1
|
Kok M, Brodsky JL. The biogenesis of potassium transporters: implications of disease-associated mutations. Crit Rev Biochem Mol Biol 2024; 59:154-198. [PMID: 38946646 PMCID: PMC11444911 DOI: 10.1080/10409238.2024.2369986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
The concentration of intracellular and extracellular potassium is tightly regulated due to the action of various ion transporters, channels, and pumps, which reside primarily in the kidney. Yet, potassium transporters and cotransporters play vital roles in all organs and cell types. Perhaps not surprisingly, defects in the biogenesis, function, and/or regulation of these proteins are linked to range of catastrophic human diseases, but to date, few drugs have been approved to treat these maladies. In this review, we discuss the structure, function, and activity of a group of potassium-chloride cotransporters, the KCCs, as well as the related sodium-potassium-chloride cotransporters, the NKCCs. Diseases associated with each of the four KCCs and two NKCCs are also discussed. Particular emphasis is placed on how these complex membrane proteins fold and mature in the endoplasmic reticulum, how non-native forms of the cotransporters are destroyed in the cell, and which cellular factors oversee their maturation and transport to the cell surface. When known, we also outline how the levels and activities of each cotransporter are regulated. Open questions in the field and avenues for future investigations are further outlined.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Ehret E, Stroh S, Auberson M, Ino F, Jäger Y, Maillard M, Szabo R, Bugge TH, Frateschi S, Hummler E. Kidney-Specific Membrane-Bound Serine Proteases CAP1/Prss8 and CAP3/St14 Affect ENaC Subunit Abundances but Not Its Activity. Cells 2023; 12:2342. [PMID: 37830556 PMCID: PMC10572026 DOI: 10.3390/cells12192342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
The serine proteases CAP1/Prss8 and CAP3/St14 are identified as ENaC channel-activating proteases in vitro, highly suggesting that they are required for proteolytic activation of ENaC in vivo. The present study tested whether CAP3/St14 is relevant for renal proteolytic ENaC activation and affects ENaC-mediated Na+ absorption following Na+ deprivation conditions. CAP3/St14 knockout mice exhibit a significant decrease in CAP1/Prss8 protein expression with altered ENaC subunit and decreased pNCC protein abundances but overall maintain sodium balance. RNAscope-based analyses reveal co-expression of CAP3/St14 and CAP1/Prss8 with alpha ENaC in distal tubules of the cortex from wild-type mice. Double CAP1/Prss8; CAP3/St14-deficiency maintained Na+ and K+ balance on a Na+-deprived diet, restored ENaC subunit protein abundances but showed reduced NCC activity under Na+ deprivation. Overall, our data clearly show that CAP3/St14 is not required for direct proteolytic activation of ENaC but for its protein abundance. Our study reveals a complex regulation of ENaC by these serine proteases on the expression level rather than on its proteolytic activation.
Collapse
Affiliation(s)
- Elodie Ehret
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
- National Center of Competence in Research “Kidney.CH”, 1011 Lausanne, Switzerland
| | - Sévan Stroh
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
| | - Muriel Auberson
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
| | - Frédérique Ino
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
- Department of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Yannick Jäger
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Marc Maillard
- Service of Nephrology, Department of Medicine, Lausanne University Hospital (CHUV), 1005 Lausanne, Switzerland
| | - Roman Szabo
- National Institutes of Health/NIDCR, Bethesda, MD 20892, USA
| | - Thomas H. Bugge
- National Institutes of Health/NIDCR, Bethesda, MD 20892, USA
| | - Simona Frateschi
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
| | - Edith Hummler
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (E.E.)
- National Center of Competence in Research “Kidney.CH”, 1011 Lausanne, Switzerland
| |
Collapse
|
3
|
Hao S, Zhao H, Hao DH, Ferreri NR. MicroRNA-195a-5p Regulates Blood Pressure by Inhibiting NKCC2A. Hypertension 2023; 80:426-439. [PMID: 36448465 PMCID: PMC9852070 DOI: 10.1161/hypertensionaha.122.19794] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Previous studies showed that miR-195a-5p was among the most abundant microRNAs (miRNAs) expressed in the kidney. METHODS Lentivirus silencing of tumor necrosis factor-α (TNF) was performed in vivo and in vitro. Luciferase reporter assays confirmed that bumetanide-sensitive Na+-K+-2Cl- cotransporter isoform A (NKCC2A) mRNA is targeted and repressed by miR-195a-5p. Radiotelemetry was used to measure mean arterial pressure. RESULTS TNF upregulates mmu-miR-195a-5p, and -203 and downregulates mmu-miR-30c and -100 in the medullary thick ascending limb of male mice. miR-195a-5p was >3-fold higher in the renal outer medulla of mice given an intrarenal injection of murine recombinant TNF, whereas silencing TNF inhibited miR-195a-5p expression by ≈51%. Transient transfection of a miR-195a-5p mimic into medullary thick ascending limb cells suppressed NKCC2A mRNA by ≈83%, whereas transfection with Anti-miR-195a-5p increased NKCC2A mRNA. Silencing TNF in medullary thick ascending limb cells prevented increases in miR-195 induced by 400 mosmol/kg H2O medium, an effect reversed by transfection with a miR-195a-5p mimic. Expression of phosphorylated NKCC2 increased 1.5-fold in medullary thick ascending limb cells transfected with Anti-miR-195a-5p and a miR-195a-5p mimic prevented the increase, which was induced by silencing TNF in cells exposed to 400 mosmol/kg H2O medium after osmolality was increased by adding NaCl. Intrarenal injection of TNF suppressed NKCC2A mRNA, whereas injection of miR-195a-5p prevented the increase of NKCC2A mRNA abundance and phosphorylated NKCC2 expression when TNF was silenced. Intrarenal injection with miR-195a-5p markedly attenuated MAP after renal silencing of TNF in mice given 1% NaCl. CONCLUSIONS The study identifies miR-195a-5p as a salt-sensitive and TNF-inducible miRNA that attenuates NaCl-mediated increases in blood pressure by inhibiting NKCC2A.
Collapse
Affiliation(s)
- Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla
| | - Hong Zhao
- Department of Pharmacology, New York Medical College, Valhalla
| | - David H Hao
- Department of Pharmacology, New York Medical College, Valhalla
| | | |
Collapse
|
4
|
Wang B, Wang Y, Wen Y, Zhang YL, Ni WJ, Tang TT, Cao JY, Yin Q, Jiang W, Yin D, Li ZL, Lv LL, Liu BC. Tubular-specific CDK12 knockout causes a defect in urine concentration due to premature cleavage of the slc12a1 gene. Mol Ther 2022; 30:3300-3312. [PMID: 35581939 PMCID: PMC9552909 DOI: 10.1016/j.ymthe.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/21/2022] [Accepted: 05/11/2022] [Indexed: 11/27/2022] Open
Abstract
Cyclin-dependent kinase 12 (CDK12) plays a critical role in regulating gene transcription. CDK12 inhibition is a potential anticancer therapeutic strategy. However, several clinical trials have shown that CDK inhibitors might cause renal dysfunction and electrolyte disorders. CDK12 is abundant in renal tubular epithelial cells (RTECs), but the exact role of CDK12 in renal physiology remains unclear. Genetic knockout of CDK12 in mouse RTECs causes polydipsia, polyuria, and hydronephrosis. This phenotype is caused by defects in water reabsorption that are the result of reduced Na-K-2Cl cotransporter 2 (NKCC2) levels in the kidney. In addition, CKD12 knockout causes an increase in Slc12a1 (which encodes NKCC2) intronic polyadenylation events, which results in Slc12a1 truncated transcript production and NKCC2 downregulation. These findings provide novel insight into CDK12 being necessary for maintaining renal homeostasis by regulating NKCC2 transcription, which explains the critical water and electrolyte disturbance that occurs during the application of CDK12 inhibitors for cancer treatment. Therefore, there are safety concerns about the clinical use of these new anticancer drugs.
Collapse
Affiliation(s)
- Bin Wang
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Yao Wang
- Nanjing Medical University, Nanjing, Jiangsu, China; Department of Nephrology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yi Wen
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China.
| | - Yi-Lin Zhang
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Wei-Jie Ni
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Jing-Yuan Cao
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Qing Yin
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Wei Jiang
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Di Yin
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong da Hospital, Southeast University School of Medicine, No. 87, Dingjiaqiao Road, Gulou District, Nanjing, Jiangsu Province, China; Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
5
|
Kidney-Specific CAP1/Prss8-Deficient Mice Maintain ENaC-Mediated Sodium Balance through an Aldosterone Independent Pathway. Int J Mol Sci 2022; 23:ijms23126745. [PMID: 35743186 PMCID: PMC9224322 DOI: 10.3390/ijms23126745] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 12/14/2022] Open
Abstract
The serine protease prostasin (CAP1/Prss8, channel-activating protease-1) is a confirmed in vitro and in vivo activator of the epithelial sodium channel ENaC. To test whether proteolytic activity or CAP1/Prss8 abundance itself are required for ENaC activation in the kidney, we studied animals either hetero- or homozygous mutant at serine 238 (S238A; Prss8cat/+ and Prss8cat/cat), and renal tubule-specific CAP1/Prss8 knockout (Prss8PaxLC1) mice. When exposed to varying Na+-containing diets, no changes in Na+ and K+ handling and only minor changes in the expression of Na+ and K+ transporting protein were found in both models. Similarly, the α- or γENaC subunit cleavage pattern did not differ from control mice. On standard and low Na+ diet, Prss8cat/+ and Prss8cat/cat mice exhibited standard plasma aldosterone levels and unchanged amiloride-sensitive rectal potential difference indicating adapted ENaC activity. Upon Na+ deprivation, mice lacking the renal CAP1/Prss8 expression (Prss8PaxLC1) exhibit significantly decreased plasma aldosterone and lower K+ levels but compensate by showing significantly higher plasma renin activity. Our data clearly demonstrated that the catalytic activity of CAP1/Prss8 is dispensable for proteolytic ENaC activation. CAP1/Prss8-deficiency uncoupled ENaC activation from its aldosterone dependence, but Na+ homeostasis is maintained through alternative pathways.
Collapse
|
6
|
Clinical and Genetic Characterization of Patients with Bartter and Gitelman Syndrome. Int J Mol Sci 2022; 23:ijms23105641. [PMID: 35628451 PMCID: PMC9144947 DOI: 10.3390/ijms23105641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Bartter (BS) and Gitelman (GS) syndrome are autosomal recessive inherited tubulopathies, whose clinical diagnosis can be challenging, due to rarity and phenotypic overlap. Genotype-phenotype correlations have important implications in defining kidney and global outcomes. The aim of our study was to assess the diagnostic rate of whole-exome sequencing (WES) coupled with a bioinformatic analysis of copy number variations in a population of 63 patients with BS and GS from a single institution, and to explore genotype-phenotype correlations. We obtained a diagnostic yield of 86% (54/63 patients), allowing disease reclassification in about 14% of patients. Although some clinical and laboratory features were more commonly reported in patients with BS or GS, a significant overlap does exist, and age at onset, preterm birth, gestational age and nephro-calcinosis are frequently misleading. Finally, chronic kidney disease (CKD) occurs in about 30% of patients with BS or GS, suggesting that the long-term prognosis can be unfavorable. In our cohort the features associated with CKD were lower gestational age at birth and a molecular diagnosis of BS, especially BS type 1. The results of our study demonstrate that WES is useful in dealing with the phenotypic heterogeneity of these disorders, improving differential diagnosis and genotype-phenotype correlation.
