1
|
Nguyen NH, Sheng S, Banerjee A, Guerriero CJ, Chen J, Wang X, Mackie TD, Welling PA, Kleyman TR, Bahar I, Carlson AE, Brodsky JL. Characterization of hyperactive mutations in the renal potassium channel ROMK uncovers unique effects on channel biogenesis and ion conductance. Mol Biol Cell 2024; 35:ar119. [PMID: 39024255 PMCID: PMC11449386 DOI: 10.1091/mbc.e23-12-0494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024] Open
Abstract
Hypertension affects one billion people worldwide and is the most common risk factor for cardiovascular disease, yet a comprehensive picture of its underlying genetic factors is incomplete. Amongst regulators of blood pressure is the renal outer medullary potassium (ROMK) channel. While select ROMK mutants are prone to premature degradation and lead to disease, heterozygous carriers of some of these same alleles are protected from hypertension. Therefore, we hypothesized that gain-of-function (GoF) ROMK variants which increase potassium flux may predispose people to hypertension. To begin to test this hypothesis, we employed genetic screens and a candidate-based approach to identify six GoF variants in yeast. Subsequent functional assays in higher cells revealed two variant classes. The first group exhibited greater stability in the endoplasmic reticulum, enhanced channel assembly, and/or increased protein at the cell surface. The second group of variants resided in the PIP2-binding pocket, and computational modeling coupled with patch-clamp studies demonstrated lower free energy for channel opening and slowed current rundown, consistent with an acquired PIP2-activated state. Together, these findings advance our understanding of ROMK structure-function, suggest the existence of hyperactive ROMK alleles in humans, and establish a system to facilitate the development of ROMK-targeted antihypertensives.
Collapse
Affiliation(s)
- Nga H. Nguyen
- Department of Biological Sciences, School of Medicine, University of Pittsburgh, PA 15260
| | - Shaohu Sheng
- Renal-Electrolyte Division, Department of Medicine, School of Medicine, University of Pittsburgh, PA 15260
| | - Anupam Banerjee
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, PA 15260
| | | | - Jingxin Chen
- Renal-Electrolyte Division, Department of Medicine, School of Medicine, University of Pittsburgh, PA 15260
| | - Xueqi Wang
- Renal-Electrolyte Division, Department of Medicine, School of Medicine, University of Pittsburgh, PA 15260
| | - Timothy D. Mackie
- Department of Biological Sciences, School of Medicine, University of Pittsburgh, PA 15260
| | - Paul A. Welling
- Division of Nephrology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Thomas R. Kleyman
- Renal-Electrolyte Division, Department of Medicine, School of Medicine, University of Pittsburgh, PA 15260
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, PA 15260
| | - Anne E. Carlson
- Department of Biological Sciences, School of Medicine, University of Pittsburgh, PA 15260
| | - Jeffrey L. Brodsky
- Department of Biological Sciences, School of Medicine, University of Pittsburgh, PA 15260
| |
Collapse
|
2
|
Nguyen NH, Sarangi S, McChesney EM, Sheng S, Durrant JD, Porter AW, Kleyman TR, Pitluk ZW, Brodsky JL. Genome mining yields putative disease-associated ROMK variants with distinct defects. PLoS Genet 2023; 19:e1011051. [PMID: 37956218 PMCID: PMC10695394 DOI: 10.1371/journal.pgen.1011051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 12/04/2023] [Accepted: 11/04/2023] [Indexed: 11/15/2023] Open
Abstract
Bartter syndrome is a group of rare genetic disorders that compromise kidney function by impairing electrolyte reabsorption. Left untreated, the resulting hyponatremia, hypokalemia, and dehydration can be fatal, and there is currently no cure. Bartter syndrome type II specifically arises from mutations in KCNJ1, which encodes the renal outer medullary potassium channel, ROMK. Over 40 Bartter syndrome-associated mutations in KCNJ1 have been identified, yet their molecular defects are mostly uncharacterized. Nevertheless, a subset of disease-linked mutations compromise ROMK folding in the endoplasmic reticulum (ER), which in turn results in premature degradation via the ER associated degradation (ERAD) pathway. To identify uncharacterized human variants that might similarly lead to premature degradation and thus disease, we mined three genomic databases. First, phenotypic data in the UK Biobank were analyzed using a recently developed computational platform to identify individuals carrying KCNJ1 variants with clinical features consistent with Bartter syndrome type II. In parallel, we examined genomic data in both the NIH TOPMed and ClinVar databases with the aid of Rhapsody, a verified computational algorithm that predicts mutation pathogenicity and disease severity. Subsequent phenotypic studies using a yeast screen to assess ROMK function-and analyses of ROMK biogenesis in yeast and human cells-identified four previously uncharacterized mutations. Among these, one mutation uncovered from the two parallel approaches (G228E) destabilized ROMK and targeted it for ERAD, resulting in reduced cell surface expression. Another mutation (T300R) was ERAD-resistant, but defects in channel activity were apparent based on two-electrode voltage clamp measurements in X. laevis oocytes. Together, our results outline a new computational and experimental pipeline that can be applied to identify disease-associated alleles linked to a range of other potassium channels, and further our understanding of the ROMK structure-function relationship that may aid future therapeutic strategies to advance precision medicine.
Collapse
Affiliation(s)
- Nga H. Nguyen
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Srikant Sarangi
- Paradigm4, Inc., Waltham, Massachusetts, United States of America
| | - Erin M. McChesney
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Shaohu Sheng
- Renal-Electrolyte Division, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jacob D. Durrant
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Aidan W. Porter
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Thomas R. Kleyman
- Renal-Electrolyte Division, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | | | - Jeffrey L. Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
3
|
Nguyen NH, Sarangi S, McChesney EM, Sheng S, Porter AW, Kleyman TR, Pitluk ZW, Brodsky JL. Genome mining yields new disease-associated ROMK variants with distinct defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539609. [PMID: 37214976 PMCID: PMC10197530 DOI: 10.1101/2023.05.05.539609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Bartter syndrome is a group of rare genetic disorders that compromise kidney function by impairing electrolyte reabsorption. Left untreated, the resulting hyponatremia, hypokalemia, and dehydration can be fatal. Although there is no cure for this disease, specific genes that lead to different Bartter syndrome subtypes have been identified. Bartter syndrome type II specifically arises from mutations in the KCNJ1 gene, which encodes the renal outer medullary potassium channel, ROMK. To date, over 40 Bartter syndrome-associated mutations in KCNJ1 have been identified. Yet, their molecular defects are mostly uncharacterized. Nevertheless, a subset of disease-linked mutations compromise ROMK folding in the endoplasmic reticulum (ER), which in turn results in premature degradation via the ER associated degradation (ERAD) pathway. To identify uncharacterized human variants that might similarly lead to premature degradation and thus disease, we mined three genomic databases. First, phenotypic data in the UK Biobank were analyzed using a recently developed computational platform to identify individuals carrying KCNJ1 variants with clinical features consistent with Bartter syndrome type II. In parallel, we examined ROMK genomic data in both the NIH TOPMed and ClinVar databases with the aid of a computational algorithm that predicts protein misfolding and disease severity. Subsequent phenotypic studies using a high throughput yeast screen to assess ROMK function-and analyses of ROMK biogenesis in yeast and human cells-identified four previously uncharacterized mutations. Among these, one mutation uncovered from the two parallel approaches (G228E) destabilized ROMK and targeted it for ERAD, resulting in reduced protein expression at the cell surface. Another ERAD-targeted ROMK mutant (L320P) was found in only one of the screens. In contrast, another mutation (T300R) was ERAD-resistant, but defects in ROMK activity were apparent after expression and two-electrode voltage clamp measurements in Xenopus oocytes. Together, our results outline a new computational and experimental pipeline that can be applied to identify disease-associated alleles linked to a range of other potassium channels, and further our understanding of the ROMK structure-function relationship that may aid future therapeutic strategies. Author Summary Bartter syndrome is a rare genetic disorder characterized by defective renal electrolyte handing, leading to debilitating symptoms and, in some patients, death in infancy. Currently, there is no cure for this disease. Bartter syndrome is divided into five types based on the causative gene. Bartter syndrome type II results from genetic variants in the gene encoding the ROMK protein, which is expressed in the kidney and assists in regulating sodium, potassium, and water homeostasis. Prior work established that some disease-associated ROMK mutants misfold and are destroyed soon after their synthesis in the endoplasmic reticulum (ER). Because a growing number of drugs have been identified that correct defective protein folding, we wished to identify an expanded cohort of similarly misshapen and unstable disease-associated ROMK variants. To this end, we developed a pipeline that employs computational analyses of human genome databases with genetic and biochemical assays. Next, we both confirmed the identity of known variants and uncovered previously uncharacterized ROMK variants associated with Bartter syndrome type II. Further analyses indicated that select mutants are targeted for ER-associated degradation, while another mutant compromises ROMK function. This work sets-the-stage for continued mining for ROMK loss of function alleles as well as other potassium channels, and positions select Bartter syndrome mutations for correction using emerging pharmaceuticals.
Collapse
|
4
|
Padhy B, Xie J, Wang R, Lin F, Huang CL. Channel Function of Polycystin-2 in the Endoplasmic Reticulum Protects against Autosomal Dominant Polycystic Kidney Disease. J Am Soc Nephrol 2022; 33:1501-1516. [PMID: 35835458 PMCID: PMC9342640 DOI: 10.1681/asn.2022010053] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/03/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Mutations of PKD2, which encodes polycystin-2, cause autosomal dominant polycystic kidney disease (ADPKD). The prevailing view is that defects in polycystin-2-mediated calcium ion influx in the primary cilia play a central role in the pathogenesis of cyst growth. However, polycystin-2 is predominantly expressed in the endoplasmic reticulum (ER) and more permeable to potassium ions than to calcium ions. METHODS The trimeric intracellular cation (TRIC) channel TRIC-B is an ER-resident potassium channel that mediates potassium-calcium counterion exchange for inositol trisphosphate-mediated calcium ion release. Using TRIC-B as a tool, we examined the function of ER-localized polycystin-2 and its role in ADPKD pathogenesis in cultured cells, zebrafish, and mouse models. RESULTS Agonist-induced ER calcium ion release was defective in cells lacking polycystin-2 and reversed by exogenous expression of TRIC-B. Vice versa, exogenous polycystin-2 reversed an ER calcium-release defect in cells lacking TRIC-B. In a zebrafish model, expression of wild-type but not nonfunctional TRIC-B suppressed polycystin-2-deficient phenotypes. Similarly, these phenotypes were suppressed by targeting the ROMK potassium channel (normally expressed on the cell surface) to the ER. In cultured cells and polycystin-2-deficient zebrafish phenotypes, polycystin-2 remained capable of reversing the ER calcium release defect even when it was not present in the cilia. Transgenic expression of Tric-b ameliorated cystogenesis in the kidneys of conditional Pkd2-inactivated mice, whereas Tric-b deletion enhanced cystogenesis in Pkd2-heterozygous kidneys. CONCLUSIONS Polycystin-2 in the ER appears to be critical for anticystogenesis and likely functions as a potassium ion channel to facilitate potassium-calcium counterion exchange for inositol trisphosphate-mediated calcium release. The results advance the understanding of ADPKD pathogenesis and provides proof of principle for pharmacotherapy by TRIC-B activators.
