNK4)-SPAK, must be turned off for rapid NCC dephosphorylation by high [K+]. Longer-term WNK-SPAK-mediated stimulation, however, altered the sensitivity of the system, as it attenuated rapid NCC dephosphorylation due to acute K+ loading. Although blockade of protein phosphatase (PP)1 increased NCC phosphorylation at baseline, neither PP1 nor PP3, singly or in combination, was essential for NCC dephosphorylation. Overall, our data suggest that NCC phosphorylation is regulated by a dynamic equilibrium between activating kinases and inactivating phosphatases, with kinase inactivation playing a key role in the rapid NCC dephosphorylation by high extracellular K+.NEW & NOTEWORTHY Although a great deal is known about mechanisms by which thiazide-sensitive NaCl cotransporter is phosphorylated and activated, much less is known about dephosphorylation. Here, we show that rapid dephosphorylation by high K+ depends on the Cl- sensitivity of with no lysine kinase 4 and the rapid dephosphorylation of STE20/SPS1-related proline-alanine-rich protein kinase, primarily along the early distal convoluted tubule.


Subject(s)
Chlorides/metabolism , Kidney Tubules, Distal/enzymology , Potassium, Dietary/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , HEK293 Cells , Humans , Kinetics , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Transport , Solute Carrier Family 12, Member 3/metabolism
2.
Am J Physiol Renal Physiol ; 319(3): F423-F435, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32657158

ABSTRACT

Cre-lox technology has revolutionized research in renal physiology by allowing site-specific genetic recombination in individual nephron segments. The distal convoluted tubule (DCT), consisting of distinct early (DCT1) and late (DCT2) segments, plays a central role in Na+ and K+ homeostasis. The only established Cre line targeting the DCT is Pvalb-Cre, which is limited by noninducibility, activity along DCT1 only, and activity in neurons. Here, we report the characterization of the first Cre line specific to the entire DCT. CRISPR/Cas9 targeting was used to introduce a tamoxifen-inducible IRES-Cre-ERT2 cassette downstream of the coding region of the Slc12a3 gene encoding the NaCl cotransporter (NCC). The resulting Slc12a3-Cre-ERT2 mice were crossed with R26R-YFP reporter mice, which revealed minimal leakiness with 6.3% of NCC-positive cells expressing yellow fluorescent protein (YFP) in the absence of tamoxifen. After tamoxifen injection, YFP expression was observed in 91.2% of NCC-positive cells and only in NCC-positive cells, revealing high recombination efficiency and DCT specificity. Crossing to R26R-TdTomato mice revealed higher leakiness (64.5%), suggesting differential sensitivity of the floxed site. Western blot analysis revealed no differences in abundances of total NCC or the active phosphorylated form of NCC in Slc12a3-Cre-ERT2 mice of either sex compared with controls. Plasma K+ and Mg2+ concentrations and thiazide-sensitive Na+ and K+ excretion did not differ in Slc12a3-Cre-ERT2 mice compared with controls when sex matched. These data suggest genetic modification had no obvious effect on NCC function. Slc12a3-Cre-ERT2 mice are the first line generated demonstrating inducible Cre recombinase activity along the entire DCT and will be a useful tool to study DCT function.


Subject(s)
Kidney Tubules, Distal/enzymology , Recombinases/metabolism , Sodium Chloride Symporters/metabolism , Animals , Estrogen Antagonists/pharmacology , Gene Expression Regulation/drug effects , Mice , Recombinases/genetics , Sodium Chloride Symporters/genetics , Solute Carrier Family 12, Member 3/genetics , Solute Carrier Family 12, Member 3/metabolism , Tamoxifen/pharmacology
3.
Am J Physiol Renal Physiol ; 316(2): F292-F300, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30484345

ABSTRACT

Hypokalemia contributes to the progression of chronic kidney disease, although a definitive pathophysiological theory to explain this remains to be established. K+ deficiency results in profound alterations in renal epithelial transport. These include an increase in salt reabsorption via the Na+-Cl- cotransporter (NCC) of the distal convoluted tubule (DCT), which minimizes electroneutral K+ loss in downstream nephron segments. In experimental conditions of dietary K+ depletion, punctate structures in the DCT containing crucial NCC-regulating kinases have been discovered in the murine DCT and termed "WNK bodies," referring to their component, with no K (lysine) kinases (WNKs). We hypothesized that in humans, WNK bodies occur in hypokalemia as well. Renal needle biopsies of patients with chronic hypokalemic nephropathy and appropriate controls were examined by histological stains and immunofluorescence. Segment- and organelle-specific marker proteins were used to characterize the intrarenal and subcellular distribution of established WNK body constituents, namely, WNKs and Ste20-related proline-alanine-rich kinase (SPAK). In both patients with hypokalemia, WNKs and SPAK concentrated in non-membrane-bound cytoplasmic regions in the DCT, consistent with prior descriptions of WNK bodies. The putative WNK bodies were located in the perinuclear region close to, but not within, the endoplasmic reticulum. They were closely adjacent to microtubules but not clustered in aggresomes. Notably, we provide the first report of WNK bodies, which are functionally challenging structures associated with K+ deficiency, in human patients.


