Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Physiol Renal Physiol ; 307(7): F833-43, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25100281

ABSTRACT

The kidney adjusts K⁺ excretion to match intake in part by regulation of the activity of apical K⁺ secretory channels, including renal outer medullary K⁺ (ROMK)-like K⁺ channels, in the cortical collecting duct (CCD). ANG II inhibits ROMK channels via the ANG II type 1 receptor (AT1R) during dietary K⁺ restriction. Because AT1Rs and ANG II type 2 receptors (AT2Rs) generally function in an antagonistic manner, we sought to characterize the regulation of ROMK channels by the AT2R. Patch-clamp experiments revealed that ANG II increased ROMK channel activity in CCDs isolated from high-K⁺ (HK)-fed but not normal K⁺ (NK)-fed rats. This response was blocked by PD-123319, an AT2R antagonist, but not by losartan, an AT1R antagonist, and was mimicked by the AT2R agonist CGP-42112. Nitric oxide (NO) synthase is present in CCD cells that express ROMK channels. Blockade of NO synthase with N-nitro-l-arginine methyl ester and free NO with 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide potassium salt completely abolished ANG II-stimulated ROMK channel activity. NO enhances the synthesis of cGMP, which inhibits phosphodiesterases (PDEs) that normally degrade cAMP; cAMP increases ROMK channel activity. Pretreatment of CCDs with IBMX, a broad-spectrum PDE inhibitor, or cilostamide, a PDE3 inhibitor, abolished the stimulatory effect of ANG II on ROMK channels. Furthermore, PKA inhibitor peptide, but not an activator of the exchange protein directly activated by cAMP (Epac), also prevented the stimulatory effect of ANG II. We conclude that ANG II acts at the AT2R to stimulate ROMK channel activity in CCDs from HK-fed rats, a response opposite to that mediated by the AT1R in dietary K⁺-restricted animals, via a NO/cGMP pathway linked to a cAMP-PKA pathway.


Subject(s)
Kidney Tubules, Collecting/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Potassium, Dietary/metabolism , Receptor, Angiotensin, Type 2/metabolism , Adaptation, Physiological , Animals , Female , Male , Patch-Clamp Techniques , Rats, Sprague-Dawley
2.
Am J Physiol Renal Physiol ; 302(1): F205-15, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21957178

ABSTRACT

Epithelial Na(+) channel (ENaC)-mediated Na(+) absorption and BK channel-mediated K(+) secretion in the cortical collecting duct (CCD) are modulated by flow, the latter requiring an increase in intracellular Ca(2+) concentration ([Ca(2+)](i)), microtubule integrity, and exocytic insertion of preformed channels into the apical membrane. As axial flow modulates HCO(3)(-) reabsorption in the proximal tubule due to changes in both luminal Na(+)/H(+) exchanger 3 and H(+)-ATPase activity (Du Z, Yan Q, Duan Y, Weinbaum S, Weinstein AM, Wang T. Am J Physiol Renal Physiol 290: F289-F296, 2006), we sought to test the hypothesis that flow also regulates H(+)-ATPase activity in the CCD. H(+)-ATPase activity was assayed in individually identified cells in microperfused CCDs isolated from New Zealand White rabbits, loaded with the pH-sensitive dye BCECF, and then subjected to an acute intracellular acid load (NH(4)Cl prepulse technique). H(+)-ATPase activity was defined as the initial rate of bafilomycin-inhibitable cell pH (pH(i)) recovery in the absence of luminal K(+), bilateral Na(+), and CO(2)/HCO(3)(-), from a nadir pH of ∼6.2. We found that 1) an increase in luminal flow rate from ∼1 to 5 nl·min(-1)·mm(-1) stimulated H(+)-ATPase activity, 2) flow-stimulated H(+) pumping was Ca(2+) dependent and required microtubule integrity, and 3) basal and flow-stimulated pH(i) recovery was detected in cells that labeled with the apical principal cell marker rhodamine Dolichos biflorus agglutinin as well as cells that did not. We conclude that luminal flow modulates H(+)-ATPase activity in the rabbit CCD and that H(+)-ATPases therein are present in both principal and intercalated cells.