Collapse
|
7
|
Marcoux AA, Tremblay LE, Slimani S, Fiola MJ, Mac-Way F, Haydock L, Garneau AP, Isenring P. Anatomophysiology of the Henle's Loop: Emphasis on the Thick Ascending Limb. Compr Physiol 2021; 12:3119-3139. [PMID: 34964111 DOI: 10.1002/cphy.c210021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The loop of Henle plays a variety of important physiological roles through the concerted actions of ion transport systems in both its apical and basolateral membranes. It is involved most notably in extracellular fluid volume and blood pressure regulation as well as Ca2+ , Mg2+ , and acid-base homeostasis because of its ability to reclaim a large fraction of the ultrafiltered solute load. This nephron segment is also involved in urinary concentration by energizing several of the steps that are required to generate a gradient of increasing osmolality from cortex to medulla. Another important role of the loop of Henle is to sustain a process known as tubuloglomerular feedback through the presence of specialized renal tubular cells that lie next to the juxtaglomerular arterioles. This article aims at describing these physiological roles and at discussing a number of the molecular mechanisms involved. It will also report on novel findings and uncertainties regarding the realization of certain processes and on the pathophysiological consequences of perturbed salt handling by the thick ascending limb of the loop of Henle. Since its discovery 150 years ago, the loop of Henle has remained in the spotlight and is now generating further interest because of its role in the renal-sparing effect of SGLT2 inhibitors. © 2022 American Physiological Society. Compr Physiol 12:1-21, 2022.
Collapse
Affiliation(s)
- Andrée-Anne Marcoux
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Laurence E Tremblay
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Samira Slimani
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Marie-Jeanne Fiola
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Fabrice Mac-Way
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Ludwig Haydock
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Alexandre P Garneau
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada.,Cardiometabolic Axis, School of Kinesiology and Physical Activity Sciences, University of Montréal, Montréal, QC, Canada
| | - Paul Isenring
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| |
Collapse
|
8
|
Marcoux A, Tremblay LE, Slimani S, Fiola M, Mac‐Way F, Garneau AP, Isenring P. Molecular characteristics and physiological roles of Na + -K + -Cl - cotransporter 2. J Cell Physiol 2021; 236:1712-1729. [PMID: 32776569 PMCID: PMC7818487 DOI: 10.1002/jcp.29997] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/28/2020] [Accepted: 07/24/2020] [Indexed: 12/23/2022]
Abstract
Na+ -K+ -Cl- cotransporter 2 (NKCC2; SLC12A1) is an integral membrane protein that comes as three splice variants and mediates the cotranslocation of Na+ , K+ , and Cl- ions through the apical membrane of the thick ascending loop of Henle (TALH). In doing so, and through the involvement of other ion transport systems, it allows this nephron segment to reclaim a large fraction of the ultrafiltered Na+ , Cl- , Ca2+ , Mg2+ , and HCO3- loads. The functional relevance of NKCC2 in human is illustrated by the many abnormalities that result from the inactivation of this transport system through the use of loop diuretics or in the setting of inherited disorders. The following presentation aims at discussing the physiological roles and molecular characteristics of Na+ -K+ -Cl- cotransport in the TALH and those of the individual NKCC2 splice variants more specifically. Many of the historical and recent data that have emerged from the experiments conducted will be outlined and their larger meaning will also be placed into perspective with the aid of various hypotheses.
Collapse
Affiliation(s)
- Andree‐Anne Marcoux
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| | - Laurence E. Tremblay
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| | - Samira Slimani
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| | - Marie‐Jeanne Fiola
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| | - Fabrice Mac‐Way
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| | - Alexandre P. Garneau
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
- Cardiometabolic Axis, School of Kinesiology and Physical Activity SciencesUniversity of MontréalMontréalQuebecCanada
| | - Paul Isenring
- Department of Medicine, Nephrology Research GroupLaval UniversityQuebec CityQuébecCanada
| |
Collapse
|
9
|
Garneau AP, Slimani S, Fiola MJ, Tremblay LE, Isenring P. Multiple Facets and Roles of Na+-K+-Cl−Cotransport: Mechanisms and Therapeutic Implications. Physiology (Bethesda) 2020; 35:415-429. [DOI: 10.1152/physiol.00012.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The Na+-K+-Cl−cotransporters play key physiological and pathophysiological roles by regulating the membrane potential of many cell types and the movement of fluid across a variety of epithelial or endothelial structures. As such, they should soon become invaluable targets for the treatment of various disorders including pain, epilepsy, brain edema, and hypertension. This review highlights the nature of these roles, the mechanisms at play, and the unresolved issues in the field.
Collapse
Affiliation(s)
- A. P. Garneau
- Department of Medicine, Nephrology Research Group, Laval University, Québec, Canada; and
- Cardiometabolic Axis, School of Kinesiology and Physical Activity Sciences, University of Montréal, Montréal, Canada
| | - S. Slimani
- Department of Medicine, Nephrology Research Group, Laval University, Québec, Canada; and
| | - M. J. Fiola
- Department of Medicine, Nephrology Research Group, Laval University, Québec, Canada; and
| | - L. E. Tremblay
- Department of Medicine, Nephrology Research Group, Laval University, Québec, Canada; and
| | - P. Isenring
- Department of Medicine, Nephrology Research Group, Laval University, Québec, Canada; and
| |
Collapse
|
10
|
Limbutara K, Chou CL, Knepper MA. Quantitative Proteomics of All 14 Renal Tubule Segments in Rat. J Am Soc Nephrol 2020; 31:1255-1266. [PMID: 32358040 DOI: 10.1681/asn.2020010071] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Previous research has used RNA sequencing in microdissected kidney tubules or single cells isolated from the kidney to profile gene expression in each type of kidney tubule epithelial cell. However, because proteins, not mRNA molecules, mediate most cellular functions, it is desirable to know the identity and amounts of each protein species to understand function. Recent improvements in the sensitivity of mass spectrometers offered us the ability to quantify the proteins expressed in each of 14 different renal tubule segments from rat. METHODS We manually dissected kidney tubules from rat kidneys and subjected samples to protein mass spectrometry. We used the "proteomic ruler" technique to estimate the number of molecules of each protein per cell. RESULTS Over the 44 samples analyzed, the average number of quantified proteins per segment was 4234, accounting for at least 99% of protein molecules in each cell. We have made the data publicly available online at the Kidney Tubule Expression Atlas website (https://esbl.nhlbi.nih.gov/KTEA/). Protein abundance along the renal tubule for many commonly studied water and solute transport proteins and metabolic enzymes matched expectations from prior localization studies, demonstrating the overall reliability of the data. The site features a "correlated protein" function, which we used to identify cell type-specific transcription factors expressed along the renal tubule. CONCLUSIONS We identified and quantified proteins expressed in each of the 14 segments of rat kidney tubules and used the proteomic data that we obtained to create an online information resource, the Kidney Tubule Expression Atlas. This resource will allow users throughout the world to browse segment-specific protein expression data and download them for their own research.
Collapse
Affiliation(s)
- Kavee Limbutara
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
11
|
Natochin YV, Golosova DV. Vasopressin receptor subtypes and renal sodium transport. VITAMINS AND HORMONES 2019; 113:239-258. [PMID: 32138950 DOI: 10.1016/bs.vh.2019.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In mammals, three subtypes of V-receptors have been identified in the kidney. The effects of vasopressin, a hormone synthesized in the hypothalamus, are triggered by three distinct receptor isoforms: V2, V1a, and V1b. Stimulation of V2-receptors regulates urine osmotic concentration by increasing sodium reabsorption in the thick ascending limb of the loop of Henle and enhancing osmotic permeability of the epithelium cells in the collecting duct. Stimulation of V1a-receptors inhibits renal sodium reabsorption and induces natriuresis, comparable to the effect of the diuretic furosemide, in the thick ascending limb of the loop of Henle. Stimulation of V1b-receptors induces potassium secretion in the final parts of the distal segments and initial parts of the collecting ducts. In this review, we discuss the role of vasopressin and its interaction with V-receptor subtypes in natriuresis and for stabilizing the physicochemical parameters of the internal environment and water-salt homeostasis in humans. A better understanding of these systems and their regulation is necessary to facilitate identification of additional system components and mechanisms, clarify their contribution during various normal and pathological functional states, and suggest novel strategies for the development of therapeutic interventions.
Collapse
Affiliation(s)
- Yu V Natochin
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia.
| | - D V Golosova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
12
|
Canonica J, Frateschi S, Boscardin E, Ebering A, Sergi C, Jäger Y, Peyrollaz T, Mérillat AM, Maillard M, Klusonova P, Odermatt A, Koesters R, Debonneville A, Staub O, Verouti SN, Hummler E. Lack of Renal Tubular Glucocorticoid Receptor Decreases the Thiazide-Sensitive Na +/Cl - Cotransporter NCC and Transiently Affects Sodium Handling. Front Physiol 2019; 10:989. [PMID: 31474871 PMCID: PMC6702950 DOI: 10.3389/fphys.2019.00989] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic glucocorticoid infusion impairs NCC activity and induces a non-dipping profile in mice, suggesting that glucocorticoids are essential for daily blood pressure variations. In this paper, we studied mice lacking the renal tubular glucocorticoid receptor (GR) in adulthood (GR knockouts, Nr3c1Pax8/LC1). Upon standard salt diet, Nr3c1Pax8/LC1 mice grow normally, but show reduced NCC activity despite normal plasma aldosterone levels. Following diet switch to low sodium, Nr3c1Pax8/LC1 mice exhibit a transient but significant reduction in the activity of NCC and expression of NHE3 and NKCC2 accompanied by significant increased Spak activity. This is followed by transiently increased urinary sodium excretion and higher plasma aldosterone concentrations. Plasma corticosterone levels and 11βHSD2 mRNA expression and activity in the whole kidney remain unchanged. High salt diet does not affect whole body Na+ and/or K+ balance and NCC activity is not reduced, but leads to a significant increase in diastolic blood pressure dipping in Nr3c1Pax8/LC1 mice. When high sodium treatment is followed by 48 h of darkness, NCC abundance is reduced in knockout mice although activity is not different. Our data show that upon Na+ restriction renal tubular GR-deficiency transiently affects Na+ handling and transport pathways. Overall, upon standard, low Na+ and high Na+ diet exposure Na+ and K+ balance is maintained as evidenced by normal plasma and urinary Na+ and K+ and aldosterone concentrations.
Collapse
Affiliation(s)
- Jérémie Canonica
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland.,National Centre of Competence in Research "Kidney.CH", Lausanne, Switzerland
| | - Simona Frateschi
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland.,National Centre of Competence in Research "Kidney.CH", Lausanne, Switzerland
| | - Emilie Boscardin
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland.,National Centre of Competence in Research "Kidney.CH", Lausanne, Switzerland
| | - Anna Ebering
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Chloé Sergi
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Yannick Jäger
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Thibaud Peyrollaz
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Anne-Marie Mérillat
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Marc Maillard
- Department of Nephrology Service, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Petra Klusonova
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Alex Odermatt
- National Centre of Competence in Research "Kidney.CH", Lausanne, Switzerland.,Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Robert Koesters
- Hôpital Tenon, Université Pierre et Marie Curie, Paris, France
| | - Anne Debonneville
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Olivier Staub
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland.,National Centre of Competence in Research "Kidney.CH", Lausanne, Switzerland
| | - Sophia N Verouti
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Edith Hummler
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland.,National Centre of Competence in Research "Kidney.CH", Lausanne, Switzerland
| |
Collapse
|
13
|
Marcoux AA, Slimani S, Tremblay LE, Frenette-Cotton R, Garneau AP, Isenring P. Endocytic recycling of Na + -K + -Cl - cotransporter type 2: importance of exon 4. J Physiol 2019; 597:4263-4276. [PMID: 31216057 DOI: 10.1113/jp278024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/05/2019] [Indexed: 01/29/2023] Open
Abstract
KEY POINTS Na+ -K+ -Cl- cotransporter type 2 (NKCC2) is a 27-exon membrane protein that is expressed in the thick ascending limb (TAL) of Henle where it is involved in reabsorption of the ultrafiltered NaCl load. It comes as three splice variants that are identical to each other except for the residue composition of exon 4 and that differ in their transport characteristics, functional roles and distributions along the TAL. In this report, it is shown that the variants also differ in their trafficking properties and that two residues in exon 4 play a key role in this regard. One of these residues was also shown to sustain carrier internalization. Through these results, a novel function for the alternatively spliced exon of NKCC2 has been identified and a domain that is involved in carrier trafficking has been uncovered for the first time in a cation-Cl- cotransporter family member. ABSTRACT Na+ -K+ -Cl- cotransporter type 2 (NKCC2) is a 12-transmembrane (TM) domain cell surface glycoprotein that is expressed in the thick ascending limb (TAL) of Henle and stimulated during cell shrinkage. It comes as three splice variants (A, B and F) that are identical to each other except for TM2 and the following connecting segment (CS2). Yet, these variants do not share the same localization, transport characteristics and physiological roles along the TAL. We have recently found that while cell shrinkage could exert its activating effect by increasing NKCC2 expression at the cell surface, the variants also responded differentially to this stimulus. In the current work, a mutagenic approach was exploited to determine whether CS2 could play a role in carrier trafficking and identify the residues potentially involved. We found that when the residue of position 238 in NKCC2A (F) and NKCC2B (Y) was replaced by the corresponding residue in NKCC2F (V), carrier activity increased by over 3-fold and endocytosis decreased concomitantly. We also found that when the residue of position 230 in NKCC2F (M) was replaced by the one in NKCC2B (T), carrier activity and affinity for ions both increased substantially whereas expression at the membrane decreased. Taken together, these results suggest that CS2 is involved in carrier trafficking and that two of its residues, those of positions 238 and 230, are part of an internalization motif. They also indicate that the divergent residue of position 230 plays the dual role of specifying ion affinity and sustaining carrier internalization.