Collapse
Affiliation(s)
- Biswajit Padhy
- Division of Nephrology, Department of Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Jian Xie
- Division of Nephrology, Department of Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Runping Wang
- Division of Nephrology, Department of Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Fang Lin
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Chou-Long Huang
- Division of Nephrology, Department of Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
5
|
Sung CC, Chen MH, Lin YC, Lin YC, Lin YJ, Yang SS, Lin SH. Urinary Extracellular Vesicles for Renal Tubular Transporters Expression in Patients With Gitelman Syndrome. Front Med (Lausanne) 2021; 8:679171. [PMID: 34179047 PMCID: PMC8219937 DOI: 10.3389/fmed.2021.679171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/11/2021] [Indexed: 11/27/2022] Open
Abstract
Background: The utility of urinary extracellular vesicles (uEVs) to faithfully represent the changes of renal tubular protein expression remains unclear. We aimed to evaluate renal tubular sodium (Na+) or potassium (K+) associated transporters expression from uEVs and kidney tissues in patients with Gitelman syndrome (GS) caused by inactivating mutations in SLC12A3. Methods: uEVs were isolated by ultracentrifugation from 10 genetically-confirmed GS patients. Membrane transporters including Na+-hydrogen exchanger 3 (NHE3), Na+/K+/2Cl− cotransporter (NKCC2), NaCl cotransporter (NCC), phosphorylated NCC (p-NCC), epithelial Na+ channel β (ENaCβ), pendrin, renal outer medullary K1 channel (ROMK), and large-conductance, voltage-activated and Ca2+-sensitive K+ channel (Maxi-K) were examined by immunoblotting of uEVs and immunofluorescence of biopsied kidney tissues. Healthy and disease (bulimic patients) controls were also enrolled. Results: Characterization of uEVs was confirmed by nanoparticle tracking analysis, transmission electron microscopy, and immunoblotting. Compared with healthy controls, uEVs from GS patients showed NCC and p-NCC abundance were markedly attenuated but NHE3, ENaCβ, and pendrin abundance significantly increased. ROMK and Maxi-K abundance were also significantly accentuated. Immunofluorescence of the representative kidney tissues from GS patients also demonstrated the similar findings to uEVs. uEVs from bulimic patients showed an increased abundance of NCC and p-NCC as well as NHE3, NKCC2, ENaCβ, pendrin, ROMK and Maxi-K, akin to that in immunofluorescence of their kidney tissues. Conclusion: uEVs could be a non-invasive tool to diagnose and evaluate renal tubular transporter adaptation in patients with GS and may be applied to other renal tubular diseases.
Collapse
Affiliation(s)
- Chih-Chien Sung
- Division of Nephrology, Department of Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Min-Hsiu Chen
- Division of Nephrology, Department of Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Yi-Chang Lin
- Division of Cardiovascular Surgery, Department of Surgery, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Yu-Chun Lin
- Deparment of Pathology, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Yi-Jia Lin
- Deparment of Pathology, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Sung-Sen Yang
- Division of Nephrology, Department of Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Shih-Hua Lin
- Division of Nephrology, Department of Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| |
Collapse
|
6
|
Jiang J, Ding FX, Zhou X, Bateman TJ, Dong S, Gu X, Keh deJesus R, Pio B, Tang H, Chobanian HR, Levorse D, Hu M, Thomas-Fowlkes B, Margulis M, Koehler M, Weinglass A, Gibson J, Houle K, Yudkovitz J, Hampton C, Pai LY, Samuel K, Cutarelli T, Sullivan K, Parmee ER, Davies I, Pasternak A. Discovery of MK-8153, a Potent and Selective ROMK Inhibitor and Novel Diuretic/Natriuretic. J Med Chem 2021; 64:7691-7701. [PMID: 34038119 DOI: 10.1021/acs.jmedchem.1c00406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A renal outer medullary potassium channel (ROMK, Kir1.1) is a putative drug target for a novel class of diuretics with potential for treating hypertension and heart failure. Our first disclosed clinical ROMK compound, 2 (MK-7145), demonstrated robust diuresis, natriuresis, and blood pressure lowering in preclinical models, with reduced urinary potassium excretion compared to the standard of care diuretics. However, 2 projected to a short human half-life (∼5 h) that could necessitate more frequent than once a day dosing. In addition, a short half-life would confer a high peak-to-trough ratio which could evoke an excessive peak diuretic effect, a common liability associated with loop diuretics such as furosemide. This report describes the discovery of a new ROMK inhibitor 22e (MK-8153), with a longer projected human half-life (∼14 h), which should lead to a reduced peak-to-trough ratio, potentially extrapolating to more extended and better tolerated diuretic effects.
Collapse
Affiliation(s)
- Jinlong Jiang
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Fa-Xiang Ding
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Xiaoyan Zhou
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Thomas J Bateman
- Pharmacokinetics Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Shuzhi Dong
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Xin Gu
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Reynalda Keh deJesus
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Barbara Pio
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Haifeng Tang
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Harry R Chobanian
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Dorothy Levorse
- Discovery and Preclinical Sciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Mengwei Hu
- Discovery and Preclinical Sciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Brande Thomas-Fowlkes
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Michael Margulis
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Martin Koehler
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Adam Weinglass
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Jack Gibson
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Kevin Houle
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Joel Yudkovitz
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Caryn Hampton
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Lee-Yuh Pai
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Koppara Samuel
- Pharmacokinetics Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Timothy Cutarelli
- Discovery Process Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Kathleen Sullivan
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Emma R Parmee
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Ian Davies
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Alexander Pasternak
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| |
Collapse
|
7
|
Zuo J, Guo W, Wang S, Lang Y, Wang S, Shi X, Zhang R, Zhao X, Han Y, Shao L. Eight novel KCNJ1 variants and parathyroid hormone overaction or resistance in 5 probands with Bartter syndrome type 2. Clin Chim Acta 2020; 511:248-254. [PMID: 33058840 DOI: 10.1016/j.cca.2020.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/07/2020] [Accepted: 10/02/2020] [Indexed: 01/19/2023]
Abstract
PURPOSE Bartter syndrome type 2 (BS2) is an autosomal recessive renal tubular disorder, which is caused by the mutations in KCNJ1. This study was designed to analyze and describe the genotype and clinical features of five Chinese probands with BS2. METHODS Identify KCNJ1 gene variants by the next generation sequencing and evaluate their mutation effects according to 2015 American College of Medical Genetics and Genomics (ACMG) standards and guidelines. RESULTS Ten variants including eight novel ones of KCNJ1 gene were found, the most common type was missense variant. The common symptoms and signs from high to low incidence were: polydipsia and polyuria (5/5), one of them (1/5) presented with diabetes insipidus; maternal polyhydramnios and premature delivery (4/5); growth retardation (3/5). Two patients presented with hypochloremic metabolic alkalosis and hypokalemia; whereas the acid-base disturbance was absent in the others. One patient had evident parathyroid hormone (PTH) resistance (hypocalcemia, hyperphosphatemia and markedly elevated PTH levels), three presented with PTH overacting (hypercalcemia, hypophosphatemia and mild elevated PTH levels), and one showed normal blood calcium and phosphorus concentrations with high-normal PTH levels. All patients had nephrocalcinosis and/or hypercalciuria, and one of them complicated with nephrolithiasis. Indomethacin has significant therapeutic effect on the growth retardation, polydipsia and polyuria and treatment was associated with a decrease in urine calcium excretion, normalization of electrolyte disturbance and PTH parameters. CONCLUSIONS Ten variants of KCNJ1 gene were identified in five Chinese probands. These patients had atypical BS phenotype lacking evident metabolic alkalosis and/or manifesting with PTH overaction/resistance, which reminds clinicians to carefully differentiate BS2 with other parathyroid disorders. This is the first report of BS2 from Chinese populations.
Collapse
Affiliation(s)
- Jianxin Zuo
- Department of Obstetrics, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China
| | - Wencong Guo
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Shujuan Wang
- Department of Nursing, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Yanhua Lang
- Department of Nursing, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Sai Wang
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Xiaomeng Shi
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Ruixiao Zhang
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Xiangzhong Zhao
- Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Yue Han
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China.
| | - Leping Shao
- Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Qingdao University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Department of Nephrology, the Affiliated Qingdao Municipal Hospital of Shandong University, No.5 Donghai Middle Road, Qingdao 266071, People's Republic of China; Central Laboratory, the Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China.
| |
Collapse
|
8
|
Role of potassium channels in female reproductive system. Obstet Gynecol Sci 2020; 63:565-576. [PMID: 32838485 PMCID: PMC7494774 DOI: 10.5468/ogs.20064] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/26/2020] [Indexed: 12/26/2022] Open
Abstract
Potassium channels are widely expressed in most types of cells in living organisms and regulate the functions of a variety of organs, including kidneys, neurons, cardiovascular organs, and pancreas among others. However, the functional roles of potassium channels in the reproductive system is less understood. This mini-review provides information about the localization and functions of potassium channels in the female reproductive system. Five types of potassium channels, which include inward-rectifying (Kir), voltage-gated (Kv), calcium-activated (KCa), 2-pore domain (K2P), and rapidly-gating sodium-activated (Slo) potassium channels are expressed in the hypothalamus, ovaries, and uterus. Their functions include the regulation of hormone release and feedback by Kir6.1 and Kir6.2, which are expressed in the luteal granulosa cells and gonadotropin-releasing hormone neurons respectively, and regulate the functioning of the hypothalamus–pituitary–ovarian axis and the production of progesterone. Both channels are regulated by subtypes of the sulfonylurea receptor (SUR), Kir6.1/SUR2B and Kir6.2/SUR1. Kv and Slo2.1 affect the transition from uterine quiescence in late pregnancy to the state of strong myometrial contractions in labor. Intermediate- and small-conductance KCa modulate the vasodilatation of the placental chorionic plate resistance arteries via the secretion of nitric oxide and endothelium-derived hyperpolarizing factors. Treatment with specific channel activators and inhibitors provides information relevant for clinical use that could help alter the functions of the female reproductive system.