Subject(s)
Hypokalemia/enzymology , Kidney Diseases/enzymology , Kidney Tubules, Distal/enzymology , Potassium/blood , Protein Serine-Threonine Kinases/analysis , Biomarkers/blood , Case-Control Studies , Humans , Hypokalemia/blood , Hypokalemia/pathology , Kidney Diseases/blood , Kidney Diseases/pathology , Kidney Tubules, Distal/ultrastructure , Multienzyme Complexes , WNK Lysine-Deficient Protein Kinase 1/analysis
4.
Am J Physiol Renal Physiol ; 315(3): F429-F444, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29993276

ABSTRACT

The vacuolar-type H+-ATPase B1 subunit is heavily expressed in the intercalated cells of the collecting system, where it contributes to H+ transport, but has also been described in other segments of the renal tubule. This study aimed to determine the localization of the B1 subunit of the vacuolar-type H+-ATPase in the early distal nephron, encompassing thick ascending limbs (TAL) and distal convoluted tubules (DCT), in human kidney and determine whether the localization differs between rodents and humans. Antibodies directed against the H+-ATPase B1 subunit were used to determine its localization in paraffin-embedded formalin-fixed mouse, rat, and human kidneys by light microscopy and in sections of Lowicryl-embedded rat kidneys by electron microscopy. Abundant H+-ATPase B1 subunit immunoreactivity was observed in the human kidney. As expected, intercalated cells showed the strongest signal, but significant signal was also observed in apical membrane domains of the distal nephron, including TAL, macula densa, and DCT. In mouse and rat, H+-ATPase B1 subunit expression could also be detected in apical membrane domains of these segments. In rat, electron microscopy revealed that the H+-ATPase B1 subunit was located in the apical membrane. Furthermore, the H+-ATPase B1 subunit colocalized with other H+-ATPase subunits in the TAL and DCT. In conclusion, the B1 subunit is expressed in the early distal nephron. The physiological importance of H+-ATPase expression in these segments remains to be delineated in detail. The phenotype of disease-causing mutations in the B1 subunit may also relate to its presence in the TAL and DCT.


Subject(s)
Kidney Tubules, Distal/enzymology , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Cell Polarity , Humans , Immunohistochemistry , Kidney Tubules, Distal/ultrastructure , Mice, Knockout , Microscopy, Electron, Transmission , Species Specificity , Vacuolar Proton-Translocating ATPases/deficiency , Vacuolar Proton-Translocating ATPases/genetics
5.
Am J Physiol Renal Physiol ; 315(2): F223-F230, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29667910

ABSTRACT

With-no-lysine kinase 4 (WNK4) and kidney-specific (KS)-WNK1 regulate ROMK (Kir1.1) channels in a variety of cell models. We now explore the role of WNK4 and KS-WNK1 in regulating ROMK in the native distal convoluted tubule (DCT)/connecting tubule (CNT) by measuring tertiapin-Q (TPNQ; ROMK inhibitor)-sensitive K+ currents with whole cell recording. TPNQ-sensitive K+ currents in DCT2/CNT of KS- WNK1-/- and WNK4-/- mice were significantly smaller than that of WT mice. In contrast, the basolateral K+ channels (a Kir4.1/5.1 heterotetramer) in the DCT were not inhibited. Moreover, WNK4-/- mice were hypokalemic, while KS- WNK1-/- mice had normal plasma K+ levels. High K+ (HK) intake significantly increased TPNQ-sensitive K+ currents in DCT2/CNT of WT and WNK4-/- mice but not in KS- WNK1-/- mice. However, TPNQ-sensitive K+ currents in the cortical collecting duct (CCD) were normal not only under control conditions but also significantly increased in response to HK in KS- WNK1-/- mice. This suggests that the deletion of KS-WNK1-induced inhibition of ROMK occurs only in the DCT2/CNT. Renal clearance study further demonstrated that the deletion of KS-WNK1 did not affect the renal ability of K+ excretion under control conditions and during increasing K+ intake. Also, HK intake did not cause hyperkalemia in KS- WNK1-/- mice. We conclude that KS-WNK1 but not WNK4 is required for HK intake-induced stimulation of ROMK activity in DCT2/CNT. However, KS-WNK1 is not essential for HK-induced stimulation of ROMK in the CCD, and the lack of KS-WNK1 does not affect net renal K+ excretion.


