Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Hypertension ; 81(5): 1044-1054, 2024 May.
Article in English | MEDLINE | ID: mdl-38465625

ABSTRACT

BACKGROUND: Potassium (K+)-deficient diets, typical of modern processed foods, increase blood pressure (BP) and NaCl sensitivity. A K+-dependent signaling pathway in the kidney distal convoluted tubule, coined the K+ switch, that couples extracellular K+ sensing to activation of the thiazide-sensitive NaCl cotransporter (NCC) and NaCl retention has been implicated, but causality has not been established. METHODS: To test the hypothesis that small, physiological changes in plasma K+ (PK+) are translated to BP through the switch pathway, a genetic approach was used to activate the downstream switch kinase, SPAK (SPS1-related proline/alanine-rich kinase), within the distal convoluted tubule. The CA-SPAK (constitutively active SPS1-related proline/alanine-rich kinase mice) were compared with control mice over a 4-day PK+ titration (3.8-5.1 mmol) induced by changes in dietary K+. Arterial BP was monitored using radiotelemetry, and renal function measurements, NCC abundance, phosphorylation, and activity were made. RESULTS: As PK+ decreased in control mice, BP progressively increased and became sensitive to dietary NaCl and hydrochlorothiazide, coincident with increased NCC phosphorylation and urinary sodium retention. By contrast, BP in CA-SPAK mice was elevated, resistant to the PK+ titration, and sensitive to hydrochlorothiazide and salt at all PK+ levels, concomitant with sustained and elevated urinary sodium retention and NCC phosphorylation and activity. Thus, genetically locking the switch on drives NaCl sensitivity and prevents the response of BP to potassium. CONCLUSIONS: Low K+, common in modern ultraprocessed diets, presses the K+-switch pathway to turn on NCC activity, increasing sodium retention, BP, and salt sensitivity.


Subject(s)
Potassium , Protein Serine-Threonine Kinases , Animals , Mice , Potassium/metabolism , Protein Serine-Threonine Kinases/metabolism , Potassium, Dietary/metabolism , Blood Pressure/physiology , Sodium Chloride/metabolism , Solute Carrier Family 12, Member 3/metabolism , Signal Transduction , Phosphorylation , Kidney Tubules, Distal/metabolism , Hydrochlorothiazide , Sodium/metabolism , Alanine/metabolism , Proline/metabolism
2.
J Clin Invest ; 133(21)2023 11 01.
Article in English | MEDLINE | ID: mdl-37676724

ABSTRACT

Consumption of low dietary potassium, common with ultraprocessed foods, activates the thiazide-sensitive sodium chloride cotransporter (NCC) via the with no (K) lysine kinase/STE20/SPS1-related proline-alanine-rich protein kinase (WNK/SPAK) pathway to induce salt retention and elevate blood pressure (BP). However, it remains unclear how high-potassium "DASH-like" diets (dietary approaches to stop hypertension) inactivate the cotransporter and whether this decreases BP. A transcriptomics screen identified Ppp1Ca, encoding PP1A, as a potassium-upregulated gene, and its negative regulator Ppp1r1a, as a potassium-suppressed gene in the kidney. PP1A directly binds to and dephosphorylates NCC when extracellular potassium is elevated. Using mice genetically engineered to constitutively activate the NCC-regulatory kinase SPAK and thereby eliminate the effects of the WNK/SPAK kinase cascade, we confirmed that PP1A dephosphorylated NCC directly in a potassium-regulated manner. Prior adaptation to a high-potassium diet was required to maximally dephosphorylate NCC and lower BP in constitutively active SPAK mice, and this was associated with potassium-dependent suppression of Ppp1r1a and dephosphorylation of its cognate protein, inhibitory subunit 1 (I1). In conclusion, potassium-dependent activation of PP1A and inhibition of I1 drove NCC dephosphorylation, providing a mechanism to explain how high dietary K+ lowers BP. Shifting signaling of PP1A in favor of activation of WNK/SPAK may provide an improved therapeutic approach for treating salt-sensitive hypertension.


Subject(s)
Hypertension , Protein Serine-Threonine Kinases , Animals , Mice , Blood Pressure/physiology , Solute Carrier Family 12, Member 3/genetics , Solute Carrier Family 12, Member 3/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sodium Chloride/metabolism , Sodium Chloride/pharmacology , Potassium, Dietary/metabolism , Potassium, Dietary/pharmacology , Kidney/metabolism , Hypertension/genetics , Hypertension/metabolism , Potassium/metabolism , Potassium/pharmacology , Phosphorylation
3.
Am J Physiol Renal Physiol ; 325(3): F377-F393, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37498547