Collapse
|
9
|
The Urinary Excretion of Uromodulin is Regulated by the Potassium Channel ROMK. Sci Rep 2019; 9:19517. [PMID: 31863061 PMCID: PMC6925250 DOI: 10.1038/s41598-019-55771-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/28/2019] [Indexed: 12/20/2022] Open
Abstract
Uromodulin, the most abundant protein in normal urine, is produced by cells lining the thick ascending limb (TAL) of the loop of Henle. Uromodulin regulates the activity of the potassium channel ROMK in TAL cells. Common variants in KCNJ1, the gene encoding ROMK, are associated with urinary levels of uromodulin in population studies. Here, we investigated the functional link between ROMK and uromodulin in Kcnj1 knock-out mouse models, in primary cultures of mouse TAL (mTAL) cells, and in patients with Bartter syndrome due to KCNJ1 mutations. Both global and kidney-specific Kcnj1 knock-out mice showed reduced urinary levels of uromodulin paralleled by increased levels in the kidney, compared to wild-type controls. Pharmacological inhibition and genetic deletion of ROMK in mTAL cells caused a reduction in apical uromodulin excretion, reflected by cellular accumulation. In contrast, NKCC2 inhibition showed no effect on uromodulin processing. Patients with Bartter syndrome type 2 showed reduced urinary uromodulin levels compared to age and gender matched controls. These results demonstrate that ROMK directly regulates processing and release of uromodulin by TAL cells, independently from NKCC2. They support the functional link between transport activity and uromodulin in the TAL, relevant for blood pressure control and urinary concentrating ability.
Collapse
|
10
|
Gonzalez-Vicente A, Saez F, Monzon CM, Asirwatham J, Garvin JL. Thick Ascending Limb Sodium Transport in the Pathogenesis of Hypertension. Physiol Rev 2019; 99:235-309. [PMID: 30354966 DOI: 10.1152/physrev.00055.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The thick ascending limb plays a key role in maintaining water and electrolyte balance. The importance of this segment in regulating blood pressure is evidenced by the effect of loop diuretics or local genetic defects on this parameter. Hormones and factors produced by thick ascending limbs have both autocrine and paracrine effects, which can extend prohypertensive signaling to other structures of the nephron. In this review, we discuss the role of the thick ascending limb in the development of hypertension, not as a sole participant, but one that works within the rich biological context of the renal medulla. We first provide an overview of the basic physiology of the segment and the anatomical considerations necessary to understand its relationship with other renal structures. We explore the physiopathological changes in thick ascending limbs occurring in both genetic and induced animal models of hypertension. We then discuss the racial differences and genetic defects that affect blood pressure in humans through changes in thick ascending limb transport rates. Throughout the text, we scrutinize methodologies and discuss the limitations of research techniques that, when overlooked, can lead investigators to make erroneous conclusions. Thus, in addition to advancing an understanding of the basic mechanisms of physiology, the ultimate goal of this work is to understand our research tools, to make better use of them, and to contextualize research data. Future advances in renal hypertension research will require not only collection of new experimental data, but also integration of our current knowledge.
Collapse
Affiliation(s)
| | - Fara Saez
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Casandra M Monzon
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jessica Asirwatham
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|
11
|
Lu M, Li JR, Alvarez-Lugo L, Li Y, Yu S, Li X, Shi B, Chai TC. Lipopolysaccharide stimulates BK channel activity in bladder umbrella cells. Am J Physiol Cell Physiol 2018; 314:C643-C653. [PMID: 29466671 DOI: 10.1152/ajpcell.00339.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bladder urothelium plays an active role in response to bacterial infection. There is little known about the electrophysiological activity in urothelial cells in this process. We used a nonenzymatic method to isolate bladder urothelial tissue and to patch clamp umbrella cells in situ. A 200 pS conductance potassium (K+) channel was detected from female C57BL6 mice. Of 58 total patches, 17.2% patches displayed the 200 pS K+ conductance channel. This K+ conductance channel showed Ca2+ sensitivity and voltage dependence. Specific big-conductance potassium channel (BK) inhibitors (paxilline, iberiotoxin) blocked the 200 pS K+ conductance channel activity. RT-PCR and immunoblot confirmed BK channel pore-forming α-subunit (BK-α) mRNA and protein in urothelium. Immunohistochemistry also showed the BK-α located in urothelium. The above data provided evidence that the 200 pS K+ conductance channel was a BK channel. Lipopolysaccharide (LPS), a component of uropathogenic Escherichia coli, was used to investigate the role of BK channel in the pathogenesis of urinary tract infection. BK channel activity as NPo increased threefold within 30 min of exposure to LPS. mRNAs for LPS receptors (TLR4, CD14, MD-2) were expressed in the urothelium but not in lamina propria or detrusor. Blockade of the receptors by an antagonist (polymyxin B) abrogated LPS's effect on BK channel. The involvement of protein kinase A (PKA) on BK channel activity was demonstrated by applying PKA blockers (H89 and PKI). Both PKA inhibitors abolished the BK channel activity induced by LPS. In conclusion, BK channel was identified in bladder umbrella cells, and its activity was significantly increased by LPS.
Collapse
Affiliation(s)
- Ming Lu
- Department of Urology, Yale University School of Medicine , New Haven, Connecticut
| | - Jian-Ri Li
- Department of Urology, Yale University School of Medicine , New Haven, Connecticut
| | - Lery Alvarez-Lugo
- Department of Urology, Yale University School of Medicine , New Haven, Connecticut
| | - Yan Li
- Department of Urology, Qilu Hospital, Shandong University , Jinan , China
| | - Shan Yu
- Department of Urology, Yale University School of Medicine , New Haven, Connecticut
| | - XuanHao Li
- Department of Urology, Yale University School of Medicine , New Haven, Connecticut
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Shandong University , Jinan , China
| | - Toby C Chai
- Department of Urology, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
12
|
Zhou X, Forrest MJ, Sharif-Rodriguez W, Forrest G, Szeto D, Urosevic-Price O, Zhu Y, Stevenson AS, Zhou Y, Stribling S, Dajee M, Walsh SP, Pasternak A, Sullivan KA. Chronic Inhibition of Renal Outer Medullary Potassium Channel Not Only Prevented but Also Reversed Development of Hypertension and End-Organ Damage in Dahl Salt-Sensitive Rats. Hypertension 2017; 69:332-338. [DOI: 10.1161/hypertensionaha.116.08358] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 09/05/2016] [Accepted: 11/13/2016] [Indexed: 01/21/2023]
Abstract
The renal outer medullary potassium (ROMK) channel mediates potassium recycling and facilitates sodium reabsorption through the Na
+
/K
+
/2Cl
−
cotransporter in the loop of Henle and potassium secretion at the cortical collecting duct. Evidence from the phenotype of humans and rodents with functional ROMK deficiency supports the contention that selective ROMK inhibitors (ROMKi) will represent a novel diuretic with potential of therapeutic benefit for hypertension. ROMKi have recently been synthesized by Merck & Co, Inc. The present studies were designed to examine the effects of ROMKi B on systemic hemodynamics, renal function and structure, and vascular function in Dahl salt-sensitive rats. Four experimental groups—control, high-salt diet alone; ROMKi B 3 mg·kg
−
1
·d
−
1
; ROMKi B 10 mg·kg
−
1
·d
−
1
; and hydrochlorothiazide 25 mg·kg
−
1
·d
−
1
—were included in prophylactic (from week 1 to week 9 on high-salt diet) and therapeutic studies (from week 5 to week 9 on high-salt diet), respectively. ROMKi B produced sustained blood pressure reduction and improved renal and vascular function and histological alterations induced by a high-salt diet. ROMKi B was superior to hydrochlorothiazide at reducing blood pressure. Furthermore, ROMKi B provided beneficial effects on both the plasma lipid profile and bone mineral density. Chronic ROMK inhibition not only prevented but also reversed the development of hypertension and end-organ damage in Dahl salt-sensitive rats. Our findings suggest a potential utility of ROMKi B as a novel antihypertensive agent, particularly for the treatment of the salt-sensitive hypertension patient population.
Collapse
Affiliation(s)
- Xiaoyan Zhou
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Michael J. Forrest
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Wanda Sharif-Rodriguez
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Gail Forrest
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Daphne Szeto
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Olga Urosevic-Price
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Yonghua Zhu
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Andra S. Stevenson
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Yuchen Zhou
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Sloan Stribling
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Maya Dajee
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Shawn P. Walsh
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Alexander Pasternak
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Kathleen A. Sullivan
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| |
Collapse
|
13
|
Sopjani M, Dërmaku-Sopjani M. Klotho-Dependent Cellular Transport Regulation. VITAMINS AND HORMONES 2016; 101:59-84. [PMID: 27125738 DOI: 10.1016/bs.vh.2016.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Klotho is a transmembrane protein that in humans is encoded by the hKL gene. This protein is known to have aging suppressor effects and is predominantly expressed in the distal convoluted tubule of the kidney, parathyroid glands, and choroid plexus of the brain. The Klotho protein exists in both full-length membrane form and a soluble secreted form, which exerts numerous distinct functions. The extracellular domain of Klotho can be enzymatically cleaved off and released into the systemic circulation where it functions as β-glucuronidase and a hormone. Soluble Klotho is a multifunction protein present in the biological fluids including blood, urine, and cerebrospinal fluid of mammals. Klotho deficiency leads to multiple organ failure accompanied by early appearance of multiple age-related disorders and early death, whereas overexpression of Klotho results in the opposite effects. Klotho, an enzyme and hormone, has been reported to participate in the regulation of cellular transport processes across the plasma membrane either indirectly through inhibiting calcitriol (1,25(OH)2D3) formation or other mechanism, or by directly affecting transporter proteins, including ion channels, cellular carriers, and Na(+)/K(+)-ATPase. Accordingly, Klotho protein serves as a powerful regulator of cellular transport across the plasma membrane. Importantly, Klotho-dependent cellular transport regulation implies stimulatory or inhibitory effects. Klotho has been shown to play a key role in the regulation of multiple calcium and potassium ion channels, and various cellular carriers including the Na(+)-coupled cotransporters such as NaPi-IIa, NaPi-IIb, EAAT3, and EAAT4, CreaT1 as well as Na(+)/K(+)-ATPase. These regulations are parts of the antiaging function of Klotho, which will be discussing throughout this chapter. Clearly, further experimental efforts are required to investigate the effect of Klotho on other transport proteins and underlying molecular mechanisms by which Klotho exerts its effect.