Subject(s)
Kidney Tubules, Distal/enzymology , Potassium Channels, Inwardly Rectifying/metabolism , Potassium, Dietary/metabolism , Protein Serine-Threonine Kinases/metabolism , WNK Lysine-Deficient Protein Kinase 1/metabolism , Animals , Female , Genotype , Hyperkalemia/enzymology , Hyperkalemia/genetics , Hypokalemia/enzymology , Hypokalemia/genetics , In Vitro Techniques , Male , Membrane Potentials , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Potassium Channels, Inwardly Rectifying/genetics , Potassium, Dietary/urine , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Renal Elimination , WNK Lysine-Deficient Protein Kinase 1/deficiency , WNK Lysine-Deficient Protein Kinase 1/genetics
6.
Am J Physiol Renal Physiol ; 314(5): F999-F1007, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29384416

ABSTRACT

The Na+-Cl- cotransporter (NCC) in distal convoluted tubule (DCT) plays important roles in renal NaCl reabsorption. The current hypothesis for the mechanism of regulation of NCC focuses on WNK4 and intracellular Cl- concentration ([Cl-]i). WNK kinases bind Cl-, and Cl- binding decreases the catalytic activity. It is believed that hypokalemia under low K+ intake decreases [Cl-]i to activate WNK4, which thereby phosphorylates and stimulates NCC through activation of SPAK. However, increased NCC activity and apical NaCl entry would mitigate the fall in [Cl-]i. Whether [Cl-]i in DCT under low-K+ diet is sufficiently low to activate WNK4 is unknown. Furthermore, increased luminal NaCl delivery also stimulates NCC and causes upregulation of the transporter. Unlike low K+ intake, increased luminal NaCl delivery would tend to increase [Cl-]i. Thus we investigated the role of WNK4 and [Cl-]i in regulating NCC. We generated Wnk4-knockout mice and examined regulation of NCC by low K+ intake and by increased luminal NaCl delivery in knockout (KO) and wild-type mice. Wnk4-KO mice have marked reduction in the abundance, phosphorylation, and functional activity of NCC vs. wild type. Low K+ intake increases NCC phosphorylation and functional activity in wild-type mice, but not in Wnk4-KO mice. Increased luminal NaCl delivery similarly upregulates NCC, which, contrary to low K+ intake, is not abolished in Wnk4-KO mice. The results reveal that modulation of WNK4 activity by [Cl-]i is not the sole mechanism for regulating NCC. Increased luminal NaCl delivery upregulates NCC via yet unknown mechanism(s) that may override inhibition of WNK4 by high [Cl-]i.


Subject(s)
Kidney Tubules, Distal/enzymology , Potassium, Dietary/metabolism , Protein Serine-Threonine Kinases/metabolism , Sodium Chloride/metabolism , Animals , Biological Transport , Gene Expression Regulation, Enzymologic , Injections, Subcutaneous , Kidney Tubules, Distal/drug effects , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Renal Elimination , Renal Reabsorption , Sodium Chloride/administration & dosage , Sodium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 3/deficiency , Solute Carrier Family 12, Member 3/genetics , Solute Carrier Family 12, Member 3/metabolism
7.
J Am Soc Nephrol ; 28(8): 2431-2442, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28289184

ABSTRACT

Adaptation of the organism to potassium (K+) deficiency requires precise coordination among organs involved in K+ homeostasis, including muscle, liver, and kidney. How the latter performs functional and molecular changes to ensure K+ retention is not well understood. Here, we investigated the role of ubiquitin-protein ligase NEDD4-2, which negatively regulates the epithelial sodium channel (ENaC), Na+/Cl- cotransporter (NCC), and with no-lysine-kinase 1 (WNK1). After dietary K+ restriction for 2 weeks, compared with control littermates, inducible renal tubular NEDD4-2 knockout (Nedd4LPax8/LC1 ) mice exhibited severe hypokalemia and urinary K+ wasting. Notably, expression of the ROMK K+ channel did not change in the distal convoluted tubule and decreased slightly in the cortical/medullary collecting duct, whereas BK channel abundance increased in principal cells of the connecting tubule/collecting ducts. However, K+ restriction also enhanced ENaC expression in Nedd4LPax8/LC1 mice, and treatment with the ENaC inhibitor, benzamil, reversed excessive K+ wasting. Moreover, K+ restriction increased WNK1 and WNK4 expression and enhanced SPAK-mediated NCC phosphorylation in Nedd4LPax8/LC1 mice, with no change in total NCC. We propose a mechanism in which NEDD4-2 deficiency exacerbates hypokalemia during dietary K+ restriction primarily through direct upregulation of ENaC, whereas increased BK channel expression has a less significant role. These changes outweigh the compensatory antikaliuretic effects of diminished ROMK expression, increased NCC phosphorylation, and enhanced WNK pathway activity in the distal convoluted tubule. Thus, NEDD4-2 has a crucial role in K+ conservation through direct and indirect effects on ENaC, distal nephron K+ channels, and WNK signaling.