ABSTRACT

The urinary potassium (K+) excretion machinery is upregulated with increasing dietary K+, but the role of accompanying dietary anions remains inadequately characterized. Poorly absorbable anions, including [Formula: see text], are thought to increase K+ secretion through a transepithelial voltage effect. Here, we tested if they also influence the K+ secretion machinery. Wild-type mice, aldosterone synthase (AS) knockout (KO) mice, or pendrin KO mice were randomized to control, high-KCl, or high-KHCO3 diets. The K+ secretory capacity was assessed in balance experiments. Protein abundance, modification, and localization of K+-secretory transporters were evaluated by Western blot analysis and confocal microscopy. Feeding the high-KHCO3 diet increased urinary K+ excretion and the transtubular K+ gradient significantly more than the high-KCl diet, coincident with more pronounced upregulation of epithelial Na+ channels (ENaC) and renal outer medullary K+ (ROMK) channels and apical localization in the distal nephron. Experiments in AS KO mice revealed that the enhanced effects of [Formula: see text] were aldosterone independent. The high-KHCO3 diet also uniquely increased the large-conductance Ca2+-activated K+ (BK) channel ß4-subunit, stabilizing BKα on the apical membrane, the Cl-/[Formula: see text] exchanger, pendrin, and the apical KCl cotransporter (KCC3a), all of which are expressed specifically in pendrin-positive intercalated cells. Experiments in pendrin KO mice revealed that pendrin was required to increase K+ excretion with the high-KHCO3 diet. In summary, [Formula: see text] stimulates K+ excretion beyond a poorly absorbable anion effect, upregulating ENaC and ROMK in principal cells and BK, pendrin, and KCC3a in pendrin-positive intercalated cells. The adaptive mechanism prevents hyperkalemia and alkalosis with the consumption of alkaline ash-rich diets but may drive K+ wasting and hypokalemia in alkalosis.NEW & NOTEWORTHY Dietary anions profoundly impact K+ homeostasis. Here, we found that a K+-rich diet, containing [Formula: see text] as the counteranion, enhances the electrogenic K+ excretory machinery, epithelial Na+ channels, and renal outer medullary K+ channels, much more than a high-KCl diet. It also uniquely induces KCC3a and pendrin, in B-intercalated cells, providing an electroneutral KHCO3 secretion pathway. These findings reveal new K+ balance mechanisms that drive adaption to alkaline and K+-rich foods, which should guide new treatment strategies for K+ disorders.


Subject(s)
Alkalosis , Potassium , Animals , Mice , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Anions/metabolism , Diet , Mice, Knockout , Potassium/metabolism , Potassium, Dietary/metabolism , Sodium/metabolism , Sulfate Transporters/genetics
4.
Adv Kidney Dis Health ; 30(2): 137-147, 2023 03.
Article in English | MEDLINE | ID: mdl-36868729

ABSTRACT

Potassium channels are expressed in virtually all cell types, and their activity is the dominant determinant of cellular membrane potential. As such, potassium flux is a key regulator of many cellular processes including the regulation of action potentials in excitable cells. Subtle changes in extracellular potassium can initiate signaling processes vital for survival (insulin signaling) while more extreme and chronic changes may lead to pathological states (acid-base disturbances and cardiac arrhythmia). While many factors acutely influence extracellular potassium levels, it is principally the role of the kidneys to maintain potassium balance by matching urinary excretion with dietary intake. When this balance is disrupted, human health is negatively impacted. In this review, we discuss evolving views of dietary potassium intake as means of preventing and mitigating diseases. We also provide an update on a molecular pathway called the potassium switch, a mechanism by which extracellular potassium regulates distal nephron sodium reabsorption. Finally, we review recent literature describing how several popular therapeutics influence potassium homeostasis.


Subject(s)
Kidney , Urinary Tract Physiological Phenomena , Humans , Action Potentials , Biological Transport , Potassium
5.
Am J Physiol Renal Physiol ; 320(5): F748-F760, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33749322

ABSTRACT

The association between diabetes insipidus (DI) and chronic dietary K+ deprivation is well known, but it remains uncertain how the disorder develops and whether it is influenced by the sexual dimorphism in K+ handling. Here, we determined the plasma K+ (PK) threshold for DI in male and female mice and ascertained if DI is initiated by polydipsia or by a central or nephrogenic defect. C57BL6J mice were randomized to a control diet or to graded reductions in dietary K+ for 8 days, and kidney function and transporters involved in water balance were characterized. We found that male and female mice develop polyuria and secondary polydipsia. Altered water balance coincided with a decrease in aquaporin-2 (AQP2) phosphorylation and apical localization despite increased levels of the vasopressin surrogate marker copeptin. No change in the protein abundance of urea transporter-A1 was observed. The Na+-K+-2Cl- cotransporter decreased only in males. Desmopressin treatment failed to reverse water diuresis in K+-restricted mice. These findings indicate that even a small fall in PK is associated with nephrogenic DI (NDI), coincident with the development of altered AQP2 regulation, implicating low PK as a causal trigger of NDI. We found that PK decreased more in females, and, consequently, females were more prone to develop NDI. Together, these data indicate that AQP2 regulation is disrupted by a small decrease in PK and that the response is influenced by sexual dimorphism in K+ handling. These findings provide new insights into the mechanisms linking water and K+ balances and support defining the disorder as "potassium-dependent NDI."NEW & NOTEWORTHY This study shows that aquaporin-2 regulation is disrupted by a small fall in plasma potassium levels and the response is influenced by sexual dimorphism in renal potassium handling. The findings provided new insights into the mechanisms by which water balance is altered in dietary potassium deficiency and support defining the disorder as "potassium-dependent nephrogenic diabetes insipidus."