Collapse
Affiliation(s)
- M Sopjani
- University of Prishtina, Prishtinë, Republic of Kosova.
| | | |
Collapse
|
14
|
Glaaser IW, Slesinger PA. Structural Insights into GIRK Channel Function. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 123:117-60. [PMID: 26422984 DOI: 10.1016/bs.irn.2015.05.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
G protein-gated inwardly rectifying potassium (GIRK; Kir3) channels, which are members of the large family of inwardly rectifying potassium channels (Kir1-Kir7), regulate excitability in the heart and brain. GIRK channels are activated following stimulation of G protein-coupled receptors that couple to the G(i/o) (pertussis toxin-sensitive) G proteins. GIRK channels, like all other Kir channels, possess an extrinsic mechanism of inward rectification involving intracellular Mg(2+) and polyamines that occlude the conduction pathway at membrane potentials positive to E(K). In the past 17 years, more than 20 high-resolution atomic structures containing GIRK channel cytoplasmic domains and transmembrane domains have been solved. These structures have provided valuable insights into the structural determinants of many of the properties common to all inward rectifiers, such as permeation and rectification, as well as revealing the structural bases for GIRK channel gating. In this chapter, we describe advances in our understanding of GIRK channel function based on recent high-resolution atomic structures of inwardly rectifying K(+) channels discussed in the context of classical structure-function experiments.
Collapse
Affiliation(s)
- Ian W Glaaser
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paul A Slesinger
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
15
|
Glaudemans B, Terryn S, Gölz N, Brunati M, Cattaneo A, Bachi A, Al-Qusairi L, Ziegler U, Staub O, Rampoldi L, Devuyst O. A primary culture system of mouse thick ascending limb cells with preserved function and uromodulin processing. Pflugers Arch 2013; 466:343-56. [DOI: 10.1007/s00424-013-1321-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 06/23/2013] [Accepted: 06/23/2013] [Indexed: 11/28/2022]
|
16
|
Abstract
Sensing of sugar by specialized 'glucose-inhibited' cells helps organisms to counteract swings in their internal energy levels. Evidence from several cell types in both vertebrates and invertebrates suggests that this process involves sugar-induced stimulation of plasma membrane K(+) currents. However, the molecular composition and the mechanism of activation of the underlying channel(s) remain controversial. In mouse hypothalamic neurones and neurosecretory cells of the crab Cancer borealis, glucose stimulates K(+) currents displaying leak-like properties. Yet knockout of some of the candidate 'leak' channel subunits encoded by the KCNK gene family so far failed to abolish glucose inhibition of hypothalamic cells. Moreover, in other tissues, such as the carotid body, glucose-stimulated K(+) channels appear to be not leak-like but voltage-gated, suggesting that glucose-induced inhibition may engage multiple types of K(+) channels. Other mechanisms of glucose-induced inhibition, such as hyperpolarization mediated by opening of Cl(-) channels and depolarization block caused by closure of K(ATP) channels have also been proposed. Here we review known ionic and pharmacological features of glucose-induced inhibition in different cell types, which may help to identify its molecular correlates.
Collapse
Affiliation(s)
- D Burdakov
- Department of Pharmacology, University of Cambridge, Cambridge, UK.
| | | |
Collapse
|
17
|
Cha SK, Hu MC, Kurosu H, Kuro-o M, Moe O, Huang CL. Regulation of renal outer medullary potassium channel and renal K(+) excretion by Klotho. Mol Pharmacol 2009; 76:38-46. [PMID: 19349416 PMCID: PMC2701452 DOI: 10.1124/mol.109.055780] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Accepted: 04/06/2009] [Indexed: 11/22/2022] Open
Abstract
Klotho is an aging-suppression protein predominantly expressed in kidney, parathyroid glands, and choroids plexus of the brain. The extracellular domain of Klotho, a type-1 membrane protein, is secreted into urine and blood and may function as an endocrine or paracrine hormone. The functional role of Klotho in the kidney remains largely unknown. Recent studies reported that treatment by the extracellular domain of Klotho (KLe) increases cell-surface abundance of transient receptor potential vanilloid type isoform 5, an epithelial Ca(2+) channel critical for Ca(2+) reabsorption in the kidney. Whether Klotho regulates surface expression of other channels in the kidney is not known. Here, we report that KLe treatment increases the cell-membrane abundance of the renal K(+) channel renal outer medullary potassium channel 1 (ROMK1) by removing terminal sialic acids from N-glycan of the channel. Removal of sialic acids exposes underlying disaccharide galactose-N-acetylglucosamine, a ligand for a ubiquitous galactoside-binding lectin galectin-1. Binding to galectin-1 at the extracellular surface prevents clathrin-mediated endocytosis of ROMK1 and leads to accumulation of functional channel on the plasma membrane. Intravenous administration of KLe increases the level of Klotho in urine and increases urinary excretion of K(+). These results suggest that Klotho may have a broader function in the regulation of ion transport in the kidney.
Collapse
Affiliation(s)
- Seung-Kuy Cha
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8856, USA
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Studies on the mechanisms of distal K+ secretion have highlighted the importance of the renal outer-medullary K+ (ROMK) and maxi-K channels. This review considers several human disorders characterized by hypokalemia and hyperkalemia, as well as mouse models of these disorders, and the mechanisms by which ROMK and maxi-K may be dysregulated. RECENT FINDINGS Analysis of knockout mice lacking ROMK, a model for type II Bartter's syndrome, has shown a role for maxi-K in distal K+ secretion. Knockout mice lacking either the alpha or beta1 subunits of maxi-K also show deficits in flow-dependent K+ secretion. Analysis of transgenic and knock-in mouse models of pseudohypoaldosteronism type II, in which mutant forms of with-no-lysine kinase 4 are expressed, suggests ways in which ROMK and maxi-K may be dysregulated to result in hyperkalemia. Modeling studies also provide insights into the role of Na+ delivery vs. flow in K+ secretion. SUMMARY The importance of both ROMK and maxi-K to distal K+ secretion is now well established, but the relative role that each of these two channels plays in normal and diseased states has not been definitively established. Analysis of human and animal model data can generate hypotheses for future experiments.
Collapse
Affiliation(s)
- Aylin R Rodan
- Division of Nephrology, Department of Medicine, UT Southwestern Medical Center, Dallas, Texas 75390-8856, USA.
| | | |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW This review summarizes recent advances in the understanding of the mechanism of regulation of renal ion transport by WNK kinases. RECENT FINDINGS There are four mammalian WNK [with-no-lysine (K)] kinases: WNK1-WNK4. Mutations of WNK1 and WNK4 in humans cause hypertension and hyperkalemia at least partly by altering renal sodium and potassium transport. WNK1 and WNK4 stimulate endocytosis of ROMK1 by recruiting an endocytic scaffold protein, intersectin. The recruitment is independent of the kinase activity and occurs between the PXXP motif of WNKs and the SH3 domain of intersectin. Regulation of cation-chloride-coupled cotransporters, Na+-K+-2Cl(-) cotransporter (NKCC) 1 and NKCC2 [and the Na-Cl co-transporter (NCC), under some conditions] by WNKs requires kinase activity. WNK1 and WNK4 bind with and phosphorylate two Ste20-related protein kinases, OSR1 and SPAK, which in turn bind with and phosphorylate NKCCs and NCC to increase their activity. Binding of OSR1/SPAK to upstream activators (WNKs) and downstream substrates (NKCCs and NCC) are both mediated by a docking site in the C-terminus of OSR1/SPAK and RFX[V/I] motifs present in WNKs or in NKCCs and NCC. SUMMARY WNKs regulate ion transport via both catalytic and noncatalytic mechanisms. We discuss hypotheses that WNKs, contrasting with aldosterone, play important roles in dissociating sodium reabsorption from potassium secretion.
Collapse
|
20
|
|
21
|
Theilig F, Goranova I, Hirsch JR, Wieske M, Ünsal S, Bachmann S, Veh RW, Derst C. Cellular Localization of THIK-1 (K 2P13.1) and THIK-2 (K 2P12.1) K + Channels in the Mammalian Kidney. Cell Physiol Biochem 2008; 21:63-74. [DOI: 10.1159/000113748] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2007] [Indexed: 11/19/2022] Open
|
22
|
Xie LH, John SA, Ribalet B, Weiss JN. Activation of inwardly rectifying potassium (Kir) channels by phosphatidylinosital-4,5-bisphosphate (PIP2): Interaction with other regulatory ligands. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2007; 94:320-35. [PMID: 16837026 DOI: 10.1016/j.pbiomolbio.2006.04.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
All members of the inwardly rectifying potassium channels (Kir1-7) are regulated by the membrane phospholipid, phosphatidylinosital-4,5-bisphosphate (PIP(2)). Some are also modulated by other regulatory factors or ligands such as ATP and G-proteins, which give them their common names, such as the ATP sensitive potassium (K(ATP)) channel and the G-protein gated potassium channel. Other more non-specific regulators include polyamines, kinases, pH and Na(+) ions. Recent studies have demonstrated that PIP(2) acts cooperatively with other regulatory factors to modulate Kir channels. Here we review how PIP(2) and co-factors modulate channel activities in each subfamily of the Kir channels.
Collapse
Affiliation(s)
- Lai-Hua Xie
- Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | | | | | | |
Collapse
|
23
|
Ariceta G, Rodríguez-Soriano J. Inherited Renal Tubulopathies Associated With Metabolic Alkalosis: Effects on Blood Pressure. Semin Nephrol 2006; 26:422-33. [PMID: 17275579 DOI: 10.1016/j.semnephrol.2006.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inherited tubular disorders associated with metabolic alkalosis are caused by several gene mutations encoding different tubular transporters responsible for NaCl renal handling. Body volume and renin-angiotensin-aldosterone system status are determined by NaCl reabsorption in the distal nephron. Two common hallmarks in affected individuals: hypokalemia and normal / high blood pressure, support the differential diagnosis. Bartter's syndrome, characterized by hypokalemia and normal blood pressure, is a heterogenic disease caused by the loss of function of SLC12A1 (type 1), KCNJ1 (type 2), CLCNKB (type 3), or BSND genes (type 4). As a result, patients present with renal salt wasting and hypercalciuria. Gitelman's syndrome is caused by the loss of funcion of the SLC12A3 gene and may resemble Bartter's syndrome, though is associated with the very low urinary calcium. Liddle's syndrome, also with similar phenotype but with hypertension, is produced by the gain of function of the SNCC1B or SNCC1G genes, and must be distinguished from other entities of inherited hypertension such as Apparently Mineralocorticoid Excess, of glucocorticoid remediable hypertension.