Subject(s)
Kidney Tubules, Collecting/metabolism , Proton-Translocating ATPases/metabolism , Animals , Calcium/metabolism , Female , Fluoresceins , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/drug effects , Macrolides/pharmacology , Mechanotransduction, Cellular , Proton-Translocating ATPases/antagonists & inhibitors , Rabbits , Urodynamics
3.
Am J Physiol Renal Physiol ; 301(5): F1088-97, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21816753

ABSTRACT

Apical SK/ROMK and BK channels mediate baseline and flow-induced K secretion (FIKS), respectively, in the cortical collecting duct (CCD). BK channels are detected in acid-base transporting intercalated (IC) and Na-absorbing principal (PC) cells. Although the density of BK channels is greater in IC than PC, Na-K-ATPase activity in IC is considered inadequate to sustain high rates of urinary K secretion. To test the hypothesis that basolateral NKCC in the CCD contributes to BK channel-mediated FIKS, we measured net K secretion (J(K)) and Na absorption (J(Na)) at slow (∼1) and fast (∼5 nl·min(-1)·mm(-1)) flow rates in rabbit CCDs microperfused in vitro in the absence and presence of bumetanide, an inhibitor of NKCC, added to the bath. Bumetanide inhibited FIKS but not basal J(K), J(Na), or the flow-induced [Ca(2+)](i) transient necessary for BK channel activation. Addition of luminal iberiotoxin, a BK channel inhibitor, to bumetanide-treated CCDs did not further reduce J(K). Basolateral Cl removal reversibly inhibited FIKS but not basal J(K) or J(Na). Quantitative PCR performed on single CCD samples using NKCC1- and 18S-specific primers and probes and the TaqMan assay confirmed the presence of the transcript in this nephron segment. To identify the specific cell type to which basolateral NKCC is localized, we exploited the ability of NKCC to accept NH(4)(+) at its K-binding site to monitor the rate of bumetanide-sensitive cytosolic acidification after NH(4)(+) addition to the bath in CCDs loaded with the pH indicator dye BCECF. Both IC and PC were found to have a basolateral bumetanide-sensitive NH(4)(+) entry step and NKCC1-specific antibodies labeled the basolateral surfaces of both cell types in CCDs. These results suggest that BK channel-mediated FIKS is dependent on a basolateral bumetanide-sensitive, Cl-dependent transport pathway, proposed to be NKCC1, in both IC and PC in the CCD.


Subject(s)
Kidney Cortex/metabolism , Kidney Tubules, Collecting/metabolism , Large-Conductance Calcium-Activated Potassium Channels/physiology , Potassium/metabolism , Sodium-Potassium-Chloride Symporters/physiology , Animals , Bumetanide/pharmacology , Calcium/metabolism , Cations/metabolism , Diuretics/pharmacology , Fluorescent Dyes , Immunohistochemistry , Kidney Cortex/cytology , Kidney Tubules, Collecting/cytology , Male , Nephrons/metabolism , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Quaternary Ammonium Compounds/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rabbits , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 2
4.
Kidney Int ; 80(8): 832-40, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21796099

ABSTRACT

Recent studies have identified Rhesus proteins as important molecules for ammonia transport in acid-secreting intercalated cells in the distal nephron. Here, we provide evidence for an additional molecule that can mediate NH3/NH4 excretion, the subtype 2 of the hyperpolarization-activated cyclic nucleotide-gated channel family (HCN2), in collecting ducts in rat renal cortex and medulla. Chronic metabolic acidosis in rats did not alter HCN2 protein expression but downregulated the relative abundance of HCN2 mRNA. Its cDNA was identical to the homolog from the brain and the protein was post-translationally modified by N-type glycosylation. Electrophysiological recordings in Xenopus oocytes injected with HCN2 cRNA found that potassium was transported better than ammonium, each of which was transported significantly better than sodium, criteria that are compatible with a role for HCN2 in ammonium transport. In microperfused rat outer medullary collecting duct segments, the initial rate of acidification, upon exposure to a basolateral ammonium chloride pulse, was higher in intercalated than in principal cells. A specific inhibitor of HCN2 (ZD7288) decreased acidification only in intercalated cells from control rats. In rats with chronic metabolic acidosis, the rate of acidification doubled in both intercalated and principal cells; however, ZD7288 had no significant inhibitory effect. Thus, HCN2 is a basolateral ammonium transport pathway of intercalated cells and may contribute to the renal regulation of body pH under basal conditions.