Collapse
Affiliation(s)
- Andrée-Anne Marcoux
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6
| | - Samira Slimani
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6
| | - Laurence E Tremblay
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6
| | - Rachelle Frenette-Cotton
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6
| | - Alexandre P Garneau
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6.,Cardiometabolic Research Group, Department of Kinesiology, Faculty of Medicine, University of Montréal, Montréal, QC, Canada, H3T 1J4
| | - Paul Isenring
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Québec, QC, Canada, G1R 2J6
| |
Collapse
|
14
|
Marcoux AA, Slimani S, Tremblay LE, Frenette-Cotton R, Garneau AP, Isenring P. Regulation of Na +-K +-Cl - cotransporter type 2 by the with no lysine kinase-dependent signaling pathway. Am J Physiol Cell Physiol 2019; 317:C20-C30. [PMID: 30917032 DOI: 10.1152/ajpcell.00041.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Na+-K+-Cl- cotransporter type 2 (NKCC2) is confined to the apical membrane of the thick ascending limb of Henle, where it reabsorbs a substantial fraction of the ultrafiltered NaCl load. It is expressed along this nephron segment as three main splice variants (called NKCC2A, NKCC2B, and NKCC2F) that differ in residue composition along their second transmembrane domain and first intracellular cytosolic connecting segment (CS2). NKCC2 is known to be activated by cell shrinkage and intracellular [Cl-] reduction. Although the with no lysine (WNK) kinases could play a role in this response, the mechanisms involved are ill defined, and the possibility of variant-specific responses has not been tested thus far. In this study, we have used the Xenopus laevis oocyte expression system to gain further insight in these regards. We have found for the first time that cell shrinkage could stimulate NKCC2A- and NKCC2B-mediated ion transport by increasing carrier abundance at the cell surface and that this response was achieved (at least in part) by the enzymatic function of a WNK kinase. Interestingly, we have also found that the activity and cell surface abundance of NKCC2F were less affected by cell shrinkage compared with the other variants and that ion transport by certain variants could be stimulated through WNK kinase expression in the absence of carrier redistribution. Taken together, these results suggest that the WNK kinase-dependent pathway can affect both the trafficking as well as intrinsic activity of NKCC2 and that CS2 plays an important role in carrier regulation.
Collapse
Affiliation(s)
- Andrée-Anne Marcoux
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada
| | - Samira Slimani
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada
| | - Laurence E Tremblay
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada
| | - Rachelle Frenette-Cotton
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada
| | - Alexandre P Garneau
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada.,Cardiometabolic Research Group, Department of Kinesiology, Faculty of Medicine, University of Montreal , Montreal, Quebec , Canada
| | - Paul Isenring
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University , Quebec City, Quebec , Canada
| |
Collapse
|
15
|
Koumangoye R, Omer S, Delpire E. Mistargeting of a truncated Na-K-2Cl cotransporter in epithelial cells. Am J Physiol Cell Physiol 2018; 315:C258-C276. [PMID: 29719172 DOI: 10.1152/ajpcell.00130.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We recently reported the case of a young patient with multisystem failure carrying a de novo mutation in SLC12A2, the gene encoding the Na-K-2Cl cotransporter-1 (NKCC1). Heterologous expression studies in nonepithelial cells failed to demonstrate dominant-negative effects. In this study, we examined expression of the mutant cotransporter in epithelial cells. Using Madin-Darby canine kidney (MDCK) cells grown on glass coverslips, permeabilized support, and Matrigel, we show that the fluorescently tagged mutant cotransporter is expressed in cytoplasm and at the apical membrane and affects epithelium integrity. Expression of the mutant transporter at the apical membrane also results in the mislocalization of some of the wild-type transporter to the apical membrane. This mistargeting is specific to NKCC1 as the Na+-K+-ATPase remains localized on the basolateral membrane. To assess transporter localization in vivo, we created a mouse model using CRISPR/cas9 that reproduces the 11 bp deletion in exon 22 of Slc12a2. Although the mice do not display an overt phenotype, we show that the colon and salivary gland expresses wild-type NKCC1 abundantly at the apical pole, confirming the data obtained in cultured epithelial cells. Enough cotransporter must remain, however, on the basolateral membrane to participate in saliva secretion, as no significant decrease in saliva production was observed in the mutant mice.
Collapse
Affiliation(s)
- Rainelli Koumangoye
- Department of Anesthesiology, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Salma Omer
- Department of Anesthesiology, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine , Nashville, Tennessee
| |
Collapse
|
16
|
Dimke H, Schnermann J. Axial and cellular heterogeneity in electrolyte transport pathways along the thick ascending limb. Acta Physiol (Oxf) 2018; 223:e13057. [PMID: 29476644 DOI: 10.1111/apha.13057] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/27/2018] [Accepted: 02/17/2018] [Indexed: 12/21/2022]
Abstract
The thick ascending limb (TAL) extends from the border of the inner medulla to the renal cortex, thus ascending through regions with wide differences in tissue solute and electrolyte concentrations. Structural and functional differences between TAL cells in the medulla (mTAL) and the cortex (cTAL) would therefore be useful to adapt TAL transport function to a changing external fluid composition. While mechanisms common to all TAL cells play a central role in the reclamation of about 25% of the NaCl filtered by the kidney, morphological features, Na+ / K+ -ATPase activity, NKCC2 splicing and phosphorylation do vary between segments and cells. The TAL contributes to K+ homeostasis and TAL cells with high or low basolateral K+ conductances have been identified which may be involved in K+ reabsorption and secretion respectively. Although transport rates for HCO3- do not differ between mTAL and cTAL, divergent axial and cellular expression of H+ transport proteins in TAL have been documented. The reabsorption of the divalent cations Ca2+ and Mg2+ is highest in cTAL and paralleled by differences in divalent cation permeability and the expression of select claudins. Morphologically, two cell types with different cell surface phenotypes have been described that still need to be linked to specific functional characteristics. The unique external environment and its change along the longitudinal axis require an axial functional heterogeneity for the TAL to optimally participate in conserving electrolyte homeostasis. Despite substantial progress in understanding TAL function, there are still considerable knowledge gaps that are just beginning to become bridged.
Collapse
Affiliation(s)
- H. Dimke
- Department of Cardiovascular and Renal Research; Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
| | - J. Schnermann
- National Institute of Diabetes and Digestive and Kidney Diseases; Bethesda MD USA
| |
Collapse
|
17
|
Delpire E, Gagnon KB. Na + -K + -2Cl - Cotransporter (NKCC) Physiological Function in Nonpolarized Cells and Transporting Epithelia. Compr Physiol 2018; 8:871-901. [PMID: 29687903 DOI: 10.1002/cphy.c170018] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Two genes encode the Na+ -K+ -2Cl- cotransporters, NKCC1 and NKCC2, that mediate the tightly coupled movement of 1Na+ , 1K+ , and 2Cl- across the plasma membrane of cells. Na+ -K+ -2Cl- cotransport is driven by the chemical gradient of the three ionic species across the membrane, two of them maintained by the action of the Na+ /K+ pump. In many cells, NKCC1 accumulates Cl- above its electrochemical potential equilibrium, thereby facilitating Cl- channel-mediated membrane depolarization. In smooth muscle cells, this depolarization facilitates the opening of voltage-sensitive Ca2+ channels, leading to Ca2+ influx, and cell contraction. In immature neurons, the depolarization due to a GABA-mediated Cl- conductance produces an excitatory rather than inhibitory response. In many cell types that have lost water, NKCC is activated to help the cells recover their volume. This is specially the case if the cells have also lost Cl- . In combination with the Na+ /K+ pump, the NKCC's move ions across various specialized epithelia. NKCC1 is involved in Cl- -driven fluid secretion in many exocrine glands, such as sweat, lacrimal, salivary, stomach, pancreas, and intestine. NKCC1 is also involved in K+ -driven fluid secretion in inner ear, and possibly in Na+ -driven fluid secretion in choroid plexus. In the thick ascending limb of Henle, NKCC2 activity in combination with the Na+ /K+ pump participates in reabsorbing 30% of the glomerular-filtered Na+ . Overall, many critical physiological functions are maintained by the activity of the two Na+ -K+ -2Cl- cotransporters. In this overview article, we focus on the functional roles of the cotransporters in nonpolarized cells and in epithelia. © 2018 American Physiological Society. Compr Physiol 8:871-901, 2018.
Collapse
Affiliation(s)
- Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, Tennessee, USA
| | - Kenneth B Gagnon
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Keystone, USA
| |
Collapse
|
18
|
Asico LD, Cuevas S, Ma X, Jose PA, Armando I, Konkalmatt PR. Nephron segment-specific gene expression using AAV vectors. Biochem Biophys Res Commun 2018; 497:19-24. [PMID: 29407172 PMCID: PMC5893140 DOI: 10.1016/j.bbrc.2018.01.169] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 01/27/2018] [Indexed: 11/02/2022]
Abstract
AAV9 vector provides efficient gene transfer in all segments of the renal nephron, with minimum expression in non-renal cells, when administered retrogradely via the ureter. It is important to restrict the transgene expression to the desired cell type within the kidney, so that the physiological endpoints represent the function of the transgene expressed in that specific cell type within kidney. We hypothesized that segment-specific gene expression within the kidney can be accomplished using the highly efficient AAV9 vectors carrying the promoters of genes that are expressed exclusively in the desired segment of the nephron in combination with administration by retrograde infusion into the kidney via the ureter. We constructed AAV vectors carrying eGFP under the control of: kidney-specific cadherin (KSPC) gene promoter for expression in the entire nephron; Na+/glucose co-transporter (SGLT2) gene promoter for expression in the S1 and S2 segments of the proximal tubule; sodium, potassium, 2 chloride co-transporter (NKCC2) gene promoter for expression in the thick ascending limb of Henle's loop (TALH); E-cadherin (ECAD) gene promoter for expression in the collecting duct (CD); and cytomegalovirus (CMV) early promoter that provides expression in most of the mammalian cells, as control. We tested the specificity of the promoter constructs in vitro for cell type-specific expression in mouse kidney cells in primary culture, followed by retrograde infusion of the AAV vectors via the ureter in the mouse. Our data show that AAV9 vector, in combination with the segment-specific promoters administered by retrograde infusion via the ureter, provides renal nephron segment-specific gene expression.