Subject(s)
Antidiuretic Agents/pharmacology , Deamino Arginine Vasopressin/pharmacology , Diabetes Insipidus, Nephrogenic/drug therapy , Drug Resistance , Kidney/drug effects , Potassium Deficiency/complications , Potassium, Dietary/metabolism , Animals , Aquaporin 2/metabolism , Diabetes Insipidus, Nephrogenic/etiology , Diabetes Insipidus, Nephrogenic/metabolism , Diabetes Insipidus, Nephrogenic/physiopathology , Disease Models, Animal , Female , Kidney/metabolism , Kidney/physiopathology , Male , Mice, Inbred C57BL , Phosphorylation , Potassium Deficiency/metabolism , Potassium Deficiency/physiopathology , Potassium, Dietary/blood , Risk Factors , Sex Characteristics , Water-Electrolyte Balance/drug effects
6.
J Am Soc Nephrol ; 29(5): 1411-1425, 2018 05.
Article in English | MEDLINE | ID: mdl-29483157

ABSTRACT

Background Hyperkalemia in association with metabolic acidosis that are out of proportion to changes in glomerular filtration rate defines type 4 renal tubular acidosis (RTA), the most common RTA observed, but the molecular mechanisms underlying the associated metabolic acidosis are incompletely understood. We sought to determine whether hyperkalemia directly causes metabolic acidosis and, if so, the mechanisms through which this occurs.Methods We studied a genetic model of hyperkalemia that results from early distal convoluted tubule (DCT)-specific overexpression of constitutively active Ste20/SPS1-related proline-alanine-rich kinase (DCT-CA-SPAK).Results DCT-CA-SPAK mice developed hyperkalemia in association with metabolic acidosis and suppressed ammonia excretion; however, titratable acid excretion and urine pH were unchanged compared with those in wild-type mice. Abnormal ammonia excretion in DCT-CA-SPAK mice associated with decreased proximal tubule expression of the ammonia-generating enzymes phosphate-dependent glutaminase and phosphoenolpyruvate carboxykinase and overexpression of the ammonia-recycling enzyme glutamine synthetase. These mice also had decreased expression of the ammonia transporter family member Rhcg and decreased apical polarization of H+-ATPase in the inner stripe of the outer medullary collecting duct. Correcting the hyperkalemia by treatment with hydrochlorothiazide corrected the metabolic acidosis, increased ammonia excretion, and normalized ammoniagenic enzyme and Rhcg expression in DCT-CA-SPAK mice. In wild-type mice, induction of hyperkalemia by administration of the epithelial sodium channel blocker benzamil caused hyperkalemia and suppressed ammonia excretion.Conclusions Hyperkalemia decreases proximal tubule ammonia generation and collecting duct ammonia transport, leading to impaired ammonia excretion that causes metabolic acidosis.


Subject(s)
Ammonia/urine , Hyperkalemia/genetics , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/metabolism , Protein Serine-Threonine Kinases/genetics , Acidosis/etiology , Aldosterone/urine , Amiloride/analogs & derivatives , Animals , Cation Transport Proteins/metabolism , Diuretics/therapeutic use , Glutaminase/metabolism , Hydrochlorothiazide/therapeutic use , Hydrogen-Ion Concentration , Hyperkalemia/blood , Hyperkalemia/complications , Hyperkalemia/drug therapy , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Proton-Translocating ATPases/metabolism , Urinalysis
7.
J Am Soc Nephrol ; 28(9): 2597-2606, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28442491

ABSTRACT

Aberrant activation of with no lysine (WNK) kinases causes familial hyperkalemic hypertension (FHHt). Thiazide diuretics treat the disease, fostering the view that hyperactivation of the thiazide-sensitive sodium-chloride cotransporter (NCC) in the distal convoluted tubule (DCT) is solely responsible. However, aberrant signaling in the aldosterone-sensitive distal nephron (ASDN) and inhibition of the potassium-excretory renal outer medullary potassium (ROMK) channel have also been implicated. To test these ideas, we introduced kinase-activating mutations after Lox-P sites in the mouse Stk39 gene, which encodes the terminal kinase in the WNK signaling pathway, Ste20-related proline-alanine-rich kinase (SPAK). Renal expression of the constitutively active (CA)-SPAK mutant was specifically targeted to the early DCT using a DCT-driven Cre recombinase. CA-SPAK mice displayed thiazide-treatable hypertension and hyperkalemia, concurrent with NCC hyperphosphorylation. However, thiazide-mediated inhibition of NCC and consequent restoration of sodium excretion did not immediately restore urinary potassium excretion in CA-SPAK mice. Notably, CA-SPAK mice exhibited ASDN remodeling, involving a reduction in connecting tubule mass and attenuation of epithelial sodium channel (ENaC) and ROMK expression and apical localization. Blocking hyperactive NCC in the DCT gradually restored ASDN structure and ENaC and ROMK expression, concurrent with the restoration of urinary potassium excretion. These findings verify that NCC hyperactivity underlies FHHt but also reveal that NCC-dependent changes in the driving force for potassium secretion are not sufficient to explain hyperkalemia. Instead, a DCT-ASDN coupling process controls potassium balance in health and becomes aberrantly activated in FHHt.