Subject(s)
Ion Channels/physiology , Kidney Tubules, Distal/metabolism , Quaternary Ammonium Compounds/metabolism , Acidosis/metabolism , Animals , Biological Transport , Fluorescent Antibody Technique , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/analysis , Kidney Tubules/metabolism , Potassium Channels , Rats
5.
Am J Physiol Renal Physiol ; 299(1): F255-64, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20427469

ABSTRACT

The fine regulation of Na(+) and K(+) transport takes place in the cortical distal nephron. It is well established that K(+) secretion occurs through apical K(+) channels: the ROMK and the Ca(2+)- and voltage-dependent maxi-K. Previously, we identified the voltage-gated Kv1.3 channel in the inner medulla of the rat kidney (Escobar LI, Martínez-Téllez JC, Salas M, Castilla SA, Carrisoza R, Tapia D, Vázquez M, Bargas J, Bolívar JJ. Am J Physiol Cell Physiol 286: C965-C974, 2004). To examine the role of Kv1.3 in the renal regulation of K(+) homeostasis, we characterized the effect of dietary K(+) on the molecular and functional expression of this channel. We performed real-time-PCR and immunoblot assays in kidneys from rats fed a control (CK; 1.2% wt/wt) or high-K(+) (HK; 10% wt/wt) diet for 5-15 days. Kv1.3 mRNA and protein expression did not change with HK in the whole kidney. However, dietary K(+) loading provoked a change in the cellular distribution of Kv1.3 from the cytoplasm to apical membranes. Immunolocalization of Kv1.3 detected the channel exclusively in the intercalated cells. We investigated whether Kv1.3 mediated K(+) transport in microperfused cortical collecting ducts (CCDs). The HK diet led to an increase in net K(+) transport from 7.4 +/- 1.1 (CK) to 11.4 +/- 1.0 (HK) pmol x min(-1.) mm(-1). Luminal margatoxin, a specific blocker of Kv1.3, decreased net K(+) secretion in HK CCDs to 6.0 +/- 1.6 pmol x min(-1.) mm(-1). Our data provide the first evidence that Kv1.3 channels participate in K(+) secretion and that apical membrane localization of Kv1.3 is enhanced in the intercalated cells by dietary K(+) loading.


Subject(s)
Ion Channel Gating , Kidney/metabolism , Kv1.3 Potassium Channel/metabolism , Potassium, Dietary/metabolism , Animals , Blotting, Western , Homeostasis , Hydrogen-Ion Concentration , Immunohistochemistry , Kidney/cytology , Kidney/drug effects , Kinetics , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Male , Membrane Potentials , Microscopy, Fluorescence , Perfusion , Potassium Channel Blockers/pharmacology , Potassium, Dietary/urine , Protein Transport , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Scorpion Venoms/pharmacology , Time Factors , Urinalysis
6.
J Biol Chem ; 282(9): 6455-62, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17194699