Collapse
Affiliation(s)
- Laureano D Asico
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - Santiago Cuevas
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - Xiaobo Ma
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - Pedro A Jose
- Department of Medicine, The George Washington University, Washington, DC, USA; Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
| | - Ines Armando
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - Prasad R Konkalmatt
- Department of Medicine, The George Washington University, Washington, DC, USA.
| |
Collapse
|
19
|
Johnson BG, Dang LT, Marsh G, Roach AM, Levine ZG, Monti A, Reyon D, Feigenbaum L, Duffield JS. Uromodulin p.Cys147Trp mutation drives kidney disease by activating ER stress and apoptosis. J Clin Invest 2017; 127:3954-3969. [PMID: 28990932 DOI: 10.1172/jci93817] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 08/24/2017] [Indexed: 12/13/2022] Open
Abstract
Uromodulin-associated kidney disease (UAKD) is caused by mutations in the uromodulin (UMOD) gene that result in a misfolded form of UMOD protein, which is normally secreted by nephrons. In UAKD patients, mutant UMOD is poorly secreted and accumulates in the ER of distal kidney epithelium, but its role in disease progression is largely unknown. Here, we modeled UMOD accumulation in mice by expressing the murine equivalent of the human UMOD p.Cys148Trp point mutation (UmodC147W/+ mice). Like affected humans, these UmodC147W/+ mice developed spontaneous and progressive kidney disease with organ failure over 24 weeks. Analysis of diseased kidneys and purified UMOD-producing cells revealed early activation of the PKR-like ER kinase/activating transcription factor 4 (PERK/ATF4) ER stress pathway, innate immune mediators, and increased apoptotic signaling, including caspase-3 activation. Unexpectedly, we also detected autophagy deficiency. Human cells expressing UMOD p.Cys147Trp recapitulated the findings in UmodC147W/+ mice, and autophagy activation with mTOR inhibitors stimulated the intracellular removal of aggregated mutant UMOD. Human cells producing mutant UMOD were susceptible to TNF-α- and TRAIL-mediated apoptosis due to increased expression of the ER stress mediator tribbles-3. Blocking TNF-α in vivo with the soluble recombinant fusion protein TNFR:Fc slowed disease progression in UmodC147W/+ mice by reducing active caspase-3, thereby preventing tubule cell death and loss of epithelial function. These findings reveal a targetable mechanism for disease processes involved in UAKD.
Collapse
Affiliation(s)
- Bryce G Johnson
- Research and Development, Biogen, Cambridge, Massachusetts, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Lan T Dang
- Research and Development, Biogen, Cambridge, Massachusetts, USA
| | - Graham Marsh
- Research and Development, Biogen, Cambridge, Massachusetts, USA
| | - Allie M Roach
- Research and Development, Biogen, Cambridge, Massachusetts, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Anthony Monti
- Research and Development, Biogen, Cambridge, Massachusetts, USA
| | - Deepak Reyon
- Research and Development, Biogen, Cambridge, Massachusetts, USA
| | | | - Jeremy S Duffield
- Research and Development, Biogen, Cambridge, Massachusetts, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Yu G, Cheng M, Wang W, Zhao R, Liu Z. Involvement of WNK1-mediated potassium channels in the sexual dimorphism of blood pressure. Biochem Biophys Res Commun 2017; 485:255-260. [PMID: 28237360 DOI: 10.1016/j.bbrc.2017.02.098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 02/18/2017] [Indexed: 11/25/2022]
Abstract
Potassium homeostasis plays an essential role in the control of blood pressure. It is unknown, however, whether potassium balance is involved in the gender-associated blood pressure differences. We therefore investigated the possible mechanism of sexual dimorphism in blood pressure regulation by measuring the blood pressure, plasma potassium, renal actions of potassium channels and upstream regulator in male and female mice. Here we found that female mice exhibited lower blood pressure and higher plasma K+ level as compared to male littermates. Western blot analyses of mouse kidney extract revealed a significant decrease in renal outer medullary potassium (ROMK) channel expression, while large-conductance Ca2+-activated K+ (BK) channel and Na-K-2Cl cotransporter (NKCC2) as well as the upstream regulator with-no-lysine kinase 1 (WNK1) enhanced in female mice under normal condition. Surprisingly, both dietary K+ loading and K+ depletion eliminated the differences in plasma K+ and blood pressure between females and males, and the differences of renal K+ channels and WNK1 also attenuated in both groups of mice. These findings indicated the existence of a close correlation between K+ homeostasis and sex-associated blood pressure. Moreover, the differential regulation of ROMK, BK-α and NKCC2 between female and male mice, at least, were partly mediated via WNK1 pathway, which may contribute to the sexual dimorphism of plasma K+ and blood pressure control.
Collapse
Affiliation(s)
- Guofeng Yu
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325003, China
| | - Mengting Cheng
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325003, China
| | - Wei Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325003, China
| | - Rong Zhao
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325003, China
| | - Zhen Liu
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325003, China.
| |
Collapse
|
21
|
Ure ME, Heydari E, Pan W, Ramesh A, Rehman S, Morgan C, Pinsk M, Erickson R, Herrmann JM, Dimke H, Cordat E, Lemaire M, Walter M, Alexander RT. A variant in a cis-regulatory element enhances claudin-14 expression and is associated with pediatric-onset hypercalciuria and kidney stones. Hum Mutat 2017; 38:649-657. [PMID: 28229505 DOI: 10.1002/humu.23202] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 01/26/2017] [Accepted: 02/18/2017] [Indexed: 12/19/2022]
Abstract
The greatest risk factor for kidney stones is hypercalciuria, the etiology of which is largely unknown. A recent genome-wide association study (GWAS) linked hypercalciuria and kidney stones to a claudin-14 (CLDN14) risk haplotype. However, the underlying molecular mechanism was not delineated. Recently, renal CLDN14 expression was found to increase in response to increased plasma calcium, thereby inducing calciuria. We hypothesized therefore that some children with hypercalciuria and kidney stones harbor a CLDN14 variant that inappropriately increases gene expression. To test this hypothesis, we sequenced the CLDN14 risk haplotype in a cohort of children with idiopathic hypercalciuria and kidney stones. An intronic SNP was more frequent in affected children. Dual luciferase and cell-based assays demonstrated increased reporter or CLDN14 expression when this polymorphism was introduced. In silico studies predicted the SNP introduced a novel insulinoma-associated 1 (INSM1) transcription factor binding site. Consistent with this, repeating the dual luciferase assay in the presence of INSM1 further increased reporter expression. Our data suggest that children with the INSM1 binding site within the CLDN14 risk haplotype have a higher likelihood of hypercalciuria and kidney stones. Enhanced CLDN14 expression may play a role in the pathophysiology of their hypercalciuria.
Collapse
Affiliation(s)
- Megan E Ure
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Emma Heydari
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Wanling Pan
- Department of Physiology, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Ajay Ramesh
- Depratment of Cell Biology, University of Kaiserslautern, Kaiserslautern, 67663, Germany
| | - Sabah Rehman
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Catherine Morgan
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Maury Pinsk
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Robin Erickson
- Department of Pediatrics, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 0W8, Canada
| | - Johannes M Herrmann
- Depratment of Cell Biology, University of Kaiserslautern, Kaiserslautern, 67663, Germany
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, 5000, Denmark
| | - Emmanuelle Cordat
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Mathieu Lemaire
- The Hospital for Sick Children, Toronto, Ontario, M5G 1×8, Canada
| | - Michael Walter
- Department of Medical Genetics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - R Todd Alexander
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada.,Department of Physiology, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| |
Collapse
|
22
|
Delpire E, Wolfe L, Flores B, Koumangoye R, Schornak CC, Omer S, Pusey B, Lau C, Markello T, Adams DR. A patient with multisystem dysfunction carries a truncation mutation in human SLC12A2, the gene encoding the Na-K-2Cl cotransporter, NKCC1. Cold Spring Harb Mol Case Stud 2016; 2:a001289. [PMID: 27900370 PMCID: PMC5111002 DOI: 10.1101/mcs.a001289] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/10/2016] [Indexed: 12/30/2022] Open
Abstract
This study describes a 13-yr-old girl with orthostatic intolerance, respiratory weakness, multiple endocrine abnormalities, pancreatic insufficiency, and multiorgan failure involving the gut and bladder. Exome sequencing revealed a de novo, loss-of-function allele in SLC12A2, the gene encoding the Na-K-2Cl cotransporter-1. The 11-bp deletion in exon 22 results in frameshift (p.Val1026Phefs*2) and truncation of the carboxy-terminal tail of the cotransporter. Preliminary studies in heterologous expression systems demonstrate that the mutation leads to a nonfunctional transporter, which is expressed and trafficked to the plasma membrane alongside wild-type NKCC1. The truncated protein, visible at higher molecular sizes, indicates either enhanced dimerization or misfolded aggregate. No significant dominant-negative effect was observed. K+ transport experiments performed in fibroblasts from the patient showed reduced total and NKCC1-mediated K+ influx. The absence of a bumetanide effect on K+ influx in patient fibroblasts only under hypertonic conditions suggests a deficit in NKCC1 regulation. We propose that disruption in NKCC1 function might affect sensory afferents and/or smooth muscle cells, as their functions depend on NKCC1 creating a Cl- gradient across the plasma membrane. This Cl- gradient allows the γ-aminobutyric acid (GABA) receptor or other Cl- channels to depolarize the membrane affecting processes such as neurotransmission or cell contraction. Under this hypothesis, disrupted sensory and smooth muscle function in a diverse set of tissues could explain the patient's phenotype.
Collapse
Affiliation(s)
- Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Lynne Wolfe
- Undiagnosed Diseases Program, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Bianca Flores
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Rainelli Koumangoye
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Cara C Schornak
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Salma Omer
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Barbara Pusey
- Undiagnosed Diseases Program, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Christopher Lau
- Undiagnosed Diseases Program, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Thomas Markello
- Undiagnosed Diseases Program, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - David R Adams
- Undiagnosed Diseases Program, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
23
|
Bazúa-Valenti S, Castañeda-Bueno M, Gamba G. Physiological role of SLC12 family members in the kidney. Am J Physiol Renal Physiol 2016; 311:F131-44. [DOI: 10.1152/ajprenal.00071.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/12/2016] [Indexed: 12/30/2022] Open
Abstract
The solute carrier family 12, as numbered according to Human Genome Organisation (HUGO) nomenclature, encodes the electroneutral cation-coupled chloride cotransporters that are expressed in many cells and tissues; they play key roles in important physiological events, such as cell volume regulation, modulation of the intracellular chloride concentration, and transepithelial ion transport. Most of these family members are expressed in specific regions of the nephron. The Na-K-2Cl cotransporter NKCC2, which is located in the thick ascending limb, and the Na-Cl cotransporter, which is located in the distal convoluted tubule, play important roles in salt reabsorption and serve as the receptors for loop and thiazide diuretics, respectively (Thiazide diuretics are among the most commonly prescribed drugs in the world.). The activity of these transporters correlates with blood pressure levels; thus, their regulation has been a subject of intense research for more than a decade. The K-Cl cotransporters KCC1, KCC3, and KCC4 are expressed in several nephron segments, and their role in renal physiology is less understood but nevertheless important. Evidence suggests that they are involved in modulating proximal tubule glucose reabsorption, thick ascending limb salt reabsorption and collecting duct proton secretion. In this work, we present an overview of the physiological roles of these transporters in the kidney, with particular emphasis on the knowledge gained in the past few years.