Subject(s)
Hydrochlorothiazide/pharmacology , Kidney Tubules, Distal/pathology , Protein Serine-Threonine Kinases/metabolism , Pseudohypoaldosteronism/metabolism , Sodium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 3/metabolism , Aldosterone/metabolism , Animals , Blood Pressure/drug effects , Epithelial Sodium Channels/metabolism , Hydrochlorothiazide/therapeutic use , Kidney Tubules, Distal/metabolism , Mice , Natriuresis/drug effects , Phosphorylation , Potassium/urine , Potassium Channels, Inwardly Rectifying/metabolism , Protein Serine-Threonine Kinases/genetics , Pseudohypoaldosteronism/drug therapy , Pseudohypoaldosteronism/genetics , Pseudohypoaldosteronism/urine , Signal Transduction , Sodium Chloride Symporter Inhibitors/therapeutic use
8.
J Clin Invest ; 125(5): 2136-50, 2015 May.
Article in English | MEDLINE | ID: mdl-25893600

ABSTRACT

Thiazide diuretics are used to treat hypertension; however, compensatory processes in the kidney can limit antihypertensive responses to this class of drugs. Here, we evaluated compensatory pathways in SPAK kinase-deficient mice, which are unable to activate the thiazide-sensitive sodium chloride cotransporter NCC (encoded by Slc12a3). Global transcriptional profiling, combined with biochemical, cell biological, and physiological phenotyping, identified the gene expression signature of the response and revealed how it establishes an adaptive physiology. Salt reabsorption pathways were created by the coordinate induction of a multigene transport system, involving solute carriers (encoded by Slc26a4, Slc4a8, and Slc4a9), carbonic anhydrase isoforms, and V-type H⁺-ATPase subunits in pendrin-positive intercalated cells (PP-ICs) and ENaC subunits in principal cells (PCs). A distal nephron remodeling process and induction of jagged 1/NOTCH signaling, which expands the cortical connecting tubule with PCs and replaces acid-secreting α-ICs with PP-ICs, were partly responsible for the compensation. Salt reabsorption was also activated by induction of an α-ketoglutarate (α-KG) paracrine signaling system. Coordinate regulation of a multigene α-KG synthesis and transport pathway resulted in α-KG secretion into pro-urine, as the α-KG-activated GPCR (Oxgr1) increased on the PP-IC apical surface, allowing paracrine delivery of α-KG to stimulate salt transport. Identification of the integrated compensatory NaCl reabsorption mechanisms provides insight into thiazide diuretic efficacy.


Subject(s)
Blood Pressure/physiology , Chlorides/urine , Gitelman Syndrome/physiopathology , Natriuresis/physiology , Nephrons/metabolism , Renal Reabsorption/physiology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Ammonia/metabolism , Animals , Biological Transport , Carbonic Anhydrases/genetics , Carbonic Anhydrases/physiology , Disease Models, Animal , Enzyme Activation , Epithelial Sodium Channels/physiology , Gene Expression Profiling , Gene Regulatory Networks , Gitelman Syndrome/genetics , Ketoglutaric Acids/metabolism , Kidney Glomerulus/metabolism , Male , Mice , Mice, Knockout , Natriuresis/genetics , Paracrine Communication , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Notch/physiology , Receptors, Purinergic P2/physiology , Signal Transduction , Sodium Chloride/pharmacokinetics , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/physiology , Solute Carrier Family 12, Member 3/metabolism
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 Biol Chem ; 289(13): 9121-35, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24550400

ABSTRACT

The highly sialylated vascular endothelial surface undergoes changes in sialylation upon adopting the migratory/angiogenic phenotype. We recently established endothelial cell (EC) expression of NEU1 sialidase (Cross, A. S., Hyun, S. W., Miranda-Ribera, A., Feng, C., Liu, A., Nguyen, C., Zhang, L., Luzina, I. G., Atamas, S. P., Twaddell, W. S., Guang, W., Lillehoj, E. P., Puché, A. C., Huang, W., Wang, L. X., Passaniti, A., and Goldblum, S. E. (2012) NEU1 and NEU3 sialidase activity expressed in human lung microvascular endothelia. NEU1 restrains endothelial cell migration whereas NEU3 does not. J. Biol. Chem. 287, 15966-15980). We asked whether NEU1 might regulate EC capillary-like tube formation on a Matrigel substrate. In human pulmonary microvascular ECs (HPMECs), prior silencing of NEU1 did not alter tube formation. Infection of HPMECs with increasing multiplicities of infection of an adenovirus encoding for catalytically active WT NEU1 dose-dependently impaired tube formation, whereas overexpression of either a catalytically dead NEU1 mutant, NEU1-G68V, or another human sialidase, NEU3, did not. NEU1 overexpression also diminished EC adhesion to the Matrigel substrate and restrained EC migration in a wounding assay. In HPMECs, the adhesion molecule, CD31, also known as platelet endothelial cell adhesion molecule-1, was sialylated via α2,6-linkages, as shown by Sambucus nigra agglutinin lectin blotting. NEU1 overexpression increased CD31 binding to Arachis hypogaea or peanut agglutinin lectin, indicating CD31 desialylation. In the postconfluent state, when CD31 ectodomains are homophilically engaged, NEU1 was recruited to and desialylated CD31. In postconfluent ECs, CD31 was desialylated compared with subconfluent cells, and prior NEU1 silencing completely protected against CD31 desialylation. Prior CD31 silencing and the use of CD31-null ECs each abrogated the NEU1 inhibitory effect on EC tube formation. Sialyltransferase 6 GAL-I overexpression increased α2,6-linked CD31 sialylation and dose-dependently counteracted NEU1-mediated inhibition of EC tube formation. These combined data indicate that catalytically active NEU1 inhibits in vitro angiogenesis through desialylation of its substrate, CD31.