Collapse
Affiliation(s)
- Gema Ariceta
- Division of Pediatric Nephrology, Department of Pediatrics, Hospital de Cruces and Basque University School of Medicine, Bilbao, Spain.
| | | |
Collapse
|
24
|
Xie J, Craig L, Cobb MH, Huang CL. Role of with-no-lysine [K] kinases in the pathogenesis of Gordon's syndrome. Pediatr Nephrol 2006; 21:1231-6. [PMID: 16683163 DOI: 10.1007/s00467-006-0106-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Accepted: 01/23/2006] [Indexed: 11/28/2022]
Abstract
Gordon's syndrome, also known as pseudohypoaldosteronism type II (PHA II) or familial hypertension with hyperkalemia, is an autosomal-dominant disease characterized by hypertension, hyperkalemia, hyperchloremic metabolic acidosis, and normal glomerular filtration rate. Recent positional cloning has linked mutations of WNK1 and WNK4 to Gordon's syndrome. With-no-lysine [K] (WNK) kinases are a new family of large serine-threonine protein kinases with an atypical placement of the catalytic lysine. Here, we review the pathogenesis of PHA II based on current understanding of the actions of WNK1 and WNK4 on Na+ and K+ handling in the renal distal tubule.
Collapse
Affiliation(s)
- Jian Xie
- Department of Medicine, Room J5-104, MC-8856, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8856, USA
| | | | | | | |
Collapse
|
25
|
Satlin LM, Carattino MD, Liu W, Kleyman TR. Regulation of cation transport in the distal nephron by mechanical forces. Am J Physiol Renal Physiol 2006; 291:F923-31. [PMID: 16849691 DOI: 10.1152/ajprenal.00192.2006] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Thiazide and loop diuretics induce renal K(+) secretion, often leading to renal K(+) wasting and hypokalemia. This phenomenon has been proposed to reflect an increase in delivery to and reabsorption of Na(+) by the distal nephron, with a resultant increase in the driving force for passive K(+) efflux across the apical membrane. Recent studies suggest that cellular mechanisms that lead to enhanced rates of Na(+) reabsorption as well as K(+) secretion in response to increases tubular flow rates are more complex. Increases in tubular flow rates directly enhance the activity of apical membrane Na(+) channels and indirectly activate a class of K(+) channels, referred to as maxi-K, that are functionally inactive under low flow states. This review addresses the role of biomechanical forces, generated by variations in urinary flow rate and tubular fluid volume, in the regulation of transepithelial Na(+) and K(+) transport in the distal nephron. The question of why the distal nephron has evolved to include a component of flow-dependent K(+) secretion is also addressed.
Collapse
Affiliation(s)
- Lisa M Satlin
- Renal-Electrolyte Div, Univ. of Pittsburgh, A919 Scaife Hall, 3550 Terrace St, Pittsburgh, PA 15261, USA
| | | | | | | |
Collapse
|
26
|
Lazrak A, Liu Z, Huang CL. Antagonistic regulation of ROMK by long and kidney-specific WNK1 isoforms. Proc Natl Acad Sci U S A 2006; 103:1615-20. [PMID: 16428287 PMCID: PMC1360592 DOI: 10.1073/pnas.0510609103] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
WNK kinases are serine-threonine kinases with an atypical placement of the catalytic lysine. Intronic deletions with increased expression of a ubiquitous long WNK1 transcript cause pseudohypoaldosteronism type 2 (PHA II), characterized by hypertension and hyperkalemia. Here, we report that long WNK1 inhibited ROMK1 by stimulating its endocytosis. Inhibition of ROMK by long WNK1 was synergistic with, but not dependent on, WNK4. A smaller transcript of WNK1 lacking the N-terminal 1-437 amino acids is expressed highly in the kidney. Whether expression of the KS-WNK1 (kidney-specific, KS) is altered in PHA II is not known. We found that KS-WNK1 did not inhibit ROMK1 but reversed the inhibition of ROMK1 caused by long WNK1. Consistent with the lack of inhibition by KS-WNK1, we found that amino acids 1-491 of the long WNK1 were sufficient for inhibiting ROMK. Dietary K(+) restriction decreases ROMK abundance in the renal cortical-collecting ducts by stimulating endocytosis, an adaptative response important for conservation of K(+) during K(+) deficiency. We found that K(+) restriction in rats increased whole-kidney transcript of long WNK1 while decreasing that of KS-WNK1. Thus, KS-WNK1 is a physiological antagonist of long WNK1. Hyperkalemia in PHA II patients with PHA II mutations may be caused, at least partially, by increased expression of long WNK1 with or without decreased expression of KS-WNK1.
Collapse
Affiliation(s)
- Ahmed Lazrak
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8856, USA
| | | | | |
Collapse
|
27
|
Lang F, Capasso G, Schwab M, Waldegger S. Renal tubular transport and the genetic basis of hypertensive disease. Clin Exp Nephrol 2005; 9:91-9. [PMID: 15980941 DOI: 10.1007/s10157-005-0355-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Accepted: 03/11/2005] [Indexed: 10/25/2022]
Abstract
Several monogenic hypertensive disorders are caused by genetic mutations leading to the deranged function and/or regulation of renal tubular NaCl transport, such as mutations of the renal epithelial Na+ channel (ENaC) in Liddle syndrome, of the kinase WNK1 (with no K) in Gordon syndrome, and of the mineralocorticoid receptor, or of 11beta-hydroxysteroid dehydrogenase. Moreover, excessive formation of aldosterone in glucocorticoid-remediable hypertension leads to severe hypertension. Conversely, impaired function of the Na+,K+,2Cl- cotransporter (NKCC2), the renal outer medullary K+ channel (ROMK1), and the renal epithelial Cl- channel ClCKb/Barttin causes Bartter syndrome and defective Na+,Cl+ cotransporter (NCCT) Gitelman syndrome, salt-wasting disorders with hypotension. These monogenic disorders are rare, but illustrate the significance of renal tubular transport in blood pressure regulation. There is little doubt, however, that deranged renal salt reabsorption significantly contributes to essential hypertension polymorphisms of several genes participating in the regulation of renal Na+ transport have been shown to be associated with blood pressure and prevalence of hypertension. Two common genes will be discussed in more detail. The first encodes the renal Cl- channel ClCKb. A gain-of-function mutation of ClCKb, increasing channel activity by 7- to 20-fold is found in approximately 20% of unselected Caucasians and 40% of an unselected African population. The second common gene variant (prevalence, 3%-5% in unselected Caucasians), to be discussed in more detail, affects the serum and glucocorticoid inducible kinase SGK1, a kinase upregulated by mineralocorticoids and enhancing the activity of ENaC, ROMK, and Na+/K+ATPase. Both gene variants are associated with slightly increased blood pressure. SGK1 further stimulates the glucose transporter SGLT1, and the SGK1 gene variant correlates, in addition, with increased body mass index.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany.
| | | | | | | |
Collapse
|
28
|
Abstract
ROMK potassium channels are present in the cortical collecting duct (CCD) of the kidney and serve as apical exit pathways for K+secretion in this nephron segment. K+secretion in the CCD is regulated by multiple factors. In this study, we show that syntaxin 1A, but not syntaxin 3 or 4, inhibited whole cell ROMK currents in Xenopus laevis oocytes. Syntaxin 1A, but not syntaxin 3 or 4, interacted with the COOH-terminal cytoplasmic domain of ROMK in intro. Coexpression with synaptobrevin 2 reversed inhibition of whole cell ROMK currents by syntaxin 1A. In excised inside-out membranes of oocytes, application of fusion proteins containing the cytoplasmic region of syntaxin 1A to the cytoplasmic face caused a dose-dependent inhibition of ROMK. We further examined regulation of the K+channels in the CCD by syntaxin 1A. Application of botulinum toxin C1 to the excised inside-out membranes of the CCD caused an increase in the activity of K+channels. In contrast, application of toxin B had no effects. These results suggest that syntaxin 1A causes a tonic inhibition of the K+channels in the apical membrane of the CCD. Binding of synaptobrevin 2 to syntaxin 1A during docking and fusion of transport vesicles to the plasma membranes of CCD may lead to activation of these channels.
Collapse
Affiliation(s)
- Tie-Jun Sun
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8856, USA
| | | | | |
Collapse
|
29
|
Giebisch G. Challenges to potassium metabolism: internal distribution and external balance. Wien Klin Wochenschr 2004; 116:353-66. [PMID: 15291287 DOI: 10.1007/bf03040914] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
A complex pump-leak system involving both active and passive transport mechanisms is responsible for the appropriate distribution of potassium (K) between the intra- and extracellular fluid compartments. In addition, the kidneys, and to a lesser extent the colon, safeguard maintenance of the narrow range of low K concentrations in the extracellular fluid. Early renal clearance studies showed that K is normally both reabsorbed and secreted by renal tubules, and that regulated secretion is the major source of K excretion. Net K secretion occurs mainly in principal cells while K absorption takes place in intercalated cells. Studies on single tubules and principal and intercalated cells have defined the determinants of K secretion and reabsorption including the electrochemical driving forces, specific carriers, ATPases, and K channels. Recent studies on the properties and molecular identity of renal K channels have also contributed significantly to understanding the renal mechanisms that transport and regulate K excretion.
Collapse
Affiliation(s)
- Gerhard Giebisch
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
30
|
Jeck N, Waldegger S, Lampert A, Boehmer C, Waldegger P, Lang PA, Wissinger B, Friedrich B, Risler T, Moehle R, Lang UE, Zill P, Bondy B, Schaeffeler E, Asante-Poku S, Seyberth H, Schwab M, Lang F. Activating Mutation of the Renal Epithelial Chloride Channel ClC-Kb Predisposing to Hypertension. Hypertension 2004; 43:1175-81. [PMID: 15148291 DOI: 10.1161/01.hyp.0000129824.12959.f0] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The chloride channel ClC-Kb is expressed in the basolateral cell membrane of the distal nephron and participates in renal NaCl reabsorption. Loss-of-function mutations of ClC-Kb lead to classic Bartter syndrome, a rare salt-wasting disorder. Recently, we identified the ClC-Kb
T481S
polymorphism, which confers a strong gain-of-function effect on the ClC-Kb chloride channel. The present study has been performed to explore the prevalence of the mutation and its functional significance in renal salt handling and blood pressure regulation. As evident from electrophysiological analysis with the 2-electrode voltage-clamp technique, heterologous expression of ClC-Kb
T481S
in Xenopus oocytes gave rise to a current that was 7-fold larger than the current produced by wild-type ClC-Kb. The prevalence of the mutant allele was significantly higher in an African population from Ghana (22%) than in whites (12%). As tested in 1 white population, carriers of ClC-Kb
T481S
were associated with significantly higher systolic (by ≈6.0 mm Hg) and diastolic (by ≈4.2 mm Hg) blood pressures and significantly higher prevalence (45% versus 25%) of hypertensive (≥140/90 mm Hg) blood pressure levels. Individuals carrying ClC-Kb
T481S
had significantly higher plasma Na
+
concentrations and significantly decreased glomerular filtration rate. In conclusion, the mutation ClC-Kb
T481S
of the renal epithelial Cl
−
channel ClC-Kb strongly activates ClC-Kb chloride channel function in vitro and may predispose to the development of essential hypertension in vivo.