Collapse
Affiliation(s)
- Silvana Bazúa-Valenti
- Molecular Physiology Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| | - María Castañeda-Bueno
- Molecular Physiology Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| | - Gerardo Gamba
- Molecular Physiology Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| |
Collapse
|
24
|
Jaykumar AB, Caceres PS, Sablaban I, Tannous BA, Ortiz PA. Real-time monitoring of NKCC2 endocytosis by total internal reflection fluorescence (TIRF) microscopy. Am J Physiol Renal Physiol 2015; 310:F183-91. [PMID: 26538436 DOI: 10.1152/ajprenal.00104.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 10/30/2015] [Indexed: 11/22/2022] Open
Abstract
The apical Na-K-2Cl cotransporter (NKCC2) mediates NaCl reabsorption by the thick ascending limb (TAL). The amount of NKCC2 at the apical membrane of TAL cells is determined by exocytic delivery, recycling, and endocytosis. Surface biotinylation allows measurement of NKCC2 endocytosis, but it has low time resolution and does not allow imaging of the dynamic process of endocytosis. We hypothesized that total internal reflection fluorescence (TIRF) microscopy imaging of labeled NKCC2 would allow monitoring of NKCC2 endocytosis in polarized Madin-Darby canine kidney (MDCK) and TAL cells. Thus we generated a NKCC2 construct containing a biotin acceptor domain (BAD) sequence between the transmembrane domains 5 and 6. Once expressed in polarized MDCK or TAL cells, surface NKCC2 was specifically biotinylated by exogenous biotin ligase (BirA). We also demonstrate that expression of a secretory form of BirA in TAL cells induces metabolic biotinylation of NKCC2. Labeling biotinylated surface NKCC2 with fluorescent streptavidin showed that most apical NKCC2 was located within small discrete domains or clusters referred to as "puncta" on the TIRF field. NKCC2 puncta were observed to disappear from the TIRF field, indicating an endocytic event which led to a decrease in the number of surface puncta at a rate of 1.18 ± 0.16%/min in MDCK cells, and a rate 1.09 ± 0.08%/min in TAL cells (n = 5). Treating cells with a cholesterol-chelating agent (methyl-β-cyclodextrin) completely blocked NKCC2 endocytosis. We conclude that TIRF microscopy of labeled NKCC2 allows the dynamic imaging of individual endocytic events at the apical membrane of TAL cells.
Collapse
Affiliation(s)
- Ankita Bachhawat Jaykumar
- Hypertension and Vascular Research, Henry Ford Hospital, Detroit, Michigan; Department of Physiology, Wayne State University, Detroit, Michigan; and
| | - Paulo S Caceres
- Hypertension and Vascular Research, Henry Ford Hospital, Detroit, Michigan; Department of Physiology, Wayne State University, Detroit, Michigan; and
| | - Ibrahim Sablaban
- Hypertension and Vascular Research, Henry Ford Hospital, Detroit, Michigan
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Laboratory, Neuroscience Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pablo A Ortiz
- Hypertension and Vascular Research, Henry Ford Hospital, Detroit, Michigan; Department of Physiology, Wayne State University, Detroit, Michigan; and
| |
Collapse
|
25
|
Kortenoeven MLA, Pedersen NB, Rosenbaek LL, Fenton RA. Vasopressin regulation of sodium transport in the distal nephron and collecting duct. Am J Physiol Renal Physiol 2015; 309:F280-99. [DOI: 10.1152/ajprenal.00093.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/27/2015] [Indexed: 12/22/2022] Open
Abstract
Arginine vasopressin (AVP) is released from the posterior pituitary gland during states of hyperosmolality or hypovolemia. AVP is a peptide hormone, with antidiuretic and antinatriuretic properties. It allows the kidneys to increase body water retention predominantly by increasing the cell surface expression of aquaporin water channels in the collecting duct alongside increasing the osmotic driving forces for water reabsorption. The antinatriuretic effects of AVP are mediated by the regulation of sodium transport throughout the distal nephron, from the thick ascending limb through to the collecting duct, which in turn partially facilitates osmotic movement of water. In this review, we will discuss the regulatory role of AVP in sodium transport and summarize the effects of AVP on various molecular targets, including the sodium-potassium-chloride cotransporter NKCC2, the thiazide-sensitive sodium-chloride cotransporter NCC, and the epithelial sodium channel ENaC.
Collapse
Affiliation(s)
- M. L. A. Kortenoeven
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus, Denmark
| | - N. B. Pedersen
- Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; and
| | - L. L. Rosenbaek
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - R. A. Fenton
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus, Denmark
| |
Collapse
|
26
|
Na+-H+ exchanger-1 (NHE1) regulation in kidney proximal tubule. Cell Mol Life Sci 2015; 72:2061-74. [PMID: 25680790 DOI: 10.1007/s00018-015-1848-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 01/28/2015] [Accepted: 01/29/2015] [Indexed: 01/17/2023]
Abstract
The ubiquitously expressed plasma membrane Na(+)-H(+) exchanger NHE1 is a 12 transmembrane-spanning protein that directs important cell functions such as homeostatic intracellular volume and pH control. The 315 amino acid cytosolic tail of NHE1 binds plasma membrane phospholipids and multiple proteins that regulate additional, ion-translocation independent functions. This review focuses on NHE1 structure/function relationships, as well as the role of NHE1 in kidney proximal tubule functions, including pH regulation, vectorial Na(+) transport, cell volume control and cell survival. The implications of these functions are particularly critical in the setting of progressive, albuminuric kidney diseases, where the accumulation of reabsorbed fatty acids leads to disruption of NHE1-membrane phospholipid interactions and tubular atrophy, which is a poor prognostic factor for progression to end stage renal disease. This review amplifies the vital role of the proximal tubule NHE1 Na(+)-H(+) exchanger as a kidney cell survival factor.
Collapse
|
27
|
Guetg A, Mariotta L, Bock L, Herzog B, Fingerhut R, Camargo SMR, Verrey F. Essential amino acid transporter Lat4 (Slc43a2) is required for mouse development. J Physiol 2015; 593:1273-89. [PMID: 25480797 DOI: 10.1113/jphysiol.2014.283960] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/25/2014] [Indexed: 12/23/2022] Open
Abstract
Amino acid (AA) uniporter Lat4 (Slc43a2) mediates facilitated diffusion of branched-chain AAs, methionine and phenylalanine, although its physiological role and subcellular localization are not known. We report that Slc43a2 knockout mice were born at expected Mendelian frequency but displayed an ∼10% intrauterine growth retardation and low amniotic fluid AAs, suggesting defective transplacental transport. Postnatal growth was strongly reduced, with premature death occurring within 9 days such that further investigations were made within 3 days of birth. Lat4 immunofluorescence showed a strong basolateral signal in the small intestine, kidney proximal tubule and thick ascending limb epithelial cells of wild-type but not Slc43a2 null littermates and no signal in liver and skeletal muscle. Experiments using Xenopus laevis oocytes demonstrated that Lat4 functioned as a symmetrical low affinity uniporter with a K₀.₅ of ∼5 mm for both in- and efflux. Plasma AA concentration was decreased in Slc43a2 null pups, in particular that of non-essential AAs alanine, serine, histidine and proline. Together with an increased level of plasma long chain acylcarnitines and a strong alteration of liver gene expression, this indicates malnutrition. Attempts to rescue pups by decreasing the litter size or by nutrients injected i.p. did not succeed. Radioactively labelled leucine but not lysine given per os accumulated in the small intestine of Slc43a2null pups, suggesting the defective transcellular transport of Lat4 substrates. In summary, Lat4 is a symmetrical uniporter for neutral essential AAs localizing at the basolateral side of (re)absorbing epithelia and is necessary for early nutrition and development.
Collapse
Affiliation(s)
- Adriano Guetg
- Institute of Physiology and Zurich Center of Integrative Human Physiology, University of Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
28
|
Junttila S, Saarela U, Halt K, Manninen A, Pärssinen H, Lecca MR, Brändli AW, Sims-Lucas S, Skovorodkin I, Vainio SJ. Functional genetic targeting of embryonic kidney progenitor cells ex vivo. J Am Soc Nephrol 2014; 26:1126-37. [PMID: 25201883 DOI: 10.1681/asn.2013060584] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 07/23/2014] [Indexed: 01/11/2023] Open
Abstract
The embryonic mammalian metanephric mesenchyme (MM) is a unique tissue because it is competent to generate the nephrons in response to Wnt signaling. An ex vivo culture in which the MM is separated from the ureteric bud (UB), the natural inducer, can be used as a classic tubule induction model for studying nephrogenesis. However, technological restrictions currently prevent using this model to study the molecular genetic details before or during tubule induction. Using nephron segment-specific markers, we now show that tubule induction in the MM ex vivo also leads to the assembly of highly segmented nephrons. This induction capacity was reconstituted when MM tissue was dissociated into a cell suspension and then reaggregated (drMM) in the presence of human recombinant bone morphogenetic protein 7/human recombinant fibroblast growth factor 2 for 24 hours before induction. Growth factor-treated drMM also recovered the capacity for organogenesis when recombined with the UB. Cell tracking and time-lapse imaging of chimeric drMM cultures indicated that the nephron is not derived from a single progenitor cell. Furthermore, viral vector-mediated transduction of green fluorescent protein was much more efficient in dissociated MM cells than in intact mesenchyme, and the nephrogenic competence of transduced drMM progenitor cells was preserved. Moreover, drMM cells transduced with viral vectors mediating Lhx1 knockdown were excluded from the nephric tubules, whereas cells transduced with control vectors were incorporated. In summary, these techniques allow reproducible cellular and molecular examinations of the mechanisms behind nephrogenesis and kidney organogenesis in an ex vivo organ culture/organoid setting.
Collapse
Affiliation(s)
- Sanna Junttila
- Biocenter Oulu, Infotech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Ulla Saarela
- Biocenter Oulu, Infotech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Kimmo Halt
- Biocenter Oulu, Infotech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Aki Manninen
- Biocenter Oulu, Infotech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Heikki Pärssinen
- Biocenter Oulu, Infotech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - M Rita Lecca
- Functional Genomics Center Zurich, University of Zurich/ETH Zurich, Zurich, Switzerland
| | - André W Brändli
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany; and
| | - Sunder Sims-Lucas
- Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ilya Skovorodkin
- Biocenter Oulu, Infotech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Seppo J Vainio
- Biocenter Oulu, Infotech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland;
| |
Collapse
|
29
|
Caceres PS, Mendez M, Ortiz PA. Vesicle-associated membrane protein 2 (VAMP2) but Not VAMP3 mediates cAMP-stimulated trafficking of the renal Na+-K+-2Cl- co-transporter NKCC2 in thick ascending limbs. J Biol Chem 2014; 289:23951-62. [PMID: 25008321 PMCID: PMC4156046 DOI: 10.1074/jbc.m114.589333] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In the kidney, epithelial cells of the thick ascending limb (TAL) reabsorb NaCl via the apical Na+/K+/2Cl− co-transporter NKCC2. Steady-state surface NKCC2 levels in the apical membrane are maintained by a balance between exocytic delivery, endocytosis, and recycling. cAMP is the second messenger of hormones that enhance NaCl absorption. cAMP stimulates NKCC2 exocytic delivery via protein kinase A (PKA), increasing steady-state surface NKCC2. However, the molecular mechanism involved has not been studied. We found that several members of the SNARE family of membrane fusion proteins are expressed in TALs. Here we report that NKCC2 co-immunoprecipitates with VAMP2 in rat TALs, and they co-localize in discrete domains at the apical surface. cAMP stimulation enhanced VAMP2 exocytic delivery to the plasma membrane of renal cells, and stimulation of PKA enhanced VAMP2-NKCC2 co-immunoprecipitation in TALs. In vivo silencing of VAMP2 but not VAMP3 in TALs blunted cAMP-stimulated steady-state surface NKCC2 expression and completely blocked cAMP-stimulated NKCC2 exocytic delivery. VAMP2 was not involved in constitutive NKCC2 delivery. We concluded that VAMP2 but not VAMP3 selectively mediates cAMP-stimulated NKCC2 exocytic delivery and surface expression in TALs. We also demonstrated that cAMP stimulation enhances VAMP2 exocytosis and promotes VAMP2 interaction with NKCC2.