Subject(s)
Capillaries/cytology , Endothelial Cells/metabolism , Lung/blood supply , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Animals , Antigens, CD/genetics , Capillaries/physiology , Cell Adhesion , Cell Movement , Endothelial Cells/cytology , Humans , Mice , Neovascularization, Physiologic , Protein Transport , Sialyltransferases/genetics
11.
J Biol Chem ; 287(45): 37673-90, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-22977235

ABSTRACT

STE20/SPS-1-related proline-alanine-rich protein kinase (SPAK) and oxidative stress-related kinase (OSR1) activate the potassium-dependent sodium-chloride co-transporter, NKCC2, and thiazide-sensitive sodium-chloride cotransporter, NCC, in vitro, and both co-localize with a kinase regulatory molecule, Cab39/MO25α, at the apical membrane of the thick ascending limb (TAL) and distal convoluted tubule (DCT). Yet genetic ablation of SPAK in mice causes a selective loss of NCC function, whereas NKCC2 becomes hyperphosphorylated. Here, we explore the underlying mechanisms in wild-type and SPAK-null mice. Unlike in the DCT, OSR1 remains at the TAL apical membrane of KO mice where it is accompanied by an increase in the active, phosphorylated form of AMP-activated kinase. We found an alterative SPAK isoform (putative SPAK2 form), which modestly inhibits co-transporter activity in vitro, is more abundant in the medulla than the cortex. Thus, enhanced NKCC2 phosphorylation in the SPAK knock-out may be explained by removal of inhibitory SPAK2, sustained activity of OSR1, and activation of other kinases. By contrast, the OSR1/SPAK/M025α signaling apparatus is disrupted in the DCT. OSR1 becomes largely inactive and displaced from M025α and NCC at the apical membrane, and redistributes to dense punctate structures, containing WNK1, within the cytoplasm. These changes are paralleled by a decrease in NCC phosphorylation and a decrease in the mass of the distal convoluted tubule, exclusive to DCT1. As a result of the dependent nature of OSR1 on SPAK in the DCT, NCC is unable to be activated. Consequently, SPAK(-/-) mice are highly sensitive to dietary salt restriction, displaying prolonged negative sodium balance and hypotension.


Subject(s)
Nephrons/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Drug/metabolism , Symporters/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , Calcium-Binding Proteins , Gene Expression , Isoenzymes/genetics , Isoenzymes/metabolism , Kidney Cortex/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Distal/metabolism , Loop of Henle/metabolism , Mice , Mice, Knockout , Phosphorylation , Potassium, Dietary/administration & dosage , Potassium, Dietary/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Drug/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sodium Chloride Symporters/genetics , Sodium Chloride Symporters/metabolism , Sodium Chloride, Dietary/administration & dosage , Sodium Chloride, Dietary/metabolism , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1 , Solute Carrier Family 12, Member 3 , Symporters/genetics
12.
Curr Opin Nephrol Hypertens ; 20(5): 512-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21670674

ABSTRACT

PURPOSE OF REVIEW: This review summarizes recent studies of hypertension associated with a defect in renal K excretion due to genetic deletions of various components of the large, Ca-activated K channel (BK), and describes new evidence and theories regarding K secretory roles of BK in intercalated cells. RECENT FINDINGS: Isolated perfused tubule methods have revealed the importance of BK in flow-induced K secretion. Subsequently, mice with genetically deleted BK subunits revealed the complexities of BK-mediated K secretion. Deletion of BKα results in extreme aldosteronism, hypertension, and an absence of flow-induced K secretion. Deletion of the BKß1 ancillary subunit results in decreased handling of a K load, increased plasma K, mild aldosteronism and hypertension that is exacerbated by a high K diet. Deletion of BKß4 (ß4KO) leads to insufficient K handling, high plasma K, fluid retention, but with milder hypertension. Fluid retention in ß4KO may be the result of insufficient flow-induced secretion of adenosine triphosphate (ATP), which normally inhibits epithelial Na channels (ENaCs). SUMMARY: Classical physiological analysis of electrolyte handling in knockout mice has enlightened our understanding of the mechanism of handling K loads by renal K channels. Studies have focused on the different roles of BK-α/ß1 and BK-α/ß4 in the kidney. BKß1 hypertension may be a 'three-hit' hypertension, involving a K secretory defect, elevated production of aldosterone, and increased vascular tone. The disorders observed in BK knockout mice have shed new insights on the importance of proper renal K handling for maintaining volume balance and blood pressure.