Collapse
Affiliation(s)
- Nikola Jeck
- Department of Pediatrics, University of Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Chu PY, Quigley R, Babich V, Huang CL. Dietary potassium restriction stimulates endocytosis of ROMK channel in rat cortical collecting duct. Am J Physiol Renal Physiol 2003; 285:F1179-87. [PMID: 12952855 DOI: 10.1152/ajprenal.00150.2003] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ROMK potassium channels are present in the cortical collecting ducts (CCDs) of the kidney and serve as the exit pathways for K+ secretion in this nephron segment. Dietary K+ restriction reduces the abundance of ROMK in the kidney. We have previously shown that ROMK undergoes endocytosis via clathrin-coated vesicles in Xenopus laevis oocytes and in cultured cells. Here, we examined the effect of dietary K+ restriction on endocytosis of ROMK in CCDs using double-labeling immunofluorescent staining and confocal microscopic imaging in whole kidney sections as well as in individually isolated tubules. We found that ROMK abundance in kidney cortex and CCDs was reduced in rats fed a K+-restricted diet compared with rats fed the control K+ diet. In the control animals, ROMK staining was preferentially localized to the apical membrane of CCDs. Compared with control tubules, ROMK staining in CCDs was markedly shifted toward intracellular locations in animals fed a K+-deficient diet for 48 h. Some of the intracellular distribution of ROMK colocalized with an early endosomal marker, early endosomal antigen-1 or with a late endosomal/lysosomal marker, lysosomal membrane glycoprotein-120. These results suggest that K+ restriction reduces the abundance of ROMK in CCDs by increasing endocytosis and degradation of the channel protein. This decrease in the abundance of ROMK is likely important for maintaining K+ homeostasis during K+ deficiency.
Collapse
Affiliation(s)
- Po-Yin Chu
- Department of Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8856, USA
| | | | | | | |
Collapse
|
32
|
Yoo D, Flagg TP, Olsen O, Raghuram V, Foskett JK, Welling PA. Assembly and trafficking of a multiprotein ROMK (Kir 1.1) channel complex by PDZ interactions. J Biol Chem 2003; 279:6863-73. [PMID: 14604981 DOI: 10.1074/jbc.m311599200] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The ROMK subtypes of inward rectifier K+ channels (Kir 1.1, KCNJ1) mediate potassium secretion and regulate NaCl reabsorption in the kidney. In the present study, the role of the PDZ binding motif in ROMK function is explored. Here we identify the Na/H exchange regulatory factors, NHERF-1 and NHERF-2, as PDZ domain interaction partners of the ROMK channel. Characterization of the basis and consequences of NHERF association with ROMK reveals a PDZ interaction-dependent trafficking process and a coupling mechanism for linking ROMK to a channel modifier protein, the cystic fibrosis transmembrane regulator (CFTR). As measured by antibody binding of external epitope-tagged forms of Kir 1.1 in intact cells, NHERF-1 or NHERF-2 coexpression increased cell surface expression of ROMK. Channel interaction with NHERF proteins and effects of NHERF on ROMK localization were dependent on the presence of the PDZ domain binding motif in ROMK. Both NHERF proteins contain two PDZ domains; recombinant protein-protein binding assays and yeast-two-hybrid studies revealed that ROMK preferentially associates with the second PDZ domain of NHERF-1 and with the first PDZ domain of NHERF-2, precisely opposite of what has been reported for CFTR. Consistent with the scaffolding capacity of the NHERF proteins, coexpression of NHERF-2 with ROMK and CFTR dramatically increases the amount of ROMK protein that coimmunopurifies and functionally interacts with CFTR. Thus NHERF facilitates assembly of a ternary complex containing ROMK and CFTR. These observations raise the possibility that PDZ-based interactions may underscore physiological regulation and membrane targeting of ROMK in the kidney.
Collapse
Affiliation(s)
- Dana Yoo
- Department of Physiology, University of Maryland School of Medicine, Baltimore 21201, USA
| | | | | | | | | | | |
Collapse
|
33
|
Starremans PGJF, Kersten FFJ, Knoers NVAM, van den Heuvel LPWJ, Bindels RJM. Mutations in the human Na-K-2Cl cotransporter (NKCC2) identified in Bartter syndrome type I consistently result in nonfunctional transporters. J Am Soc Nephrol 2003; 14:1419-26. [PMID: 12761241 DOI: 10.1097/01.asn.0000064948.39199.a0] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Bartter syndrome (BS) is a heterogeneous renal tubular disorder affecting Na-K-Cl reabsorption in the thick ascending limb of Henle's loop. BS type I patients typically present with profound hypokalemia and metabolic alkalosis. The main goal of the present study was to elucidate the functional implications of six homozygous mutations (G193R, A267S, G319R, A508T, del526N, and Y998X) in the bumetanide-sensitive Na-K-2Cl cotransporter (hNKCC2) identified in patients diagnosed with BS type I. To this end, capped RNA (cRNA) of FLAG-tagged hNKCC2 and the corresponding mutants was injected in Xenopus laevis oocytes and transporter activity was measured after 72 h by means of a bumetanide-sensitive (22)Na(+) uptake assay at 30 degrees C. Injection of 25 ng of hNKCC2 cRNA resulted in bumetanide-sensitive (22)Na(+) uptake of 2.5 +/- 0.5 nmol/oocyte per 30 min. Injection of 25 ng of mutant cRNA yielded no significant bumetanide-sensitive (22)Na(+) uptake. Expression of wild-type and mutant transporters was confirmed by immunoblotting, showing significantly less mutant protein compared with wild-type at the same cRNA injection levels. However, when the wild-type cRNA injection level was reduced to obtain a protein expression level equal to that of the mutants, the wild-type still exhibited a significant bumetanide-sensitive (22)Na(+) uptake. Immunocytochemical analysis showed immunopositive staining of hNKCC2 at the plasma membrane for wild-type and all studied mutants. In conclusion, mutations in hNKCC2 identified in type I BS patients, when expressed in Xenopus oocytes, result in a low expression of normally routed but functionally impaired transporters. These results are in line with the hypothesis that the mutations in hNKCC2 are the underlying cause of the clinical abnormalities seen in patients with type I BS.
Collapse
|
34
|
Zeng WZ, Li XJ, Hilgemann DW, Huang CL. Protein kinase C inhibits ROMK1 channel activity via a phosphatidylinositol 4,5-bisphosphate-dependent mechanism. J Biol Chem 2003; 278:16852-6. [PMID: 12615924 DOI: 10.1074/jbc.m300619200] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The activity of apical K(+) channels in cortical collecting duct (CCD) is stimulated and inhibited by protein kinase A (PKA) and C (PKC), respectively. Direct interaction between phosphatidylinositol 4,5-bisphosphate (PIP(2)) and the cloned CCD K(+) channel, ROMK1, is critical for channel opening. We have found previously that phosphorylation of ROMK1 by PKA increases affinity of the channel for PIP(2) and mutation of PKA sites reduces the affinity of ROMK1 for PIP(2). In this study we investigate the molecular mechanism for PKC regulation of ROMK and report that mutants of ROMK1 with reduced PIP(2) affinity exhibit an increased sensitivity to inhibition by phorbol myristate acetate (PMA). The effect of PMA can be prevented by pretreatment with calphostin-C. Activation of PKC by carbachol in Xenopus oocytes co-expressing M1 muscarinic receptors also causes inhibition of the channels. Calphostin-C prevents carbachol-induced inhibition, suggesting that activation of PKC is necessary for inhibition of the channels. PMA reduces open probability of the channel in cell-attached patch clamp recordings. After inhibition by PMA in cell-attached recordings, application of PIP(2) to the cytoplasmic face of excised inside-out membranes restores channel activity. PMA reduces PIP(2) content in oocyte membrane and calphostin-C prevents the reduction. These results suggest that reduction of membrane PIP(2) content contributes to the inhibition of ROMK1 channels by PKC. This mechanism may underscore the inhibition of K(+) secretion in CCD by hormones that activate PKC.
Collapse
Affiliation(s)
- Wei-Zhong Zeng
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8856, USA
| | | | | | | |
Collapse
|
35
|
Kishore BK, Kran CM, Reif M, Menon AG. Molecular physiology of urinary concentration defect in elderly population. Int Urol Nephrol 2002; 33:235-48. [PMID: 12092636 DOI: 10.1023/a:1015239915543] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
It is estimated that by the year 2050 one in five Americans will be 65 years or older. This mandates the growing need for clinical and basic research in the field of geriatric medicine to understand age-related maladies. The most prominent abnormality in renal function in the aging population is the inability to handle water, frequently resulting in hypo- or hyperosmolar states, and the associated electrolyte imbalances. During the past decade, thanks to the advent of powerful molecular techniques, rapid strides have been made in the approaches employed to understand and dissect the physiology of renal function in general and the urinary concentration mechanism in particular. Using an integrated approach of clinical observations, experimental model systems, molecular analysis, and functional genomics, a more comprehensive picture of the interplay of physiological systems in the genesis of urinary concentration defect in the elderly is beginning to emerge. Much remains to be deciphered regarding the complex interactions between the role of environment, genetics, diet, pharmacological agents and the general effects of aging on kidney function. The emerging importance of socio-economic and quality of life issues surrounding geriatric medicine encourage public and private support and funding for research in the area of age-related diseases, especially as they are related to the kidney.
Collapse
Affiliation(s)
- B K Kishore
- Department of Internal Medicine, University of Cincinnati Medical Center, OH, USA.
| | | | | | | |
Collapse
|
36
|
Abstract
A complex pump-leak system involving both active and passive transport mechanisms is responsible for the appropriate distribution of potassium (K) between the intra- and extracellular fluid compartments. In addition, the kidneys, and to a lesser extent the colon, safeguard maintenance of the narrow range of low K concentrations in the extracellular fluid. Early renal clearance studies showed that K is normally both reabsorbed and secreted by renal tubules, and that regulated secretion is the major source of K excretion. Studies at the tubule and cell level have localized secretion and reabsorption of K to principal and intercalated cells in the collecting ducts. Measurements of the electrochemical driving forces across individual cell membranes have permitted the characterization of specific ATPases, K channels and K cotransporters and also provided insights into the molecular structure of individual transporters that regulate K excretion.