Collapse
Affiliation(s)
- Paulo S Caceres
- From the Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202 and the Department of Physiology, Wayne State University, Detroit, Michigan 48202
| | - Mariela Mendez
- From the Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202 and
| | - Pablo A Ortiz
- From the Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202 and the Department of Physiology, Wayne State University, Detroit, Michigan 48202
| |
Collapse
|
30
|
Kumar R, Vallon V. Reduced renal calcium excretion in the absence of sclerostin expression: evidence for a novel calcium-regulating bone kidney axis. J Am Soc Nephrol 2014; 25:2159-68. [PMID: 24876121 DOI: 10.1681/asn.2014020166] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The kidneys contribute to calcium homeostasis by adjusting the reabsorption and excretion of filtered calcium through processes that are regulated by parathyroid hormone (PTH) and 1α,25-dihydroxyvitamin D3 (1α,25[OH]2D3). Most of the filtered calcium is reabsorbed in the proximal tubule, primarily by paracellular mechanisms that are not sensitive to calcium-regulating hormones in physiologically relevant ways. In the distal tubule, however, calcium is reabsorbed by channels and transporters, the activity or expression of which is highly regulated and increased by PTH and 1α,25(OH)2D3. Recent research suggests that other, heretofore unrecognized factors, such as the osteocyte-specific protein sclerostin, also regulate renal calcium excretion. Clues in this regard have come from the study of humans and mice with inactivating mutations of the sclerostin gene that both have increased skeletal density, which would necessitate an increase in intestinal absorption and/or renal reabsorption of calcium. Deletion of the sclerostin gene in mice significantly diminishes urinary calcium excretion and increases fractional renal calcium reabsorption. This is associated with increased circulating 1α,25(OH)2D3 levels, whereas sclerostin directly suppresses 1α-hydroxylase in immortalized proximal tubular cells. Thus, evidence is accumulating that sclerostin directly or indirectly reduces renal calcium reabsorption, suggesting the presence of a novel calcium-excreting bone-kidney axis.
Collapse
Affiliation(s)
- Rajiv Kumar
- Division of Nephrology and Hypertension, Department of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota; and
| | - Volker Vallon
- Division of Nephrology and Hypertension, Departments of Medicine and Pharmacology, University of California San Diego, and Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
31
|
Markadieu N, Delpire E. Physiology and pathophysiology of SLC12A1/2 transporters. Pflugers Arch 2014; 466:91-105. [PMID: 24097229 PMCID: PMC3877717 DOI: 10.1007/s00424-013-1370-5] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 09/21/2013] [Accepted: 09/23/2013] [Indexed: 01/14/2023]
Abstract
The electroneutral Na(+)-K(+)-Cl(-) cotransporters NKCC1 (encoded by the SLC12A2 gene) and NKCC2 (SLC12A1 gene) belong to the Na(+)-dependent subgroup of solute carrier 12 (SLC12) family of transporters. They mediate the electroneutral movement of Na(+) and K(+), tightly coupled to the movement of Cl(-) across cell membranes. As they use the energy of the ion gradients generated by the Na(+)/K(+)-ATPase to transport Na(+), K(+), and Cl(-) from the outside to the inside of a cell, they are considered secondary active transport mechanisms. NKCC-mediated transport occurs in a 1Na(+), 1K(+), and 2Cl(-) ratio, although NKCC1 has been shown to sometimes mediate partial reactions. Both transporters are blocked by bumetanide and furosemide, drugs which are commonly used in clinical medicine. NKCC2 is the molecular target of loop diuretics as it is expressed on the apical membrane of thick ascending limb of Henle epithelial cells, where it mediates NaCl reabsorption. NKCC1, in contrast, is found on the basolateral membrane of Cl(-) secretory epithelial cells, as well as in a variety of non-epithelial cells, where it mediates cell volume regulation and participates in Cl(-) homeostasis. Following their molecular identification two decades ago, much has been learned about their biophysical properties, their mode of operation, their regulation by kinases and phosphatases, and their physiological relevance. However, despite this tremendous amount of new information, there are still so many gaps in our knowledge. This review summarizes information that constitutes consensus in the field, but it also discusses current points of controversy and highlights many unanswered questions.
Collapse
Affiliation(s)
- Nicolas Markadieu
- Department of Anesthesiology, Vanderbilt University School of Medicine, MCN T-4202, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | | |
Collapse
|
32
|
Wright PA, Wood CM, Wilson JM. Rh vs pH: the role of Rhesus glycoproteins in renal ammonia excretion during metabolic acidosis in a freshwater teleost fish. J Exp Biol 2014; 217:2855-65. [DOI: 10.1242/jeb.098640] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Abstract
Increased renal ammonia excretion in response to metabolic acidosis is thought to be a conserved response in vertebrates. We tested the hypothesis that Rhesus (Rh) glycoproteins in the kidney of the freshwater common carp Cyprinus carpio play a critical role in regulating renal ammonia excretion during chronic metabolic acidosis. Exposure to water pH 4.0 (72 h) resulted in a classic metabolic acidosis with reduced plasma pHa, [HCO3-], no change in PCO2, and large changes in renal function. Urine [NH4+] as well as [titratable acidity–HCO3-] rose significantly over the acid exposure, but the profound reduction (5-fold) in urine flow rates eliminated the expected elevations in renal ammonia excretion. Low urine flow rates may be a primary strategy to conserve ions, as urinary excretion of Na+, Cl- and Ca2+ were significantly lower during the acid exposure relative to the control period. Interestingly, renal Rhcg1 mRNA and protein levels were elevated in acid relative to control groups, along with mRNA levels of several ion transporters, including the Na+/H+ exchanger (NHE3), H+ATPase and Na+/K+ATPase (NKA). Immunofluorescence microscopy showed a strong apical Rhcg1 signal in distal tubules. Taken together, these data show that renal Rh glycoproteins and associated ion transporters are responsive to metabolic acidosis, but conservation of ions through reduced urine flow rates takes primacy over renal acid-base regulation in the freshwater C. carpio. We propose that an “acid/base-ion balance” compromise explains the variable renal responses to metabolic acidosis in freshwater teleosts.
Collapse
|
33
|
Gagnon KB, Delpire E. Molecular physiology of SPAK and OSR1: two Ste20-related protein kinases regulating ion transport. Physiol Rev 2013; 92:1577-617. [PMID: 23073627 DOI: 10.1152/physrev.00009.2012] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
SPAK (Ste20-related proline alanine rich kinase) and OSR1 (oxidative stress responsive kinase) are members of the germinal center kinase VI subfamily of the mammalian Ste20 (Sterile20)-related protein kinase family. Although there are 30 enzymes in this protein kinase family, their conservation across the fungi, plant, and animal kingdom confirms their evolutionary importance. Already, a large volume of work has accumulated on the tissue distribution, binding partners, signaling cascades, and physiological roles of mammalian SPAK and OSR1 in multiple organ systems. After reviewing this basic information, we will examine newer studies that demonstrate the pathophysiological consequences to SPAK and/or OSR1 disruption, discuss the development and analysis of genetically engineered mouse models, and address the possible role these serine/threonine kinases might have in cancer proliferation and migration.
Collapse
Affiliation(s)
- Kenneth B Gagnon
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-2520, USA
| | | |
Collapse
|
34
|
Cellular distribution of NKCC2 in the gastric mucosa and its response to short-term osmotic shock. Cell Tissue Res 2012; 348:155-65. [PMID: 22388656 DOI: 10.1007/s00441-012-1359-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Accepted: 02/02/2012] [Indexed: 01/03/2023]
Abstract
The Na(+)-K(+)-2Cl(-) cotransporter-2 (NKCC2) has long been recognized as a "kidney-specific" transporter and is important in salt reabsorption. NKCC2 has been found in the gastric mucosa; however, its cellular distribution and function remain obscure. The present study characterized the distribution pattern of NKCC2 in mammalian gastric mucosa and investigated its response to osmotic challenge. Reverse transcription with the polymerase chain reaction, Western blot and immunofluorescence were used to determine NKCC2 expression and localization. The effect of osmotic shock on NKCC2 expression was studied in isolated gastric mucosa with variable osmolarity treatment. Results from all of the above studies were compared with those of NKCC1. Our data indicated that NKCC1 and NKCC2 were expressed in the gastric mucosa of rat, mouse and human. The mRNA transcripts and proteins for NKCC1 and NKCC2 were broadly expressed in the rat gastric mucosa. In rat and mouse, NKCC1 was largely confined to the lower part of the oxyntic and pyloric gland areas, whereas NKCC2 extended throughout the gastric glands. NKCC1 immunoreactivity was strongly expressed in the parietal and chief cells but was weaker in the mucous cells. NKCC2 was abundantly located in the parietal and mucous cells but faintly distributed in the chief cells. Hypertonic treatment increased the protein level of NKCC1 and caused evident membrane translocation. In contrast, NKCC2 was significantly downregulated and no obvious membrane translocation was observed. Thus, NKCC2 displayed a more ubiquitous distribution in the gastric mucosa and might work coordinately with NKCC1 to maintain cell volume homeostasis under hypertonic conditions.
Collapse
|
35
|
Ferreri NR, Hao S, Pedraza PL, Escalante B, Vio CP. Eicosanoids and tumor necrosis factor-alpha in the kidney. Prostaglandins Other Lipid Mediat 2011; 98:101-6. [PMID: 22101002 DOI: 10.1016/j.prostaglandins.2011.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 10/31/2011] [Accepted: 11/03/2011] [Indexed: 12/14/2022]
Abstract
The thick ascending limb of Henle's loop (TAL) is capable of metabolizing arachidonic acid (AA) by cytochrome P450 (CYP450) and cyclooxygenase (COX) pathways and has been identified as a nephron segment that contributes to salt-sensitive hypertension. Previous studies demonstrated a prominent role for CYP450-dependent metabolism of AA to products that inhibited ion transport pathways in the TAL. However, COX-2 is constitutively expressed along all segments of the TAL and is increased in response to diverse stimuli. The ability of Tamm-Horsfall glycoprotein, a selective marker of cortical TAL (cTAL) and medullary (mTAL), to bind TNF and localize it to this nephron segment prompted studies to determine the capacity of mTAL cells to produce TNF and determine its effects on mTAL function. The colocalization of calcium-sensing receptor (CaR) and COX-2 in the TAL supports the notion that activation of CaR induces TNF-dependent COX-2 expression and PGE₂ synthesis in mTAL cells. Additional studies showed that TNF produced by mTAL cells inhibits ⁸⁶Rb uptake, an in vitro correlate of natriuresis, in an autocrine- and COX-2-dependent manner. The molecular mechanism for these effects likely includes inhibition of Na⁺-K⁺-2Cl⁻ cotransporter (NKCC2) expression and trafficking.
Collapse
Affiliation(s)
- Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA.
| | | | | | | | | |
Collapse
|
36
|
Ares GR, Caceres PS, Ortiz PA. Molecular regulation of NKCC2 in the thick ascending limb. Am J Physiol Renal Physiol 2011; 301:F1143-59. [PMID: 21900458 DOI: 10.1152/ajprenal.00396.2011] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The kidney plays an essential role in blood pressure regulation by controlling short-term and long-term NaCl and water balance. The thick ascending limb of the loop of Henle (TAL) reabsorbs 25-30% of the NaCl filtered by the glomeruli in a process mediated by the apical Na(+)-K(+)-2Cl(-) cotransporter NKCC2, which allows Na(+) and Cl(-) entry from the tubule lumen into TAL cells. In humans, mutations in the gene coding for NKCC2 result in decreased or absent activity characterized by severe salt and volume loss and decreased blood pressure (Bartter syndrome type 1). Opposite to Bartter's syndrome, enhanced NaCl absorption by the TAL is associated with human hypertension and animal models of salt-sensitive hypertension. TAL NaCl reabsorption is subject to exquisite control by hormones like vasopressin, parathyroid, glucagon, and adrenergic agonists (epinephrine and norepinephrine) that stimulate NaCl reabsorption. Atrial natriuretic peptides or autacoids like nitric oxide and prostaglandins inhibit NaCl reabsorption, promoting salt excretion. In general, the mechanism by which hormones control NaCl reabsorption is mediated directly or indirectly by altering the activity of NKCC2 in the TAL. Despite the importance of NKCC2 in renal physiology, the molecular mechanisms by which hormones, autacoids, physical factors, and intracellular ions regulate NKCC2 activity are largely unknown. During the last 5 years, it has become apparent that at least three molecular mechanisms determine NKCC2 activity. As such, membrane trafficking, phosphorylation, and protein-protein interactions have recently been described in TALs and heterologous expression systems as mechanisms that modulate NKCC2 activity. The focus of this review is to summarize recent data regarding NKCC2 regulation and discuss their potential implications in physiological control of TAL function, renal physiology, and blood pressure regulation.