Subject(s)
Adaptation, Physiological , Endosomal Sorting Complexes Required for Transport/physiology , Hypokalemia/physiopathology , Kidney Tubules, Distal/enzymology , Ubiquitin-Protein Ligases/physiology , Animals , Kidney/physiopathology , Mice , Nedd4 Ubiquitin Protein Ligases , Time Factors
8.
J Biol Chem ; 289(17): 11791-11806, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24610784

ABSTRACT

It has been well established that blood pressure and renal function undergo circadian fluctuations. We have demonstrated that the circadian protein Per1 regulates multiple genes involved in sodium transport in the collecting duct of the kidney. However, the role of Per1 in other parts of the nephron has not been investigated. The distal convoluted tubule (DCT) plays a critical role in renal sodium reabsorption. Sodium is reabsorbed in this segment through the actions of the NaCl co-transporter (NCC), which is regulated by the with-no-lysine kinases (WNKs). The goal of this study was to test if Per1 regulates sodium transport in the DCT through modulation of NCC and the WNK kinases, WNK1 and WNK4. Pharmacological blockade of nuclear Per1 entry resulted in decreased mRNA expression of NCC and WNK1 but increased expression of WNK4 in the renal cortex of mice. These findings were confirmed by using Per1 siRNA and pharmacological blockade of Per1 nuclear entry in mDCT15 cells, a model of the mouse distal convoluted tubule. Transcriptional regulation was demonstrated by changes in short lived heterogeneous nuclear RNA. Chromatin immunoprecipitation experiments demonstrated interaction of Per1 and CLOCK with the promoters of NCC, WNK1, and WNK4. This interaction was modulated by blockade of Per1 nuclear entry. Importantly, NCC protein expression and NCC activity, as measured by thiazide-sensitive, chloride-dependent (22)Na uptake, were decreased upon pharmacological inhibition of Per1 nuclear entry. Taken together, these data demonstrate a role for Per1 in the transcriptional regulation of NCC, WNK1, and WNK4.


Subject(s)
Kidney Tubules, Distal/metabolism , Period Circadian Proteins/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Base Sequence , Cell Line , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , DNA Primers , Gene Knockdown Techniques , Kidney Tubules, Distal/enzymology , Mice , Mice, Knockout , Period Circadian Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Solute Carrier Family 12, Member 3/genetics
9.
Am J Physiol Renal Physiol ; 308(8): F923-31, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25651563

ABSTRACT

The NaCl cotransporter (NCC) of the renal distal convoluted tubule is stimulated by low-K(+) diet by an unknown mechanism. Since recent work has shown that the STE20/SPS-1-related proline-alanine-rich protein kinase (SPAK) can function to stimulate NCC by phosphorylation of specific N-terminal sites, we investigated whether the NCC response to low-K(+) diet is mediated by SPAK. Using phospho-specific antibodies in Western blot and immunolocalization studies of wild-type and SPAK knockout (SPAK(-/-)) mice fed a low-K(+) or control diet for 4 days, we found that low-K(+) diet strongly increased total NCC expression and phosphorylation of NCC. This was associated with an increase in total SPAK expression in cortical homogenates and an increase in phosphorylation of SPAK at the S383 activation site. The increased pNCC in response to low-K(+) diet was blunted but not completely inhibited in SPAK(-/-) mice. These findings reveal that SPAK is an important mediator of the increased NCC activation by phosphorylation that occurs in the distal convoluted tubule in response to a low-K(+) diet, but other low-potassium-activated kinases are likely to be involved.


Subject(s)
Kidney Tubules, Distal/enzymology , Potassium Deficiency/enzymology , Potassium, Dietary/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Mice, Knockout , Phosphorylation , Potassium Deficiency/genetics , Potassium, Dietary/administration & dosage , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Solute Carrier Family 12, Member 3/metabolism , Up-Regulation
10.
J Am Soc Nephrol ; 25(3): 511-22, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24231659

ABSTRACT

The thiazide-sensitive NaCl cotransporter (NCC) of the renal distal convoluted tubule (DCT) controls ion homeostasis and arterial BP. Loss-of-function mutations of NCC cause renal salt wasting with arterial hypotension (Gitelman syndrome). Conversely, mutations in the NCC-regulating WNK kinases or kelch-like 3 protein cause familial hyperkalemic hypertension. Here, we performed automated sorting of mouse DCTs and microarray analysis for comprehensive identification of novel DCT-enriched gene products, which may potentially regulate DCT and NCC function. This approach identified protein phosphatase 1 inhibitor-1 (I-1) as a DCT-enriched transcript, and immunohistochemistry revealed I-1 expression in mouse and human DCTs and thick ascending limbs. In heterologous expression systems, coexpression of NCC with I-1 increased thiazide-dependent Na(+) uptake, whereas RNAi-mediated knockdown of endogenous I-1 reduced NCC phosphorylation. Likewise, levels of phosphorylated NCC decreased by approximately 50% in I-1 (I-1(-/-)) knockout mice without changes in total NCC expression. The abundance and phosphorylation of other renal sodium-transporting proteins, including NaPi-IIa, NKCC2, and ENaC, did not change, although the abundance of pendrin increased in these mice. The abundance, phosphorylation, and subcellular localization of SPAK were similar in wild-type (WT) and I-1(-/-) mice. Compared with WT mice, I-1(-/-) mice exhibited significantly lower arterial BP but did not display other metabolic features of NCC dysregulation. Thus, I-1 is a DCT-enriched gene product that controls arterial BP, possibly through regulation of NCC activity.