ABSTRACT

Base-line urinary potassium secretion in the distal nephron is mediated by small conductance rat outer medullary K (ROMK)-like channels. We used the patch clamp technique applied to split-open cortical collecting ducts (CCDs) isolated from rats fed a normal potassium (NK) or low potassium (LK) diet to test the hypothesis that AngII directly inhibits ROMK channel activity. We found that AngII inhibited ROMK channel activity in LK but not NK rats in a dose-dependent manner. The AngII-induced reduction in channel activity was mediated by AT1 receptor (AT1R) binding, because pretreatment of CCDs with losartan but not PD123319 AT1 and AT2 receptor antagonists, respectively, blocked the response. Pretreatment of CCDs with U73122 and calphostin C, inhibitors of phospholipase C (PLC) and protein kinase C (PKC), respectively, abolished the AngII-induced decrease in ROMK channel activity, confirming a role of the PLC-PKC pathway in this response. Studies by others suggest that AngII stimulates an Src family protein-tyrosine kinase (PTK) via PKC-NADPH oxidase. PTK has been shown to regulate the ROMK channel. Inhibition of NADPH oxidase with diphenyliodonium abolished the inhibitory effect of AngII or the PKC activator phorbol 12-myristate 13-acetate on ROMK channels. Suppression of PTK by herbimycin A significantly attenuated the inhibitory effect of AngII on ROMK channel activity. We conclude that AngII inhibits ROMK channel activity through PKC-, NADPH oxidase-, and PTK-dependent pathways under conditions of dietary potassium restriction.


Subject(s)
Angiotensin II/pharmacology , Kidney Tubules, Collecting/metabolism , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium/administration & dosage , Animals , Diet , Kidney Tubules, Collecting/chemistry , NADPH Oxidases/metabolism , Patch-Clamp Techniques , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism
8.
Am J Physiol Renal Physiol ; 285(4): F629-39, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12824078

ABSTRACT

High urinary flow rates stimulate K secretion in the fully differentiated but not neonatal or weanling rabbit cortical collecting duct (CCD). Both small-conductance secretory K and high-conductance Ca2+/stretch-activated maxi-K channels have been identified in the apical membrane of the mature CCD by patch-clamp analysis. We reported that flow-stimulated net K secretion in the adult rabbit CCD is 1) blocked by TEA and charybdotoxin, inhibitors of intermediate- and high-conductance (maxi-K) Ca2+-activated K channels, and 2) associated with increases in net Na absorption and intracellular Ca2+ concentration ([Ca2+]i). The present study examined whether the absence of flow-stimulated K secretion early in life is due to a 1) limited flow-induced rise in net Na absorption and/or [Ca2+]i and/or 2) paucity of apical maxi-K channels. An approximately sixfold increase in tubular fluid flow rate in CCDs isolated from 4-wk-old rabbits and microperfused in vitro led to an increase in net Na absorption and [Ca2+]i, similar in magnitude to the response observed in 6-wk-old tubules, but it failed to generate an increase in net K secretion. By 5 wk of age, there was a small, but significant, flow-stimulated rise in net K secretion that increased further by 6 wk of life. Luminal perfusion with iberiotoxin blocked the flow stimulation of net K secretion in the adult CCD, confirming the identity of the maxi-K channel in this response. Maxi-K channel alpha-subunit message was consistently detected in single CCDs from animals >/=4 wk of age by RT-PCR. Indirect immunofluorescence microscopy using antibodies directed against the alpha-subunit revealed apical labeling of intercalated cells in cryosections from animals >/=5 wk of age; principal cell labeling was generally intracellular and punctate. We speculate that the postnatal appearance of flow-dependent K secretion is determined by the transcriptional/translational regulation of expression of maxi-K channels. Furthermore, our studies suggest a novel function for intercalated cells in mediating flow-stimulated K secretion.


Subject(s)
Kidney Tubules, Collecting/metabolism , Potassium/metabolism , Renal Circulation/physiology , Absorption , Aging/physiology , Animals , Blotting, Western , Calcium/metabolism , Fluorescent Antibody Technique , In Vitro Techniques , Intracellular Membranes/metabolism , Kidney Cortex , Kidney Tubules, Proximal/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Large-Conductance Calcium-Activated Potassium Channels , Osmolar Concentration , Peptides/pharmacology , Potassium Channels/metabolism , Potassium Channels, Calcium-Activated/genetics , RNA, Messenger/metabolism , Rabbits , Sodium/metabolism , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...