Collapse
Affiliation(s)
- Gustavo R Ares
- Hypertension and Vascular Research Division, Dept. of Internal Medicine, Henry Ford Hospital, 2799 West Grand Blvd., Detroit, MI 48202, USA
| | | | | |
Collapse
|
37
|
Ohno M, Uchida K, Ohashi T, Nitta K, Ohta A, Chiga M, Sasaki S, Uchida S. Immunolocalization of WNK4 in mouse kidney. Histochem Cell Biol 2011; 136:25-35. [PMID: 21660484 DOI: 10.1007/s00418-011-0827-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2011] [Indexed: 11/29/2022]
Abstract
Initial reports claim that WNK4 localization is mainly at intercellular junctions of distal convoluted tubules (DCT) and cortical collecting ducts (CCD) in the kidney. However, we recently clarified the major targets of WNK4 kinase to be the OSR1/SPAK kinases and the Na-Cl co-transporter (NCC), an apical membrane protein in the DCT, thus raising the question of whether the cellular localization of WNK4 is at intercellular junctions. In this study, we re-evaluate the intrarenal and intracellular immunolocalization of WNK4 in the mouse kidney using a newly generated anti-WNK4 antibody. By performing double immunofluorescence of WNK4 with several nephron-segment-specific markers, we have found that WNK4 is present in podocytes in glomeruli, the cortical thick ascending limb of Henle's loop including macula densa, and the medullary collecting ducts (MCD), in addition to the previously identified nephron segments, i.e., DCT and CCD. These results are consistent with the finding that WNK4 constitutes a kinase cascade with OSR1/SPAK and NCC in the DCT, and highlights a novel role for WNK4 in nephron segments newly identified as being WNK4-positive in this study.
Collapse
Affiliation(s)
- Mayuko Ohno
- Department of Medicine, Kidney Center, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjyuku-ku, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Watanabe S, Mekuchi M, Ideuchi H, Kim YK, Kaneko T. Electroneutral cation-Cl- cotransporters NKCC2β and NCCβ expressed in the intestinal tract of Japanese eel Anguilla japonica. Comp Biochem Physiol A Mol Integr Physiol 2011; 159:427-35. [PMID: 21539929 DOI: 10.1016/j.cbpa.2011.04.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 04/14/2011] [Accepted: 04/17/2011] [Indexed: 11/29/2022]
Abstract
In the present study, we aimed to elucidate the mechanisms of intestinal Na(+) and Cl(-) absorption in Japanese eel, focusing on electroneutral cation-Cl(-) cotransporters, NKCC2β and NCCβ, expressed in the intestinal tract. First, we cloned cDNAs encoding NKCC2β and NCCβ from the intestinal tract of Japanese eel. In both freshwater- and seawater-acclimated eels, quantitative PCR analysis showed that NKCC2β was predominantly expressed in the anterior and posterior intestines, and that NCCβ expression was specifically high in the rectum. According to immunohistochemistry with anti-eel NKCC2β (reacting with NKCC2β but not with NCCβ) and T4 antibody (reacting with both NKCC2β and NCCβ), NKCC2β was localized in the apical surface of the epithelial cells in the anterior and posterior intestines, whereas NCCβ was likely to be distributed to that in the rectum. Furthermore, a specific NCC inhibitor, hydrochlorothiazide, inhibited of Na(+) and Cl(-) absorption, as well as water absorption, in the rectal sac preparations from seawater eel, indicating the involvement of NCCβ in ion absorption in the rectum. Our findings indicate that NKCC2β expressed in the anterior and posterior intestines and NCCβ in the rectum are importantly involved in ion absorption to reduce osmolality of ingested seawater prior to water absorption in seawater-acclimated eel.
Collapse
Affiliation(s)
- Soichi Watanabe
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
39
|
Tresguerres M, Levin LR, Buck J. Intracellular cAMP signaling by soluble adenylyl cyclase. Kidney Int 2011; 79:1277-88. [PMID: 21490586 DOI: 10.1038/ki.2011.95] [Citation(s) in RCA: 161] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Soluble adenylyl cyclase (sAC) is a recently identified source of the ubiquitous second messenger cyclic adenosine 3',5' monophosphate (cAMP). sAC is distinct from the more widely studied source of cAMP, the transmembrane adenylyl cyclases (tmACs); its activity is uniquely regulated by bicarbonate anions, and it is distributed throughout the cytoplasm and in cellular organelles. Due to its unique localization and regulation, sAC has various functions in a variety of physiological systems that are distinct from tmACs. In this review, we detail the known functions of sAC, and we reassess commonly held views of cAMP signaling inside cells.
Collapse
Affiliation(s)
- Martin Tresguerres
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | | | | |
Collapse
|
40
|
|
41
|
Gagnon KB, Delpire E. Molecular determinants of hyperosmotically activated NKCC1-mediated K+/K+ exchange. J Physiol 2010; 588:3385-96. [PMID: 20530115 DOI: 10.1113/jphysiol.2010.191932] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Na(+)-K(+)-2Cl(-) cotransport (NKCC) mediates the movement of two Cl(-) ions for one Na(+) and one K(+) ion. Under isosmotic conditions or with activation of the kinases SPAK/WNK4, the NKCC1-mediated Cl(-) uptake in Xenopus laevis oocytes, as measured using (36)Cl, is twice the value of K(+) uptake, as determined using (86)Rb. Under hyperosmotic conditions, there is a significant activation of the bumetanide-sensitive K(+) uptake with only a minimal increase in bumetanide-sensitive Cl(-) uptake. This suggests that when stimulated by hypertonicity, the cotransporter mediates K(+)/K(+) and Cl(-)/Cl(-) exchange. Although significant stimulation of K(+)/K(+) exchange was observed with NKCC1, a significantly smaller hyperosmotic stimulatory effect was observed with NKCC2. In order to identify the molecular determinant(s) of this NKCC1-specific activation, we created chimeras of the mouse NKCC1 and the rat NKCC2. Swapping the regulatory amino termini of the cotransporters neither conferred activation to NKCC2 nor prevented activation of NKCC1. Using unique restrictions sites, we created additional chimeric molecules and determined that the first intracellular loop between membrane-spanning domains one and two and the second extracellular loop between membrane-spanning domains three and four of NKCC1 are necessary components of the hyperosmotic stimulation of K(+)/K(+) exchange.
Collapse
Affiliation(s)
- Kenneth B Gagnon
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | |
Collapse
|
42
|
Tresguerres M, Levin LR, Buck J, Grosell M. Modulation of NaCl absorption by [HCO(3)(-)] in the marine teleost intestine is mediated by soluble adenylyl cyclase. Am J Physiol Regul Integr Comp Physiol 2010; 299:R62-71. [PMID: 20410468 DOI: 10.1152/ajpregu.00761.2009] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intestinal HCO(3)(-) secretion and NaCl absorption are essential for counteracting dehydration in marine teleost fish. We investigated how these two processes are coordinated in toadfish. HCO(3)(-) stimulated a luminal positive short-circuit current (I(sc)) in intestine mounted in Ussing chamber, bathed with the same saline solution on the external and internal sides of the epithelium. The I(sc) increased proportionally to the [HCO(3)(-)] in the bath up to 80 mM NaHCO(3), and it did not occur when NaHCO(3) was replaced with Na(+)-gluconate or with NaHCO(3) in Cl(-)-free saline. HCO(3)(-) (20 mM) induced a approximately 2.5-fold stimulation of I(sc), and this [HCO(3)(-)] was used in all subsequent experiments. The HCO(3)(-)-stimulated I(sc) was prevented or abolished by apical application of 10 muM bumetanide (a specific inhibitor of NKCC) and by 30 microM 4-catechol estrogen [CE; an inhibitor of soluble adenylyl cyclase (sAC)]. The inhibitory effects of bumetanide and CE were not additive. The HCO(3)(-)-stimulated I(sc) was prevented by apical bafilomycin (1 microM) and etoxolamide (1 mM), indicating involvement of V-H(+)-ATPase and carbonic anhydrases, respectively. Immunohistochemistry and Western blot analysis confirmed the presence of an NKCC2-like protein in the apical membrane and subapical area of epithelial intestinal cells, of Na(+)/K(+)-ATPase in basolateral membranes, and of an sAC-like protein in the cytoplasm. We propose that sAC regulates NKCC activity in response to luminal HCO(3)(-), and that V-H(+)-ATPase and intracellular carbonic anhydrase are essential for transducing luminal HCO(3)(-) into the cell by CO(2)/HCO(3)(-) hydration/dehydration. This mechanism putatively coordinates HCO(3)(-) secretion with NaCl and water absorption in toadfish intestine.
Collapse
Affiliation(s)
- Martin Tresguerres
- Department of Pharmacology, Weill Cornell Medical College, New York, New York 10065, USA.
| | | | | | | |
Collapse
|
43
|
da Silva CA, de Bragança AC, Shimizu MHM, Sanches TR, Fortes MAZ, Giorgi RR, Andrade L, Seguro AC. Rosiglitazone prevents sirolimus-induced hypomagnesemia, hypokalemia, and downregulation of NKCC2 protein expression. Am J Physiol Renal Physiol 2009; 297:F916-22. [PMID: 19656910 DOI: 10.1152/ajprenal.90256.2008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Sirolimus, an antiproliferative immunosuppressant, induces hypomagnesemia and hypokalemia. Rosiglitazone activates renal sodium- and water-reabsorptive pathways. We evaluated whether sirolimus induces renal wasting of magnesium and potassium, attempting to identify the tubule segments in which this occurs. We tested the hypothesis that reduced expression of the cotransporter NKCC2 forms the molecular basis of this effect and evaluated the possible association between increased urinary excretion of magnesium and renal expression of the epithelial Mg2+ channel TRPM6. We then analyzed whether rosiglitazone attenuates these sirolimus-induced tubular effects. Wistar rats were treated for 14 days with sirolimus (3 mg/kg body wt in drinking water), with or without rosiglitazone (92 mg/kg body wt in food). Protein abundance of NKCC2, aquaporin-2 (AQP2), and TRPM6 was assessed using immunoblotting. Sirolimus-treated animals presented no change in glomerular filtration rate, although there were marked decreases in plasma potassium and magnesium. Sirolimus treatment reduced expression of NKCC2, and this was accompanied by greater urinary excretion of sodium, potassium, and magnesium. In sirolimus-treated animals, AQP2 expression was reduced. Expression of TRPM6 was increased, which might represent a direct stimulatory effect of sirolimus or a compensatory response. The finding that rosiglitazone prevented or attenuated all sirolimus-induced renal tubular defects has potential clinical implications.
Collapse
Affiliation(s)
- Cristianne Alexandre da Silva
- Laboratório de Pesquisa Básica LIM-12, Faculdade de Medicina da USP, Av. Dr. Arnaldo 455, Sala 3310, CEP 01246-903, São Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Gamba G, Friedman PA. Thick ascending limb: the Na(+):K (+):2Cl (-) co-transporter, NKCC2, and the calcium-sensing receptor, CaSR. Pflugers Arch 2009; 458:61-76. [PMID: 18982348 PMCID: PMC3584568 DOI: 10.1007/s00424-008-0607-1] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 10/21/2008] [Indexed: 01/12/2023]
Abstract
The thick ascending limb of Henle's loop is a nephron segment that is vital to the formation of dilute and concentrated urine. This ability is accomplished by a consortium of functionally coupled proteins consisting of the apical Na(+):K(+):2Cl(-) co-transporter, the K(+) channel, and basolateral Cl(-) channel that mediate electroneutral salt absorption. In thick ascending limbs, salt absorption is importantly regulated by the calcium-sensing receptor. Genetic or pharmacological disruption impairing the function of any of these proteins results in Bartter syndrome. The thick ascending limb is also an important site of Ca(2+) and Mg(2+) absorption. Calcium-sensing receptor activation inhibits cellular Ca(2+) absorption induced by parathyroid hormone, as well as passive paracellular Ca(2+) transport. The present review discusses these functions and their genetic and molecular regulation.