Subject(s)
Hypotension/enzymology , Kidney Tubules, Distal/enzymology , Proteins/metabolism , Solute Carrier Family 12, Member 3/metabolism , Animals , Anion Transport Proteins/metabolism , Blood Pressure , Female , Humans , Loop of Henle/enzymology , Male , Mice , Mice, Transgenic , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proteins/genetics , Solute Carrier Family 12, Member 1/metabolism , Sulfate Transporters , Up-Regulation , Xenopus
11.
Am J Physiol Cell Physiol ; 307(6): C532-41, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25031022

ABSTRACT

Cumulative evidence suggests that guanylin peptides play an important role on electrolyte homeostasis. We have previously reported that uroguanylin (UGN) inhibits bicarbonate reabsorption in a renal distal tubule. In the present study, we tested the hypothesis that the bicarbonaturic effect of UGN is at least in part attributable to inhibition of H(+)-ATPase-mediated hydrogen secretion in the distal nephron. By in vivo stationary microperfusion experiments, we were able to show that UGN inhibits H(+)-ATPase activity by a PKG-dependent pathway because KT5823 (PKG inhibitor) abolished the UGN effect on distal bicarbonate reabsorption and H89 (PKA inhibitor) was unable to prevent it. The in vivo results were confirmed by the in vitro experiments, where we used fluorescence microscopy to measure intracellular pH (pHi) recovery after an acid pulse with NH4Cl. By this technique, we observed that UGN and 8 bromoguanosine-cGMP (8Br-cGMP) inhibited H(+)-ATPase-dependent pHi recovery and that the UGN inhibitory effect was abolished in the presence of the PKG inhibitor. In addition, by using RT-PCR technique, we verified that Madin-Darby canine kidney (MDCK)-C11 cells express guanylate cyclase-C. Besides, UGN stimulated an increase of both cGMP content and PKG activity but was unable to increase the production of cellular cAMP content and PKA activity. Furthermore, we found that UGN reduced cell surface abundance of H+-ATPase B1 subunit in MDCK-C11 and that this effect was abolished by the PKG inhibitor. Taken together, our data suggest that UGN inhibits H(+)-ATPase activity and surface expression in renal distal cells by a cGMP/PKG-dependent pathway.


Subject(s)
Cell Membrane/drug effects , Cyclic GMP-Dependent Protein Kinases/metabolism , Kidney Tubules, Distal/drug effects , Natriuretic Peptides/pharmacology , Proton-Translocating ATPases/metabolism , Animals , Bicarbonates/metabolism , Cell Membrane/enzymology , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Dogs , Hydrogen-Ion Concentration , Kidney Tubules, Distal/enzymology , Madin Darby Canine Kidney Cells , Male , Perfusion , Protein Kinase Inhibitors/pharmacology , Protein Transport , Rats , Rats, Wistar , Receptors, Guanylate Cyclase-Coupled/drug effects , Receptors, Guanylate Cyclase-Coupled/genetics , Receptors, Guanylate Cyclase-Coupled/metabolism , Signal Transduction/drug effects , Time Factors
12.
Am J Physiol Renal Physiol ; 305(7): F995-F1005, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23863470

ABSTRACT

Nadph oxidase 4 is an important cellular source of reactive oxygen species (ROS) generation in the kidney. Novel antioxidant drugs, such as Nox4 inhibitor compounds, are being developed. There is, however, very little experimental evidence for the biological role and regulation of Nadph oxidase isoforms in the kidney. Herein, we show that Fulvene-5 is an effective inhibitor of Nox-generated ROS and report the role of Nox isoforms in activating epithelial sodium channels (ENaC) in A6 distal nephron cells via oxidant signaling and cell stretch activation. Using single-channel patch-clamp analysis, we report that Fulvene-5 blocked the increase in ENaC activity that is typically observed with H2O2 treatment of A6 cells: average ENaC NPo values decreased from a baseline level of 1.04 ± 0.18 (means ± SE) to 0.25 ± 0.08 following Fulvene-5 treatment. H2O2 treatment failed to increase ENaC activity in the presence of Fulvene-5. Moreover, Fulvene-5 treatment of A6 cells blocked the osmotic cell stretch response of A6 cells, indicating that stretch activation of Nox-derived ROS plays an important role in ENaC regulation. Together, these findings indicate that Fulvene-5, and perhaps other classes of antioxidant inhibitors, may represent a novel class of compounds useful for the treatment of pathological disorders stemming from inappropriate ion channel activity, such as hypertension.