Collapse
Affiliation(s)
- Gerhard H Giebisch
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| |
Collapse
|
37
|
Zeng WZ, Babich V, Ortega B, Quigley R, White SJ, Welling PA, Huang CL. Evidence for endocytosis of ROMK potassium channel via clathrin-coated vesicles. Am J Physiol Renal Physiol 2002; 283:F630-9. [PMID: 12217853 DOI: 10.1152/ajprenal.00378.2001] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ROMK channels are present in the cortical collecting ducts of kidney and are responsible for K(+) secretion in this nephron segment. Recent studies suggest that endocytosis of ROMK channels is important for regulation of K(+) secretion in cortical collecting ducts. We investigated the molecular mechanisms for endocytosis of ROMK channels expressed in Xenopus laevis oocytes and cultured Madin-Darby canine kidney cells. When plasma membrane insertion of newly synthesized channel proteins was blocked by incubation with brefeldin A, ROMK currents decreased with a half-time of ~6 h. Coexpression with the Lys44-->Ala dominant-negative mutant dynamin, but not wild-type dynamin, reduced the rate of reduction of ROMK in the presence of brefeldin A. Mutation of Asn371 to Ile in the putative NPXY internalization motif of ROMK1 abolished the effect of the Lys44-->Ala dynamin mutant on endocytosis of the channel. Coimmunoprecipitation study and confocal fluorescent imaging revealed that ROMK channels associated with clathrin coat proteins in Madin-Darby canine kidney cells. These results provide compelling evidence for endocytosis of ROMK channels via clathrin-coated vesicles.
Collapse
Affiliation(s)
- Wei-Zhong Zeng
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas 75390-8856, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Zeng WZ, Liou HH, Krishna UM, Falck JR, Huang CL. Structural determinants and specificities for ROMK1-phosphoinositide interaction. Am J Physiol Renal Physiol 2002; 282:F826-34. [PMID: 11934692 DOI: 10.1152/ajprenal.00300.2001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We have recently reported that direct interaction between phosphatidylinositol bisphosphate (PIP(2)) and the COOH-terminal cytoplasmic domain of ROMK1 is important for opening of the channel. We identified arginine-188 of ROMK1 as a critical residue for this interaction. Here, we further report that substitution of a neutral amino acid for lysine-181, arginine-217, or lysine-218 decreases single-channel open probability for the full-conductance state and increases the frequency of opening at a subconductance state. Compared with wild-type ROMK1 channels, these substitution mutants also display an increased sensitivity to the block by anti-PIP(2) antibodies and to inhibition by intracellular protons. These results indicate that, like arginine-188, lysine-181, arginine-217, and lysine-218 are also involved in interactions with PIP(2) and are critical for ROMK1 to open at full conductance. Using synthetic phosphoinositides containing phosphates at different positions in the head group, we also examined the specificities of phosphoinositides in the regulation of ROMK1 channels. We found that phosphoinositides containing phosphate at both positions 4 and 5 of the inositol head group have the highest efficacy in activating ROMK1 channels. These results suggest that phosphatidylinositol 4,5-bisphosphate is likely the important phosphoinositide in the regulation of ROMK1 channels in a physiological membrane milieu.
Collapse
Affiliation(s)
- Wei-Zhong Zeng
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8856, USA
| | | | | | | | | |
Collapse
|
39
|
Abstract
Many transport functions in renal tubules depend on potassium (K) channels. Not only does K secretion and the maintenance of external K balance depend on K channel activity in principal tubule cells, but K channels also regulate cell volume; they are an integral party of cell function in all tubule cells because of their key role in the generation of the cell-negative electrical potential that affects the transmembrane movement of many charged solutes. Moreover, the recycling of K across the apical membrane of the thick ascending limb (TAL) plays an important role in the control of NaCl reabsorption in this tubule segment. Significant progress in our understanding of the structure and function of renal K channels has become possible by combining several strategies. These include transport studies in single tubules, application of the patch-clamp technique for exploring the properties of single K channels in native tubules and the cloning, and expression of diverse K channels of renal origin. Insights from these investigations promise to provide a deeper understanding of the mechanism by which K channels participate in many diverse tubule functions.
Collapse
Affiliation(s)
- G Giebisch
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520-8026, USA
| |
Collapse
|
40
|
Derst C, Hirsch JR, Preisig-Müller R, Wischmeyer E, Karschin A, Döring F, Thomzig A, Veh RW, Schlatter E, Kummer W, Daut J. Cellular localization of the potassium channel Kir7.1 in guinea pig and human kidney. Kidney Int 2001; 59:2197-205. [PMID: 11380822 DOI: 10.1046/j.1523-1755.2001.00735.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND K(+) channels have important functions in the kidney, such as maintenance of the membrane potential, volume regulation, recirculation, and secretion of potassium ions. The aim of this study was to obtain more information on the localization and possible functional role of the inwardly rectifying K(+) channel, Kir7.1. METHODS Kir7.1 cDNA (1114 bp) was isolated from guinea pig kidney (gpKir7.1), and its tissue distribution was analyzed by reverse transcriptase-polymerase chain reaction (RT-PCR). In addition, a genomic DNA fragment (6153 bp) was isolated from a genomic library. cRNA was expressed in Xenopus laevis oocytes for functional studies. Immunohistochemistry and RT-PCR were used to localize Kir7.1 in guinea pig and human kidney. RESULTS The expression of gpKir7.1 in Xenopus laevis oocytes revealed inwardly rectifying K(+) currents. The reversal potential was strongly dependent on the extracellular K(+) concentration, shifting from -14 mV at 96 mmol/L K(+) to -90 mV at 1 mmol/L K(+). gpKir7.1 showed a low affinity for Ba(2+). Significant expression of gpKir7.1 was found in brain, kidney, and lung, but not in heart, skeletal muscle, liver, or spleen. Immunocytochemical detection in guinea pig identified the gpKir7.1 protein in the basolateral membrane of epithelial cells of the proximal tubule. RT-PCR analysis identified strong gpKir7.1 expression in the proximal tubule and weak expression in glomeruli and thick ascending limb. In isolated human tubule fragments, RT-PCR showed expression in proximal tubule and thick ascending limb. CONCLUSION Our results suggest that Kir7.1 may contribute to basolateral K(+) recycling in the proximal tubule and in the thick ascending limb.
Collapse
Affiliation(s)
- C Derst
- Institut für Normale und Pathologische Physiologie, Philipps-Universität, Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Giebisch G, Wang W. Renal tubule potassium channels: function, regulation and structure. ACTA PHYSIOLOGICA SCANDINAVICA 2000; 170:153-73. [PMID: 11114953 DOI: 10.1046/j.1365-201x.2000.00770.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- G Giebisch
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
42
|
Keren-Raifman T, Ivanina T, Bismuth Y, Dascal N. Expression cloning of KCRF, a potassium channel regulatory factor. Biochem Biophys Res Commun 2000; 274:852-8. [PMID: 10924366 DOI: 10.1006/bbrc.2000.3240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
By functional coexpression screening of a rat cDNA library in Xenopus oocytes, we have cloned a protein (KCRF: K Channel Regulatory Factor) that reduces currents of several K(+) channels: G protein-activated GIRK1/4 (K(ir)3.1/K(ir)3.4), inward rectifier IRK1 (K(ir)2.1), and voltage-dependent K(V)1.1/K(V)beta1.1. KCRF did not modulate two other K(+) channels: ROMK1 (K(ir)1.1) and GIRK1/2 (K(ir)3.1/K(ir)3.2) and the voltage-dependent L-type Ca(2+) channels. Western blot analysis showed that KCRF is ubiquitous in rat tissues. Biochemical and electrophysiological experiments revealed that coexpression of KCRF causes a decrease in the level of expression of IRK1 and K(V)1.1/K(V)beta1.1 proteins in the oocytes.
Collapse
Affiliation(s)
- T Keren-Raifman
- Department of Physiology and Pharmacology, Sackler School of Medicine, Ramat Aviv, 69978, Israel
| | | | | | | |
Collapse
|
43
|
Wei Y, Bloom P, Gu R, Wang W. Protein-tyrosine phosphatase reduces the number of apical small conductance K+ channels in the rat cortical collecting duct. J Biol Chem 2000; 275:20502-7. [PMID: 10787405 DOI: 10.1074/jbc.m000783200] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous studies have demonstrated that an increase in the activity of protein-tyrosine kinase (PTK) is involved in the down-regulation of the activity of apical small conductance K(+) (SK) channels in the cortical collecting duct (CCD) from rats on a K(+)-deficient diet (). We used the patch clamp technique to investigate the role of protein-tyrosine phosphatase (PTP) in the regulation of the activity of SK channels in the CCD from rats on a high K(+) diet. Western blot analysis indicated that PTP-1D is expressed in the renal cortex. Application of 1 microm phenylarsine oxide (PAO) or 1 mm benzylphosphonic acid, agents that inhibit PTP, reversibly reduced channel activity by 95%. Pretreatment of CCDs with PAO for 30 min decreased the mean NP(o) reversibly from control value 3.20 to 0.40. Addition of 1 microm herbimycin A, an inhibitor of PTK, had no significant effect on channel activity in the CCDs from rats on a high K(+) diet. However, herbimycin A abolished the inhibitory effect of PAO, indicating that the effect of PAO is the result of interaction between PTK and PTP. Addition of brefeldin A, an agent that blocks protein trafficking from Golgi complex to the membrane, had no effect on channel activity. Moreover, application of colchicine, a microtubule inhibitor, or paclitaxel, a microtubule stabilizer, had no effect on channel activity. In contrast, PAO still reduced channel activity in the presence of brefeldin A, colchicine, or paclitaxel. Furthermore, the effect of PAO on channel activity was absent when the tubules were bathed in 16% sucrose-containing bath solution or treated with concanavalin A. We conclude that PTP is involved in the regulation of the activity of SK channels and that inhibition of PTP may facilitate the internalization of the SK channels.
Collapse
Affiliation(s)
- Y Wei
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | |
Collapse
|
44
|
Abstract
BACKGROUND The calcineurin inhibitors cyclosporine and FK506 are widely used for immunosuppression in solid organ transplantation. One of the side effects of these agents is renal magnesium wasting. The site of action and molecular mechanism of this effect are not known. We hypothesized that agents such as diuretics that cause renal magnesium wasting through a similar action would not have an additive effect on magnesium deficiency with calcineurin inhibitors. METHODS The records of 50 heart transplant patients on calcineurin inhibitors were reviewed to determine levels of serum magnesium and required replacement dose of magnesium, diuretic usage, and other laboratory values. RESULTS Loop diuretics did not change either the magnesium level or magnesium replacement requirements in patients on calcineurin inhibitors. In contrast, the thiazide diuretic resulted in an increase in serum magnesium and a decrease in magnesium replacement. Results were similar when the cyclosporine or FK506 groups were evaluated alone. Patients taking FK506 had lower serum magnesium values and higher requirements for magnesium replacement compared with patients taking cyclosporine. CONCLUSION We conclude that calcineurin inhibitors and loop diuretics have a similar site of action.