Collapse
Affiliation(s)
- Gerardo Gamba
- Molecular Physiology Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, 14000 Mexico City, Mexico
| | - Peter A. Friedman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
45
|
Katoh F, Cozzi RRF, Marshall WS, Goss GG. Distinct Na+/K+/2Cl- cotransporter localization in kidneys and gills of two euryhaline species, rainbow trout and killifish. Cell Tissue Res 2008; 334:265-81. [DOI: 10.1007/s00441-008-0679-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Accepted: 08/18/2008] [Indexed: 10/21/2022]
|
46
|
Ares GR, Caceres P, Alvarez-Leefmans FJ, Ortiz PA. cGMP decreases surface NKCC2 levels in the thick ascending limb: role of phosphodiesterase 2 (PDE2). Am J Physiol Renal Physiol 2008; 295:F877-87. [PMID: 18684888 DOI: 10.1152/ajprenal.00449.2007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
NaCl absorption in the medullary thick ascending limb of the loop of Henle (THAL) is mediated by the apical Na/K/2Cl cotransporter (NKCC2). Hormones that increase cGMP, such as nitric oxide (NO) and natriuretic peptides, decrease NaCl absorption by the THAL. However, the mechanism by which cGMP decreases NaCl absorption in THALs is not known. We hypothesized that cGMP decreases surface NKCC2 levels in the THAL. We used surface biotinylation to measure surface NKCC2 levels in rat THAL suspensions. We tested the effect of the membrane-permeant cGMP analog dibutyryl-cGMP (db-cGMP) on surface NKCC2 levels. Incubating THALs with db-cGMP for 20 min decreased surface NKCC2 levels in a concentration-dependent manner (basal=100%; db-cGMP 100 microM=77+/-7%; 500 microM=54+/-10% and 1,000 microM=61+/-8%). A different cGMP analog 8-bromo-cGMP (8-Br-cGMP) also decreased surface NKCC2 levels by 25%, (basal=100%; 8-Br-cGMP=75+/-5%). Incubation of isolated, perfused THALs with db-cGMP decreased apical surface NKCC2 labeling levels as measured by immunofluorescence and confocal microscopy. cGMP-stimulated phosphodiesterase 2 (PDE2) mediates the inhibitory effect of NO on NaCl absorption by THALs. Thus we examined the role of PDE2 and found that PDE2 inhibitors blocked the effect of db-cGMP on surface NKCC2. Also, a nonstimulatory concentration of db-cAMP blocked the cGMP-induced decrease in surface NKCC2. Finally, db-cGMP inhibited THAL net Cl absorption by 48+/-4%, and this effect was completely blocked by PDE2 inhibition. We conclude that cGMP decreases NKCC2 levels in the apical membrane of THALs and that this effect is mediated by PDE2. This is an important mechanism by which cGMP inhibits NaCl absorption by the THAL.
Collapse
Affiliation(s)
- Gustavo R Ares
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, and Department of Physiology, Wayne State University, 2799 West Grand Blvd., Detroit, MI 48202, USA
| | | | | | | |
Collapse
|
47
|
Wagner CA, Loffing-Cueni D, Yan Q, Schulz N, Fakitsas P, Carrel M, Wang T, Verrey F, Geibel JP, Giebisch G, Hebert SC, Loffing J. Mouse model of type II Bartter's syndrome. II. Altered expression of renal sodium- and water-transporting proteins. Am J Physiol Renal Physiol 2008; 294:F1373-80. [DOI: 10.1152/ajprenal.00613.2007] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Bartter's syndrome represents a group of hereditary salt- and water-losing renal tubulopathies caused by loss-of-function mutations in proteins mediating or regulating salt transport in the thick ascending limb (TAL) of Henle's loop. Mutations in the ROMK channel cause type II antenatal Bartter's syndrome that presents with maternal polyhydramnios and postnatal life-threatening volume depletion. We have developed a colony of Romk null mice showing a Bartter-like phenotype and with increased survival to adulthood, suggesting the activation of compensatory mechanisms. To test the hypothesis that upregulation of Na+-transporting proteins in segments distal to the TAL contributes to compensation, we studied expression of salt-transporting proteins in ROMK-deficient ( Romk−/−) mice. Plasma aldosterone was 40% higher and urinary PGE2 excretion was 1.5-fold higher in Romk−/− compared with wild-type littermates. Semiquantitative immunoblotting of kidney homogenates revealed decreased abundances of proximal tubule Na+/H+ exchanger (NHE3) and Na+-Pi cotransporter (NaPi-IIa) and TAL-specific Na+-K+-2Cl−-cotransporter (NKCC2/BSC1) in Romk−/− mice, while the distal convoluted tubule (DCT)-specific Na+-Cl− cotransporter (NCC/TSC) was markedly increased. The abundance of the α-,β-, and γ-subunits of the epithelial Na+ channel (ENaC) was slightly increased, although only differences for γ-ENaC reached statistical significance. Morphometry revealed a fourfold increase in the fractional volume of DCT but not of connecting tubule (CNT) and collecting duct (CCD). Consistently, CNT and CD of Romk−/− mice revealed no apparent increase in the luminal abundance of the ENaC compared with those of wild-type mice. These data suggest that the loss of ROMK-dependent Na+ absorption in the TAL is compensated predominately by upregulation of Na+ transport in downstream DCT cells. These adaptive changes in Romk−/− mice may help to limit renal Na+ loss, and thereby, contribute to survival of these mice.
Collapse
|
48
|
Castrop H, Schnermann J. Isoforms of renal Na-K-2Cl cotransporter NKCC2: expression and functional significance. Am J Physiol Renal Physiol 2008; 295:F859-66. [PMID: 18495801 DOI: 10.1152/ajprenal.00106.2008] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renal Na-K-2Cl cotransporter (NKCC2, BSC1) is selectively expressed in the apical membrane of cells of the thick ascending limb of the loop of Henle (TAL) and macula densa. NKCC2-dependent salt transport constitutes the major apical entry pathway for transepithelial salt reabsorption in the TAL. Although NKCC2 is encoded by a single gene (Slc12a1), differential splicing of the NKCC2 pre-mRNA results in the formation of several alternate transcripts. Thus three full-length splice isoforms of NKCC2 differ in their variable exon 4, resulting in transcripts for NKCC2B, NKCC2A, and NKCC2F. In addition to full-length isoforms, variants with truncated COOH-terminal ends have been described. The various splice isoforms of NKCC2 differ in their localization along the TAL and in their transport characteristics. Data in the literature are reviewed to assess the principles of NKCC2 differential splicing, the localization of NKCC2 splice isoforms along the TAL in various species, and the functional characteristics of the splice isoforms. In addition, we discuss the functional significance of NKCC2 isoforms for TAL salt retrieval and for the specific salt sensor function of macula densa cells based on studies using isoform-specific NKCC2-knockout mice. We suggest that different NKCC2 splice variants cooperate in salt retrieval along the TAL and that the coexpression of two splice variants (NKCC2B and NKCC2A) in the macula densa cells facilitates efficient salt sensing over wide ranges of fluctuating salt concentrations.
Collapse
Affiliation(s)
- Hayo Castrop
- Physiologisches Institut der Universität Regensburg, Universitätsstr. 31, D-93040 Regensburg, Germany.
| | | |
Collapse
|
49
|
Capasso G, Rizzo M, Garavaglia ML, Trepiccione F, Zacchia M, Mugione A, Ferrari P, Paulmichl M, Lang F, Loffing J, Carrel M, Damiano S, Wagner CA, Bianchi G, Meyer G. Upregulation of apical sodium-chloride cotransporter and basolateral chloride channels is responsible for the maintenance of salt-sensitive hypertension. Am J Physiol Renal Physiol 2008; 295:F556-67. [PMID: 18480177 DOI: 10.1152/ajprenal.00340.2007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We investigated which of the NaCl transporters are involved in the maintenance of salt-sensitive hypertension. Milan hypertensive (MHS) rats were studied 3 mo after birth. In MHS, compared with normotensive strain (MNS), mRNA abundance, quantified by competitive PCR on isolated tubules, was unchanged, both for Na+/H+ isoform 3 (NHE3) and Na+-K+-2Cl- (NKCC2), but higher (119%, n = 5, P < 0.005) for Na+-Cl- (NCC) in distal convoluted tubules (DCT). These results were confirmed by Western blots, which revealed: 1) unchanged NHE3 in the cortex and NKCC2 in the outer medulla; 2) a significant increase (52%, n = 6, P < 0.001) of NCC in the cortex; 3) alpha- and beta-sodium channels [epithelial Na+ channel (ENaC)] unaffected in renal cortex and slightly reduced in the outer medulla, while gamma-ENaC remained unchanged. Pendrin protein expression was unaffected. The role of NCC was reinforced by immunocytochemical studies showing increased NCC on the apical membrane of DCT cells of MHS animals, and by clearance experiments demonstrating a larger sensitivity (P < 0.001) to bendroflumethiazide in MHS rats. Kidney-specific chloride channels (ClC-K) were studied by Western blot experiments on renal cortex and by patch-clamp studies on primary culture of DCT dissected from MNS and MHS animals. Electrophysiological characteristics of ClC-K channels were unchanged in MHS rats, but the number of active channels in a patch was 0.60 +/- 0.21 (n = 35) in MNS rats and 2.17 +/- 0.59 (n = 23) in MHS rats (P < 0.05). The data indicate that, in salt-sensitive hypertension, there is a strong upregulation, both of NCC and ClC-K along the DCT, which explains the persistence of hypertension.
Collapse
Affiliation(s)
- Giovambattista Capasso
- Department of Internal Medicine, Chair of Nephrology, Faculty of Medicine, Second University of Napoli, Napoli, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fenton RA, Knepper MA. Mouse models and the urinary concentrating mechanism in the new millennium. Physiol Rev 2007; 87:1083-112. [PMID: 17928581 DOI: 10.1152/physrev.00053.2006] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Our understanding of urinary concentrating and diluting mechanisms at the end of the 20th century was based largely on data from renal micropuncture studies, isolated perfused tubule studies, tissue analysis studies and anatomical studies, combined with mathematical modeling. Despite extensive data, several key questions remained to be answered. With the advent of the 21st century, a new approach, transgenic and knockout mouse technology, is providing critical new information about urinary concentrating processes. The central goal of this review is to summarize findings in transgenic and knockout mice pertinent to our understanding of the urinary concentrating mechanism, focusing chiefly on mice in which expression of specific renal transporters or receptors has been deleted. These include the major renal water channels (aquaporins), urea transporters, ion transporters and channels (NHE3, NKCC2, NCC, ENaC, ROMK, ClC-K1), G protein-coupled receptors (type 2 vasopressin receptor, prostaglandin receptors, endothelin receptors, angiotensin II receptors), and signaling molecules. These studies shed new light on several key questions concerning the urinary concentrating mechanism including: 1) elucidation of the role of water absorption from the descending limb of Henle in countercurrent multiplication, 2) an evaluation of the feasibility of the passive model of Kokko-Rector and Stephenson, 3) explication of the role of inner medullary collecting duct urea transport in water conservation, 4) an evaluation of the role of tubuloglomerular feedback in maintenance of appropriate distal delivery rates for effective regulation of urinary water excretion, and 5) elucidation of the importance of water reabsorption in the connecting tubule versus the collecting duct for maintenance of water balance.
Collapse
Affiliation(s)
- Robert A Fenton
- Water and Salt Research Center, Institute of Anatomy, University of Aarhus, Aarhus, Denmark.
| | | |
Collapse
|