Subject(s)
Cyclopentanes/pharmacology , Epithelial Sodium Channels/metabolism , Kidney Tubules, Distal/enzymology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Line , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/drug effects , Osmosis/drug effects , Xenopus
13.
EMBO J ; 27(12): 1747-57, 2008 Jun 18.
Article in English | MEDLINE | ID: mdl-18497742

ABSTRACT

In patients with von Hippel-Lindau (VHL) disease, renal cysts and clear cell renal cell carcinoma (ccRCC) arise from renal tubular epithelial cells containing biallelic inactivation of the VHL tumour suppressor gene. However, it is presumed that formation of renal cysts and their conversion to ccRCC involve additional genetic changes at other loci. Here, we show that cystic lesions in the kidneys of patients with VHL disease also demonstrate activation of the phosphatidylinositol-3-kinase (PI3K) pathway. Strikingly, combined conditional inactivation of Vhlh and the Pten tumour suppressor gene, which normally antagonises PI3K signalling, in the mouse kidney, elicits cyst formation after short latency, whereas inactivation of either tumour suppressor gene alone failed to produce such a phenotype. Interestingly, cells lining these cysts frequently lack a primary cilium, a microtubule-based cellular antenna important for suppression of uncontrolled kidney epithelial cell proliferation and cyst formation. Our results support a model in which the PTEN tumour suppressor protein cooperates with pVHL to suppress cyst development in the kidney.


Subject(s)
Cysts/enzymology , Cysts/pathology , Kidney Diseases, Cystic/enzymology , Kidney Diseases, Cystic/pathology , PTEN Phosphohydrolase/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Cell Proliferation , Cilia/enzymology , Cilia/pathology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Tubules, Distal/enzymology , Kidney Tubules, Distal/pathology , MAP Kinase Signaling System , Mice , Mutation/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
14.
J Am Soc Nephrol ; 22(9): 1707-19, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21852580

ABSTRACT

Regulation of renal Na(+) transport is essential for controlling blood pressure, as well as Na(+) and K(+) homeostasis. Aldosterone stimulates Na(+) reabsorption by the Na(+)-Cl(-) cotransporter (NCC) in the distal convoluted tubule (DCT) and by the epithelial Na(+) channel (ENaC) in the late DCT, connecting tubule, and collecting duct. Aldosterone increases ENaC expression by inhibiting the channel's ubiquitylation and degradation; aldosterone promotes serum-glucocorticoid-regulated kinase SGK1-mediated phosphorylation of the ubiquitin-protein ligase Nedd4-2 on serine 328, which prevents the Nedd4-2/ENaC interaction. It is important to note that aldosterone increases NCC protein expression by an unknown post-translational mechanism. Here, we present evidence that Nedd4-2 coimmunoprecipitated with NCC and stimulated NCC ubiquitylation at the surface of transfected HEK293 cells. In Xenopus laevis oocytes, coexpression of NCC with wild-type Nedd4-2, but not its catalytically inactive mutant, strongly decreased NCC activity and surface expression. SGK1 prevented this inhibition in a kinase-dependent manner. Furthermore, deficiency of Nedd4-2 in the renal tubules of mice and in cultured mDCT(15) cells upregulated NCC. In contrast to ENaC, Nedd4-2-mediated inhibition of NCC did not require the PY-like motif of NCC. Moreover, the mutation of Nedd4-2 at either serine 328 or 222 did not affect SGK1 action, and mutation at both sites enhanced Nedd4-2 activity and abolished SGK1-dependent inhibition. Taken together, these results suggest that aldosterone modulates NCC protein expression via a pathway involving SGK1 and Nedd4-2 and provides an explanation for the well-known aldosterone-induced increase in NCC protein expression.


Subject(s)
Aldosterone/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Immediate-Early Proteins/metabolism , Kidney Tubules, Distal/enzymology , Protein Serine-Threonine Kinases/metabolism , Sodium Chloride Symporters/metabolism , Ubiquitin-Protein Ligases/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Animals , Down-Regulation , HEK293 Cells , Humans , Mice , Mice, Knockout , Nedd4 Ubiquitin Protein Ligases , Phosphorylation , Signal Transduction , Ubiquitination , Xenopus Proteins , Xenopus laevis
15.
Biochem Biophys Res Commun ; 404(1): 284-90, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21130072

ABSTRACT

Hepatocyte nuclear factor-1B (HNF-1B) is a transcription factor involved in embryonic development and tissue-specific gene expression in several organs, including the kidney. Recently heterozygous mutations in the HNF1B gene have been identified in patients with hypomagnesemia due to renal Mg(2+) wasting. Interestingly, ChIP-chip data revealed HNF-1B binding sites in the FXYD2 gene, encoding the γ-subunit of the Na(+)/K(+)-ATPase. The γ-subunit has been described as one of the molecular players in the renal Mg(2+) reabsorption in the distal convoluted tubule (DCT). Of note, the FXYD2 gene can be alternatively transcribed into two main variants, namely γa and γb. In the present study, we demonstrated via two different reporter gene assays that HNF-1B specifically acts as an activator of the γa-subunit, whereas the γb-subunit expression was not affected. Moreover, the HNF-1B mutations H69fsdelAC, H324S325fsdelCA, Y352finsA and K156E, previously identified in patients with hypomagnesemia, prevented transcription activation of γa-subunit via a dominant negative effect on wild type HNF1-B. By immunohistochemistry, it was shown that the γa- and γb-subunits colocalize at the basolateral membrane of the DCT segment of mouse kidney. On the basis of these data, we suggest that abnormalities involving the HNF-1B gene may impair the relative abundance of γa and γb, thus affecting the transcellular Mg(2+) reabsorption in the DCT.