Collapse
Affiliation(s)
- J M Arthur
- Departments of Medicine and Biochemistry and Molecular Biology, The University of Louisville, KY 40202-1764, USA.
| | | |
Collapse
|
45
|
Huang DY, Osswald H, Vallon V. Sodium reabsorption in thick ascending limb of Henle's loop: effect of potassium channel blockade in vivo. Br J Pharmacol 2000; 130:1255-62. [PMID: 10903963 PMCID: PMC1572189 DOI: 10.1038/sj.bjp.0703429] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
1. Based on previous in vitro studies, inhibition of K(+) recycling in thick ascending limb (TAL) is expected to lower Na(+) reabsorption through (i) reducing the luminal availability of K(+) to reload the Na(+)-2Cl(-)-K(+) cotransporter and (ii) diminishing the lumen positive transepithelial potential difference which drives paracellular cation transport. 2. This issue was investigated in anaesthetized rats employing microperfusion of Henle's loop downstream from late proximal tubular site with K(+)-free artificial tubular fluid in nephrons with superficial glomeruli. 3. The unselective K(+) channel blocker Cs(+) (5 - 40 mM) dose-dependently increased early distal tubular delivery of fluid and Na(+) with a maximum increase of approximately 20 and 185%, respectively, indicating predominant effects on water-impermeable TAL. 4. The modest inhibition of Na(+) reabsorption in response to the 15 mM of Cs(+) but not the enhanced inhibition by 20 mM Cs(+) was prevented by luminal K(+) supplementation. Furthermore, pretreatment with 20 mM Cs(+) did not attenuate the inhibitory effect of furosemide (100 microM) on Na(+)-2Cl(-)-K(+) cotransport. 5. Neither inhibitors of large (charybdotoxin 1 microM) nor low (glibenclamide 250 microM; U37883A 100 microM) conductance K(+) channels altered loop of Henle fluid or Na(+) reabsorption. 6. The intermediate conductance K(+) channel blockers verapamil and quinine (100 microM) modestly increased early distal tubular Na(+) but not fluid delivery, indicating a role for this K(+) channel in Na(+) reabsorption in TAL. As observed for equieffective concentrations of Cs(+) (15 mM), Na(+) reabsorption was preserved by K(+) supplementation. 7. The results indicate that modest inhibition of K(+) channels lowers the luminal availability of K(+) and thus transcellular Na(+) reabsorption in TAL. More complete inhibition lowers paracellular Na(+) transport probably by reducing or even abolishing the lumen positive transepithelial potential difference. Under the latter conditions, transcellular Na(+) transport may be restored by paracellular K(+) backleak.
Collapse
Affiliation(s)
- D Y Huang
- Department of Pharmacology, University of Tübingen, Germany
| | | | | |
Collapse
|
46
|
Tan HL, Alings M, Van Olden RW, Wilde AA. Long-term (subacute) potassium treatment in congenital HERG-related long QT syndrome (LQTS2). J Cardiovasc Electrophysiol 1999; 10:229-33. [PMID: 10090227 DOI: 10.1111/j.1540-8167.1999.tb00665.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Congenital long QT syndrome (LQTS) is subdivided according to the underlying gene defect. In LQTS2, an aberrant HERG gene that encodes the potassium channel IKr leads to insufficient IKr activity and delayed repolarization, causing ECG abnormalities and torsades de pointes (TdP). Increasing serum potassium levels by potassium infusion normalizes the ECG in LQTS2 because IKr activity varies with serum potassium levels. METHODS AND RESULTS In an LQTS2 patient who presented with TdP, we attempted to achieve a long-term (subacute) elevation of serum potassium by increased potassium intake and potassium-sparing drugs. However, due to renal potassium homeostasis, it was impossible to achieve a long-lasting rise of serum potassium above 4.0 mmol/L. CONCLUSION Although raising serum potassium reverses the ECG abnormalities in LQTS2, a long-lasting rise of serum potassium is only partially achievable because in the presence of normal renal function, potassium homeostasis limits the amount of serum potassium increase.
Collapse
Affiliation(s)
- H L Tan
- Department of Cardiology, University of Amsterdam, Academic Medical Center, The Netherlands
| | | | | | | |
Collapse
|
47
|
Wolkoff AW, Suchy FJ, Moseley RH, Meier PJ, Gollan JL, Freimer N, Fitz JG, Boyer JL, Berk PD, Scharschmidt BF. Advances in hepatic transport: molecular mechanisms, genetic disorders, and treatment. A summary of the 1998 AASLD single topic conference. Hepatology 1998; 28:1713-9. [PMID: 9828241 DOI: 10.1002/hep.510280637] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- A W Wolkoff
- Albert Einstein College of Medicine, Bronx, NY, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kubokawa M, Nakaya S, Yoshioka Y, Nakamura K, Sato F, Mori Y, Kubota T. Activation of inwardly rectifying K+ channel in OK proximal tubule cells involves cGMP-dependent phosphorylation process. THE JAPANESE JOURNAL OF PHYSIOLOGY 1998; 48:467-76. [PMID: 10021500 DOI: 10.2170/jjphysiol.48.467] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The inwardly rectifying K+ channel with an inward conductance of about 90 pS in the surface membrane of cultured opossum kidney proximal tubule (OKP) cell is activated by cyclic AMP-dependent protein kinase (PKA). In this study, we further examined the involvement of the guanosine 3',5'-cyclic monophosphate (cGMP)-dependent process in modulation of this K+ channel by using the patch-clamp technique. In cell-attached patches, channel activity was increased by the application of either N2, 2'-O-dibutyrylguanosine 3',5'-cyclic monophosphate (DBcGMP, 100 microM) or 8-bromoguanosine 3',5'-cyclic monophosphate (8BrcGMP, 100 microM), and it was inhibited by KT5823 (10 microM), a membrane-permeable specific inhibitor of cGMP-dependent protein kinase (PKG). The effect of DBcGMP on channel activity was abolished by the pretreatment of cells with KT5823 (10 microM), but it was observed in the presence of KT5720 (200 nM), a specific inhibitor of PKA. Furthermore, atrial natriuretic peptide (ANP, 10 nM) increased channel activity, which was also prevented by the application of KT5823 (10 microM). In inside-out patches, ATP (3 mM) was required to maintain channel activity, which was inhibited by KT5823 (10 microM), but it was not increased by cGMP (100 microM) alone. The channel activity was increased by the coapplication of PKG (500 U/ml) and cGMP (100 microM). These results suggest that cGMP activates the inwardly rectifying K+ channel in OKP cells through PKG-mediated phosphorylation processes independent of PKA-mediated processes, and that ANP is an agonist which stimulates PKG-mediated processes in the proximal tubule cell. Furthermore, it is suggested that the ATP-dependent channel activity in inside-out patches is maintained at least in part by PKG, which is the membrane-bound catalytic domain.
Collapse
Affiliation(s)
- M Kubokawa
- Department of Physiology II, Iwate Medical University, School of Medicine, Morioka, 020-8505, Japan.
| | | | | | | | | | | | | |
Collapse
|
49
|
Cluzeaud F, Reyes R, Escoubet B, Fay M, Lazdunski M, Bonvalet JP, Lesage F, Farman N. Expression of TWIK-1, a novel weakly inward rectifying potassium channel in rat kidney. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:C1602-9. [PMID: 9843722 DOI: 10.1152/ajpcell.1998.275.6.c1602] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Several K+ conductances have been identified in the kidney, with specific properties and localization in distinct cell types and membrane domains. On the other hand, several K+ channels have been characterized at the molecular level. By immunolocalization, we show that a new inward rectifying K+ channel, TWIK-1, is specifically expressed in distinct tubular segments and cell types of the rat kidney. In the proximal tubule, TWIK-1 prevails in the initial portions (convoluted part), where it is restricted to the apical (brush-border) membrane. In the collecting duct, immunofluorescence was intracellular or confined to the apical membrane and restricted to intercalated cells, i.e., in cells lacking aquaporin-2, as shown by double immunofluorescence. TWIK was also expressed in medullary and cortical parts of the thick limb of the loop of Henle, identified with an anti-Tamm-Horsfall protein antibody (double immunofluorescence). The intensity of TWIK-1 immunolabeling was unchanged in rats fed a low-Na+ or a low-K+ diet. Because TWIK-1 shares common properties with the low-conductance apical K+ channel of the collecting duct, we propose that it could play a role in K+ secretion, complementary to ROMK, another recently characterized K+ channel located in principal cells of the cortical collecting duct and in the loop of Henle.
Collapse
Affiliation(s)
- F Cluzeaud
- Unité 478, Institut National de la Santé et de la Recherche Médicale, Faculté de Médecine X. Bichat, F-75870 Paris cedex 18, France
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kohda Y, Ding W, Phan E, Housini I, Wang J, Star RA, Huang CL. Localization of the ROMK potassium channel to the apical membrane of distal nephron in rat kidney. Kidney Int 1998; 54:1214-23. [PMID: 9767537 DOI: 10.1046/j.1523-1755.1998.00120.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The apical potassium (K+) channels mediate K+ recycling in thick ascending limb (TAL) and K+ secretion in cortical collecting duct (CCD). Recently, the cDNAs for a family of renal K+ channels, ROMK1, -2 and -3, were identified. Based on the biophysical properties and mRNA distribution, it is believed that these ROMK cDNAs encode the apical K+ channels of TAL and CCD. However, the information for cellular and subcellular localization of the ROMK proteins in these tubules is still not available. METHODS Paraffin or frozen kidney sections from adult Sprague-Dawley rats were stained by polyclonal antibodies against the N- and C-terminal domain of ROMK. Immunoreactive staining was visualized by color development from horseradish peroxidase reaction. Membrane homogenates from kidney were analyzed by Western blot analysis. RESULTS The polyclonal antibodies against cytoplasmic epitope of ROMK recognized a approximately 42 kD protein in the membrane homogenates from kidney, but not from liver. Staining by immunocytochemistry revealed that ROMK channels were localized to the apical membranes of the distal nephron in cortex and outer medulla, including thick ascending limb and collecting tubule. ROMK staining was absent in glomerulus, proximal tubule and inner medulla. Double staining of the tissue section with both ROMK-specific and H+-ATPase-specific antibodies revealed labeling of ROMK in the principal cells of the collecting tubules. CONCLUSIONS These results further strengthen the idea that ROMK channels play important roles in the recycling of K+ in TAL and the secretion of K+ in CCD.
Collapse
Affiliation(s)
- Y Kohda
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas 75235-8856, USA
| | | | | | | | | | | | | |
Collapse
|