Subject(s)
Hypertension/metabolism , Kidney Tubules/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Potassium/metabolism , Aldosterone/metabolism , Animals , Blood Pressure , Humans , Hypertension/physiopathology , Ion Transport , Kidney Tubules/physiopathology , Large-Conductance Calcium-Activated Potassium Channels/deficiency , Large-Conductance Calcium-Activated Potassium Channels/genetics , Mice , Mice, Knockout , Potassium/blood , Water-Electrolyte Balance
13.
Am J Physiol Renal Physiol ; 300(6): F1385-93, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21454252

ABSTRACT

ROMK channels are well-known to play a central role in renal K secretion, but the absence of highly specific and avid-ROMK antibodies has presented significant roadblocks toward mapping the extent of expression along the entire distal nephron and determining whether surface density of these channels is regulated in response to physiological stimuli. Here, we prepared new ROMK antibodies verified to be highly specific, using ROMK knockout mice as a control. Characterization with segmental markers revealed a more extensive pattern of ROMK expression along the entire distal nephron than previously thought, localizing to distal convoluted tubule regions, DCT1 and DCT2; the connecting tubule (CNT); and cortical collecting duct (CD). ROMK was diffusely distributed in intracellular compartments and at the apical membrane of each tubular region. Apical labeling was significantly increased by high-K diet in DCT2, CNT1, CNT2, and CD (P < 0.05) but not in DCT1. Consistent with the large increase in apical ROMK, dramatically increased mature glycosylation was observed following dietary potassium augmentation. We conclude 1) our new antibody provides a unique tool to characterize ROMK channel localization and expression and 2) high-K diet causes a large increase in apical expression of ROMK in DCT2, CNT, and CD but not in DCT1, indicating that different regulatory mechanisms are involved in K diet-regulated ROMK channel functions in the distal nephron.


Subject(s)
Nephrons/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Potassium, Dietary/metabolism , Analysis of Variance , Animals , Blotting, Western , Mice , Mice, Knockout , Potassium Channels, Inwardly Rectifying/genetics
14.
Kidney Int ; 78(10): 956-62, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20720523

ABSTRACT

Large, Ca-activated K channels (BK) are comprised of an α pore (BKα) and one of four ß subunits (BKß1-4). When the gene for BKß1 is knocked out (BKß1-KO), the result is increased myogenic tone of vascular smooth muscle and hypertension. We reexamined whether the hypertension is entirely due to increased vascular tone, because most monogenic forms of hypertension have renal origins and BKß1 resides in renal connecting tubule (CNT) cells. Moreover, BKß1 is localized in the adrenal glands, where it may control production of aldosterone. This review will summarize our report that a majority of the hypertension of BKß1-KO is the result of insufficient handling of dietary K, resulting in increased plasma K and hyperaldosteronism, the latter promoting Na and fluid retention. The fluid retention and hypertension are exacerbated by a high-K diet and reduced by eplerenone, an aldosterone receptor inhibitor. Genetic knockout of BKß4 (BKß4-KO), which resides in intercalated cells, also exhibits deficient K excretion, fluid retention, and mild hypertension that is not exacerbated when animals are treated with a high-K diet. These results show that the hypertension associated with BKß1-KO occurs because of enhanced fluid retention, as well as because of the previously described vascular dysfunction.


Subject(s)
Hyperaldosteronism/physiopathology , Hypertension/physiopathology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Adrenal Medulla/physiopathology , Animals , Disease Models, Animal , Humans , Hypertension/genetics , Kidney/physiopathology , Large-Conductance Calcium-Activated Potassium Channels/genetics , Mice , Mice, Knockout , Muscle, Smooth, Vascular/physiopathology , Potassium/metabolism
15.
Am J Physiol Renal Physiol ; 299(3): F507-16, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20576683

ABSTRACT

Large-conductance, calcium-activated potassium channels (BK) are expressed in principal cells (PC) and intercalated cells (IC) in mammalian nephrons as BK-alpha/beta1 and BK-alpha/beta4, respectively. IC, which protrude into the lumens of tubules, express substantially more BK than PC despite lacking sufficient Na-K-ATPase to support K secretion. We previously showed in mice that IC exhibit size reduction when experiencing high distal flows induced by a high-K diet. We therefore tested the hypothesis that BK-alpha/beta4 are regulators of IC volume via a shear stress (tau)-induced, calcium-dependent mechanism, resulting in a reduction in intracellular K content. We determined by Western blot and immunocytochemical analysis that C11-Madin-Darby canine kidney cells contained a predominance of BK-alpha/beta4. To determine the role of BK-alpha/beta4 in tau-induced volume reduction, we exposed C11 cells to tau and measured K efflux by flame photometry and cell volume by calcein staining, which changes inversely to cell volume. With 10 dynes/cm(2), calcein intensity significantly increased 39% and monovalent cationic content decreased significantly by 37% compared with static conditions. Furthermore, the shear-induced K loss from C11 was abolished by the reduction of extracellular calcium, addition of 5 mM TEA, or BK-beta4 small interfering (si) RNA, but not by addition of nontarget siRNA. These results show that BK-alpha/beta4 plays a role in shear-induced K loss from IC, suggesting that BK-alpha/beta4 regulate IC volume during high-flow conditions. Furthermore, these results support the use of C11 cells as in vitro models for studying BK-related functions in IC of the kidney.