Subject(s)
Gene Expression Regulation , Hepatocyte Nuclear Factor 1-beta/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Green Fluorescent Proteins/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Kidney/enzymology , Kidney Tubules, Distal/enzymology , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Sodium-Potassium-Exchanging ATPase/metabolism , Transcription, Genetic
16.
Histochem Cell Biol ; 136(1): 25-35, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21660484

ABSTRACT

Initial reports claim that WNK4 localization is mainly at intercellular junctions of distal convoluted tubules (DCT) and cortical collecting ducts (CCD) in the kidney. However, we recently clarified the major targets of WNK4 kinase to be the OSR1/SPAK kinases and the Na-Cl co-transporter (NCC), an apical membrane protein in the DCT, thus raising the question of whether the cellular localization of WNK4 is at intercellular junctions. In this study, we re-evaluate the intrarenal and intracellular immunolocalization of WNK4 in the mouse kidney using a newly generated anti-WNK4 antibody. By performing double immunofluorescence of WNK4 with several nephron-segment-specific markers, we have found that WNK4 is present in podocytes in glomeruli, the cortical thick ascending limb of Henle's loop including macula densa, and the medullary collecting ducts (MCD), in addition to the previously identified nephron segments, i.e., DCT and CCD. These results are consistent with the finding that WNK4 constitutes a kinase cascade with OSR1/SPAK and NCC in the DCT, and highlights a novel role for WNK4 in nephron segments newly identified as being WNK4-positive in this study.


Subject(s)
Kidney/enzymology , Protein Serine-Threonine Kinases/analysis , Animals , COS Cells , Chlorocebus aethiops , Fluorescent Antibody Technique , Kidney/metabolism , Kidney Tubules, Distal/enzymology , Kidney Tubules, Distal/metabolism , Mice , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Receptors, Drug/metabolism , Solute Carrier Family 12, Member 3 , Symporters/metabolism
17.
Vet Pathol ; 48(6): 1125-33, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21160023

ABSTRACT

The objective of this study was to investigate the effects of chronic inhibition of nitric oxide synthase (NOS) on cyclooxygenase-2 (COX-2) expression in the macula densa (MD) of swine, as well as the effects on expression of related proteins. Adult female Yucatan swine were given either tap water (control, n = 6) or water with N (G)-nitro-L-arginine methyl ester (L-NAME, 100 mg/liter, n = 5) for a minimum of 30 days. Duplicate samples of kidney were fixed or snap frozen. There was a significant (P = .0082) upregulation of COX-2 mRNA expression in the MD of L-NAME, as well as an apparent increase in COX-2 protein. Plasma renin activity also increased with L-NAME treatment (control, 0.34 ± 0.08 ng/ml; L-NAME, 1.26 ± 0.03 ng/ml; P = .00000003). There were no differences between groups in expression of either inducible NOS or renin protein or in serum electrolyte concentrations. In conclusion, with chronic inhibition of NOS, COX-2 in MD is upregulated, perhaps to compensate for loss of nitric oxide. Increases in COX-2 products may counteract renal arteriolar constriction and sustain renin release.


Subject(s)
Cyclooxygenase 2/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Kidney/enzymology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Animals , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Disease Models, Animal , Electrolytes/blood , Euthanasia, Animal , Female , Humans , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/enzymology , Kidney/cytology , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/enzymology , Microdissection/methods , Nitric Oxide/blood , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Protein Isoforms , RNA, Messenger/genetics , Rabbits , Renin/metabolism , Swine , Up-Regulation/drug effects
18.
Am J Physiol Regul Integr Comp Physiol ; 298(3): R707-12, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20053956

ABSTRACT

Macula densa (MD)-mediated regulation of renal hemodynamics via tubuloglomerular feedback is regulated by interactions between factors such as superoxide (O(2)(-)) and angiotensin II (ANG II). We have reported that NaCl-induced O(2)(-) in the MD is produced by the NOX2 isoform of NADPH oxidase (NOX); however, the source of ANG II-induced O(2)(-) in MD is unknown. Thus we determined the pathways by which ANG II increased O(2)(-) in the MD by measuring O(2)(-) in ANG II-treated MMDD1 cells, a MD-like cell line. ANG II caused MMDD1 O(2)(-) levels to increase by more than twofold (P < 0.01). This increase was blocked by losartan (AT(1) receptor blocker) but not PD-123319 (AT(2) receptor antagonist). Apocynin (a NOX inhibitor) decreased O(2)(-) by 86% (P < 0.01), whereas oxypurinol (a xanthine oxidase inhibitor) and NS-398 (a cyclooxygenase-2 inhibitor) had no significant effect. The NOX-dependent increase in O(2)(-) was due to the NOX2 isoform; a short interfering (si)RNA against NOX2 blunted ANG II-induced increases in O(2)(-), whereas the NOX4/siRNA did not. Finally, we found that inhibiting the Rac1 subunit of NOX blunted ANG II-induced O(2)(-) production in NOX4/siRNA-treated cells but did not further decrease it in NOX2/siRNA-treated cells. Our results indicate that ANG II stimulates O(2)(-) production in the MD primarily via AT(1)-dependent activation of NOX2. Rac1 is required for the full activation of NOX2. This pathway may be an important component of ANG II enhancement of tubuloglomerular feedback.