Subject(s)
Cell Size , Kidney/cytology , Kidney/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Stress, Mechanical , Animals , Calcium/metabolism , Cell Line , Dogs , Models, Animal , Potassium/metabolism , Potassium Channel Blockers/pharmacology , RNA, Small Interfering/pharmacology , Tetraethylammonium/pharmacology
16.
J Am Soc Nephrol ; 21(4): 634-45, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20299355

ABSTRACT

The large-conductance, calcium-activated potassium (BK) channels help eliminate potassium in mammals consuming potassium-rich diets. In the distal nephron, principal cells contain BK-alpha/beta1 channels and intercalated cells contain BK-alpha/beta4 channels. We studied whether BK-beta4-deficient mice (Kcnmb4(-/-)) have altered renal sodium and potassium clearances compared with wild-type mice when fed a regular or potassium-rich diet for ten days. We did not detect differences in urinary flow or fractional excretions of potassium (FE(K)) or sodium (FE(Na)) between Kcnmb4-deficient and wild-type mice fed a regular diet. However, a potassium-rich diet led to >4-fold increases in urinary flows for both groups of mice, although Kcnmb4-deficient mice exhibited less urinary flow, higher plasma potassium concentration, more fluid retention, and significantly lower FE(K) and FE(Na) than wild-type mice despite similar plasma aldosterone levels. Immunohistochemical analysis revealed increased basolateral Na-K-ATPase in principal cells of all potassium-adapted mice, but expression of Na-K-ATPase in intercalated cells was >10-fold lower. The size of intercalated cells reduced and luminal volume increased among potassium-adapted wild-type but not Kcnmb4-deficient mice. Paradoxically, this led to increased urinary fluid velocity in potassium-adapted Kcnmb4-deficient mice compared with wild-type mice. Taken together, these data suggest that BK-alpha/beta4 channels in intercalated cells reduce cell size, increasing luminal volume to accommodate higher distal flow rates during potassium adaptation. These changes streamline flow across the principal cells, producing gradients more favorable for potassium secretion and less favorable for sodium reabsorption.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/physiology , Potassium/metabolism , Sodium/metabolism , Adaptation, Physiological , Animals , Mice
17.
Proc Natl Acad Sci U S A ; 106(28): 11800-5, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19556540

ABSTRACT

Mice lacking the beta1-subunit (gene, Kcnmb1; protein, BK-beta1) of the large Ca-activated K channel (BK) are hypertensive. This phenotype is thought to result from diminished BK currents in vascular smooth muscle where BK-beta1 is an ancillary subunit. However, the beta1-subunit is also expressed in the renal connecting tubule (CNT), a segment of the aldosterone-sensitive distal nephron, where it associates with BK and facilitates K secretion. Because of the correlation between certain forms of hypertension and renal defects, particularly in the distal nephron, it was determined whether the hypertension of Kcnmb1(-/-) has a renal origin. We found that Kcnmb1(-/-) are hypertensive, volume expanded, and have reduced urinary K and Na clearances. These conditions are exacerbated when the animals are fed a high K diet (5% K; HK). Supplementing HK-fed Kcnmb1(-/-) with eplerenone (mineralocorticoid receptor antagonist) corrected the fluid imbalance and more than 70% of the hypertension. Finally, plasma [aldo] was elevated in Kcnmb1(-/-) under basal conditions (control diet, 0.6% K) and increased significantly more than wild type when fed the HK diet. We conclude that the majority of the hypertension of Kcnmb1(-/-) is due to aldosteronism, resulting from renal potassium retention and hyperkalemia.


Subject(s)
Hyperaldosteronism/complications , Hyperkalemia/complications , Hypertension/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/deficiency , Potassium/metabolism , Analysis of Variance , Animals , Eplerenone , Hyperaldosteronism/etiology , Hypertension/etiology , Hypertension/metabolism , Kidney Tubules, Collecting/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Mice , Mice, Knockout , Spironolactone/analogs & derivatives
18.
Am J Physiol Renal Physiol ; 297(2): F420-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19458125

ABSTRACT

On a low-Na(+) diet (LNa(+)), urinary Na(+) loss is prevented by aldosterone-induced Na(+) reabsorption through epithelial Na(+) channels (ENaC) in the connecting tubules (CNT) and cortical collecting ducts (CCD). However, the mechanism whereby K(+) loss is minimized and Na(+) reabsorption is maximized in the face of a reduced lumen-to-bath Na(+) gradient is not fully understood. The large-conductance calcium-activated potassium channel (BK)beta1 subunit (gene: Kcnmb1), which has a role in K(+) secretion in the CNT, is absent in the CCD in mice on a control diet. We hypothesized that BKalpha/beta1 helps to maximize Na(+) reabsorption during Na(+) deficiency. With LNa(+), the Na(+) clearance of Kcnmb1-mutant mice (Kcnmb1(-/-)) was 45% greater and the plasma Na(+) concentration and osmolality were significantly reduced compared with wild-type mouse (WT) controls. On LNa(+), Kcnmb1(-/-) exhibited exacerbated volume depletion (higher Hct and weight loss) compared with WT. LNa(+), which did not affect the mean arterial blood pressure (MAP) of WT, significantly reduced MAP of Kcnmb1(-/-). The plasma aldosterone concentration of Kcnmb1(-/-) on LNa(+) was significantly elevated compared with Kcnmb1(-/-) on a control diet but was not different from WT on LNa(+). Immunohistochemical staining revealed that BKalpha and BKbeta1, which were absent in the principal cells (PCs) of the CCD, were localized on the basolateral membrane (BSM) of PCs of WT on LNa(+). Moreover, BKalpha was absent from the BSM of PCs of Na(+)-deficient Kcnmb1(-/-). We conclude that part of the mechanism to maximize Na(+) reabsorption during Na(+) deficiency is the placement of BKalpha/beta1 channels in the BSM of CCD PCs.


Subject(s)
Diet, Sodium-Restricted , Hyponatremia/blood , Kidney Tubules, Collecting/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Potassium/metabolism , Sodium/metabolism , Aldosterone/blood , Animals , Biological Transport , Blood Pressure , Cell Membrane/metabolism , Creatinine/blood , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmolar Concentration , Plasma Volume , Potassium/blood , Potassium/urine , Protein Transport , Sodium/blood , Sodium/urine , Sodium, Dietary/metabolism , Urodynamics , Water-Electrolyte Balance
19.
Am J Physiol Renal Physiol ; 294(6): F1465-72, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18367663

ABSTRACT

Glomerular hyperfiltration and mesangial expansion have been described in mouse models of a hyperinsulinemic early stage of type 2 diabetes mellitus (DM). Large-conductance Ca(2+)-activated K(+) channels (BK) have been linked to relaxation of human mesangial cells (MC) and may contribute to MC expansion and hyperfiltration. We hypothesized that high insulin levels increase BK activity in MC by increasing the number and/or open probability (P(o)) of BK in the plasma membrane. With the use of the patch-clamp technique, BK activity was analyzed in cultured MC exposed to normal insulin (1 nM) and high insulin (100 nM) for a 48-h period. The mean P(o) and the percentage of patches (cell attached) with detected BK increased by 100% in the insulin-treated cells. Real-time PCR revealed that insulin increased mRNA of BK-alpha. Western blot revealed an insulin-stimulated increase in BK-alpha from both total cellular and plasma membrane protein fractions. The mitogen-activated protein kinase (MAPK) inhibitors PD-098059 and U-0126 attenuated the insulin-induced increase in BK-alpha expression. PD-098059 inhibited insulin-stimulated phosphorylation of extracellular signal-regulated kinase 1/2 in MC. An insulin-stimulated increase also was found for total cellular BK-beta(1), the accessory subunit of BK in MC. A similar increase in BK-alpha mRNA and protein was evoked by an insulin-like growth factor I analog. Glomeruli, isolated from hyperinsulinemic early stage type 2 DM mice, exhibited increased BK-alpha mRNA by real-time PCR and protein by immunohistochemical staining and Western blot. These results indicate that insulin activates BK in the plasma membrane of MC and stimulates, via MAPK, an increase in cellular and plasma membrane BK-alpha.


Subject(s)
Hypoglycemic Agents/metabolism , Insulin/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , MAP Kinase Signaling System/physiology , Mesangial Cells/metabolism , Animals , Butadienes/pharmacology , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/metabolism , Dietary Fats/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Hyperinsulinism/complications , Hyperinsulinism/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , MAP Kinase Signaling System/drug effects , Male , Mesangial Cells/cytology , Mesangial Cells/drug effects , Mice , Mice, Inbred C57BL , Nitriles/pharmacology , Patch-Clamp Techniques , RNA, Messenger/metabolism
20.
Curr Opin Nephrol Hypertens ; 16(5): 430-6, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17693758

ABSTRACT

PURPOSE OF REVIEW: Large, BK (calcium-activated potassium) channels are now regarded as relevant players in many aspects of renal physiology, including potassium secretion. This review will highlight recent discoveries regarding the function and localization of BK in the kidney. RECENT FINDINGS: Patch clamp electrophysiology has revealed BK in cultured podocytes, glomerular mesangial cells, and in several tubule segments including principal cells (connecting tubules/principal cells), and intercalated cells of connecting tubules and cortical collecting ducts. Flow-induced potassium secretion is mediated by BK in the distal nephron and may be partly the result of shear stress-induced increases in cell calcium concentrations. ROMK-/- and wild-type mice on a high potassium diet exhibit BK-mediated potassium secretion, and studies of BK-alpha-/- and BK-beta1-/- mice suggest that flow-induced potassium secretion is mediated by BK-alpha/beta1, which is specifically localized in the apical membrane of the connecting tubule of the mouse and connecting tubule plus initial cortical collecting duct of the rabbit. SUMMARY: BK channels, located in glomerular cells and in many nephron segments, especially mediate potassium secretion in the combined condition of potassium adaptation and high flow. Understanding the molecular makeup of BK in specific renal cells and the dietary and physiological conditions for their expression can yield improved potassium-sparing compounds.


Subject(s)
Kidney/metabolism , Large-Conductance Calcium-Activated Potassium Channels/physiology , Animals , Calcium/metabolism , Cyclic GMP/metabolism , Electrophysiology , Guanylate Kinases/metabolism , Humans , Kidney Glomerulus/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Mesangial Cells/metabolism , Mice , Mice, Transgenic , Patch-Clamp Techniques , Potassium/chemistry , Potassium/metabolism , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...