Subject(s)
Angiotensin II/pharmacology , Kidney Tubules, Distal/enzymology , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Renal Circulation/physiology , Superoxides/metabolism , Acetophenones/pharmacology , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Epithelial Cells/enzymology , Feedback, Physiological/physiology , Kidney Tubules, Distal/blood supply , Kidney Tubules, Distal/cytology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Neuropeptides/metabolism , RNA, Small Interfering , Receptor, Angiotensin, Type 1/metabolism , Renal Circulation/drug effects , Vasoconstrictor Agents/pharmacology , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
19.
Can J Physiol Pharmacol ; 88(10): 986-95, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20962898

ABSTRACT

Cation transport in the distal mammalian nephron relies on the SLC12 family of membrane cotransporters that include the thiazide-sensitive Na(+)-Cl⁻ cotransporter (NCC). NCC is regulated through a scaffold of interacting proteins, including the WNK kinases, WNK 1 and WNK 4, which are mutated in the hypertensive Gordon's syndrome. Dynamic regulation of NCC function by kinases must involve dephosphorylation by phosphatases, as illustrated by the role of PP1 and PP2B in the regulation of KCC members of the SLC12 family. There are 2 phosphorylation-controlled regulatory pathways for NCC: type 1, mediated by WNK4 and affecting trafficking to the surface membrane, and type 2, affecting intrinsic transporter kinetics by phosphorylation of conserved N-terminal S/T amino acids. Using the Xenopus oocyte expression system, we show that PP4 inhibits NCC activity - but not trafficking to the surface membrane - by a mechanism that requires phosphatase activity and a conserved N-terminal amino acid of NCC, threonine 58. This action is distinct from WNK4 regulation of membrane trafficking. In the mouse kidney, PP4 is selectively expressed in the distal nephron, including cells of the distal convoluted tubule cells, suggesting that PP4 may have a physiological role in regulating NCC and hence NaCl reabsorption in vivo.


Subject(s)
Kidney/metabolism , Phosphoprotein Phosphatases/metabolism , Receptors, Drug/metabolism , Sodium Chloride Symporters/metabolism , Animals , Cell Membrane/enzymology , Cell Membrane/metabolism , Cells, Cultured , Immunohistochemistry , Kidney/enzymology , Kidney Tubules, Distal/enzymology , Kidney Tubules, Distal/metabolism , Male , Mice , Mice, Inbred C57BL , Nephrons/enzymology , Nephrons/metabolism , Oocytes , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Receptors, Drug/genetics , Sodium Chloride Symporters/genetics , Transfection , Xenopus Proteins/metabolism , Xenopus laevis
20.
Biochemistry (Mosc) ; 75(8): 971-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-21073417

ABSTRACT

Ouabain and other cardiotonic steroids (CTS) kill renal epithelial cells from distal tubules (C7-MDCK) via interaction with Na,K-ATPase but independently of inhibition of Na,K-ATPase-mediated ion fluxes. Recently, we demonstrated that modest intracellular acidification and inhibition of p38 MAPK suppress death of C7-MDCK cells triggered by ouabain. In the present study we investigate the mechanism of p38 MAPK activation in renal epithelial cell from distal tubules evoked by cardiotonic steroids. Using Na+/K+ ionophores (monensin, nigericin) and media with different content of monovalent cations, we revealed that p38 MAPK phosphorylation in ouabain-treated renal epithelial cells is not caused by Na,K-ATPase inhibition and inversion of the [Na+](i)/[K+](i) ratio. We also demonstrated that attenuation of pH from 7.45 to 6.75 did not alter the level of p38 MAPK phosphorylation observed in ouabain-treated cells. Inhibitors of PKA, PKC, and PKG as well as protein phosphatases were unable to abolish p38 MAPK activation triggered by ouabain. Using phosphotyrosine antibodies we did not detect any effect of ouabain on activation of tyrosine kinases. Thus, our results show that activation of p38 MAPK and cytotoxic action of CTS are independent of intracellular Na+, K+, and H+ concentrations. The molecular origin of intermediates of death signaling induced by CTS via conformation changes of Na,K-ATPase with following activation of p38 MAPK should be examined further.


Subject(s)
Cardiac Glycosides/pharmacology , Epithelial Cells/enzymology , Kidney Tubules, Distal/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Death , Cell Line , Dogs , Epithelial Cells/drug effects , Hydrogen-Ion Concentration , Kidney Tubules, Distal/drug effects , Ouabain/pharmacology , Phosphorylation , Signal Transduction , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL